Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Virol ; 98(2): e0137223, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38214525

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV) are pathogenic paramyxoviruses that cause mild-to-severe disease in humans. As members of the Henipavirus genus, NiV and HeV use an attachment (G) glycoprotein and a class I fusion (F) glycoprotein to invade host cells. The F protein rearranges from a metastable prefusion form to an extended postfusion form to facilitate host cell entry. Prefusion NiV F elicits higher neutralizing antibody titers than postfusion NiV F, indicating that stabilization of prefusion F may aid vaccine development. A combination of amino acid substitutions (L104C/I114C, L172F, and S191P) is known to stabilize NiV F in its prefusion conformation, although the extent to which substitutions transfer to other henipavirus F proteins is not known. Here, we perform biophysical and structural studies to investigate the mechanism of prefusion stabilization in F proteins from three henipaviruses: NiV, HeV, and Langya virus (LayV). Three known stabilizing substitutions from NiV F transfer to HeV F and exert similar structural and functional effects. One engineered disulfide bond, located near the fusion peptide, is sufficient to stabilize the prefusion conformations of both HeV F and LayV F. Although LayV F shares low overall sequence identity with NiV F and HeV F, the region around the fusion peptide exhibits high sequence conservation across all henipaviruses. Our findings indicate that substitutions targeting this site of conformational change might be applicable to prefusion stabilization of other henipavirus F proteins and support the use of NiV as a prototypical pathogen for henipavirus vaccine antigen design.IMPORTANCEPathogenic henipaviruses such as Nipah virus (NiV) and Hendra virus (HeV) cause respiratory symptoms, with severe cases resulting in encephalitis, seizures, and coma. The work described here shows that the NiV and HeV fusion (F) proteins share common structural features with the F protein from an emerging henipavirus, Langya virus (LayV). Sequence alignment alone was sufficient to predict which known prefusion-stabilizing amino acid substitutions from NiV F would stabilize the prefusion conformations of HeV F and LayV F. This work also reveals an unexpected oligomeric interface shared by prefusion HeV F and NiV F. Together, these advances lay a foundation for future antigen design targeting henipavirus F proteins. In this way, Nipah virus can serve as a prototypical pathogen for the development of protective vaccines and monoclonal antibodies to prepare for potential henipavirus outbreaks.


Subject(s)
Hendra Virus , Henipavirus Infections , Henipavirus , Nipah Virus , Viral Proteins , Humans , Glycoproteins/metabolism , Hendra Virus/physiology , Henipavirus/physiology , Nipah Virus/genetics , Nipah Virus/metabolism , Peptides/metabolism , Viral Fusion Proteins , Viral Proteins/metabolism
2.
Nat Commun ; 14(1): 1494, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36932063

ABSTRACT

Nipah virus (NiV) is a pathogenic paramyxovirus that causes fatal encephalitis in humans. Two envelope glycoproteins, the attachment protein (G/RBP) and fusion protein (F), facilitate entry into host cells. Due to its vital role, NiV F presents an attractive target for developing vaccines and therapeutics. Several neutralization-sensitive epitopes on the NiV F apex have been described, however the antigenicity of most of the F protein's surface remains uncharacterized. Here, we immunize mice with prefusion-stabilized NiV F and isolate ten monoclonal antibodies that neutralize pseudotyped virus. Cryo-electron microscopy reveals eight neutralization-sensitive epitopes on NiV F, four of which have not previously been described. Novel sites span the lateral and basal faces of NiV F, expanding the known library of vulnerable epitopes. Seven of ten antibodies bind the Hendra virus (HeV) F protein. Multiple sequence alignment suggests that some of these newly identified neutralizing antibodies may also bind F proteins across the Henipavirus genus. This work identifies new epitopes as targets for therapeutics, provides a molecular basis for NiV neutralization, and lays a foundation for development of new cross-reactive antibodies targeting Henipavirus F proteins.


