Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 15448, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37723180

ABSTRACT

The hepatitis E virus (HEV) is an emergent zoonotic virus causing viral hepatitis worldwide. Clinically, hepatitis E is not easily distinguished from other types of acute viral hepatitis. There is a need for HEV diagnostic assays to detect and prevent interspecies transmission among susceptible populations. Nanobodies (Nbs) are expressed recombinantly in different systems, produced with high yields, and have superior physicochemical properties compared with conventional antibodies (Ab). Several Nbs against ORF2, the capsid protein and main antigen, were selected and produced in E. coli. Nb39 and Nb74 specifically recognized HEV ORF2 (genotypes 3 and 4). A competitive ELISA (cELISA) was developed and validated using a reference panel of human (n = 86) and swine sera (n = 116) tested in comparison with a commercial kit. The optimal cutoff values determined by ROC analysis were 69.16% (human) and 58.76% (swine); the sensitivity and specificity were high: 97.4% (95% CI 86.5-99.5%) and 95.8% (95% CI 86.0-98.8%) for human vs. 100% (95% CI 93.5-100%) and 98.3% (95% CI 91.0-99.7%) for swine. Further, the cELISA detected total anti-HEV antibodies in wild boar, deer, and mice. To our knowledge, this is the first report of production of Nbs against HEV-3 ORF2 for diagnostic purposes.


Subject(s)
Deer , Hepatitis E virus , Single-Domain Antibodies , Humans , Animals , Mice , Swine , Escherichia coli , Antibodies , Enzyme-Linked Immunosorbent Assay
2.
Biochem J ; 480(19): 1533-1551, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37721041

ABSTRACT

In this paper, we describe the development of a Dictyostelium discoideum strain deficient in frataxin protein (FXN). We investigated the conservation of function between humans and D. discoideum and showed that DdFXN can substitute the human version in the interaction and activation of the Fe-S assembly supercomplex. We edited the D. discoideum fxn locus and isolated a defective mutant, clone 8, which presents landmarks of frataxin deficiency, such as a decrease in Fe-S cluster-dependent enzymatic functions, growth rate reduction, and increased sensitivity to oxidative stress. In addition, the multicellular development is affected as well as growing on bacterial lawn. We also assessed the rescuing capacity of DdFXN-G122V, a version that mimics a human variant present in some FA patients. While the expression of DdFXN-G122V rescues growth and enzymatic activity defects, as DdFXN does, multicellular development defects were only partially rescued. The results of the study suggest that this new D. discoideum strain offers a wide range of possibilities to easily explore diverse FA FXN variants. This can facilitate the development of straightforward drug screenings to look for new therapeutic strategies.

4.
J Immunol Methods ; 518: 113503, 2023 07.
Article in English | MEDLINE | ID: mdl-37263391

ABSTRACT

In recent years dengue has become a rapidly growing public health problem worldwide, however, the availability of accurate and affordable diagnostic immunoassays is limited, partly due to the difficulty of producing large quantities of purified antigen. Non-structural protein 1 (NS1) has shown to be a good candidate for inclusion in diagnostic assays and for serosurveys, particularly in endemic countries as a prerequisite for vaccination. In this work the NS1 antigen derived from dengue virus type-1 (DENV1) was expressed in HEK293-T cells and purified by affinity chromatography. The recombinant protein was recovered properly folded as dimers, highly purified and with good yield (1.5 mg/L). It was applied as a serological probe in an indirect ELISA developed in this work to detect human IgG antibodies. Preliminary comparative performance values of 81.1% sensitivity and 83.0% specificity of the developed and preliminary validated iELISA, relative to a commercial kit were obtained, suggesting that the purified recombinant DENV1 NS1 antigen is suitable to detect IgG antibodies, indicative of past DENV infection.


