Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (132)2018 02 23.
Article in English | MEDLINE | ID: mdl-29553549

ABSTRACT

Antibodies play a crucial role in coupling the innate and adaptive immune responses against viral pathogens through their antigen binding domains and Fc-regions. Here, we describe how to measure the activation of Fc effector functions by monoclonal antibodies targeting the influenza virus hemagglutinin with the use of a genetically engineered Jurkat cell line expressing an activating type 1 Fc-FcγR. Using this method, the contribution of specific Fc-FcγR interactions conferred by immunoglobulins can be determined using an in vitro assay.


Subject(s)
Hemagglutinins/metabolism , Influenza, Human/virology , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Antibodies, Monoclonal/immunology , Humans
2.
Nat Commun ; 8(1): 846, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29018261

ABSTRACT

The aim of candidate universal influenza vaccines is to provide broad protection against influenza A and B viruses. Studies have demonstrated that broadly reactive antibodies require Fc-Fc gamma receptor interactions for optimal protection; however, the innate effector cells responsible for mediating this protection remain largely unknown. Here, we examine the roles of alveolar macrophages, natural killer cells, and neutrophils in antibody-mediated protection. We demonstrate that alveolar macrophages play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies in mice. Our data also reveal the potential mechanisms by which alveolar macrophages mediate protection in vivo, namely antibody-induced inflammation and antibody-dependent cellular phagocytosis. This study highlights the importance of innate effector cells in establishing a broad-spectrum antiviral state, as well as providing a better understanding of how multiple arms of the immune system cooperate to achieve an optimal antiviral response following influenza virus infection or immunization.Broadly reactive antibodies that recognize influenza A virus HA can be protective, but the mechanism is not completely understood. Here, He et al. show that the inflammatory response and phagocytosis mediated by the interaction between protective antibodies and macrophages are essential for protection.


Subject(s)
Antibodies, Neutralizing/physiology , Influenza A virus/immunology , Influenza, Human/immunology , Macrophage Activation , Macrophages, Alveolar/physiology , A549 Cells , Animals , Dogs , Female , HEK293 Cells , Hemagglutinins/immunology , Humans , Killer Cells, Natural/physiology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/physiology , Orthomyxoviridae Infections/immunology , Phagocytosis , Receptors, IgG/metabolism
3.
J Vis Exp ; (126)2017 08 29.
Article in English | MEDLINE | ID: mdl-28872136

ABSTRACT

Influenza viruses exhibit a remarkable ability to adapt and evade the host immune response. One way is through antigenic changes that occur on the surface glycoproteins of the virus. The generation of escape variants is a powerful method in elucidating how viruses escape immune detection and in identifying critical residues required for antibody binding. Here, we describe a protocol on how to generate influenza A virus escape variants by utilizing human or murine monoclonal antibodies (mAbs) directed against the viral hemagglutinin (HA). With the use of our technique, we previously characterized critical residues required for the binding of antibodies targeting either the head or stalk of the novel avian H7N9 HA. The protocol can be easily adapted for other virus systems. Analyses of escape variants are important for modeling antigenic drift, determining single nucleotide polymorphisms (SNPs) conferring resistance and virus fitness, and in the designing of vaccines and/or therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Influenza, Human/virology , Humans
4.
Proc Natl Acad Sci U S A ; 113(40): E5944-E5951, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647907

ABSTRACT

Influenza virus strain-specific monoclonal antibodies (mAbs) provide protection independent of Fc gamma receptor (FcγR) engagement. In contrast, optimal in vivo protection achieved by broadly reactive mAbs requires Fc-FcγR engagement. Most strain-specific mAbs target the head domain of the viral hemagglutinin (HA), whereas broadly reactive mAbs typically recognize epitopes within the HA stalk. This observation has led to questions regarding the mechanism regulating the activation of Fc-dependent effector functions by broadly reactive antibodies. To dissect the molecular mechanism responsible for this dichotomy, we inserted the FLAG epitope into discrete locations on HAs. By characterizing the interactions of several FLAG-tagged HAs with a FLAG-specific antibody, we show that in addition to Fc-FcγR engagement mediated by the FLAG-specific antibody, a second intermolecular bridge between the receptor-binding region of the HA and sialic acid on effector cells is required for optimal activation. Inhibition of this second molecular bridge, through the use of an F(ab')2 or the mutation of the sialic acid-binding site, renders the Fc-FcγR interaction unable to optimally activate effector cells. Our findings indicate that broadly reactive mAbs require two molecular contacts to possibly stabilize the immunologic synapse and potently induce antibody-dependent cell-mediated antiviral responses: (i) the interaction between the Fc of a mAb bound to HA with the FcγR of the effector cell and (ii) the interaction between the HA and its sialic acid receptor on the effector cell. This concept might be broadly applicable for protective antibody responses to viral pathogens that have suitable receptors on effector cells.


