Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 182(4): 1027-1043.e17, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32822567

ABSTRACT

Cell-surface protein-protein interactions (PPIs) mediate cell-cell communication, recognition, and responses. We executed an interactome screen of 564 human cell-surface and secreted proteins, most of which are immunoglobulin superfamily (IgSF) proteins, using a high-throughput, automated ELISA-based screening platform employing a pooled-protein strategy to test all 318,096 PPI combinations. Screen results, augmented by phylogenetic homology analysis, revealed ∼380 previously unreported PPIs. We validated a subset using surface plasmon resonance and cell binding assays. Observed PPIs reveal a large and complex network of interactions both within and across biological systems. We identified new PPIs for receptors with well-characterized ligands and binding partners for "orphan" receptors. New PPIs include proteins expressed on multiple cell types and involved in diverse processes including immune and nervous system development and function, differentiation/proliferation, metabolism, vascularization, and reproduction. These PPIs provide a resource for further biological investigation into their functional relevance and may offer new therapeutic drug targets.


Subject(s)
Ligands , Protein Interaction Maps/physiology , Receptors, Cell Surface/metabolism , DCC Receptor/chemistry , DCC Receptor/metabolism , Humans , Phylogeny , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/classification , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Signaling Lymphocytic Activation Molecule Family/chemistry , Signaling Lymphocytic Activation Molecule Family/metabolism , Surface Plasmon Resonance
2.
Nature ; 563(7731): 402-406, 2018 11.
Article in English | MEDLINE | ID: mdl-30356215

ABSTRACT

While research into the biology of animal behaviour has primarily focused on the central nervous system, cues from peripheral tissues and the environment have been implicated in brain development and function1. There is emerging evidence that bidirectional communication between the gut and the brain affects behaviours including anxiety, cognition, nociception and social interaction1-9. Coordinated locomotor behaviour is critical for the survival and propagation of animals, and is regulated by internal and external sensory inputs10,11. However, little is known about how the gut microbiome influences host locomotion, or the molecular and cellular mechanisms involved. Here we report that germ-free status or antibiotic treatment results in hyperactive locomotor behaviour in the fruit fly Drosophila melanogaster. Increased walking speed and daily activity in the absence of a gut microbiome are rescued by mono-colonization with specific bacteria, including the fly commensal Lactobacillus brevis. The bacterial enzyme xylose isomerase from L. brevis recapitulates the locomotor effects of microbial colonization by modulating sugar metabolism in flies. Notably, thermogenetic activation of octopaminergic neurons or exogenous administration of octopamine, the invertebrate counterpart of noradrenaline, abrogates the effects of xylose isomerase on Drosophila locomotion. These findings reveal a previously unappreciated role for the gut microbiome in modulating locomotion, and identify octopaminergic neurons as mediators of peripheral microbial cues that regulate motor behaviour in animals.


Subject(s)
Carbohydrate Metabolism , Drosophila melanogaster/microbiology , Drosophila melanogaster/physiology , Gastrointestinal Microbiome/physiology , Levilactobacillus brevis/enzymology , Levilactobacillus brevis/metabolism , Locomotion/physiology , Aldose-Ketose Isomerases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Carbohydrate Metabolism/drug effects , Drosophila melanogaster/drug effects , Drosophila melanogaster/metabolism , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Levilactobacillus brevis/isolation & purification , Locomotion/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Neural Pathways , Neurons/drug effects , Neurons/metabolism , Octopamine/metabolism , Octopamine/pharmacology , Symbiosis
3.
Cytotherapy ; 25(2): 120-124, 2023 02.
Article in English | MEDLINE | ID: mdl-36274007

ABSTRACT

BACKGROUND AIMS: We evaluated a commercially available instrument, OCTiCell (chromologic.com/octicell), for monitoring cell growth in suspended agitated bioreactors based on optical coherence tomography. OCTiCell is an in-line, completely non-invasive instrument that can operate on any suspended-cell bioreactor with a window or transparent wall. In traditional optical coherence tomography, the imaging beam is rastered over the sample to form a three-dimensional image. OCTiCell, instead uses a fixed imaging beam and takes advantage of the motion of the media to move the cells across the interrogating optical beam. RESULTS: We found strong correlations between the non-invasive, non-contact, reagent-free OCTiCell measurements of cell concentration and viability and those obtained from the automated cell counter, and the XTT viability assay, which is a colorimetric assay for quantifying metabolic activity. CONCLUSIONS: This novel cell monitoring method can adapt to different bioreactor form factors and could reduce the labor cost and contamination risks associated with cell growth monitoring.