Subject(s)
Henipavirus Infections , Nipah Virus , Humans , Animals , Mice , Nipah Virus/metabolism , Epitopes , Cryoelectron Microscopy , Viral Envelope Proteins , Antibodies, Neutralizing/metabolism , Antibodies, Monoclonal
3.
Front Immunol ; 12: 772864, 2021.
Article in English | MEDLINE | ID: mdl-34956199

ABSTRACT

Nipah virus (NiV) represents a significant pandemic threat with zoonotic transmission from bats-to-humans with almost annual regional outbreaks characterized by documented human-to-human transmission and high fatality rates. Currently, no vaccine against NiV has been approved. Structure-based design and protein engineering principles were applied to stabilize the fusion (F) protein in its prefusion trimeric conformation (pre-F) to improve expression and increase immunogenicity. We covalently linked the stabilized pre-F through trimerization domains at the C-terminus to three attachment protein (G) monomers, forming a chimeric design. These studies detailed here focus on mRNA delivery of NiV immunogens in mice, assessment of mRNA immunogen-specific design elements and their effects on humoral and cellular immunogenicity. The pre-F/G chimera elicited a strong neutralizing antibody response and a superior NiV-specific Tfh and other effector T cell response compared to G alone across both the mRNA and protein platforms. These findings enabled final candidate selection of pre-F/G Fd for clinical development.


Subject(s)
Antigens, Viral/genetics , Liposomes/administration & dosage , Nanoparticles/administration & dosage , Nipah Virus/immunology , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Vaccines/administration & dosage , mRNA Vaccines/administration & dosage , Animals , Antigens, Viral/immunology , Female , Immunoglobulin G/blood , Mice , Public-Private Sector Partnerships , RNA, Messenger/administration & dosage , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology
4.
Immunity ; 54(8): 1869-1882.e6, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34270939

ABSTRACT

Vaccine-associated enhanced respiratory disease (VAERD) was previously observed in some preclinical models of severe acute respiratory syndrome (SARS) and MERS coronavirus vaccines. We used the SARS coronavirus 2 (SARS-CoV-2) mouse-adapted, passage 10, lethal challenge virus (MA10) mouse model of acute lung injury to evaluate the immune response and potential for immunopathology in animals vaccinated with research-grade mRNA-1273. Whole-inactivated virus or heat-denatured spike protein subunit vaccines with alum designed to elicit low-potency antibodies and Th2-skewed CD4+ T cells resulted in reduced viral titers and weight loss post challenge but more severe pathological changes in the lung compared to saline-immunized animals. In contrast, a protective dose of mRNA-1273 induced favorable humoral and cellular immune responses that protected from viral replication in the upper and lower respiratory tract upon challenge. A subprotective dose of mRNA-1273 reduced viral replication and limited histopathological manifestations compared to animals given saline. Overall, our findings demonstrate an immunological signature associated with antiviral protection without disease enhancement following vaccination with mRNA-1273.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , COVID-19/prevention & control , Host-Pathogen Interactions/immunology , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Biopsy , COVID-19 Vaccines/administration & dosage , Disease Models, Animal , Humans , Immunoglobulin G , Immunohistochemistry , Mice , Outcome Assessment, Health Care , RNA, Messenger , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccines, Synthetic/administration & dosage , mRNA Vaccines
5.
Nature ; 586(7830): 567-571, 2020 10.
Article in English | MEDLINE | ID: mdl-32756549

ABSTRACT

A vaccine for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is needed to control the coronavirus disease 2019 (COVID-19) global pandemic. Structural studies have led to the development of mutations that stabilize Betacoronavirus spike proteins in the prefusion state, improving their expression and increasing immunogenicity1. This principle has been applied to design mRNA-1273, an mRNA vaccine that encodes a SARS-CoV-2 spike protein that is stabilized in the prefusion conformation. Here we show that mRNA-1273 induces potent neutralizing antibody responses to both wild-type (D614) and D614G mutant2 SARS-CoV-2 as well as CD8+ T cell responses, and protects against SARS-CoV-2 infection in the lungs and noses of mice without evidence of immunopathology. mRNA-1273 is currently in a phase III trial to evaluate its efficacy.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Viral Vaccines/immunology , 2019-nCoV Vaccine mRNA-1273 , Animals , Antibodies, Neutralizing/immunology , Betacoronavirus/genetics , CD8-Positive T-Lymphocytes/immunology , COVID-19 , COVID-19 Vaccines , Clinical Trials, Phase III as Topic , Coronavirus Infections/genetics , Coronavirus Infections/virology , Female , Lung/immunology , Lung/virology , Mice , Mutation , Nose/immunology , Nose/virology , Pneumonia, Viral/virology , RNA, Messenger/genetics , RNA, Viral/genetics , SARS-CoV-2 , Th1 Cells/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics
6.
bioRxiv ; 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32577634