Subject(s)
Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Orthomyxoviridae/immunology , Receptors, Fc/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Antibodies, Viral/chemistry , Antibody-Dependent Cell Cytotoxicity , Epitopes/chemistry , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Immunity, Cellular , Models, Biological , Models, Molecular , N-Acetylneuraminic Acid/metabolism , Receptors, Fc/chemistry
5.
Cell Host Microbe ; 19(6): 800-13, 2016 Jun 08.
Article in English | MEDLINE | ID: mdl-27281570

ABSTRACT

Pathogenic H7N9 avian influenza viruses continue to represent a public health concern, and several candidate vaccines are currently being developed. It is vital to assess if protective antibodies are induced following vaccination and to characterize the diversity of epitopes targeted. Here we characterized the binding and functional properties of twelve H7-reactive human antibodies induced by a candidate A/Anhui/1/2013 (H7N9) vaccine. Both neutralizing and non-neutralizing antibodies protected mice in vivo during passive transfer challenge experiments. Mapping the H7 hemagglutinin antigenic sites by generating escape mutant variants against the neutralizing antibodies identified unique epitopes on the head and stalk domains. Further, the broadly cross-reactive non-neutralizing antibodies generated in this study were protective through Fc-mediated effector cell recruitment. These findings reveal important properties of vaccine-induced antibodies and provide a better understanding of the human monoclonal antibody response to influenza in the context of vaccines.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/immunology , Antigens, Viral/immunology , Disease Models, Animal , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
6.
PLoS Pathog ; 12(4): e1005578, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27081859

ABSTRACT

In the early spring of 2013, Chinese health authorities reported several cases of H7N9 influenza virus infections in humans. Since then the virus has established itself at the human-animal interface in Eastern China and continues to cause several hundred infections annually. In order to characterize the antibody response to the H7N9 virus we generated several mouse monoclonal antibodies against the hemagglutinin of the A/Shanghai/1/13 (H7N9) virus. Of particular note are two monoclonal antibodies, 1B2 and 1H5, that show broad reactivity to divergent H7 hemagglutinins. Monoclonal antibody 1B2 binds to viruses of the Eurasian and North American H7 lineages and monoclonal antibody 1H5 reacts broadly to virus isolates of the Eurasian lineage. Interestingly, 1B2 shows broad hemagglutination inhibiting and neutralizing activity, while 1H5 fails to inhibit hemagglutination and demonstrates no neutralizing activity in vitro. However, both monoclonal antibodies were highly protective in an in vivo passive transfer challenge model in mice, even at low doses. Experiments using mutant antibodies that lack the ability for Fc/Fc-receptor and Fc/complement interactions suggest that the protection provided by mAb 1H5 is, at least in part, mediated by the Fc-fragment of the mAb. These findings highlight that a protective response to a pathogen may not only be due to neutralizing antibodies, but can also be the result of highly efficacious non-neutralizing antibodies not readily detected by classical in vitro neutralization or hemagglutination inhibition assays. This is of interest because H7 influenza virus vaccines induce only low hemagglutination inhibiting antibody titers while eliciting robust antibody titers as measured by ELISA. Our data suggest that these binding but non-neutralizing antibodies contribute to protection in vivo.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Flow Cytometry , Humans , Influenza A Virus, H7N9 Subtype , Mice , Mice, Inbred BALB C
7.
Immunity ; 44(1): 46-58, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26789921

ABSTRACT

Viruses are obligate parasites and thus require the machinery of the host cell to replicate. Inhibition of host factors co-opted during active infection is a strategy hosts use to suppress viral replication and a potential pan-antiviral therapy. To define the cellular proteins and processes required for a virus during infection is thus crucial to understanding the mechanisms of virally induced disease. In this report, we generated fully infectious tagged influenza viruses and used infection-based proteomics to identify pivotal arms of cellular signaling required for influenza virus growth and infectivity. Using mathematical modeling and genetic and pharmacologic approaches, we revealed that modulation of Sec61-mediated cotranslational translocation selectively impaired glycoprotein proteostasis of influenza as well as HIV and dengue viruses and led to inhibition of viral growth and infectivity. Thus, by studying virus-human protein-protein interactions in the context of active replication, we have identified targetable host factors for broad-spectrum antiviral therapies.


Subject(s)
Host-Parasite Interactions/physiology , Influenza A virus/physiology , Influenza A virus/pathogenicity , Models, Theoretical , Virus Replication/physiology , Dengue Virus/pathogenicity , Dengue Virus/physiology , HIV/pathogenicity , HIV/physiology , Humans , Immunoprecipitation , Mass Spectrometry , Protein Folding , Proteomics
8.
Antimicrob Agents Chemother ; 59(7): 4162-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941218

