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










Publication year range
1.
Protein Sci ; 33(7): e5081, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38924648

ABSTRACT

It has been shown previously that a set of three modifications-termed S1, Crystal Kappa, and elbow-act synergistically to improve the crystallizability of an antigen-binding fragment (Fab) framework. Here, we prepared a phage-displayed library and performed crystallization screenings to identify additional substitutions-located near the heavy-chain elbow region-which cooperate with the S1, Crystal Kappa, and elbow modifications to increase expression and improve crystallizability of the Fab framework even further. One substitution (K141Q) supports the signature Crystal Kappa-mediated Fab:Fab crystal lattice packing interaction. Another substitution (E172G) improves the compatibility of the elbow modification with the Fab framework by alleviating some of the strain incurred by the shortened and bulkier elbow linker region. A third substitution (F170W) generates a split-Fab conformation, resulting in a powerful crystal lattice packing interaction comprising the biological interaction interface between the variable heavy and light chain domains. In sum, we have used K141Q, E172G, and F170W substitutions-which complement the S1, Crystal Kappa, and elbow modifications-to generate a set of highly crystallizable Fab frameworks that can be used as chaperones to enable facile elucidation of Fab:antigen complex structures by x-ray crystallography.


Subject(s)
Immunoglobulin Fab Fragments , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Crystallography, X-Ray , Crystallization , Models, Molecular , Protein Conformation , Humans , Amino Acid Substitution
2.
ACS Chem Biol ; 17(7): 1978-1988, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35731947

ABSTRACT

The spread of COVID-19 has been exacerbated by the emergence of variants of concern (VoC). Many VoC contain mutations in the spike protein (S-protein) and are implicated in infection and response to therapeutics. Bivalent neutralizing antibodies (nAbs) targeting the S-protein receptor-binding domain (RBD) are promising therapeutics for COVID-19, but they are limited by low potency and vulnerability to RBD mutations in VoC. To address these issues, we used naïve phage-displayed peptide libraries to isolate and optimize 16-residue peptides that bind to the RBD or the N-terminal domain (NTD) of the S-protein. We fused these peptides to the N-terminus of a moderate-affinity nAb to generate tetravalent peptide-IgG fusions, and we showed that both classes of peptides were able to improve affinities for the S-protein trimer by >100-fold (apparent KD < 1 pM). Critically, cell-based infection assays with a panel of six SARS-CoV-2 variants demonstrated that an RBD-binding peptide was able to enhance the neutralization potency of a high-affinity nAb >100-fold. Moreover, this peptide-IgG was able to neutralize variants that were resistant to the same nAb in the bivalent IgG format, including the dominant B.1.1.529 (Omicron) variant that is resistant to most clinically approved therapeutic nAbs. To show that this approach is general, we fused the same peptide to a clinically approved nAb drug and showed that it enabled the neutralization of a resistant variant. Taken together, these results establish minimal peptide fusions as a modular means to greatly enhance affinities, potencies, and breadth of coverage of nAbs as therapeutics for SARS-CoV-2.


Subject(s)
Bacteriophages , COVID-19 Drug Treatment , Antibodies, Neutralizing , Antibodies, Viral/genetics , Bacteriophages/genetics , Humans , Immunoglobulin G/genetics , Neutralization Tests , Peptide Library , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
3.
J Mol Biol ; 433(19): 167177, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34329642

ABSTRACT

Neutralizing antibodies (nAbs) hold promise as therapeutics against COVID-19. Here, we describe protein engineering and modular design principles that have led to the development of synthetic bivalent and tetravalent nAbs against SARS-CoV-2. The best nAb targets the host receptor binding site of the viral S-protein and tetravalent versions block entry with a potency exceeding bivalent nAbs by an order of magnitude. Structural studies show that both the bivalent and tetravalent nAbs can make multivalent interactions with a single S-protein trimer, consistent with the avidity and potency of these molecules. Significantly, we show that the tetravalent nAbs show increased tolerance to potential virus escape mutants and an emerging variant of concern. Bivalent and tetravalent nAbs can be produced at large-scale and are as stable and specific as approved antibody drugs. Our results provide a general framework for enhancing antiviral therapies against COVID-19 and related viral threats, and our strategy can be applied to virtually any antibody drug.


