Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 5939, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009599

ABSTRACT

The precise regulation of protein function is essential in biological systems and a key goal in chemical biology and protein engineering. Here, we describe a straightforward method to engineer functional control into the isopeptide bond-forming SpyTag/SpyCatcher protein ligation system. First, we perform a cysteine scan of the structured region of SpyCatcher. Except for two known reactive and catalytic residues, none of these mutations abolish reactivity. In a second screening step, we modify the cysteines with disulfide bond-forming small molecules. Here we identify 8 positions at which modifications strongly inhibit reactivity. This inhibition can be reversed by reducing agents. We call such a reversibly inhibitable SpyCatcher "SpyLock". Using "BiLockCatcher", a genetic fusion of wild-type SpyCatcher and SpyLock, and SpyTagged antibody fragments, we generate bispecific antibodies in a single, scalable format, facilitating the screening of a large number of antibody combinations. We demonstrate this approach by screening anti-PD-1/anti-PD-L1 bispecific antibodies using a cellular reporter assay.


Subject(s)
Antibodies, Bispecific , Cysteine , Protein Engineering , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Antibodies, Bispecific/chemistry , Humans , Protein Engineering/methods , Cysteine/chemistry , Cysteine/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , HEK293 Cells , Disulfides/chemistry , Animals
2.
MAbs ; 15(1): 2177978, 2023.
Article in English | MEDLINE | ID: mdl-36803166

ABSTRACT

Phage display is an established method for the in vitro selection of recombinant antibodies and other proteins or peptides from gene libraries. Here we describe SpyDisplay, a phage display method in which the display is achieved via SpyTag/SpyCatcher protein ligation instead of genetically fusing the displayed protein to a phage coat protein. In our implementation, SpyTagged antibody antigen-binding fragments (Fabs) are displayed via protein ligation on filamentous phages carrying SpyCatcher fused to the pIII coat protein. A library of genes encoding Fab antibodies was cloned in an expression vector containing an f1 replication origin, and SpyCatcher-pIII was separately expressed from a genomic locus in engineered E. coli. We demonstrate the functional, covalent display of Fab on phage, and rapidly isolate specific high-affinity clones via phage panning, confirming the robustness of this selection system. SpyTagged Fabs, the direct outcome of the panning campaign, are compatible with modular antibody assembly using prefabricated SpyCatcher modules and can be directly tested in diverse assays. Furthermore, SpyDisplay streamlines additional applications that have traditionally been challenging for phage display: we show that it can be applied to N-terminal display of the protein of interest and it enables display of cytoplasmically folding proteins exported to periplasm via the TAT pathway.


Subject(s)
Bacteriophages , Escherichia coli , Escherichia coli/genetics , Cell Surface Display Techniques , Recombinant Proteins/genetics , Bacteriophages/genetics , Technology , Peptide Library
3.
Cell Chem Biol ; 28(6): 813-824.e6, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33529581

ABSTRACT

Antibodies are essential tools in research and diagnostics. Although antibody fragments typically obtained from in vitro selection can be rapidly produced in bacteria, the generation of full-length antibodies or the modification of antibodies with probes is time and labor intensive. Protein ligation such as SpyTag technology could covalently attach domains and labels to antibody fragments equipped with a SpyTag. However, we found that the established periplasmic expression of antibody fragments in E. coli led to quantitative cleavage of the SpyTag by the proteases Tsp and OmpT. Here we report successful periplasmic expression of SpyTagged Fab fragments and demonstrate the coupling to separately prepared SpyCatcher modules. We used this modular toolbox of SpyCatcher proteins to generate reagents for a variety of immunoassays and measured their performance in comparison with traditional reagents. Furthermore, we demonstrate surface immobilization, high-throughput screening of antibody libraries, and rapid prototyping of antibodies based on modular antibody assembly.


Subject(s)
Antibodies/metabolism , Immunoglobulin Fragments/genetics , Peptide Hydrolases/genetics , Periplasmic Proteins/genetics , Antibodies/chemistry , Cell Line, Tumor , Female , Humans , Immunoglobulin Fragments/isolation & purification , Immunoglobulin Fragments/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Periplasmic Proteins/chemistry , Periplasmic Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...