ABSTRACT

The emergence of influenza virus strains resistant to approved neuraminidase inhibitors and the time constrains after infection when these drugs can be effective constitute major drawbacks for this class of drugs. This highlights a critical need to discover new therapeutic agents that can be used for the treatment of influenza virus-infected patients. The use of broadly neutralizing anti-influenza monoclonal antibodies (MAbs) has been sought as an alternative immunotherapy against influenza infection. Here, we tested in mice previously characterized broadly neutralizing anti-hemagglutinin (HA) stalk MAbs prophylactically and therapeutically using different routes of administration. The efficacy of treatment against an influenza H1N1 pandemic virus challenge was compared between two systemic routes of administration, intraperitoneal (i.p.) and intravenous (i.v.), and two local routes, intranasal (i.n.) and aerosol (a.e.). The dose of MAb required for prophylactic protection was reduced by 10-fold in animals treated locally (i.n. or a.e.) compared with those treated systemically (i.p. or i.v.). Improved therapeutic protection was observed in animals treated i.n. on day 5 postinfection (60% survival) compared with those treated via the i.p. route (20% survival). An increase in therapeutic efficacy against other influenza virus subtypes (H5N1) was also observed when a local route of administration was used. Our findings demonstrate that local administration significantly decreases the amount of broadly neutralizing monoclonal antibody required for protection against influenza, which highlights the potential use of MAbs as a therapeutic agent for influenza-associated disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/therapeutic use , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Administration, Intranasal , Administration, Intravenous , Aerosols , Animals , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents/pharmacokinetics , Biological Availability , Female , Hemagglutinins/immunology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza, Human/pathology , Influenza, Human/virology , Injections, Intraperitoneal , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Tissue Distribution
9.
J Clin Invest ; 125(3): 1255-68, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25689254

ABSTRACT

The emergence and seasonal persistence of pathogenic H7N9 influenza viruses in China have raised concerns about the pandemic potential of this strain, which, if realized, would have a substantial effect on global health and economies. H7N9 viruses are able to bind to human sialic acid receptors and are also able to develop resistance to neuraminidase inhibitors without a loss in fitness. It is not clear whether prior exposure to circulating human influenza viruses or influenza vaccination confers immunity to H7N9 strains. Here, we demonstrate that 3 of 83 H3 HA-reactive monoclonal antibodies generated by individuals that had previously undergone influenza A virus vaccination were able to neutralize H7N9 viruses and protect mice against homologous challenge. The H7N9-neutralizing antibodies bound to the HA stalk domain but exhibited a difference in their breadth of reactivity to different H7 influenza subtypes. Mapping viral escape mutations suggested that these antibodies bind at least two different epitopes on the stalk region. Together, these results indicate that these broadly neutralizing antibodies may contribute to the development of therapies against H7N9 strains and may also be effective against pathogenic H7 strains that emerge in the future.


Subject(s)
Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/prevention & control , Vaccination , Animals , Antibodies, Monoclonal/physiology , Cross Reactions , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N9 Subtype/genetics , Influenza Vaccines , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Neutralization Tests , Point Mutation
10.
J Virol ; 88(23): 13580-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25210195

ABSTRACT

UNLABELLED: Due to continuous changes to its antigenic regions, influenza viruses can evade immune detection and cause a significant amount of morbidity and mortality around the world. Influenza vaccinations can protect against disease but must be annually reformulated to match the current circulating strains. In the development of a broad-spectrum influenza vaccine, the elucidation of conserved epitopes is paramount. To this end, we designed an immunization strategy in mice to boost the humoral response against conserved regions of the hemagglutinin (HA) glycoprotein. Of note, generation and identification of broadly neutralizing antibodies that target group 2 HAs are rare and thus far have yielded only a few monoclonal antibodies (MAbs). Here, we demonstrate that mouse MAb 9H10 has broad and potent in vitro neutralizing activity against H3 and H10 group 2 influenza A subtypes. In the mouse model, MAb 9H10 protects mice against two divergent mouse-adapted H3N2 strains, in both pre- and postexposure administration regimens. In vitro and cell-free assays suggest that MAb 9H10 inhibits viral replication by blocking HA-dependent fusion of the viral and endosomal membranes early in the replication cycle and by disrupting viral particle egress in the late stage of infection. Interestingly, electron microscopy reconstructions of MAb 9H10 bound to the HA reveal that it binds a similar binding footprint to MAbs CR8020 and CR8043. IMPORTANCE: The influenza hemagglutinin is the major antigenic target of the humoral immune response. However, due to continuous antigenic changes that occur on the surface of this glycoprotein, influenza viruses can escape the immune system and cause significant disease to the host. Toward the development of broad-spectrum therapeutics and vaccines against influenza virus, elucidation of conserved regions of influenza viruses is crucial. Thus, defining these types of epitopes through the generation and characterization of broadly neutralizing monoclonal antibodies (MAbs) can greatly assist others in highlighting conserved regions of hemagglutinin. Here, we demonstrate that MAb 9H10 that targets the hemagglutinin stalk has broadly neutralizing activity against group 2 influenza A viruses in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Cell Line , Disease Models, Animal , Epitopes/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/therapeutic use , Humans , Immunization, Passive , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/therapy , Treatment Outcome , Virus Internalization/drug effects , Virus Release/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...