Subject(s)
Antibodies, Neutralizing/immunology , COVID-19 Drug Treatment , COVID-19/immunology , Mutation , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antiviral Agents/therapeutic use , Binding Sites , Chlorocebus aethiops , HEK293 Cells , Humans , Immunoglobulin G , Models, Molecular , Protein Binding , Protein Engineering , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells
4.
bioRxiv ; 2020 Dec 21.
Article in English | MEDLINE | ID: mdl-33398270

ABSTRACT

Neutralizing antibodies (nAbs) hold promise as effective therapeutics against COVID-19. Here, we describe protein engineering and modular design principles that have led to the development of synthetic bivalent and tetravalent nAbs against SARS-CoV-2. The best nAb targets the host receptor binding site of the viral S-protein and its tetravalent versions can block entry with a potency that exceeds the bivalent nAbs by an order of magnitude. Structural studies show that both the bivalent and tetravalent nAbs can make multivalent interactions with a single S-protein trimer, observations consistent with the avidity and potency of these molecules. Significantly, we show that the tetravalent nAbs show much increased tolerance to potential virus escape mutants. Bivalent and tetravalent nAbs can be produced at large-scale and are as stable and specific as approved antibody drugs. Our results provide a general framework for developing potent antiviral therapies against COVID-19 and related viral threats, and our strategy can be readily applied to any antibody drug currently in development.

5.
ACS Infect Dis ; 5(8): 1385-1396, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31120240

ABSTRACT

Marburg virus causes sporadic outbreaks of severe hemorrhagic fever with high case fatality rates. Approved, effective, and safe therapeutic or prophylactic countermeasures are lacking. To address this, we used phage display to engineer a synthetic antibody, sFab H3, which binds the Marburg virus VP35 protein (mVP35). mVP35 is a critical cofactor of the viral replication complex and a viral immune antagonist. sFab H3 displayed high specificity for mVP35 and not for the closely related Ebola virus VP35. sFab H3 inhibited viral-RNA synthesis in a minigenome assay, suggesting its potential use as an antiviral. We characterized sFab H3 by a combination of biophysical and biochemical methods, and a crystal structure of the complex solved to 1.7 Å resolution defined the molecular interface between the sFab H3 and mVP35 interferon inhibitory domain. Our study identifies mVP35 as a therapeutic target using an approach that provides a framework for generating engineered Fabs targeting other viral proteins.


Subject(s)
Antibodies, Viral/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Marburgvirus/drug effects , RNA, Viral/biosynthesis , Viral Regulatory and Accessory Proteins/immunology , Binding Sites, Antibody , Cell Surface Display Techniques , Crystallization , Crystallography, X-Ray , Humans , Marburgvirus/genetics , Marburgvirus/physiology , Models, Molecular , Virus Replication/drug effects
6.
Structure ; 26(9): 1226-1236.e3, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30033217

ABSTRACT

Skp1-Cul1-F-box (SCF) E3 ligases constitute the largest and best-characterized family of the multisubunit E3 ligases with important cellular functions and numerous disease links. The specificity of an SCF E3 ligase is established by one of the 69 human F-box proteins that are recruited to Cul1 through the Skp1 adaptor. We previously reported generation of ubiquitin variants (UbVs) targeting Fbw7 and Fbw11, which inhibit ligase activity by binding at the F-box-Skp1 interface to competitively displace Cul1. In the present study, we employed an optimized engineering strategy to generate specific binding UbVs against 17 additional Skp1-F-box complexes. We validated our design strategy and uncovered the structural basis of binding specificity by crystallographic analyses of representative UbVs bound to Skp1-Fbl10 and Skp1-Fbl11. Our study highlights the power of combining phage display with structure-based design to develop UbVs targeting specific protein surfaces.


Subject(s)
SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitins/chemistry , Ubiquitins/pharmacology , Binding Sites , Cell Surface Display Techniques , Crystallography, X-Ray , Cullin Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Female , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Protein Binding , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
7.
MAbs ; 7(4): 681-92, 2015.
Article in English | MEDLINE | ID: mdl-25879139

ABSTRACT

Polyomavirus JC (JCV) is the causative agent of progressive multifocal leukoencephalopathy (PML), a rare and frequently fatal brain disease that afflicts a small fraction of the immune-compromised population, including those affected by AIDS and transplantation recipients on immunosuppressive drug therapy. Currently there is no specific therapy for PML. The major capsid viral protein 1 (VP1) involved in binding to sialic acid cell receptors is believed to be a key player in pathogenesis. PML-specific mutations in JCV VP1 sequences present at the binding pocket of sialic acid cell receptors, such as L55F and S269F, abolish sialic acid recognition and might favor PML onset. Early diagnosis of these PML-specific mutations may help identify patients at high risk of PML, thus reducing the risks associated with immunosuppressive therapy. As a first step in the development of such early diagnostic tools, we report identification and characterization of affinity reagents that specifically recognize PML-specific mutations in VP1 variants using phage display technology. We first identified 2 peptides targeting wild type VP1 with moderate specificity. Fine-tuning via selection of biased libraries designed based on 2 parental peptides yielded peptides with different, yet still moderate, bindinspecificities. In contrast, we had great success in identifying synthetic antibodies that recognize one of the PML-specific mutations (L55F) with high specificity from the phage-displayed libraries. These peptides and synthetic antibodies represent potential candidates for developing tailored immune-based assays for PML risk stratification in addition to complementing affinity reagents currently available for the study of PML and JCV.


Subject(s)
Capsid Proteins/immunology , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/immunology , Mutation , Peptides/immunology , Single-Chain Antibodies/immunology , Capsid Proteins/genetics , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Peptides/genetics , Single-Chain Antibodies/genetics
8.
Proc Natl Acad Sci U S A ; 110(44): 17832-7, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127596

ABSTRACT

Somatic oncogenic mutations in the receptor tyrosine kinase KIT function as major drivers of gastrointestinal stromal tumors and a subset of acute myeloid leukemia, melanoma, and other cancers. Although treatment of these cancers with tyrosine kinase inhibitors shows dramatic responses and durable disease control, drug resistance followed by clinical progression of disease eventually occurs in virtually all patients. In this report, we describe inhibitory KIT antibodies that bind to the membrane-proximal Ig-like D4 of KIT with significant overlap with an epitope in D4 that mediates homotypic interactions essential for KIT activation. Crystal structures of the anti-KIT antibody in complex with KIT D4 and D5 allowed design of affinity-matured libraries that were used to isolate variants with increased affinity and efficacy. Isolated antibodies showed KIT inhibition together with suppression of cell proliferation driven by ligand-stimulated WT or constitutively activated oncogenic KIT mutant. These antibodies represent a unique therapeutic approach and a step toward the development of "naked" or toxin-conjugated KIT antibodies for the treatment of KIT-driven cancers.


Subject(s)
Antibodies, Monoclonal/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Neoplasms/drug therapy , Protein Conformation , Proto-Oncogene Proteins c-kit/chemistry , Animals , Antibodies, Monoclonal/pharmacology , Baculoviridae , Cell Surface Display Techniques , Crystallization , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Immunoprecipitation , Mutation/genetics , Neoplasms/immunology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Sf9 Cells , Spodoptera
9.
Kidney Int ; 84(3): 468-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23636173

ABSTRACT

Although the role of the erythropoietin (EPO) receptor (EpoR) in erythropoiesis has been known for decades, its role in nonhematopoietic tissues is still not well defined. Klotho has been shown and EPo has been suggested to protect against acute ischemia-reperfusion injury in the kidney. Here we found in rat kidney and in a rat renal tubular epithelial cell line (NRK cells) EpoR transcript and antigen, and EpoR activity signified as EPo-induced phosphorylation of Jak2, ErK, Akt, and Stat5 indicating the presence of functional EpoR. Transgenic overexpression of Klotho or addition of exogenous recombinant Klotho increased kidney EpoR protein and transcript. In NRK cells, Klotho increased EpoR protein, enhanced EPo-triggered phosphorylation of Jak2 and Stat5, the nuclear translocation of phospho-Stat5, and protected NRK cells from hydrogen peroxide cytotoxicity. Knockdown of endogenous EpoR rendered NRK cells more vulnerable, and overexpression of EpoR more resistant to peroxide-induced cytotoxicity, indicating that EpoR mitigates oxidative damage. Knockdown of EpoR by siRNA abolished Epo-induced Jak2, and Stat5 phosphorylation, and blunted the protective effect of Klotho against peroxide-induced cytotoxicity. Thus in the kidney, EpoR and its activity are downstream effectors of Klotho enabling it to function as a cytoprotective protein against oxidative injury.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Cytoprotection/physiology , Glucuronidase/physiology , Receptors, Erythropoietin/physiology , Animals , Cell Line , Disease Models, Animal , Glucuronidase/deficiency , Glucuronidase/genetics , Humans , Hydrogen Peroxide/adverse effects , In Vitro Techniques , Janus Kinase 2/metabolism , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Klotho Proteins , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , STAT5 Transcription Factor/metabolism
10.
Mol Syst Biol ; 9: 652, 2013.
Article in English | MEDLINE | ID: mdl-23549480

ABSTRACT

Src homology 3 (SH3) domains bind peptides to mediate protein-protein interactions that assemble and regulate dynamic biological processes. We surveyed the repertoire of SH3 binding specificity using peptide phage display in a metazoan, the worm Caenorhabditis elegans, and discovered that it structurally mirrors that of the budding yeast Saccharomyces cerevisiae. We then mapped the worm SH3 interactome using stringent yeast two-hybrid and compared it with the equivalent map for yeast. We found that the worm SH3 interactome resembles the analogous yeast network because it is significantly enriched for proteins with roles in endocytosis. Nevertheless, orthologous SH3 domain-mediated interactions are highly rewired. Our results suggest a model of network evolution where general function of the SH3 domain network is conserved over its specific form.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , src Homology Domains/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Conserved Sequence , Endocytosis/genetics , Evolution, Molecular , Molecular Sequence Data , Protein Interaction Mapping , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structural Homology, Protein , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL