Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
2.
Nat Chem Biol ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744986

ABSTRACT

G-protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small-molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue and cellular levels. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays and structural studies, we develop maternally selective heavy-chain-only antibody ('nanobody') antagonists against the angiotensin II type I receptor and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to angiotensin II type I receptor with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.

3.
Nature ; 625(7995): 572-577, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38172635

ABSTRACT

Gram-negative bacteria are extraordinarily difficult to kill because their cytoplasmic membrane is surrounded by an outer membrane that blocks the entry of most antibiotics. The impenetrable nature of the outer membrane is due to the presence of a large, amphipathic glycolipid called lipopolysaccharide (LPS) in its outer leaflet1. Assembly of the outer membrane requires transport of LPS across a protein bridge that spans from the cytoplasmic membrane to the cell surface. Maintaining outer membrane integrity is essential for bacterial cell viability, and its disruption can increase susceptibility to other antibiotics2-6. Thus, inhibitors of the seven lipopolysaccharide transport (Lpt) proteins that form this transenvelope transporter have long been sought. A new class of antibiotics that targets the LPS transport machine in Acinetobacter was recently identified. Here, using structural, biochemical and genetic approaches, we show that these antibiotics trap a substrate-bound conformation of the LPS transporter that stalls this machine. The inhibitors accomplish this by recognizing a composite binding site made up of both the Lpt transporter and its LPS substrate. Collectively, our findings identify an unusual mechanism of lipid transport inhibition, reveal a druggable conformation of the Lpt transporter and provide the foundation for extending this class of antibiotics to other Gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents , Bacterial Outer Membrane Proteins , Lipopolysaccharides , Membrane Transport Proteins , Acinetobacter/chemistry , Acinetobacter/drug effects , Acinetobacter/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites/drug effects , Biological Transport/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microbial Viability , Protein Conformation/drug effects , Substrate Specificity
5.
bioRxiv ; 2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37662341

ABSTRACT

G protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue, and cellular level. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays, and structural studies, we develop maternally selective heavy chain-only antibody ("nanobody") antagonists against the angiotensin II type I receptor (AT1R) and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to AT1R with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.

6.
Nat Commun ; 14(1): 3439, 2023 06 10.
Article in English | MEDLINE | ID: mdl-37301887

ABSTRACT

The peptidoglycan (PG) cell wall protects bacteria against osmotic lysis and determines cell shape, making this structure a key antibiotic target. Peptidoglycan is a polymer of glycan chains connected by peptide crosslinks, and its synthesis requires precise spatiotemporal coordination between glycan polymerization and crosslinking. However, the molecular mechanism by which these reactions are initiated and coupled is unclear. Here we use single-molecule FRET and cryo-EM to show that an essential PG synthase (RodA-PBP2) responsible for bacterial elongation undergoes dynamic exchange between closed and open states. Structural opening couples the activation of polymerization and crosslinking and is essential in vivo. Given the high conservation of this family of synthases, the opening motion that we uncovered likely represents a conserved regulatory mechanism that controls the activation of PG synthesis during other cellular processes, including cell division.


Subject(s)
Bacterial Proteins , Peptidoglycan , Bacterial Proteins/chemistry , Penicillin-Binding Proteins/metabolism , Allosteric Regulation , Polysaccharides/analysis , Cell Wall/metabolism
7.
J Prosthet Dent ; 129(1): 2-6, 2023 Jan.
Article in English | MEDLINE | ID: mdl-34417017

ABSTRACT

A direct composite resin placed by using digitally planned prototyped 3D guides and retained with a digitally guided fiberglass micropin was used to restore an extensively damaged maxillary left central incisor.


