Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 5735, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38459086

ABSTRACT

The spike protein (S) of SARS-CoV-2 induces neutralizing antibodies and is the key component of current COVID-19 vaccines. The most efficacious COVID-19 vaccines are genetically-encoded spikes with a double proline substitution in the hinge region to stabilize S in the prefusion conformation (S-2P). A subunit vaccine can be a valuable addition to mRNA and viral vector-based vaccines but requires high stability of spike. In addition, further stabilization of the prefusion conformation of spike might improve immunogenicity. To test this, five spike proteins were designed and characterized, ranging from low to high stability. The immunogenicity of these proteins was assessed in mice, demonstrating that a spike (S-closed-2) with a high melting temperature, which still allowed ACE2 binding, induced the highest neutralization titers against homologous and heterologous strains (up to 16-fold higher than the least stabilized spike). In contrast, the most stable spike variant (S-locked), in which the receptor binding domains (RBDs) were locked in a closed conformation and thus not able to breathe, induced relatively low neutralizing antibody titers against heterologous strains. These data demonstrate that S protein stabilization with RBDs exposing highly conserved epitopes may be needed to increase the immunogenicity of spike proteins for future COVID-19 vaccines.


Subject(s)
COVID-19 , Viral Vaccines , Mice , Humans , Animals , SARS-CoV-2 , COVID-19 Vaccines , Antibodies, Viral , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/prevention & control , Antibodies, Neutralizing
2.
NPJ Vaccines ; 9(1): 8, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184641

ABSTRACT

The quantitation of antibody responses is a critical requirement for the successful development of vaccines and therapeutics that often relies on the use of standardized reference materials to determine relative quantities within biological samples. The validity of comparing responses across assays using arbitrarily defined reference values is therefore limited. We developed a generalizable method known as MASCALE (Mass Spectrometry Enabled Conversion to Absolute Levels of ELISA Antibodies) for absolute quantitation of antibodies by calibrating ELISA reference sera using mass spectrometry. Levels of proteotypic peptides served as a proxy for human IgG, allowing the conversion of responses from arbitrary values to absolute amounts. Applications include comparison of binding assays at two separate laboratories and evaluation of cross-clade magnitude-breadth responses induced by an investigational HIV-1 vaccine regimen. MASCALE addresses current challenges in the interpretation of immune responses in clinical trials and expands current options available to make suitable comparisons across different settings.

3.
PLoS Pathog ; 19(5): e1011308, 2023 05.
Article in English | MEDLINE | ID: mdl-37126534

ABSTRACT

The global spread of the SARS-CoV-2 virus has resulted in emergence of lineages which impact the effectiveness of immunotherapies and vaccines that are based on the early Wuhan isolate. All currently approved vaccines employ the spike protein S, as it is the target for neutralizing antibodies. Here we describe two SARS-CoV-2 isolates with unusually large deletions in the N-terminal domain (NTD) of the spike. Cryo-EM structural analysis shows that the deletions result in complete reshaping of the NTD supersite, an antigenically important region of the NTD. For both spike variants the remodeling of the NTD negatively affects binding of all tested NTD-specific antibodies in and outside of the NTD supersite. For one of the variants, we observed a P9L mediated shift of the signal peptide cleavage site resulting in the loss of a disulfide-bridge; a unique escape mechanism with high antigenic impact. Although the observed deletions and disulfide mutations are rare, similar modifications have become independently established in several other lineages, indicating a possibility to become more dominant in the future. The observed plasticity of the NTD foreshadows its broad potential for immune escape with the continued spread of SARS-CoV-2.