Subject(s)
Dengue Virus , Dengue , Virus Diseases , Animals , Humans , Dengue Virus/genetics , Dengue/diagnosis , HEK293 Cells , Sensitivity and Specificity , Antibodies, Viral , Viral Nonstructural Proteins , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin G/metabolism , Mammals
5.
bioRxiv ; 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36993215

ABSTRACT

In this work, we developed llama-derived nanobodies (Nbs) directed to the receptor binding domain (RBD) and other domains of the Spike (S) protein of SARS-CoV-2. Nanobodies were selected after the biopanning of two VHH-libraries, one of which was generated after the immunization of a llama (lama glama) with the bovine coronavirus (BCoV) Mebus, and another with the full-length pre-fused locked S protein (S-2P) and the RBD from the SARS-CoV-2 Wuhan strain (WT). Most of the neutralizing Nbs selected with either RBD or S-2P from SARS-CoV-2 were directed to RBD and were able to block S-2P/ACE2 interaction. Three Nbs recognized the N-terminal domain (NTD) of the S-2P protein as measured by competition with biliverdin, while some non-neutralizing Nbs recognize epitopes in the S2 domain. One Nb from the BCoV immune library was directed to RBD but was non-neutralizing. Intranasal administration of Nbs induced protection ranging from 40% to 80% against COVID-19 death in k18-hACE2 mice challenged with the WT strain. Interestingly, protection was not only associated with a significant reduction of virus replication in nasal turbinates and lungs, but also with a reduction of virus load in the brain. Employing pseudovirus neutralization assays, we were able to identify Nbs with neutralizing capacity against the Alpha, Beta, Delta and Omicron variants. Furthermore, cocktails of different Nbs performed better than individual Nbs to neutralize two Omicron variants (B.1.529 and BA.2). Altogether, the data suggest these Nbs can potentially be used as a cocktail for intranasal treatment to prevent or treat COVID-19 encephalitis, or modified for prophylactic administration to fight this disease.

6.
Cell Death Dis ; 13(3): 280, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35351865

ABSTRACT

RIPK3 partially protects against disease caused by influenza A virus (IAV) infection in the mouse model. Here, we compared the immune protection of active vaccination with a universal influenza A vaccine candidate based on the matrix protein 2 ectodomain (M2e) and of passive immunization with anti-M2e IgG antibodies in wild type and Ripk3-/- mice. We observed that the protection against IAV after active vaccination with M2e viral antigen is lost in Ripk3-/- mice. Interestingly, M2e-specific serum IgG levels induced by M2e vaccination were not significantly different between wild type and Ripk3-/- vaccinated mice demonstrating that the at least the humoral immune response was not affected by the absence of RIPK3 during active vaccination. Moreover, following IAV challenge, lungs of M2e vaccinated Ripk3-/- mice revealed a decreased number of immune cell infiltrates and an increased accumulation of dead cells, suggesting that phagocytosis could be reduced in Ripk3-/- mice. However, neither efferocytosis nor antibody-dependent phagocytosis were affected in macrophages isolated from Ripk3-/- mice. Likewise following IAV infection of Ripk3-/- mice, active vaccination and infection resulted in decreased presence of CD8+ T-cells in the lung. However, it is unclear whether this reflects a deficiency in vaccination or an inability following infection. Finally, passively transferred anti-M2e monoclonal antibodies at higher dose than littermate wild type mice completely protected Ripk3-/- mice against an otherwise lethal IAV infection, demonstrating that the increased sensitivity of Ripk3-/- mice could be overcome by increased antibodies. Therefore we conclude that passive immunization strategies with monoclonal antibody could be useful for individuals with reduced IAV vaccine efficacy or increased IAV sensitivity, such as may be expected in patients treated with future anti-inflammatory therapeutics for chronic inflammatory diseases such as RIPK inhibitors.


Subject(s)
Influenza A virus , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Antibodies, Viral , Humans , Immunization, Passive , Immunoglobulin G , Mice , Mice, Inbred BALB C , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Vaccination , Viral Matrix Proteins
7.
Nat Commun ; 11(1): 2832, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32504029

ABSTRACT

Human amyloids have been shown to interact with viruses and interfere with viral replication. Based on this observation, we employed a synthetic biology approach in which we engineered virus-specific amyloids against influenza A and Zika proteins. Each amyloid shares a homologous aggregation-prone fragment with a specific viral target protein. For influenza we demonstrate that a designer amyloid against PB2 accumulates in influenza A-infected tissue in vivo. Moreover, this amyloid acts specifically against influenza A and its common PB2 polymorphisms, but not influenza B, which lacks the homologous fragment. Our model amyloid demonstrates that the sequence specificity of amyloid interactions has the capacity to tune amyloid-virus interactions while allowing for the flexibility to maintain activity on evolutionary diverging variants.