Subject(s)
Dental Restoration, Permanent , Incisor , Composite Resins/therapeutic use
8.
Curr Res Struct Biol ; 3: 72-84, 2021.
Article in English | MEDLINE | ID: mdl-34235487

ABSTRACT

Epoxide hydrolases catalyze the conversion of epoxides to vicinal diols in a range of cellular processes such as signaling, detoxification, and virulence. These enzymes typically utilize a pair of tyrosine residues to orient the substrate epoxide ring in the active site and stabilize the hydrolysis intermediate. A new subclass of epoxide hydrolases that utilize a histidine in place of one of the tyrosines was established with the discovery of the CFTR Inhibitory Factor (Cif) from Pseudomonas aeruginosa. Although the presence of such Cif-like epoxide hydrolases was predicted in other opportunistic pathogens based on sequence analyses, only Cif and its homolog aCif from Acinetobacter nosocomialis have been characterized. Here we report the biochemical and structural characteristics of Cfl1 and Cfl2, two Cif-like epoxide hydrolases from Burkholderia cenocepacia. Cfl1 is able to hydrolyze xenobiotic as well as biological epoxides that might be encountered in the environment or during infection. In contrast, Cfl2 shows very low activity against a diverse set of epoxides. The crystal structures of the two proteins reveal quaternary structures that build on the well-known dimeric assembly of the α/ß hydrolase domain, but broaden our understanding of the structural diversity encoded in novel oligomer interfaces. Analysis of the interfaces reveals both similarities and key differences in sequence conservation between the two assemblies, and between the canonical dimer and the novel oligomer interfaces of each assembly. Finally, we discuss the effects of these higher-order assemblies on the intra-monomer flexibility of Cfl1 and Cfl2 and their possible roles in regulating enzymatic activity.

9.
Animal ; 15(7): 100257, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34087691

ABSTRACT

The role of beef in human diets has been questioned over the last few decades, due largely to its typically high mass-based carbon footprint. However, recent advancements in sustainability literature challenge this paradigm based on the new theory that climate impacts of food commodities should be measured relative to their overall nutritional value rather than their nominal mass. This shift has opened a new opportunity for the global beef industry, and especially for pasture-based systems that can avoid food-feed competition for land and other resources, as beef is a nutritionally dense food. Nonetheless, the sector's true capability to supply a wide range of nutrients for humans, consistently across multiple systems under multiple weather patterns, has not been well-documented. Using whole-system datasets from the North Wyke Farm Platform in the South West of England, we investigated the nutritional value of beef produced from the three most common pasture systems in temperate regions: permanent pasture (PP), grass and white clover (GWC) and a short-term monoculture grass ley (MG). Beef produced from these three pasture systems was analysed for key nutrients (fatty acids, minerals and vitamin E) over three production cycles (2015-2017) to determine potential differences between systems. Fatty acid, mineral and vitamin E profiles of the pasture and silage fed to each group were also assessed, with subtle differences between pastures reported. For beef, subtle differences were also observed between systems, with GWC having higher omega-6 polyunsaturated fatty acid (PUFA) concentrations than PP and MG. However, the overall nutritional quality of beef was found to be largely comparable across all systems, suggesting that temperate pasture-based beef can be classified as a single commodity in future sustainability assessments, regardless of specific sward types. A 100 g serving of temperate pasture-based beef was found to be a high source (>20% recommended daily intake: RDI) of protein, monounsaturated fatty acids, saturated fatty acids, vitamins - B2, B3, B12 and minerals - Fe, P, Zn; a good source (10-19% RDI) of vitamin - B6 and mineral - K; and a complementary source (5-9% RDI) of omega-3 PUFA, vitamin - B9 and minerals - Cu, Mg, Se. The nutritional value of a food item should be used in defining its environmental cost (e.g. carbon footprint) to make fair comparisons across different food groups (e.g. protein sources). Here, we showed that pasture-based beef had a nutrient indexed carbon footprint of between 0.19 and 0.23 Kg CO2-eq/1% RDI of key nutrients.


Subject(s)
Diet , Meat , Animal Feed , Animals , Cattle , Diet/veterinary , England , Fatty Acids, Omega-6 , Meat/analysis , Nutritive Value
10.
Nat Chem Biol ; 17(10): 1057-1064, 2021 10.
Article in English | MEDLINE | ID: mdl-34168368

ABSTRACT

The predominant approach for antibody generation remains animal immunization, which can yield exceptionally selective and potent antibody clones owing to the powerful evolutionary process of somatic hypermutation. However, animal immunization is inherently slow, not always accessible and poorly compatible with many antigens. Here, we describe 'autonomous hypermutation yeast surface display' (AHEAD), a synthetic recombinant antibody generation technology that imitates somatic hypermutation inside engineered yeast. By encoding antibody fragments on an error-prone orthogonal DNA replication system, surface-displayed antibody repertoires continuously mutate through simple cycles of yeast culturing and enrichment for antigen binding to produce high-affinity clones in as little as two weeks. We applied AHEAD to generate potent nanobodies against the SARS-CoV-2 S glycoprotein, a G-protein-coupled receptor and other targets, offering a template for streamlined antibody generation at large.