Subject(s)
COVID-19 , Humans , SARS-CoV-2/genetics , Antibodies, Neutralizing , Disulfides , Immunotherapy , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral
4.
NPJ Vaccines ; 8(1): 40, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36927774

ABSTRACT

Since the original outbreak of the SARS-CoV-2 virus, several rapidly spreading SARS-CoV-2 variants of concern (VOC) have emerged. Here, we show that a single dose of Ad26.COV2.S (based on the Wuhan-Hu-1 spike variant) protects against the Gamma and Delta variants in naive hamsters, supporting the observed maintained vaccine efficacy in humans against these VOC. Adapted spike-based booster vaccines targeting Omicron variants have now been authorized in the absence of human efficacy data. We evaluated the immunogenicity and efficacy of Ad26.COV2.S.529 (encoding a stabilized Omicron BA.1 spike) in naive mice and in hamsters with pre-existing immunity to the Wuhan-Hu-1 spike. In naive mice, Ad26.COV2.S.529 elicited higher neutralizing antibody titers against SARS-CoV-2 Omicron BA.1 and BA.2, compared with Ad26.COV2.S. However, neutralizing titers against the SARS-CoV-2 B.1 (D614G) and Delta variants were lower after primary vaccination with Ad26.COV2.S.529 compared with Ad26.COV2.S. In contrast, we found comparable Omicron BA.1 and BA.2 neutralizing titers in hamsters with pre-existing Wuhan-Hu-1 spike immunity after vaccination with Ad26.COV2.S, Ad26.COV2.S.529 or a combination of the two vaccines. Moreover, all three vaccine modalities induced equivalent protection against Omicron BA.2 challenge in these animals. Overall, our data suggest that an Omicron BA.1-based booster in rodents does not improve immunogenicity and efficacy against Omicron BA.2 over an Ad26.COV2.S booster in a setting of pre-existing immunity to SARS-CoV-2.

5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35131851

ABSTRACT

For an efficacious vaccine immunogen, influenza hemagglutinin (HA) needs to maintain a stable quaternary structure, which is contrary to the inherently dynamic and metastable nature of class I fusion proteins. In this study, we stabilized HA with three substitutions within its pH-sensitive regions where the refolding starts. An X-ray structure reveals how these substitutions stabilize the intersubunit ß-sheet in the base and form an interprotomeric aliphatic layer across the stem while the native prefusion HA fold is retained. The identification of the stabilizing substitutions increases our understanding of how the pH sensitivity is structurally accomplished in HA and possibly other pH-sensitive class I fusion proteins. Our stabilization approach in combination with the occasional back mutation of rare amino acids to consensus results in well-expressing stable trimeric HAs. This repair and stabilization approach, which proves broadly applicable to all tested influenza A HAs of group 1 and 2, will improve the developability of influenza vaccines based on different types of platforms and formats and can potentially improve efficacy.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinins/genetics , Amino Acids/genetics , Cell Line , Humans , Hydrogen-Ion Concentration , Influenza Vaccines/genetics , Influenza, Human/virology , Mutation/genetics , Protein Conformation, beta-Strand/genetics
6.
Nat Commun ; 12(1): 244, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431842

ABSTRACT

The trimeric spike (S) protein of SARS-CoV-2 is the primary focus of most vaccine design and development efforts. Due to intrinsic instability typical of class I fusion proteins, S tends to prematurely refold to the post-fusion conformation, compromising immunogenic properties and prefusion trimer yields. To support ongoing vaccine development efforts, we report the structure-based design of soluble S trimers with increased yields and stabilities, based on introduction of single point mutations and disulfide-bridges. We identify regions critical for stability: the heptad repeat region 1, the SD1 domain and position 614 in SD2. We combine a minimal selection of mostly interprotomeric mutations to create a stable S-closed variant with a 6.4-fold higher expression than the parental construct while no longer containing a heterologous trimerization domain. The cryo-EM structure reveals a correctly folded, predominantly closed pre-fusion conformation. Highly stable and well producing S protein and the increased understanding of S protein structure will support vaccine development and serological diagnostics.


Subject(s)
SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Cryoelectron Microscopy , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Stability , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/genetics
7.
Cell Rep ; 33(8): 108432, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33238130

ABSTRACT

Soluble envelope (Env) trimers, stabilized in a prefusion-closed conformation, can elicit neutralizing responses against HIV-1 strains closely related to the immunizing trimer. However, to date such stabilization has succeeded with only a limited number of HIV-1 strains. To address this issue, here we develop ADROITrimer, an automated procedure involving structure-based stabilization and consensus repair, and generate "RnS-DS-SOSIP"-stabilized Envs from 180 diverse Env sequences. The vast majority of these RnS-DS-SOSIP Envs fold into prefusion-closed conformations as judged by antigenic analysis and size exclusion chromatography. Additionally, representative strains from clades AE, B, and C are stabilized in prefusion-closed conformations as shown by negative-stain electron microscopy, and the crystal structure of a clade A strain MI369.A5 Env trimer provides 3.5 Å resolution detail into stabilization and repair mutations. The automated procedure reported herein that yields well-behaved, soluble, prefusion-closed Env trimers from a majority of HIV-1 strains could have substantial impact on the development of an HIV-1 vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV-1/immunology , Protein Multimerization/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Consensus , Humans
8.
Cell Rep ; 30(13): 4540-4550.e3, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234486

ABSTRACT

Ebola virus causes severe hemorrhagic fever, often leading to death in humans. The trimeric fusion glycoprotein (GP) is the sole target for neutralizing antibodies and is the major focus of vaccine development. Soluble GP ectodomains are unstable and mostly monomeric when not fused to a heterologous trimerization domain. Here, we report structure-based designs of Ebola and Marburg GP trimers based on a stabilizing mutation in the hinge loop in refolding region 1 and substitution of a partially buried charge at the interface of the GP1 and GP2 subunits. The combined substitutions (T577P and K588F) substantially increased trimer expression for Ebola GP proteins. We determined the crystal structure of stabilized GP from the Makona Zaire ebolavirus strain without a trimerization domain or complexed ligand. The structure reveals that the stabilized GP adopts the same trimeric prefusion conformation, provides insight into triggering of GP conformational changes, and should inform future filovirus vaccine development.


Subject(s)
Filoviridae/metabolism , Glycoproteins/chemistry , Protein Multimerization , Amino Acid Substitution , Cell Line , Crystallography, X-Ray , Ebolavirus/metabolism , Glycoproteins/genetics , Humans , Marburgvirus/metabolism , Models, Molecular , Mutation/genetics , Perfusion , Protein Domains , Protein Stability , Structure-Activity Relationship
9.
Science ; 363(6431)2019 03 08.
Article in English | MEDLINE | ID: mdl-30846569

ABSTRACT

Recent characterization of broadly neutralizing antibodies (bnAbs) against influenza virus identified the conserved hemagglutinin (HA) stem as a target for development of universal vaccines and therapeutics. Although several stem bnAbs are being evaluated in clinical trials, antibodies are generally unsuited for oral delivery. Guided by structural knowledge of the interactions and mechanism of anti-stem bnAb CR6261, we selected and optimized small molecules that mimic the bnAb functionality. Our lead compound neutralizes influenza A group 1 viruses by inhibiting HA-mediated fusion in vitro, protects mice against lethal and sublethal influenza challenge after oral administration, and effectively neutralizes virus infection in reconstituted three-dimensional cell culture of fully differentiated human bronchial epithelial cells. Cocrystal structures with H1 and H5 HAs reveal that the lead compound recapitulates the bnAb hotspot interactions.


Subject(s)
Antibodies, Neutralizing/chemistry , Biomimetic Materials/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/prevention & control , Piperazines/pharmacology , Pyridines/pharmacology , Tetrazoles/pharmacology , Viral Fusion Protein Inhibitors/pharmacology , Virus Internalization/drug effects , Administration, Oral , Animals , Biomimetic Materials/administration & dosage , Biomimetic Materials/pharmacokinetics , Bronchi/virology , Cells, Cultured , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Madin Darby Canine Kidney Cells , Mice , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Respiratory Mucosa/virology , Tetrazoles/administration & dosage , Tetrazoles/pharmacokinetics , Viral Fusion Protein Inhibitors/administration & dosage , Viral Fusion Protein Inhibitors/pharmacokinetics
10.
Sci Rep ; 9(1): 4735, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30894620

ABSTRACT

Epitope characterization is critical for elucidating the mechanism of action of drug candidates. However, traditional high-resolution epitope mapping techniques are not well suited for screening numerous drug candidates recognizing a similar target. Here, we use Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS) to explore the conformational impact of diverse drug molecules binding on Hemagglutinin (HA), the major surface antigen of influenza viruses. We optimized a semi-automated HDX-MS workflow to systematically probe distantly related HA subtypes in complex with 4 different drug candidates, ranging from a monoclonal antibody to a small synthetic peptide. This fast, cost-effective HDX-MS epitope mapping approach accurately determined the main antigenic site in all cases. Moreover, our studies reveal distinct changes in the local conformational dynamics of HA associated to the molecular mechanism of neutralization, establishing a marker for broad anti-HA activity. Taken together, these findings highlight the potential for HDX-MS epitope mapping-based screening to identify promising candidates against HA at early stages of drug discovery.


Subject(s)
Epitope Mapping/methods , Hemagglutinins/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry/methods , Influenza, Human/drug therapy , Drug Discovery/methods , Hemagglutinins/immunology , Humans , Pharmaceutical Preparations/metabolism , Protein Binding
11.
Acta Neuropathol Commun ; 6(1): 59, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30001207

ABSTRACT

Aggregation of tau protein and spreading of tau aggregates are pivotal pathological processes in a range of neurological disorders. Accumulating evidence suggests that immunotherapy targeting tau may be a viable therapeutic strategy. We have previously described the isolation of antibody CBTAU-22.1 from the memory B-cell repertoire of healthy human donors. CBTAU-22.1 was shown to specifically bind a disease-associated phosphorylated epitope in the C-terminus of tau (Ser422) and to be able to inhibit the spreading of pathological tau aggregates from P301S spinal cord lysates in vitro, albeit with limited potency. Using a combination of rational design and random mutagenesis we have derived a variant antibody with improved affinity while maintaining the specificity of the parental antibody. This affinity improved antibody showed greatly enhanced potency in a cell-based immunodepletion assay using paired helical filaments (PHFs) derived from human Alzheimer's disease (AD) brain tissue. Moreover, the affinity improved antibody limits the in vitro aggregation propensity of full length tau species specifically phosphorylated at position 422 produced by employing a native chemical ligation approach. Together, these results indicate that in addition to being able to inhibit the spreading of pathological tau aggregates, the matured antibody can potentially also interfere with the nucleation of tau which is believed to be the first step of the pathogenic process. Finally, the functionality in a P301L transgenic mice co-injection model highlights the therapeutic potential of human antibody dmCBTAU-22.1.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Antibodies/pharmacology , Brain/metabolism , Serine/metabolism , tau Proteins/immunology , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Antibody Affinity/drug effects , Autopsy , Brain/pathology , Dose-Response Relationship, Drug , Epitopes/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Microscopy, Atomic Force , Middle Aged , Models, Molecular , Mutagenesis , Mutation/genetics , Phosphorylation/physiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/therapy
12.
Acta Neuropathol Commun ; 6(1): 43, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855358

ABSTRACT

Misfolding and aggregation of tau protein are closely associated with the onset and progression of Alzheimer's Disease (AD). By interrogating IgG+ memory B cells from asymptomatic donors with tau peptides, we have identified two somatically mutated VH5-51/VL4-1 antibodies. One of these, CBTAU-27.1, binds to the aggregation motif in the R3 repeat domain and blocks the aggregation of tau into paired helical filaments (PHFs) by sequestering monomeric tau. The other, CBTAU-28.1, binds to the N-terminal insert region and inhibits the spreading of tau seeds and mediates the uptake of tau aggregates into microglia by binding PHFs. Crystal structures revealed that the combination of VH5-51 and VL4-1 recognizes a common Pro-Xn-Lys motif driven by germline-encoded hotspot interactions while the specificity and thereby functionality of the antibodies are defined by the CDR3 regions. Affinity improvement led to improvement in functionality, identifying their epitopes as new targets for therapy and prevention of AD.


Subject(s)
B-Lymphocytes/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/metabolism , tau Proteins/immunology , tau Proteins/metabolism , Adolescent , Adult , Aged , Antibody Specificity , B-Lymphocytes/drug effects , Crystallization , Dose-Response Relationship, Drug , Female , Humans , Immunodominant Epitopes/metabolism , Male , Microglia/metabolism , Microscopy, Atomic Force , Middle Aged , Models, Molecular , Molecular Sequence Data , Protein Aggregates , Young Adult
13.
Science ; 358(6362): 496-502, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28971971

ABSTRACT

Influenza therapeutics with new targets and mechanisms of action are urgently needed to combat potential pandemics, emerging viruses, and constantly mutating strains in circulation. We report here on the design and structural characterization of potent peptidic inhibitors of influenza hemagglutinin. The peptide design was based on complementarity-determining region loops of human broadly neutralizing antibodies against the hemagglutinin (FI6v3 and CR9114). The optimized peptides exhibit nanomolar affinity and neutralization against influenza A group 1 viruses, including the 2009 H1N1 pandemic and avian H5N1 strains. The peptide inhibitors bind to the highly conserved stem epitope and block the low pH-induced conformational rearrangements associated with membrane fusion. These peptidic compounds and their advantageous biological properties should accelerate the development of new small molecule- and peptide-based therapeutics against influenza virus.


Subject(s)
Antiviral Agents/chemistry , Drug Design , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Peptides, Cyclic/chemistry , Virus Internalization/drug effects , Animals , Antibodies, Neutralizing/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Complementarity Determining Regions/chemistry , Crystallography, X-Ray , Humans , Male , Mice , Mice, Inbred BALB C , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Protein Conformation
15.
Proc Natl Acad Sci U S A ; 111(48): E5143-8, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404330

ABSTRACT

The ability of antibodies binding the influenza hemagglutinin (HA) protein to neutralize viral infectivity is of key importance in the design of next-generation vaccines and for prophylactic and therapeutic use. The two antibodies CR6261 and CR8020 have recently been shown to efficiently neutralize influenza A infection by binding to and inhibiting the influenza A HA protein that is responsible for membrane fusion in the early steps of viral infection. Here, we use single-particle fluorescence microscopy to correlate the number of antibodies or antibody fragments (Fab) bound to an individual virion with the capacity of the same virus particle to undergo membrane fusion. To this end, individual, infectious virus particles bound by fluorescently labeled antibodies/Fab are visualized as they fuse to a planar, supported lipid bilayer. The fluorescence intensity arising from the virus-bound antibodies/Fab is used to determine the number of molecules attached to viral HA while a fluorescent marker in the viral membrane is used to simultaneously obtain kinetic information on the fusion process. We experimentally determine that the stoichiometry required for fusion inhibition by both antibody and Fab leaves large numbers of unbound HA epitopes on the viral surface. Kinetic measurements of the fusion process reveal that those few particles capable of fusion at high antibody/Fab coverage display significantly slower hemifusion kinetics. Overall, our results support a membrane fusion mechanism requiring the stochastic, coordinated action of multiple HA trimers and a model of fusion inhibition by stem-binding antibodies through disruption of this coordinated action.


Subject(s)
Antibodies, Neutralizing/immunology , Influenza A virus/immunology , Membrane Fusion/immunology , Virion/immunology , Antibodies, Neutralizing/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N1 Subtype/ultrastructure , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H3N2 Subtype/ultrastructure , Influenza A virus/physiology , Influenza A virus/ultrastructure , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Kinetics , Membrane Fusion/drug effects , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Monte Carlo Method , Protein Binding , Virion/drug effects , Virion/ultrastructure , Virus Internalization/drug effects
16.
Proc Natl Acad Sci U S A ; 111(1): 445-50, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24335589

ABSTRACT

The discovery and characterization of broadly neutralizing antibodies (bnAbs) against influenza viruses have raised hopes for the development of monoclonal antibody (mAb)-based immunotherapy and the design of universal influenza vaccines. Only one human bnAb (CR8020) specifically recognizing group 2 influenza A viruses has been previously characterized that binds to a highly conserved epitope at the base of the hemagglutinin (HA) stem and has neutralizing activity against H3, H7, and H10 viruses. Here, we report a second group 2 bnAb, CR8043, which was derived from a different germ-line gene encoding a highly divergent amino acid sequence. CR8043 has in vitro neutralizing activity against H3 and H10 viruses and protects mice against challenge with a lethal dose of H3N2 and H7N7 viruses. The crystal structure and EM reconstructions of the CR8043-H3 HA complex revealed that CR8043 binds to a site similar to the CR8020 epitope but uses an alternative angle of approach and a distinct set of interactions. The identification of another antibody against the group 2 stem epitope suggests that this conserved site of vulnerability has great potential for design of therapeutics and vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Influenza A virus/chemistry , Animals , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Immunologic Memory , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Kinetics , Mice , Mice, Inbred BALB C , Microscopy, Electron , Models, Molecular , Molecular Conformation , Species Specificity
17.
PLoS One ; 8(12): e80034, 2013.
Article in English | MEDLINE | ID: mdl-24348996

ABSTRACT

Human monoclonal antibodies have been identified which neutralize broad spectra of influenza A or B viruses. Here, we dissect the mechanisms by which such antibodies interfere with infectivity. We distinguish four mechanisms that link the conserved hemagglutinin (HA) epitopes of broadly neutralizing antibodies to critical processes in the viral life cycle. HA-stem binding antibodies can act intracellularly by blocking fusion between the viral and endosomal membranes and extracellularly by preventing the proteolytic activation of HA. HA-head binding antibodies prevent viral attachment and release. These insights into newly identified ways by which the human immune system can interfere with influenza virus infection may aid the development of novel universal vaccines and antivirals.


Subject(s)
Hemagglutinins/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Dogs , Electrophoresis, Polyacrylamide Gel , Epitopes/immunology , Humans , Immunoblotting , Influenza A virus/immunology , Madin Darby Canine Kidney Cells , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission
18.
Cancer Cell ; 23(4): 489-501, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597563

ABSTRACT

The fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling network plays an important role in cell growth, survival, differentiation, and angiogenesis. Deregulation of FGFR signaling can lead to cancer development. Here, we report an FGFR inhibitor, SSR128129E (SSR), that binds to the extracellular part of the receptor. SSR does not compete with FGF for binding to FGFR but inhibits FGF-induced signaling linked to FGFR internalization in an allosteric manner, as shown by crystallography studies, nuclear magnetic resonance, Fourier transform infrared spectroscopy, molecular dynamics simulations, free energy calculations, structure-activity relationship analysis, and FGFR mutagenesis. Overall, SSR is a small molecule allosteric inhibitor of FGF/FGFR signaling, acting via binding to the extracellular part of the FGFR.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Binding, Competitive , Cell Growth Processes/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Structure, Tertiary , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
19.
J Chem Phys ; 133(17): 174109, 2010 Nov 07.
Article in English | MEDLINE | ID: mdl-21054008

ABSTRACT

We introduce a reweighting scheme for the path ensembles in the transition interface sampling framework. The reweighting allows for the analysis of free energy landscapes and committor projections in any collective variable space. We illustrate the reweighting scheme on a two dimensional potential with a nonlinear reaction coordinate and on a more realistic simulation of the Trp-cage folding process. We suggest that the reweighted path ensemble can be used to optimize possible nonlinear reaction coordinates.


Subject(s)
Molecular Dynamics Simulation , Protein Folding , Proteins/chemistry , Nonlinear Dynamics , Solutions/chemistry , Thermodynamics
20.
J Chem Phys ; 133(17): 174110, 2010 Nov 07.
Article in English | MEDLINE | ID: mdl-21054009

ABSTRACT

We present a flexible nonlinear reaction coordinate analysis method for the transition path ensemble based on the likelihood maximization approach developed by Peters and Trout [J. Chem. Phys. 125, 054108 (2006)]. By parametrizing the reaction coordinate by a string of images in a collective variable space, we can optimize the likelihood that the string correctly models the committor data obtained from a path sampling simulation. The collective variable space with the maximum likelihood is considered to contain the best description of the reaction. The use of the reweighted path ensemble [J. Rogal et al., J. Chem. Phys. 133, 174109 (2010)] allows a complete reaction coordinate description from the initial to the final state. We illustrate the method on a z-shaped two-dimensional potential. While developed for use with path sampling, this analysis method can also be applied to regular molecular dynamics trajectories.


Subject(s)
Molecular Dynamics Simulation , Nonlinear Dynamics , Proteins/analysis , Bayes Theorem , Monte Carlo Method , Proteins/chemistry , Solutions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...