Subject(s)
Bioreactors , Tomography, Optical Coherence , Tomography, Optical Coherence/methods
4.
Proc Natl Acad Sci U S A ; 117(31): 18719-18728, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690692

ABSTRACT

CD4-based decoy approaches against HIV-1 are attractive options for long-term viral control, but initial designs, including soluble CD4 (sCD4) and CD4-Ig, were ineffective. To evaluate a therapeutic that more accurately mimics HIV-1 target cells compared with monomeric sCD4 and dimeric CD4-Ig, we generated virus-like nanoparticles that present clusters of membrane-associated CD4 (CD4-VLPs) to permit high-avidity binding of trimeric HIV-1 envelope spikes. In neutralization assays, CD4-VLPs were >12,000-fold more potent than sCD4 and CD4-Ig and >100-fold more potent than the broadly neutralizing antibody (bNAb) 3BNC117, with >12,000-fold improvements against strains poorly neutralized by 3BNC117. CD4-VLPs also neutralized patient-derived viral isolates that were resistant to 3BNC117 and other bNAbs. Intraperitoneal injections of CD4-CCR5-VLP produced only subneutralizing plasma concentrations in HIV-1-infected humanized mice but elicited CD4-binding site mutations that reduced viral fitness. All mutant viruses showed reduced sensitivity to sCD4 and CD4-Ig but remained sensitive to neutralization by CD4-VLPs in vitro. In vitro evolution studies demonstrated that CD4-VLPs effectively controlled HIV-1 replication at neutralizing concentrations, and viral escape was not observed. Moreover, CD4-VLPs potently neutralized viral swarms that were completely resistant to CD4-Ig, suggesting that escape pathways that confer resistance against conventional CD4-based inhibitors are ineffective against CD4-VLPs. These findings suggest that therapeutics that mimic HIV-1 target cells could prevent viral escape by exposing a universal vulnerability of HIV-1: the requirement to bind CD4 on a target cell. We propose that therapeutic and delivery strategies that ensure durable bioavailability need to be developed to translate this concept into a clinically feasible functional cure therapy.


Subject(s)
CD4 Antigens , HIV-1 , Nanoparticles , Virion , Anti-HIV Agents , CD4 Antigens/chemistry , CD4 Antigens/metabolism , Cell Line , HIV-1/chemistry , HIV-1/genetics , HIV-1/metabolism , Humans , Molecular Mimicry , Nanomedicine/methods , Nanoparticles/chemistry , Nanoparticles/metabolism , Virion/chemistry , Virion/metabolism
5.
J Struct Biol ; 194(1): 78-89, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850169