Subject(s)
Antibody Formation/immunology , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Antibodies/immunology , Antigens , COVID-19/immunology , Humans , Peptide Library , Recombinant Proteins/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Saccharomyces cerevisiae/metabolism , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Spike Glycoprotein, Coronavirus/immunology
11.
Immunity ; 54(4): 769-780.e6, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33823129

ABSTRACT

An effective vaccine for respiratory syncytial virus (RSV) is an unrealized public health goal. A single dose of the prefusion-stabilized fusion (F) glycoprotein subunit vaccine (DS-Cav1) substantially increases serum-neutralizing activity in healthy adults. We sought to determine whether DS-Cav1 vaccination induces a repertoire mirroring the pre-existing diversity from natural infection or whether antibody lineages targeting specific epitopes predominate. We evaluated RSV F-specific B cell responses before and after vaccination in six participants using complementary B cell sequencing methodologies and identified 555 clonal lineages. DS-Cav1-induced lineages recognized the prefusion conformation of F (pre-F) and were genetically diverse. Expressed antibodies recognized all six antigenic sites on the pre-F trimer. We identified 34 public clonotypes, and structural analysis of two antibodies from a predominant clonotype revealed a common mode of recognition. Thus, vaccination with DS-Cav1 generates a diverse polyclonal response targeting the antigenic sites on pre-F, supporting the development and advanced testing of pre-F-based vaccines against RSV.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/immunology , Cell Line , Cell Line, Tumor , Child , Child, Preschool , Cohort Studies , Epitopes/immunology , Female , HEK293 Cells , Humans , Infant , Infant, Newborn , Male , Middle Aged , Vaccination/methods , Viral Fusion Proteins/immunology , Young Adult
12.
J Med Chem ; 64(7): 3658-3676, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33729773

ABSTRACT

RV521 is an orally bioavailable inhibitor of respiratory syncytial virus (RSV) fusion that was identified after a lead optimization process based upon hits that originated from a physical property directed hit profiling exercise at Reviral. This exercise encompassed collaborations with a number of contract organizations with collaborative medicinal chemistry and virology during the optimization phase in addition to those utilized as the compound proceeded through preclinical and clinical evaluation. RV521 exhibited a mean IC50 of 1.2 nM against a panel of RSV A and B laboratory strains and clinical isolates with antiviral efficacy in the Balb/C mouse model of RSV infection. Oral bioavailability in preclinical species ranged from 42 to >100% with evidence of highly efficient penetration into lung tissue. In healthy adult human volunteers experimentally infected with RSV, a potent antiviral effect was observed with a significant reduction in viral load and symptoms compared to placebo.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Virus Internalization/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacokinetics , Biological Availability , Cell Line, Tumor , Clinical Trials as Topic , Drug Discovery , Humans , Microbial Sensitivity Tests , Protein Binding , Viral Fusion Proteins/metabolism
13.
bioRxiv ; 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33200136

ABSTRACT

The predominant approach for antibody generation remains animal immunization, which can yield exceptionally selective and potent antibody clones owing to the powerful evolutionary process of somatic hypermutation. However, animal immunization is inherently slow, has poor compatibility with certain antigens ( e . g ., integral membrane proteins), and suffers from self-tolerance and immunodominance, which limit the functional spectrum of antibodies that can be obtained. Here, we describe A utonomous H ypermutation y E ast surf A ce D isplay (AHEAD), a synthetic recombinant antibody generation technology that imitates somatic hypermutation inside engineered yeast. In AHEAD, antibody fragments are encoded on an error-prone orthogonal DNA replication system, resulting in Saccharomyces cerevisiae populations that continuously mutate surface-displayed antibody repertoires. Simple cycles of yeast culturing and enrichment for antigen binding drive the evolution of high-affinity antibody clones in a readily parallelizable process that takes as little as 2 weeks. We applied AHEAD to generate nanobodies against the SARS-CoV-2 S glycoprotein, a GPCR, and other targets. The SARS-CoV-2 nanobodies, concurrently evolved from an open-source naïve nanobody library in 8 independent experiments, reached subnanomolar affinities through the sequential fixation of multiple mutations over 3-8 AHEAD cycles that saw ∼580-fold and ∼925-fold improvements in binding affinities and pseudovirus neutralization potencies, respectively. These experiments highlight the defining speed, parallelizability, and effectiveness of AHEAD and provide a template for streamlined antibody generation at large with salient utility in rapid response to current and future viral outbreaks.

14.
Preprint in English | bioRxiv | ID: ppbiorxiv-378778

ABSTRACT

The predominant approach for antibody generation remains animal immunization, which can yield exceptionally selective and potent antibody clones owing to the powerful evolutionary process of somatic hypermutation. However, animal immunization is inherently slow, has poor compatibility with certain antigens (e.g., integral membrane proteins), and suffers from self-tolerance and immunodominance, which limit the functional spectrum of antibodies that can be obtained. Here, we describe Autonomous Hypermutation yEast surfAce Display (AHEAD), a synthetic recombinant antibody generation technology that imitates somatic hypermutation inside engineered yeast. In AHEAD, antibody fragments are encoded on an error-prone orthogonal DNA replication system, resulting in Saccharomyces cerevisiae populations that continuously mutate surface-displayed antibody repertoires. Simple cycles of yeast culturing and enrichment for antigen binding drive the evolution of high-affinity antibody clones in a readily parallelizable process that takes as little as 2 weeks. We applied AHEAD to generate nanobodies against the SARS-CoV-2 S glycoprotein, a GPCR, and other targets. The SARS-CoV-2 nanobodies, concurrently evolved from an open-source naive nanobody library in 8 independent experiments, reached subnanomolar affinities through the sequential fixation of multiple mutations over 3-8 AHEAD cycles that saw [~]580-fold and [~]925-fold improvements in binding affinities and pseudovirus neutralization potencies, respectively. These experiments highlight the defining speed, parallelizability, and effectiveness of AHEAD and provide a template for streamlined antibody generation at large with salient utility in rapid response to current and future viral outbreaks.

15.
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
16.
Nat Microbiol ; 5(6): 813-820, 2020 06.
Article in English | MEDLINE | ID: mdl-32152588

ABSTRACT

The shape, elongation, division and sporulation (SEDS) proteins are a highly conserved family of transmembrane glycosyltransferases that work in concert with class B penicillin-binding proteins (bPBPs) to build the bacterial peptidoglycan cell wall1-6. How these proteins coordinate polymerization of new glycan strands with their crosslinking to the existing peptidoglycan meshwork is unclear. Here, we report the crystal structure of the prototypical SEDS protein RodA from Thermus thermophilus in complex with its cognate bPBP at 3.3 Å resolution. The structure reveals a 1:1 stoichiometric complex with two extensive interaction interfaces between the proteins: one in the membrane plane and the other at the extracytoplasmic surface. When in complex with a bPBP, RodA shows an approximately 10 Å shift of transmembrane helix 7 that exposes a large membrane-accessible cavity. Negative-stain electron microscopy reveals that the complex can adopt a variety of different conformations. These data define the bPBP pedestal domain as the key allosteric activator of RodA both in vitro and in vivo, explaining how a SEDS-bPBP complex can coordinate its dual enzymatic activities of peptidoglycan polymerization and crosslinking to build the cell wall.


Subject(s)
Models, Molecular , Multiprotein Complexes/chemistry , Penicillin-Binding Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Protein Multimerization , Binding Sites , Cell Wall/metabolism , Molecular Structure , Multiprotein Complexes/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship
17.
Cell ; 179(1): 193-204.e14, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31495574

ABSTRACT

Numerous interventions are in clinical development for respiratory syncytial virus (RSV) infection, including small molecules that target viral transcription and replication. These processes are catalyzed by a complex comprising the RNA-dependent RNA polymerase (L) and the tetrameric phosphoprotein (P). RSV P recruits multiple proteins to the polymerase complex and, with the exception of its oligomerization domain, is thought to be intrinsically disordered. Despite their critical roles in RSV transcription and replication, structures of L and P have remained elusive. Here, we describe the 3.2-Å cryo-EM structure of RSV L bound to tetrameric P. The structure reveals a striking tentacular arrangement of P, with each of the four monomers adopting a distinct conformation. The structure also rationalizes inhibitor escape mutants and mutations observed in live-attenuated vaccine candidates. These results provide a framework for determining the molecular underpinnings of RSV replication and transcription and should facilitate the design of effective RSV inhibitors.


Subject(s)
Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/ultrastructure , Acetates/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Catalytic Domain , Cryoelectron Microscopy , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Quinolines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Vaccines/chemistry , Sf9 Cells , Spodoptera , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
18.
Nat Commun ; 10(1): 2105, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068578

ABSTRACT

The respiratory syncytial virus (RSV) F glycoprotein is a class I fusion protein that mediates viral entry and is a major target of neutralizing antibodies. Structures of prefusion forms of RSV F, as well as other class I fusion proteins, have revealed compact trimeric arrangements, yet whether these trimeric forms can transiently open remains unknown. Here, we perform structural and biochemical studies on a recently isolated antibody, CR9501, and demonstrate that it enhances the opening of prefusion-stabilized RSV F trimers. The 3.3 Å crystal structure of monomeric RSV F bound to CR9501, combined with analysis of over 25 previously determined RSV F structures, reveals a breathing motion of the prefusion conformation. We also demonstrate that full-length RSV F trimers transiently open and dissociate on the cell surface. Collectively, these findings have implications for the function of class I fusion proteins, as well as antibody prophylaxis and vaccine development for RSV.


Subject(s)
Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Respiratory Syncytial Virus, Human/physiology , Viral Fusion Proteins/metabolism , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , B-Lymphocytes/virology , Chlorocebus aethiops , Computer Simulation , Crystallography, X-Ray , Drug Development , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Protein Multimerization/physiology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/isolation & purification , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology
19.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 2): 123-131, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30713164

ABSTRACT

Advances in X-ray crystallography have streamlined the process of determining high-resolution three-dimensional macromolecular structures. However, a rate-limiting step in this process continues to be the generation of crystals that are of sufficient size and quality for subsequent diffraction experiments. Here, iterative screen optimization (ISO), a highly automated process in which the precipitant concentrations of each condition in a crystallization screen are modified based on the results of a prior crystallization experiment, is described. After designing a novel high-throughput crystallization screen to take full advantage of this method, the value of ISO is demonstrated by using it to successfully crystallize a panel of six diverse proteins. The results suggest that ISO is an effective method to obtain macromolecular crystals, particularly for proteins that crystallize under a narrow range of precipitant concentrations.


Subject(s)
Crystallization/methods , Crystallization/standards , High-Throughput Screening Assays/standards , Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation
20.
Annu Rev Med ; 70: 91-104, 2019 01 27.
Article in English | MEDLINE | ID: mdl-30691364

ABSTRACT

Enabled by new approaches for rapid identification and selection of human monoclonal antibodies, atomic-level structural information for viral surface proteins, and capacity for precision engineering of protein immunogens and self-assembling nanoparticles, a new era of antigen design and display options has evolved. While HIV-1 vaccine development has been a driving force behind these technologies and concepts, clinical proof-of-concept for structure-based vaccine design may first be achieved for respiratory syncytial virus (RSV), where conformation-dependent access to neutralization-sensitive epitopes on the fusion glycoprotein determines the capacity to induce potent neutralizing activity. Success with RSV has motivated structure-based stabilization of other class I viral fusion proteins for use as immunogens and demonstrated the importance of structural information for developing vaccines against other viral pathogens, particularly difficult targets that have resisted prior vaccine development efforts. Solving viral surface protein structures also supports rapid vaccine antigen design and application of platform manufacturing approaches for emerging pathogens.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Respiratory Syncytial Viruses/immunology , Vaccines, Synthetic/pharmacology , Viral Fusion Proteins/immunology , Viral Vaccines/pharmacology , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Humans , Sensitivity and Specificity , Structure-Activity Relationship , Vaccines, Synthetic/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...