Subject(s)
Amyloid/pharmacology , Antiviral Agents/pharmacology , Reverse Genetics/methods , Synthetic Biology/methods , Amyloid/genetics , Amyloid/therapeutic use , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Dogs , Female , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Influenza A virus/drug effects , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza, Human/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Polymorphism, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
8.
Vaccine ; 36(12): 1592-1598, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29454522

ABSTRACT

H3N8 influenza virus strains have been associated with infectious disease in equine populations throughout the world. Although current vaccines for equine influenza stimulate a protective humoral immune response against the surface glycoproteins, disease in vaccinated horses has been frequently reported, probably due to poor induction of cross-reactive antibodies against non-matching strains. This work describes the performance of a recombinant protein vaccine expressed in prokaryotic cells (ΔHAp) and of a genetic vaccine (ΔHAe), both based on the conserved stem region of influenza hemagglutinin (HA) derived from A/equine/Argentina/1/93 (H3N8) virus. Sera from mice inoculated with these immunogens in different combinations and regimes presented reactivity in vitro against highly divergent influenza virus strains belonging to phylogenetic groups 1 and 2 (H1 and H3 subtypes, respectively), and conferred robust protection against a lethal challenge with both the homologous equine strain (100%) and the homosubtypic human strain A/Victoria/3/75 (H3N2) (70-100%). Animals vaccinated with the same antigens but challenged with the human strain A/PR/8/34 (H1N1), belonging to the phylogenetic group 1, were not protected (0-33%). Combination of protein and DNA immunogens showed higher reactivity to non-homologous strains than protein alone, although all vaccines were permissive for lung infection.


Subject(s)
Cross Protection/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Horse Diseases/prevention & control , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology , Animals , Cell Line , Horse Diseases/immunology , Horse Diseases/virology , Horses , Humans , Immunization , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N8 Subtype/immunology , Influenza Vaccines/administration & dosage , Recombinant Proteins , Vaccines, Subunit/administration & dosage , Vaccines, Synthetic/administration & dosage
9.
PLoS Pathog ; 12(1): e1005410, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26815999

ABSTRACT

A20 negatively regulates multiple inflammatory signalling pathways. We here addressed the role of A20 in club cells (also known as Clara cells) of the bronchial epithelium in their response to influenza A virus infection. Club cells provide a niche for influenza virus replication, but little is known about the functions of these cells in antiviral immunity. Using airway epithelial cell-specific A20 knockout (A20AEC-KO) mice, we show that A20 in club cells critically controls innate immune responses upon TNF or double stranded RNA stimulation. Surprisingly, A20AEC-KO mice are better protected against influenza A virus challenge than their wild type littermates. This phenotype is not due to decreased viral replication. Instead host innate and adaptive immune responses and lung damage are reduced in A20AEC-KO mice. These attenuated responses correlate with a dampened cytotoxic T cell (CTL) response at later stages during infection, indicating that A20AEC-KO mice are better equipped to tolerate Influenza A virus infection. Expression of the chemokine CCL2 (also named MCP-1) is particularly suppressed in the lungs of A20AEC-KO mice during later stages of infection. When A20AEC-KO mice were treated with recombinant CCL2 the protective effect was abrogated demonstrating the crucial contribution of this chemokine to the protection of A20AEC-KO mice to Influenza A virus infection. Taken together, we propose a mechanism of action by which A20 expression in club cells controls inflammation and antiviral CTL responses in response to influenza virus infection.


Subject(s)
Cysteine Endopeptidases/immunology , Cytotoxicity, Immunologic/immunology , Intracellular Signaling Peptides and Proteins/immunology , Orthomyxoviridae Infections/immunology , Respiratory Mucosa/immunology , Animals , Cysteine Endopeptidases/deficiency , Flow Cytometry , Immunity, Innate/immunology , Immunoblotting , Immunohistochemistry , Influenza A virus , Intracellular Signaling Peptides and Proteins/deficiency , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor alpha-Induced Protein 3
10.
PLoS One ; 10(5): e0126650, 2015.
Article in English | MEDLINE | ID: mdl-25973787

ABSTRACT

Human influenza viruses are responsible for annual epidemics and occasional pandemics that cause severe illness and mortality in all age groups worldwide. Matrix protein 2 (M2) of influenza A virus is a tetrameric type III membrane protein that functions as a proton-selective channel. The extracellular domain of M2 (M2e) is conserved in human and avian influenza A viruses and is being pursued as a component for a universal influenza A vaccine. To develop a M2e vaccine that is economical and easy to purify, we genetically fused M2e amino acids 2-16 to the N-terminus of pVIII, the major coat protein of filamentous bacteriophage f88. We show that the resulting recombinant f88-M2e2-16 phages are replication competent and display the introduced part of M2e on the phage surface. Immunization of mice with purified f88-M2e2-16 phages in the presence of incomplete Freund's adjuvant, induced robust M2e-specific serum IgG and protected BALB/c mice against challenge with human and avian influenza A viruses. Thus, replication competent filamentous bacteriophages can be used as efficient and economical carriers to display conserved B cell epitopes of influenza A.


Subject(s)
Capsid Proteins/genetics , Coliphages/immunology , Influenza A Virus, H1N1 Subtype/immunology , Viral Matrix Proteins/genetics , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/biosynthesis , Antigens, Viral/immunology , Capsid Proteins/chemistry , Coliphages/isolation & purification , Coliphages/physiology , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Influenza A Virus, H1N1 Subtype/metabolism , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Orthomyxoviridae Infections/prevention & control , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Viral Matrix Proteins/chemistry , Virus Replication
11.
Expert Rev Vaccines ; 14(1): 55-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25209381

ABSTRACT

Influenza A virus is a pathogen that is feared for its capacity to cause pandemics. In this review, we illustrate the clinical evidence which support the theory that bacterial co-infection is a considerable risk factor for exacerbated disease during pandemic and seasonal influenza, including infection with influenza B viruses. We provide an overview of the multiple and diverse mechanisms that help explain how influenza creates an opportunity for replication of secondary bacterial infections. Influenza vaccines and pneumococcal vaccines are widely used and often in overlapping target groups. We summarize the evidence for a protective effect of influenza immunization against bacterial infections, and vice versa of pneumococcal vaccines against influenza-associated pneumonia and lethality. It is important that future implementation of broadly protective influenza vaccines also takes into account protection against secondary bacterial infection.


Subject(s)
Bacterial Infections/prevention & control , Influenza Vaccines/administration & dosage , Influenza, Human/complications , Influenza, Human/prevention & control , Pneumococcal Vaccines/administration & dosage , Bacterial Infections/immunology , Humans , Influenza Vaccines/immunology , Influenza, Human/immunology , Pneumococcal Vaccines/immunology
12.
J Virol ; 88(15): 8278-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24829341

ABSTRACT

UNLABELLED: Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE: Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/prevention & control , Single-Domain Antibodies/therapeutic use , Animals , Antiviral Agents/immunology , Disease Models, Animal , Female , Influenza A Virus, H5N1 Subtype/immunology , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Single-Domain Antibodies/immunology , Treatment Outcome
13.
PLoS One ; 8(3): e59081, 2013.
Article in English | MEDLINE | ID: mdl-23527091

ABSTRACT

The ectodomain of influenza A matrix protein 2 (M2e) is a candidate for a universal influenza A vaccine. We used recombinant Hepatitis B core antigen to produce virus-like particles presenting M2e (M2e-VLPs). We produced the VLPs with and without entrapped nucleic acids and compared their immunogenicity and protective efficacy. Immunization of BALB/c mice with M2e-VLPs containing nucleic acids induced a stronger, Th1-biased antibody response compared to particles lacking nucleic acids. The former also induced a stronger M2e-specific CD4(+) T cell response, as determined by ELISPOT. Mice vaccinated with alum-adjuvanted M2e-VLPs containing the nucleic acid-binding domain were better protected against influenza A virus challenge than mice vaccinated with similar particles lacking this domain, as deduced from the loss in body weight following challenge with X47 (H3N2) or PR/8 virus. Challenge of mice that had been immunized with M2e-VLPs with or without nucleic acids displayed significantly lower mortality, morbidity and lung virus titers than control-immunized groups. We conclude that nucleic acids present in M2e-VLPs correlate with improved immune protection.


Subject(s)
Adaptive Immunity , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , RNA/metabolism , Th1 Cells/immunology , Vaccines, Virus-Like Particle/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Female , Humans , Immunity, Cellular , Influenza Vaccines/metabolism , Influenza, Human/immunology , Influenza, Human/prevention & control , Lung/immunology , Lung/pathology , Lung/virology , Mice , Myeloid Differentiation Factor 88/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Signal Transduction , Vaccination , Vaccines, Virus-Like Particle/metabolism , Viral Load
14.
J Infect Dis ; 204(11): 1692-701, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21998474

ABSTRACT

Despite the medical importance of respiratory syncytial virus (RSV) infections, there is no vaccine or therapeutic agent available. Prophylactic administration of palivizumab, a humanized monoclonal RSV fusion (F) protein-specific antibody, can protect high-risk children. Previously, we have demonstrated that RSV can be neutralized by picomolar concentrations of a camelid immunoglobulin single-variable domain that binds the RSV protein F (F-VHHb nanobodies). Here, we investigated the mechanism by which these nanobodies neutralize RSV and tested their antiviral activity in vivo. We demonstrate that bivalent RSV F-specific nanobodies neutralize RSV infection by inhibiting fusion without affecting viral attachment. The ability of RSV F-specific nanobodies to protect against RSV infection was investigated in vivo. Intranasal administration of bivalent RSV F-specific nanobodies protected BALB/c mice from RSV infection, and associated pulmonary inflammation. Moreover, therapeutic treatment with these nanobodies after RSV infection could reduce viral replication and reduced pulmonary inflammation. Thus, nanobodies are promising therapeutic molecules for treatment of RSV.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Virus Attachment/drug effects , Virus Internalization/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Female , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Time Factors , Viral Fusion Proteins/immunology , Viral Load/drug effects
15.
Antiviral Res ; 92(3): 389-407, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21939690

ABSTRACT

In 1989, a new type of antibody was identified, first in the sera of dromedaries and later also in all other species of the Camelidae family. These antibodies do not contain a light chain and also lack the first constant heavy domain. Today it is still unclear what the evolutionary advantage of such heavy chain-only antibodies could be. In sharp contrast, the broad applicability of the isolated variable antigen-binding domains (VHH) was rapidly recognized, especially for the development of therapeutic proteins, called Nanobodies(®). Here we summarize first some of the unique characteristics and features of VHHs. These will next be described in the context of different experimental therapeutic applications of Nanobodies against different viruses: HIV, Hepatitis B virus, influenza virus, Respiratory Syncytial virus, Rabies virus, FMDV, Poliovirus, Rotavirus, and PERVs. Next, the diagnostic application of VHHs (Vaccinia virus, Marburg virus and plant Tulip virus X), as well as an industrial application (lytic lactococcal 936 phage) will be described. In addition, the described data show that monovalent Nanobodies can possess unique characteristics not observed with conventional antibodies. The straightforward formatting into bivalent, multivalent, and/or multispecific Nanobodies allowed tailoring molecules for potency and cross-reactivity against viral targets with high sequence diversity.


Subject(s)
Antibodies, Viral/therapeutic use , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/therapeutic use , Virus Diseases/diagnosis , Virus Diseases/drug therapy , Viruses/immunology , Animals , Antibodies, Viral/immunology , Camelids, New World/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Virus Diseases/immunology
16.
J Infect Dis ; 203(8): 1063-72, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21450996

ABSTRACT

Influenza A virus infections impose a recurrent and global disease burden. Current antivirals against influenza are not always effective. We assessed the protective potential of monovalent and bivalent Nanobodies (Ablynx) against challenge with this virus. These Nanobodies were derived from llamas and target H5N1 hemagglutinin. Intranasal administration of Nanobodies effectively controlled homologous influenza A virus replication. Administration of Nanobodies before challenge strongly reduced H5N1 virus replication in the lungs and protected mice from morbidity and mortality after a lethal challenge with H5N1 virus. The bivalent Nanobody was at least 60-fold more effective than the monovalent Nanobody in controlling virus replication. In addition, Nanobody therapy after challenge strongly reduced viral replication and significantly delayed time to death. Epitope mapping revealed that the VHH Nanobody binds to antigenic site B in H5 hemagglutinin. Because Nanobodies are small, stable, and simple to produce, they are a promising, novel therapeutic agent against influenza.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibody Specificity , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Nanocapsules , Protein Conformation , Specific Pathogen-Free Organisms , Time Factors
17.
PLoS One ; 6(4): e17665, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21483777

ABSTRACT

For efficient prevention of viral infections and cross protection, simultaneous targeting of multiple viral epitopes is a powerful strategy. Llama heavy chain antibody fragments (VHH) against the trimeric envelope proteins of Respiratory Syncytial Virus (Fusion protein), Rabies virus (Glycoprotein) and H5N1 Influenza (Hemagglutinin 5) were selected from llama derived immune libraries by phage display. Neutralizing VHH recognizing different epitopes in the receptor binding sites on the spikes with affinities in the low nanomolar range were identified for all the three viruses by viral neutralization assays. By fusion of VHH with variable linker lengths, multimeric constructs were made that improved neutralization potencies up to 4,000-fold for RSV, 1,500-fold for Rabies virus and 75-fold for Influenza H5N1. The potencies of the VHH constructs were similar or better than best performing monoclonal antibodies. The cross protection capacity against different viral strains was also improved for all three viruses, both by multivalent (two or three identical VHH) and biparatopic (two different VHH) constructs. By combining a VHH neutralizing RSV subtype A, but not subtype B with a poorly neutralizing VHH with high affinity for subtype B, a biparatopic construct was made with low nanomolar neutralizing potency against both subtypes. Trivalent anti-H5N1 VHH neutralized both Influenza H5N1 clade1 and 2 in a pseudotype assay and was very potent in neutralizing the NIBRG-14 Influenza H5N1 strain with IC(50) of 9 picomolar. Bivalent and biparatopic constructs against Rabies virus cross neutralized both 10 different Genotype 1 strains and Genotype 5.The results show that multimerization of VHH fragments targeting multiple epitopes on a viral trimeric spike protein is a powerful tool for anti-viral therapy to achieve "best-in-class" and broader neutralization capacity.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Camelids, New World , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/immunology , Viruses/immunology , Animals , Antibodies, Neutralizing/isolation & purification , Antibody Specificity , Antiviral Agents/immunology , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , Cross Reactions , Dose-Response Relationship, Immunologic , Epitopes/immunology , Genotype , Immunoglobulin Heavy Chains/isolation & purification , Influenza A Virus, H5N1 Subtype/immunology , Lyssavirus/genetics , Lyssavirus/immunology , Respiratory Syncytial Viruses/immunology , Viral Proteins/immunology
18.
J Biomed Biotechnol ; 2010: 863985, 2010.
Article in English | MEDLINE | ID: mdl-20508820

ABSTRACT

Influenza is a vaccine preventable disease that causes severe illness and excess mortality in humans. Licensed influenza vaccines induce humoral immunity and protect against strains that antigenically match the major antigenic components of the vaccine, but much less against antigenically diverse influenza strains. A vaccine that protects against different influenza viruses belonging to the same subtype or even against viruses belonging to more than one subtype would be a major advance in our battle against influenza. Heterosubtypic immunity could be obtained by cytotoxic T-cell (CTL) responses against conserved influenza virus epitopes. The molecular mechanisms involved in inducing protective CTL responses are discussed here. We also focus on CTL vaccine design and point to the importance of immune-related databases and immunoinformatics tools in the quest for new vaccine candidates. Some techniques for analysis of T-cell responses are also highlighted, as they allow estimation of cellular immune responses induced by vaccine preparations and can provide correlates of protection.

SELECTION OF CITATIONS
SEARCH DETAIL
...