ABSTRACT

The Fc region of Immunoglobulin G (IgG) initiates inflammatory responses such as antibody-dependent cell-mediated cytotoxicity (ADCC) through binding to activating Fc receptors (FcγRI, FcγRIIa, FcγRIIIa). These receptors are expressed on the surface of immune cells including macrophages, dendritic cells, and natural killer cells. An inhibitory receptor, FcγRIIb, is expressed on macrophages and other myeloid leukocytes simultaneously with the activating receptor FcγRIIa, thereby setting a threshold for cell activation. The affinity of IgG Fc for binding activating Fc receptors depends on IgG subclass and the composition of N-linked glycans attached to a conserved asparagine in the Fc CH2 domain. For example, Fc regions with afucosylated glycans bind more tightly to FcγRIIIa than fucosylated Fc, and afucosylated Fcs exhibit enhanced ADCC activity in vivo and in vitro. Enhanced pro-inflammatory responses have also been seen for Fc regions with amino acid substitutions. GASDALIE Fc is an Fc mutant (G236A/S239D/A330L/I332E) that exhibits a higher affinity for FcγRIIIa and increased effector functions in vivo compared to wild-type Fc. To explore its altered functions, we compared the affinities of GASDALIE and wild-type Fc for activating and inhibitory FcγRs. We also determined the crystal structure of GASDALIE Fc alone and bound to FcγRIIIa. The overall structure of GASDALIE Fc alone was similar to wild-type Fc structures, however, increased electrostatic interactions in the GASDALIE Fc:FcγRIIIa interface compared with other Fc:FcγR structures suggest a mechanism for the increased affinity of GASDALIE Fc for FcγRIIIa.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Models, Molecular , Protein Domains , Receptors, IgG/chemistry , Amino Acid Sequence , Antibody-Dependent Cell Cytotoxicity , Binding Sites/genetics , Crystallography, X-Ray , Fucose/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Mutation , Protein Binding , Receptors, IgG/genetics , Receptors, IgG/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
6.
ACS Biomater Sci Eng ; 10(4): 2188-2199, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38479351

ABSTRACT

Artificial protein hydrogels are an emerging class of biomaterials with numerous prospective applications in tissue engineering and regenerative medicine. These materials are likely to be immunogenic due to their frequent incorporation of novel amino acid sequence domains, which often serve a functional role within the material itself. We engineered injectable "self" and "nonself" artificial protein hydrogels, which were predicted to have divergent immune outcomes in vivo on the basis of their primary amino acid sequence. Following implantation in mouse, the nonself gels raised significantly higher antigel antibody titers than the corresponding self gels. Prophylactic administration of a fusion antibody targeting the nonself hydrogel epitopes to DEC-205, an endocytic receptor involved in Treg induction, fully suppressed the elevated antibody titer against the nonself gels. These results suggest that the clinical immune response to artificial protein biomaterials, including those that contain highly antigenic sequence domains, can be tuned through the induction of antigen-specific tolerance.


Subject(s)
Biocompatible Materials , Hydrogels , Animals , Mice , Hydrogels/pharmacology , Hydrogels/chemistry , Tissue Engineering/methods
7.
J Comput Aided Mol Des ; 27(4): 337-46, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23612915

ABSTRACT

Hepatitis C is a global health problem. While many drug companies have active R&D efforts to develop new drugs for treating Hepatitis C virus (HCV), most target the viral enzymes. The HCV glycoprotein E2 has been shown to play an essential role in hepatocyte invasion by binding to CD81 and other cell surface receptors. This paper describes the use of AutoDock to identify ligand binding sites on the large extracellular loop of the open conformation of CD81 and to perform virtual screening runs to identify sets of small molecule ligands predicted to bind to two of these sites. The best sites selected by AutoLigand were located in regions identified by mutational studies to be the site of E2 binding. Thirty-six ligands predicted by AutoDock to bind to these sites were subsequently tested experimentally to determine if they bound to CD81-LEL. Binding assays conducted using surface Plasmon resonance revealed that 26 out of 36 (72 %) of the ligands bound in vitro to the recombinant CD81-LEL protein. Competition experiments performed using dual polarization interferometry showed that one of the ligands predicted to bind to the large cleft between the C and D helices was also effective in blocking E2 binding to CD81-LEL.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hepacivirus/physiology , Host-Pathogen Interactions/drug effects , Tetraspanin 28/metabolism , Viral Envelope Proteins/metabolism , Binding Sites , Drug Design , Hepatitis C/drug therapy , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Ligands , Models, Molecular , Protein Binding/drug effects , Tetraspanin 28/chemistry
8.
Sci Adv ; 9(16): eadg6618, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37075114

ABSTRACT

The blood-brain barrier (BBB) presents a major challenge for delivering large molecules to study and treat the central nervous system. This is due in part to the scarcity of targets known to mediate BBB crossing. To identify novel targets, we leverage a panel of adeno-associated viruses (AAVs) previously identified through mechanism-agnostic directed evolution for improved BBB transcytosis. Screening potential cognate receptors for enhanced BBB crossing, we identify two targets: murine-restricted LY6C1 and widely conserved carbonic anhydrase IV (CA-IV). We apply AlphaFold-based in silico methods to generate capsid-receptor binding models to predict the affinity of AAVs for these identified receptors. Demonstrating how these tools can unlock target-focused engineering strategies, we create an enhanced LY6C1-binding vector, AAV-PHP.eC, that, unlike our prior PHP.eB, also works in Ly6a-deficient mouse strains such as BALB/cJ. Combined with structural insights from computational modeling, the identification of primate-conserved CA-IV enables the design of more specific and potent human brain-penetrant chemicals and biologicals, including gene delivery vectors.


Subject(s)
Blood-Brain Barrier , Carbonic Anhydrase IV , Mice , Humans , Animals , Blood-Brain Barrier/metabolism , Carbonic Anhydrase IV/genetics , Carbonic Anhydrase IV/metabolism , Brain/metabolism , Gene Transfer Techniques , Primates/genetics , Dependovirus/genetics , Dependovirus/metabolism
9.
NPJ Vaccines ; 6(1): 126, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34697307

ABSTRACT

HIV-1 vaccine design aims to develop an immunogen that elicits broadly neutralizing antibodies against a desired epitope, while eliminating responses to off-target regions of HIV-1 Env. We report characterization of Ab1245, an off-target antibody against the Env gp120-gp41 interface, from V3-glycan patch immunogen-primed and boosted macaques. A 3.7 Å cryo-EM structure of an Ab1245-Env complex reveals one Ab1245 Fab binding asymmetrically to Env trimer at the gp120-gp41 interface using its long CDRH3 to mimic regions of gp41. The mimicry includes positioning of a CDRH3 methionine into the gp41 tryptophan clasp, resulting in displacement of the fusion peptide and fusion peptide-proximal region. Despite fusion peptide displacement, Ab1245 is non-neutralizing even at high concentrations, raising the possibility that only two fusion peptides per trimer are required for viral-host membrane fusion. These structural analyses facilitate immunogen design to prevent elicitation of Ab1245-like antibodies that block neutralizing antibodies against the fusion peptide.

10.
J Virol ; 83(1): 98-104, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945777

ABSTRACT

The antigen-binding fragment of the broadly neutralizing human immunodeficiency virus type 1 (HIV-1) antibody 2G12 has an unusual three-dimensional (3D) domain-swapped structure with two aligned combining sites that facilitates recognition of its carbohydrate epitope on gp120. When expressed as an intact immunoglobulin G (IgG), 2G12 formed typical IgG monomers containing two combining sites and a small fraction of a higher-molecular-weight species, which showed a significant increase in neutralization potency (50- to 80-fold compared to 2G12 monomer) across a range of clade A and B strains of HIV-1. Here we show that the higher-molecular-weight species corresponds to a 2G12 dimer containing four combining sites and present a model for how intermolecular 3D domain swapping could create a 2G12 dimer. Based on the structural model for a 3D domain-swapped 2G12 dimer, we designed and tested a series of 2G12 mutants predicted to increase the ratio of 2G12 dimer to monomer. We report a mutation that effectively increases the 2G12 dimer/monomer ratio without decreasing the expression yield. Increasing the proportion of 2G12 dimer compared to monomer could lead to a more potent reagent for gene therapy or passive immunization.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV-1/immunology , Amino Acid Substitution , Dimerization , Humans , Inhibitory Concentration 50 , Models, Molecular , Mutation, Missense , Neutralization Tests , Protein Binding , Protein Interaction Domains and Motifs , Surface Plasmon Resonance
11.
Elife ; 62017 08 15.
Article in English | MEDLINE | ID: mdl-28829740