ABSTRACT

A SARS-CoV-2 vaccine is needed to control the global COVID-19 public health crisis. Atomic-level structures directed the application of prefusion-stabilizing mutations that improved expression and immunogenicity of betacoronavirus spike proteins. Using this established immunogen design, the release of SARS-CoV-2 sequences triggered immediate rapid manufacturing of an mRNA vaccine expressing the prefusion-stabilized SARS-CoV-2 spike trimer (mRNA-1273). Here, we show that mRNA-1273 induces both potent neutralizing antibody and CD8 T cell responses and protects against SARS-CoV-2 infection in lungs and noses of mice without evidence of immunopathology. mRNA-1273 is currently in a Phase 2 clinical trial with a trajectory towards Phase 3 efficacy evaluation.

7.
Front Immunol ; 11: 842, 2020.
Article in English | MEDLINE | ID: mdl-32595632

ABSTRACT

Licensed vaccines or therapeutics are rarely available for pathogens with epidemic or pandemic potential. Developing interventions for specific pathogens and defining generalizable approaches for related pathogens is a global priority and inherent to the UN Sustainable Development Goals. Nipah virus (NiV) poses a significant epidemic threat, and zoonotic transmission from bats-to-humans with high fatality rates occurs almost annually. Human-to-human transmission of NiV has been documented in recent outbreaks leading public health officials and government agencies to declare an urgent need for effective vaccines and therapeutics. Here, we evaluate NiV vaccine antigen design options including the fusion glycoprotein (F) and the major attachment glycoprotein (G). A stabilized prefusion F (pre-F), multimeric G constructs, and chimeric proteins containing both pre-F and G were developed as protein subunit candidate vaccines. The proteins were evaluated for antigenicity and structural integrity using kinetic binding assays, electron microscopy, and other biophysical properties. Immunogenicity of the vaccine antigens was evaluated in mice. The stabilized pre-F trimer and hexameric G immunogens both induced serum neutralizing activity in mice, while the post-F trimer immunogen did not elicit neutralizing activity. The pre-F trimer covalently linked to three G monomers (pre-F/G) induced potent neutralizing antibody activity, elicited responses to the greatest diversity of antigenic sites, and is the lead candidate for clinical development. The specific stabilizing mutations and immunogen designs utilized for NiV were successfully applied to other henipaviruses, supporting the concept of identifying generalizable solutions for prototype pathogens as an approach to pandemic preparedness.


Subject(s)
Antigens, Viral/immunology , Henipavirus Infections/prevention & control , Immunogenicity, Vaccine , Nipah Virus/chemistry , Nipah Virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , HEK293 Cells , Henipavirus Infections/virology , Humans , Immunization/methods , Mice , Mice, Inbred C57BL , Transfection , Viral Fusion Proteins/immunology , Virus Internalization
8.
Vaccines (Basel) ; 3(2): 429-47, 2015 May 26.
Article in English | MEDLINE | ID: mdl-26343196

ABSTRACT

Vaccination has proven to be an invaluable means of preventing infectious diseases by reducing both incidence of disease and mortality. However, vaccines have not been effectively developed for many diseases including HIV-1, hepatitis C virus (HCV), tuberculosis and malaria, among others. The emergence of new technologies with a growing understanding of host-pathogen interactions and immunity may lead to efficacious vaccines against pathogens, previously thought impossible.

9.
Nature ; 507(7491): 201-6, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24499818

ABSTRACT

Vaccines prevent infectious disease largely by inducing protective neutralizing antibodies against vulnerable epitopes. Several major pathogens have resisted traditional vaccine development, although vulnerable epitopes targeted by neutralizing antibodies have been identified for several such cases. Hence, new vaccine design methods to induce epitope-specific neutralizing antibodies are needed. Here we show, with a neutralization epitope from respiratory syncytial virus, that computational protein design can generate small, thermally and conformationally stable protein scaffolds that accurately mimic the viral epitope structure and induce potent neutralizing antibodies. These scaffolds represent promising leads for the research and development of a human respiratory syncytial virus vaccine needed to protect infants, young children and the elderly. More generally, the results provide proof of principle for epitope-focused and scaffold-based vaccine design, and encourage the evaluation and further development of these strategies for a variety of other vaccine targets, including antigenically highly variable pathogens such as human immunodeficiency virus and influenza.


Subject(s)
Drug Design , Epitopes/chemistry , Epitopes/immunology , Protein Stability , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Amino Acid Motifs , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Macaca mulatta/immunology , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Neutralization Tests , Protein Conformation , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/immunology
10.
Curr Top Microbiol Immunol ; 372: 307-24, 2013.
Article in English | MEDLINE | ID: mdl-24362696

ABSTRACT

A respiratory syncytial virus (RSV) vaccine has remained elusive for decades, largely due to the failure of a formalin-inactivated RSV vaccine in the 1960s that resulted in enhanced disease upon RSV exposure in the immunized individuals. Vaccine development has also been hindered by the incomplete immunity conferred by natural infection allowing for re-infection at any time, and the immature immune system and circulating maternal antibodies present in the neonate, the primary target for a vaccine. This chapter will review the use of gene delivery, both nonviral and viral, as a potential vaccine approach for human RSV. Many of these gene-based vaccines vectors elicit protective immune responses in animal models. None of the RSV gene-based platforms have progressed into clinical trials, mostly due to uncertainty regarding the direct translation of animal model results to humans and the hesitancy to invest in costly clinical trials with the potential for unclear and complicated immune responses. The continued development of RSV vaccine gene-based approaches is warranted because of their inherent flexibility with regard to composition and administration. It is likely that multiple candidate vaccines will reach human testing in the next few years.


Subject(s)
Gene Transfer Techniques , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Vaccines, DNA/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Alphavirus/genetics , Alphavirus/immunology , Animals , Child, Preschool , Dependovirus/genetics , Dependovirus/immunology , Genetic Vectors/immunology , Host Specificity , Humans , Infant , Mice , Poxviridae/genetics , Poxviridae/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Human/pathogenicity , Vaccines, DNA/administration & dosage
11.
Vaccine ; 31(6): 919-26, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23246547

ABSTRACT

Human cytomegalovirus (hCMV) is prevalent worldwide with infection generally being asymptomatic. Nevertheless, hCMV infection can lead to significant morbidity and mortality. Primary infection of seronegative women or reactivation/re-infection of seropositive women during pregnancy can result in transmission to the fetus, leading to severe neurological defects. In addition, hCMV is the most common viral infection in immunosuppressed organ transplant recipients and can produce serious complications. Hence, a safe and effective vaccine to prevent hCMV infection is an unmet medical need. Neutralizing antibodies to several hCMV glycoproteins, and complexes thereof, have been identified in individuals following hCMV infection. Interestingly, a portion of the CMV-specific neutralizing antibody responses are directed to epitopes found on glycoprotein complexes but not the individual proteins. Using an alphavirus replicon particle (VRP) vaccine platform, we showed that bicistronic VRPs encoding hCMV gH and gL glycoproteins produce gH/gL complexes in vitro. Furthermore, mice vaccinated with these gH/gL-expressing VRPs produced broadly cross-reactive complement-independent neutralizing antibodies to hCMV. These neutralizing antibody responses were of higher titer than those elicited in mice vaccinated with monocistronic VRPs encoding gH or gL antigens, and they were substantially more potent than those raised by VRPs encoding gB. These findings underscore the utility of co-delivery of glycoprotein components such as gH and gL for eliciting potent, broadly neutralizing immune responses against hCMV, and indicate that the gH/gL complex represents a potential target for future hCMV vaccine development.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytomegalovirus Vaccines/immunology , Viral Envelope Proteins/immunology , Viral Proteins/immunology , Alphavirus/genetics , Animals , Cross Reactions , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Female , Genetic Vectors , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Proteins/genetics
12.
Mol Cell ; 33(4): 450-61, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19250906

ABSTRACT

Histone H3 serine 10 phosphorylation is a hallmark of mitotic chromosomes, but its full function remains to be elucidated. We report here that two SR protein splicing factors, SRp20 and ASF/SF2, associate with interphase chromatin, are released from hyperphosphorylated mitotic chromosomes, but reassociate with chromatin late in M-phase. Inhibition of Aurora B kinase diminished histone H3 serine 10 phosphorylation and increased SRp20 and ASF/SF2 retention on mitotic chromosomes. Unexpectedly, we also found that HP1 proteins interact with ASF/SF2 in mitotic cells. Strikingly, siRNA-mediated knockdown of ASF/SF2 caused retention of HP1 proteins on mitotic chromatin. Finally, ASF/SF2-depleted cells released from a mitotic block displayed delayed G0/G1 entry, suggesting a functional consequence of these interactions. These findings underscore the evolving role of histone H3 phosphorylation and demonstrate a direct, functional, and histone-modification-regulated association of SRp20 and ASF/SF2 with chromatin.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Mitosis , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Serine/metabolism , Animals , Binding Sites , Cells, Cultured , Chickens , HeLa Cells , Humans , Nuclear Proteins/genetics , Nucleosomes/metabolism , Phosphorylation , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors
13.
Traffic ; 7(12): 1643-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17118119

ABSTRACT

RhoGTPases play important roles in the regulation of protein transport and membrane recycling. Little is known, however, about how RhoGTPases affect HIV-1 virion production, which is dependent on the endosomal sorting pathway. We report that ectopic expression of citron kinase (citron-K), a RhoA effector, preferentially enhances HIV-1 virion production. Depletion of endogenous citron-K inhibits HIV-1 virion production. Citron-N, which lacks the kinase domain, also enhances HIV-1 virion production. The leucine zipper, Rho-binding and zinc finger domains of citron-N are necessary for the enhancement activity. Citron-K also enhances murine leukemia virion production and the HIV-1 late domain is not required for the citron-K-mediated enhancement. Ectopic expression of citron-K leads to the formation of cytoplasmic structures containing citron-K and HIV-1 Gag proteins. HIV-1 and citron-K cooperatively enhance acidic endosome and lysosome compartments. Finally, citron-K promotes exocytosis of microvesicles or exosomes that co-purify with HIV-1 virions. We conclude that citron-K enhances HIV-1 virion production by stimulating the endosomal compartments and exocytosis.


Subject(s)
Exocytosis , HIV-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Virion/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line , Endosomes/metabolism , Gene Deletion , Gene Products, gag/deficiency , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/genetics , Humans , Intracellular Signaling Peptides and Proteins , Leucine Zippers , Lysosomes/metabolism , Mice , Protein Binding , Protein Serine-Threonine Kinases/genetics , Virus Replication , Zinc Fingers
14.
Arch Biochem Biophys ; 404(2): 197-209, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12147257

ABSTRACT

We have cloned and overexpressed a truncated, recombinant form of beta-carbonic anhydrase from Arabidopsis thaliana. The wild-type enzyme and two site-directed variants, H216N and Y212F, have been kinetically characterized both at steady state by stopped-flow spectrophotometry and at chemical equilibrium by (18)O isotope exchange methods. The wild-type enzyme has a maximal k(cat) for CO2 hydration of 320 ms(-1) and is rate limited by proton transfer involving two residues with apparent pK(a) values of 6.0 and 8.7. The mutant enzyme H216N has a maximal k(cat) at high pH that is 43% that of wild type, but is only 5% that of wild type at pH 7.0. (18)O exchange studies reveal that the effect of the mutations H216N or Y212F is primarily on proton transfer steps in the catalytic mechanism and not in the rate of CO2-HCO3- exchange. These results suggest that residues His-216 and Tyr-212 are both important for efficient proton transfer in A. thaliana carbonic anhydrase.


Subject(s)
Arabidopsis/enzymology , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/genetics , Amino Acid Substitution , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Binding Sites/physiology , Carbon Dioxide/chemistry , Catalysis , Enzyme Activation/physiology , Escherichia coli/genetics , Hydrogen-Ion Concentration , Imidazoles/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Kinetics , Molecular Weight , Mutagenesis, Site-Directed , Oxygen Isotopes , Protons , Substrate Specificity , Zinc/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...