ABSTRACT

An 'interactome' screen of all Drosophila cell-surface and secreted proteins containing immunoglobulin superfamily (IgSF) domains discovered a network formed by paralogs of Beaten Path (Beat) and Sidestep (Side), a ligand-receptor pair that is central to motor axon guidance. Here we describe a new method for interactome screening, the Bio-Plex Interactome Assay (BPIA), which allows identification of many interactions in a single sample. Using the BPIA, we 'deorphanized' four more members of the Beat-Side network. We confirmed interactions using surface plasmon resonance. The expression patterns of beat and side genes suggest that Beats are neuronal receptors for Sides expressed on peripheral tissues. side-VI is expressed in muscle fibers targeted by the ISNb nerve, as well as at growth cone choice points and synaptic targets for the ISN and TN nerves. beat-V genes, encoding Side-VI receptors, are expressed in ISNb and ISN motor neurons.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Growth Cones/metabolism , Membrane Proteins/genetics , Motor Neurons/metabolism , Muscles/metabolism , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Animals , Antibodies/chemistry , Biological Assay , Computational Biology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Embryo, Nonmammalian , Fluorescent Dyes/chemistry , Gene Expression Regulation, Developmental , Growth Cones/ultrastructure , Membrane Proteins/metabolism , Motor Neurons/ultrastructure , Muscles/ultrastructure , Nerve Tissue Proteins/metabolism , Nervous System/growth & development , Nervous System/ultrastructure , Phycoerythrin/chemistry , Phylogeny , Protein Interaction Mapping/methods , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction
12.
Elife ; 52016 09 13.
Article in English | MEDLINE | ID: mdl-27623146

ABSTRACT

SynGAP is a Ras/Rap GTPase-activating protein (GAP) that is a major constituent of postsynaptic densities (PSDs) from mammalian forebrain. Its α1 isoform binds to all three PDZ (PSD-95, Discs-large, ZO-1) domains of PSD-95, the principal PSD scaffold, and can occupy as many as 15% of these PDZ domains. We present evidence that synGAP-α1 regulates the composition of the PSD by restricting binding to the PDZ domains of PSD-95. We show that phosphorylation by Ca2+/calmodulin-dependent protein kinase II (CaMKII) and Polo-like kinase-2 (PLK2) decreases its affinity for the PDZ domains by several fold, which would free PDZ domains for occupancy by other proteins. Finally, we show that three critical postsynaptic signaling proteins that bind to the PDZ domains of PSD-95 are present in higher concentration in PSDs isolated from mice with a heterozygous deletion of synGAP.


Subject(s)
Disks Large Homolog 4 Protein/metabolism , PDZ Domains , Post-Synaptic Density/chemistry , ras GTPase-Activating Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mice , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism
13.
Drug Discov Today ; 10(15): 1065-71, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16055023

ABSTRACT

Protein therapeutics, including monoclonal antibodies, are a growing focus of drug discovery research organizations. High-throughput screening of large libraries of protein variants is therefore becoming increasingly important in R&D. As a result, there is a need to link large numbers of variant protein sequences with chemical and biological assay data. This integration will allow more efficient data mining and facilitate decision-making regarding hit identification, lead optimization and drug development. In this paper, we present an implementation in which a widely used small-molecule high-throughput screening data management system has been adapted to meet the unique needs of protein drug discovery and development.


Subject(s)
Database Management Systems , Proteomics/statistics & numerical data , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Databases, Genetic , Drug Design , Drug Evaluation, Preclinical , Receptors, Drug/drug effects , Receptors, Drug/genetics
14.
Sci Rep ; 4: 5152, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24919486

ABSTRACT

Chromatin immunoprecipitation coupled with DNA sequencing (ChIP-seq) is the major contemporary method for mapping in vivo protein-DNA interactions in the genome. It identifies sites of transcription factor, cofactor and RNA polymerase occupancy, as well as the distribution of histone marks. Consortia such as the ENCyclopedia Of DNA Elements (ENCODE) have produced large datasets using manual protocols. However, future measurements of hundreds of additional factors in many cell types and physiological states call for higher throughput and consistency afforded by automation. Such automation advances, when provided by multiuser facilities, could also improve the quality and efficiency of individual small-scale projects. The immunoprecipitation process has become rate-limiting, and is a source of substantial variability when performed manually. Here we report a fully automated robotic ChIP (R-ChIP) pipeline that allows up to 96 reactions. A second bottleneck is the dearth of renewable ChIP-validated immune reagents, which do not yet exist for most mammalian transcription factors. We used R-ChIP to screen new mouse monoclonal antibodies raised against p300, a histone acetylase, well-known as a marker of active enhancers, for which ChIP-competent monoclonal reagents have been lacking. We identified, validated for ChIP-seq, and made publicly available a monoclonal reagent called ENCITp300-1.


Subject(s)
Antibodies, Monoclonal/metabolism , Chromatin Immunoprecipitation/methods , E1A-Associated p300 Protein/metabolism , Protein Interaction Mapping/methods , Sequence Analysis, DNA/methods , Animals , Automation/methods , Histone Acetyltransferases/metabolism , Histones/metabolism , Mammals , Mice , Robotics , Transcription Factors/metabolism
15.
PLoS One ; 9(10): e111333, 2014.
Article in English | MEDLINE | ID: mdl-25357246

ABSTRACT

Hepatitis C Virus (HCV) infects 200 million individuals worldwide. Although several FDA approved drugs targeting the HCV serine protease and polymerase have shown promising results, there is a need for better drugs that are effective in treating a broader range of HCV genotypes and subtypes without being used in combination with interferon and/or ribavirin. Recently, two crystal structures of the core of the HCV E2 protein (E2c) have been determined, providing structural information that can now be used to target the E2 protein and develop drugs that disrupt the early stages of HCV infection by blocking E2's interaction with different host factors. Using the E2c structure as a template, we have created a structural model of the E2 protein core (residues 421-645) that contains the three amino acid segments that are not present in either structure. Computational docking of a diverse library of 1,715 small molecules to this model led to the identification of a set of 34 ligands predicted to bind near conserved amino acid residues involved in the HCV E2: CD81 interaction. Surface plasmon resonance detection was used to screen the ligand set for binding to recombinant E2 protein, and the best binders were subsequently tested to identify compounds that inhibit the infection of Huh-7 cells by HCV. One compound, 281816, blocked E2 binding to CD81 and inhibited HCV infection in a genotype-independent manner with IC50's ranging from 2.2 µM to 4.6 µM. 281816 blocked the early and late steps of cell-free HCV entry and also abrogated the cell-to-cell transmission of HCV. Collectively the results obtained with this new structural model of E2c suggest the development of small molecule inhibitors such as 281816 that target E2 and disrupt its interaction with CD81 may provide a new paradigm for HCV treatment.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Antiviral Agents/chemistry , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Genotype , Hepacivirus/genetics , Hepatitis C/pathology , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Protein Binding/drug effects , Recombinant Proteins/metabolism , Structural Homology, Protein , Surface Plasmon Resonance , Tetraspanin 28/metabolism , Thermodynamics , Viral Envelope Proteins/chemistry , Virus Internalization/drug effects
16.
Neuron ; 78(5): 813-26, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23764287

ABSTRACT

We developed a screening method for orphan receptor ligands, in which cell-surface proteins are expressed in Drosophila embryos from GAL4-dependent insertion lines and ligand candidates identified by the presence of ectopic staining with receptor fusion proteins. Stranded at second (Sas) binds to the receptor tyrosine phosphatase Ptp10D in embryos and in vitro. Sas and Ptp10D can interact in trans when expressed in cultured cells. Interactions between Sas and Ptp10D on longitudinal axons are required to prevent them from abnormally crossing the midline. Sas is expressed on both neurons and glia, whereas Ptp10D is restricted to CNS axons. We conducted epistasis experiments by overexpressing Sas in glia and examining how the resulting phenotypes are changed by removal of Ptp10D from neurons. We find that neuronal Ptp10D restrains signaling by overexpressed glial Sas, which would otherwise produce strong glial and axonal phenotypes.


Subject(s)
Axons/physiology , Cell Communication/physiology , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Neuroglia/physiology , Neurons/physiology , Protein Tyrosine Phosphatases/metabolism , Receptors, Cell Surface/metabolism , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Communication/genetics , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/physiology , Drosophila , Drosophila Proteins/genetics , Embryo, Nonmammalian , Enzyme-Linked Immunosorbent Assay , Horseradish Peroxidase/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Tyrosine Phosphatases/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Clin Immunol ; 118(1): 42-50, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16260183

ABSTRACT

Interferon-beta (IFN-beta) is currently the first-line therapy for the treatment of multiple sclerosis (MS). However, a significant percentage of MS patients develop anti-IFN-beta antibodies, which can reduce the efficacy of the drug. We describe an association between a common MHC class II allele (DRB1*0701), present in 23% of the patients studied, and the anti-IFN-beta antibody response. We identified IFN-beta epitopes using a peptide-binding assay with B cell lines expressing this allele. Moreover, epitope-specific activation responses obtained with peripheral blood mononuclear cells (PBMCs) from IFN-beta treated patients with the DRB1*0701 allele indicated a role for T-cell activation in IFN-beta immunogenicity. These results suggest that HLA typing of MS patients may provide an accurate screen for subjects who are likely to develop anti-IFN-beta antibodies and should therefore be considered for alternative therapies. In addition, elucidation of the factors underlying the anti-IFN-beta antibody response should accelerate the engineering of less immunogenic IFN-beta therapeutics.


Subject(s)
Antibodies/blood , Genes, MHC Class II/immunology , Interferon-beta/blood , Interferon-beta/immunology , Multiple Sclerosis/immunology , HLA-DQ Antigens/immunology , HLA-DQ Antigens/metabolism , HLA-DQ alpha-Chains , HLA-DQ beta-Chains , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , HLA-DRB1 Chains , Haplotypes , Humans , Interferon beta-1b , Interferon-beta/therapeutic use , Patients , T-Lymphocytes/immunology
19.
Proc Natl Acad Sci U S A ; 103(11): 4005-10, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537476

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity, a key effector function for the clinical efficacy of monoclonal antibodies, is mediated primarily through a set of closely related Fcgamma receptors with both activating and inhibitory activities. By using computational design algorithms and high-throughput screening, we have engineered a series of Fc variants with optimized Fcgamma receptor affinity and specificity. The designed variants display >2 orders of magnitude enhancement of in vitro effector function, enable efficacy against cells expressing low levels of target antigen, and result in increased cytotoxicity in an in vivo preclinical model. Our engineered Fc regions offer a means for improving the next generation of therapeutic antibodies and have the potential to broaden the diversity of antigens that can be targeted for antibody-based tumor therapy.


Subject(s)
Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Alemtuzumab , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Antibody Affinity , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/metabolism , B-Lymphocytes/immunology , Complement System Proteins/metabolism , Cytotoxicity, Immunologic , Genetic Variation , Humans , In Vitro Techniques , Lymphocyte Depletion , Macaca fascicularis , Protein Engineering , Receptors, IgG/metabolism , Trastuzumab
20.
Proc Natl Acad Sci U S A ; 99(25): 15926-31, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12446841

ABSTRACT

We present a combined computational and experimental method for the rapid optimization of proteins. Using beta-lactamase as a test case, we redesigned the active site region using our Protein Design Automation technology as a computational screen to search the entire sequence space. By eliminating sequences incompatible with the protein fold, Protein Design Automation rapidly reduced the number of sequences to a size amenable to experimental screening, resulting in a library of approximately equal 200,000 mutants. These were then constructed and experimentally screened to select for variants with improved resistance to the antibiotic cefotaxime. In a single round, we obtained variants exhibiting a 1,280-fold increase in resistance. To our knowledge, all of the mutations were novel, i.e., they have not been identified as beneficial by random mutagenesis or DNA shuffling or seen in any of the naturally occurring TEM beta-lactamases, the most prevalent type of Gram-negative beta-lactamases. This combined approach allows for the rapid improvement of any property that can be screened experimentally and provides a powerful broadly applicable tool for protein engineering.


Subject(s)
Computational Biology/methods , Escherichia coli Proteins/chemistry , Protein Engineering/methods , beta-Lactamases/chemistry , Amino Acid Substitution , Binding Sites , Cefotaxime/pharmacology , Drug Resistance , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Hydrogen Bonding , Models, Molecular , Monte Carlo Method , Mutagenesis, Site-Directed , Peptide Library , Protein Conformation , Protein Folding , beta-Lactamases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL