1
|
Grun CN, Jain R, Schniederberend M, Shoemaker CB, Nelson B, Kazmierczak BI. Bacterial cell surface characterization by phage display coupled to high-throughput sequencing. Nat Commun 2024; 15:7502. [PMID: 39209859 PMCID: PMC11362561 DOI: 10.1038/s41467-024-51912-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The remarkable capacity of bacteria to adapt in response to selective pressures drives antimicrobial resistance. Pseudomonas aeruginosa illustrates this point, establishing chronic infections during which it evolves to survive antimicrobials and evade host defenses. Many adaptive changes occur on the P. aeruginosa cell surface but methods to identify these are limited. Here we combine phage display with high-throughput DNA sequencing to create a high throughput, multiplexed technology for surveying bacterial cell surfaces, Phage-seq. By applying phage display panning to hundreds of bacterial genotypes and analyzing the dynamics of the phage display selection process, we capture important biological information about cell surfaces. This approach also yields camelid single-domain antibodies that recognize key P. aeruginosa virulence factors on live cells. These antibodies have numerous potential applications in diagnostics and therapeutics. We propose that Phage-seq establishes a powerful paradigm for studying the bacterial cell surface by identifying and profiling many surface features in parallel.
Collapse
Affiliation(s)
- Casey N Grun
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Ruchi Jain
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
- Piton Therapeutics, Watertown, MA, USA
| | - Maren Schniederberend
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - Bryce Nelson
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Orion Corporation, Turku, Finland
| | - Barbara I Kazmierczak
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Murciano-Calles J, Rodríguez-Martínez A, Palencia A, Andújar-Sánchez M, Iglesias-Bexiga M, Corbi-Verge C, Buzón P, Ruiz-Sanz J, Martínez JC, Pérez-Sánchez H, Cámara-Artigas A, Luque I. Phage display identification of high-affinity ligands for human TSG101-UEV: A structural and thermodynamic study of PTAP recognition. Int J Biol Macromol 2024; 274:133233. [PMID: 38901510 DOI: 10.1016/j.ijbiomac.2024.133233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
The ubiquitin E2 variant domain of TSG101 (TSG101-UEV) plays a pivotal role in protein sorting and virus budding by recognizing PTAP motifs within ubiquitinated proteins. Disrupting TSG101-UEV/PTAP interactions has emerged as a promising strategy for the development of novel host-oriented antivirals with a broad spectrum of action. Nonetheless, finding inhibitors with good properties as therapeutic agents remains a challenge since the key determinants of binding affinity and specificity are still poorly understood. Here we present a detailed thermodynamic, structural, and dynamic characterization viral PTAP Late domain recognition by TSG101-UEV, combining isothermal titration calorimetry, X-ray diffraction structural studies, molecular dynamics simulations, and computational analysis of intramolecular communication pathways. Our analysis highlights key contributions from conserved hydrophobic contacts and water-mediated hydrogen bonds at the PTAP binding interface. We have identified additional electrostatic hotspots adjacent to the core motif that modulate affinity. Using competitive phage display screening we have improved affinity by 1-2 orders of magnitude, producing novel peptides with low micromolar affinities that combine critical elements found in the best natural binders. Molecular dynamics simulations revealed that optimized peptides engage new pockets on the UEV domain surface. This study provides a comprehensive view of the molecular forces directing TSG101-UEV recognition of PTAP motifs, revealing that binding is governed by conserved structural elements yet tuneable through targeted optimization. These insights open new venues to design inhibitors targeting TSG101-dependent pathways with potential application as novel broad-spectrum antivirals.
Collapse
Affiliation(s)
- Javier Murciano-Calles
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Alejandro Rodríguez-Martínez
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain; Structural Bioinformatics and High-Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, Spain
| | - Andrés Palencia
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Montserrat Andújar-Sánchez
- Department of Chemistry and Physics, Agrifood Campus of International Excellence (ceiA3) and CIAMBITAL, University of Almería, Carretera de Sacramento s/n 04120 Almería, Spain
| | - Manuel Iglesias-Bexiga
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Carles Corbi-Verge
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Pedro Buzón
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Javier Ruiz-Sanz
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Jose C Martínez
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, Spain
| | - Ana Cámara-Artigas
- Department of Chemistry and Physics, Agrifood Campus of International Excellence (ceiA3) and CIAMBITAL, University of Almería, Carretera de Sacramento s/n 04120 Almería, Spain
| | - Irene Luque
- Department of Physical Chemistry, Institute of Biotechnology and Unit of Excellence in Chemistry applied to Biomedicine and Environment, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
3
|
Kschonsak YT, Gao X, Miller SE, Hwang S, Marei H, Wu P, Li Y, Ruiz K, Dorighi K, Holokai L, Perampalam P, Tsai WTK, Kee YS, Agard NJ, Harris SF, Hannoush RN, de Sousa E Melo F. Potent and selective binders of the E3 ubiquitin ligase ZNRF3 stimulate Wnt signaling and intestinal organoid growth. Cell Chem Biol 2024; 31:1176-1187.e10. [PMID: 38056465 DOI: 10.1016/j.chembiol.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/21/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
Selective and precise activation of signaling transduction cascades is key for cellular reprogramming and tissue regeneration. However, the development of small- or large-molecule agonists for many signaling pathways has remained elusive and is rate limiting to realize the full clinical potential of regenerative medicine. Focusing on the Wnt pathway, here we describe a series of disulfide-constrained peptides (DCPs) that promote Wnt signaling activity by modulating the cell surface levels of ZNRF3, an E3 ubiquitin ligase that controls the abundance of the Wnt receptor complex FZD/LRP at the plasma membrane. Mechanistically, monomeric DCPs induce ZNRF3 ubiquitination, leading to its cell surface clearance, ultimately resulting in FZD stabilization. Furthermore, we engineered multimeric DCPs that induce expansive growth of human intestinal organoids, revealing a dependence between valency and ZNRF3 clearance. Our work highlights a strategy for the development of potent, biologically active Wnt signaling pathway agonists via targeting of ZNRF3.
Collapse
Affiliation(s)
- Yvonne T Kschonsak
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA.
| | - Xinxin Gao
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA.
| | - Stephen E Miller
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Hadir Marei
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Ping Wu
- Department of Structural Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Yanjie Li
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Karen Ruiz
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Kristel Dorighi
- Department of Molecular Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Loryn Holokai
- Department of Biomarker Discovery, Genentech Inc, South San Francisco, CA 94080, USA
| | - Pirunthan Perampalam
- ProCogia Inc. under contract to Hoffmann-La Roche Limited, Toronto, Ontario M5J2P1, Canada
| | - Wen-Ting K Tsai
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Yee-Seir Kee
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Nicholas J Agard
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Seth F Harris
- Department of Structural Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
4
|
Zhou X, Kong S, Maker A, Remesh SG, Leung KK, Verba KA, Wells JA. Antibody discovery identifies regulatory mechanisms of protein arginine deiminase 4. Nat Chem Biol 2024; 20:742-750. [PMID: 38308046 PMCID: PMC11142921 DOI: 10.1038/s41589-023-01535-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 12/20/2023] [Indexed: 02/04/2024]
Abstract
Unlocking the potential of protein arginine deiminase 4 (PAD4) as a drug target for rheumatoid arthritis requires a deeper understanding of its regulation. In this study, we use unbiased antibody selections to identify functional antibodies capable of either activating or inhibiting PAD4 activity. Through cryogenic-electron microscopy, we characterized the structures of these antibodies in complex with PAD4 and revealed insights into their mechanisms of action. Rather than steric occlusion of the substrate-binding catalytic pocket, the antibodies modulate PAD4 activity through interactions with allosteric binding sites adjacent to the catalytic pocket. These binding events lead to either alteration of the active site conformation or the enzyme oligomeric state, resulting in modulation of PAD4 activity. Our study uses antibody engineering to reveal new mechanisms for enzyme regulation and highlights the potential of using PAD4 agonist and antagonist antibodies for studying PAD4-dependency in disease models and future therapeutic development.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sophie Kong
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Allison Maker
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Soumya G Remesh
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Kliment A Verba
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Liang CT, Roscow O, Zhang W. Generation and Characterization of Engineered Ubiquitin Variants to Modulate the Ubiquitin Signaling Cascade. Cold Spring Harb Protoc 2024; 2024:107784. [PMID: 36997275 DOI: 10.1101/pdb.over107784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The ubiquitin signaling cascade plays a crucial role in human cells. Consistent with this, malfunction of ubiquitination and deubiquitination is implicated in the initiation and progression of numerous human diseases, including cancer. Therefore, the development of potent and specific modulators of ubiquitin signal transduction has been at the forefront of drug development. In the past decade, a structure-based combinatorial protein-engineering approach has been used to generate ubiquitin variants (UbVs) as protein-based modulators of multiple components in the ubiquitin-proteasome system. Here, we review the design and generation of phage-displayed UbV libraries, including the processes of binder selection and library improvement. We also provide a comprehensive overview of the general in vitro and cellular methodologies involved in characterizing UbV binders. Finally, we describe two recent applications of UbVs for developing molecules with therapeutic potential.
Collapse
Affiliation(s)
- Chen T Liang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario N1G2W1, Canada
| | - Olivia Roscow
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario N1G2W1, Canada
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario N1G2W1, Canada
- CIFAR Azrieli Global Scholars Program, Canadian Institute for Advanced Research, MaRS Centre, Toronto, Ontario M5G1M1, Canada
| |
Collapse
|
6
|
Szekér P, Bodó T, Klima K, Csóti Á, Hanh NN, Murányi J, Hajdara A, Szántó TG, Panyi G, Megyeri M, Péterfi Z, Farkas S, Gyöngyösi N, Hornyák P. KcsA-Kv1.x chimeras with complete ligand-binding sites provide improved predictivity for screening selective Kv1.x blockers. J Biol Chem 2024; 300:107155. [PMID: 38479597 PMCID: PMC11002876 DOI: 10.1016/j.jbc.2024.107155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/12/2024] [Accepted: 03/06/2024] [Indexed: 04/08/2024] Open
Abstract
Despite significant advances in the development of therapeutic interventions targeting autoimmune diseases and chronic inflammatory conditions, lack of effective treatment still poses a high unmet need. Modulating chronically activated T cells through the blockade of the Kv1.3 potassium channel is a promising therapeutic approach; however, developing selective Kv1.3 inhibitors is still an arduous task. Phage display-based high throughput peptide library screening is a rapid and robust approach to develop promising drug candidates; however, it requires solid-phase immobilization of target proteins with their binding site preserved. Historically, the KcsA bacterial channel chimera harboring only the turret region of the human Kv1.3 channel was used for screening campaigns. Nevertheless, literature data suggest that binding to this type of chimera does not correlate well with blocking potency on the native Kv1.3 channels. Therefore, we designed and successfully produced advanced KcsA-Kv1.3, KcsA-Kv1.1, and KcsA-Kv1.2 chimeric proteins in which both the turret and part of the filter regions of the human Kv1.x channels were transferred. These T+F (turret-filter) chimeras showed superior peptide ligand-binding predictivity compared to their T-only versions in novel phage ELISA assays. Phage ELISA binding and competition results supported with electrophysiological data confirmed that the filter region of KcsA-Kv1.x is essential for establishing adequate relative affinity order among selected peptide toxins (Vm24 toxin, Hongotoxin-1, Kaliotoxin-1, Maurotoxin, Stichodactyla toxin) and consequently obtaining more reliable selectivity data. These new findings provide a better screening tool for future drug development efforts and offer insight into the target-ligand interactions of these therapeutically relevant ion channels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tibor Gábor Szántó
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Hungary
| | - György Panyi
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Hungary
| | | | | | | | - Norbert Gyöngyösi
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
7
|
Veggiani G, Sidhu SS. Generation and Selection of Synthetic Human Antibody Libraries via Phage Display. Cold Spring Harb Protoc 2024; 2024:108347. [PMID: 37295821 DOI: 10.1101/pdb.prot108347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Synthetic antibody libraries enable the development of antibodies that can recognize virtually any antigen, with affinity and specificity profiles that are superior to those of natural antibodies. By using highly stable and optimized frameworks, synthetic antibody libraries can be rapidly generated by precisely designing synthetic DNA, allowing absolute control over the position and chemical diversity introduced while expanding the sequence space for antigen recognition. Here, we describe a detailed protocol for the generation of highly diverse synthetic antibody phage display libraries based on a single framework, with diversity genetically incorporated by using finely designed mutagenic oligonucleotides. This general method enables the facile construction of large antibody libraries with precisely tunable features, resulting in the rapid development of recombinant antibodies for virtually any antigen.
Collapse
Affiliation(s)
- Gianluca Veggiani
- The Anvil Institute, Kitchener, Ontario N2G 1H6, Canada
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | |
Collapse
|
8
|
Gao X, Kaluarachchi H, Zhang Y, Hwang S, Hannoush RN. A phage-displayed disulfide constrained peptide discovery platform yields novel human plasma protein binders. PLoS One 2024; 19:e0299804. [PMID: 38547072 PMCID: PMC10977726 DOI: 10.1371/journal.pone.0299804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/15/2024] [Indexed: 04/02/2024] Open
Abstract
Disulfide constrained peptides (DCPs) show great potential as templates for drug discovery. They are characterized by conserved cysteine residues that form intramolecular disulfide bonds. Taking advantage of phage display technology, we designed and generated twenty-six DCP phage libraries with enriched molecular diversity to enable the discovery of ligands against disease-causing proteins of interest. The libraries were designed based on five DCP scaffolds, namely Momordica charantia 1 (Mch1), gurmarin, Asteropsin-A, antimicrobial peptide-1 (AMP-1), and potato carboxypeptidase inhibitor (CPI). We also report optimized workflows for screening and producing synthetic and recombinant DCPs. Examples of novel DCP binders identified against various protein targets are presented, including human IgG Fc, serum albumin, vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor (PDGF). We identified DCPs against human IgG Fc and serum albumin with sub-micromolar affinity from primary panning campaigns, providing alternative tools for potential half-life extension of peptides and small protein therapeutics. Overall, the molecular diversity of the DCP scaffolds included in the designed libraries, coupled with their distinct biochemical and biophysical properties, enables efficient and robust identification of de novo binders to drug targets of therapeutic relevance.
Collapse
Affiliation(s)
- Xinxin Gao
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Harini Kaluarachchi
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Biological Chemistry, Genentech, South San Francisco, California, United States of America
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Rami N. Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| |
Collapse
|
9
|
Zhou L, Cai F, Li Y, Gao X, Wei Y, Fedorova A, Kirchhofer D, Hannoush RN, Zhang Y. Disulfide-constrained peptide scaffolds enable a robust peptide-therapeutic discovery platform. PLoS One 2024; 19:e0300135. [PMID: 38547109 PMCID: PMC10977697 DOI: 10.1371/journal.pone.0300135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024] Open
Abstract
Peptides present an alternative modality to immunoglobulin domains or small molecules for developing therapeutics to either agonize or antagonize cellular pathways associated with diseases. However, peptides often suffer from poor chemical and physical stability, limiting their therapeutic potential. Disulfide-constrained peptides (DCP) are naturally occurring and possess numerous desirable properties, such as high stability, that qualify them as drug-like scaffolds for peptide therapeutics. DCPs contain loop regions protruding from the core of the molecule that are amenable to peptide engineering via direct evolution by use of phage display technology. In this study, we have established a robust platform for the discovery of peptide therapeutics using various DCPs as scaffolds. We created diverse libraries comprising seven different DCP scaffolds, resulting in an overall diversity of 2 x 1011. The effectiveness of this platform for functional hit discovery has been extensively evaluated, demonstrating a hit rate comparable to that of synthetic antibody libraries. By utilizing chemically synthesized and in vitro folded peptides derived from selections of phage displayed DCP libraries, we have successfully generated functional inhibitors targeting the HtrA1 protease. Through affinity maturation strategies, we have transformed initially weak binders against Notch2 with micromolar Kd values to high-affinity ligands in the nanomolar range. This process highlights a viable hit-to-lead progression. Overall, our platform holds significant potential to greatly enhance the discovery of peptide therapeutics.
Collapse
Affiliation(s)
- Lijuan Zhou
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Fei Cai
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Yanjie Li
- Department of Peptide Therapeutics, Genentech, Inc., South San Francisco, California, United States of America
| | - Xinxin Gao
- Department of Peptide Therapeutics, Genentech, Inc., South San Francisco, California, United States of America
| | - Yuehua Wei
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Anna Fedorova
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Daniel Kirchhofer
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Rami N. Hannoush
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Yingnan Zhang
- Departments of Biological Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| |
Collapse
|
10
|
Eyssen LEA, Ramadurai S, Abdelkarim S, Buckle I, Cornish K, Lin H, Jones A, Stephens GJ, Owens RJ. From Llama to Nanobody: A Streamlined Workflow for the Generation of Functionalised VHHs. Bio Protoc 2024; 14:e4962. [PMID: 38841291 PMCID: PMC10958182 DOI: 10.21769/bioprotoc.4962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 06/07/2024] Open
Abstract
Nanobodies are recombinant antigen-specific single domain antibodies (VHHs) derived from the heavy chain-only subset of camelid immunoglobulins. Their small molecular size, facile expression, high affinity, and stability have combined to make them unique targeting reagents with numerous applications in the biomedical sciences. From our work in producing nanobodies to over sixty different proteins, we present a standardised workflow for nanobody discovery from llama immunisation, library building, panning, and small-scale expression for prioritisation of binding clones. In addition, we introduce our suites of mammalian and bacterial vectors, which can be used to functionalise selected nanobodies for various applications such as in imaging and purification. Key features • Standardise the process of building nanobody libraries and finding nanobody binders so that it can be repeated in any lab with reasonable equipment. • Introduce two suites of vectors to functionalise nanobodies for production in either bacterial or mammalian cells.
Collapse
Affiliation(s)
- Lauren E-A Eyssen
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
| | - Siva Ramadurai
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
| | - Sahar Abdelkarim
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
| | - Imogen Buckle
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
| | - Katy Cornish
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
| | - Hong Lin
- School of Pharmacy, University of Reading, Reading,
RG6 6UB, UK
| | - A.K. Jones
- Centre for Dairy Research, University of Reading,
Reading, RG2 9HY, UK
| | | | - Raymond J. Owens
- Structural Biology, Rosalind Franklin Institute,
Didcot, OX11 0QX, UK
- Division of Structural Biology, Nuffield Department
of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| |
Collapse
|
11
|
Li N, Ge Q, Guo Q, Tao Y. Identification and functional validation of FZD8-specific antibodies. Int J Biol Macromol 2024; 254:127846. [PMID: 37926311 DOI: 10.1016/j.ijbiomac.2023.127846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
The Wnt pathway is an evolutionarily conserved pathway involved in stem cell homeostasis and tissue regeneration. Aberrant signaling in the Wnt pathway is highly associated with cancer. Developing antibodies to block overactivation of Frizzled receptors (FZDs), the main receptors in the Wnt pathway, is one of the viable options for treating cancer. However, obtaining isoform-specific antibodies is often challenging due to the high degree of homology among the ten FZDs. In this study, by using a synthetic library, we identified an antibody named pF8_AC3 that preferentially binds to FZD8. Guided by the structure of the complex of pF8_AC3 and FZD8, a second-generation targeted library was further constructed, and finally, the FZD8-specific antibody sF8_AG6 was obtained. Cell-based assays showed that these antibodies could selectively block FZD8-mediated signaling activation. Taken together, these antibodies have the potential to be developed into therapeutic drugs in the future.
Collapse
Affiliation(s)
- Na Li
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Qiangqiang Ge
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Qiong Guo
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Yuyong Tao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
12
|
Chung K, Booth MJ. Sequence-independent, site-specific incorporation of chemical modifications to generate light-activated plasmids. Chem Sci 2023; 14:12693-12706. [PMID: 38020373 PMCID: PMC10646958 DOI: 10.1039/d3sc02761a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Plasmids are ubiquitous in biology, where they are used to study gene-function relationships and intricate molecular networks, and hold potential as therapeutic devices. Developing methods to control their function will advance their application in research and may also expedite their translation to clinical settings. Light is an attractive stimulus to conditionally regulate plasmid expression as it is non-invasive, and its properties such as wavelength, intensity, and duration can be adjusted to minimise cellular toxicity and increase penetration. Herein, we have developed a method to site-specifically introduce photocages into plasmids, by resynthesising one strand in a manner similar to Kunkel mutagenesis. Unlike alternative approaches to chemically modify plasmids, this method is sequence-independent at the site of modification and uses commercially available phosphoramidites. To generate our light-activated (LA) plasmids, photocleavable biotinylated nucleobases were introduced at specific sites across the T7 and CMV promoters on plasmids and bound to streptavidin to sterically block access. These LA-plasmids were then successfully used to control expression in both cell-free systems (T7 promoter) and mammalian cells (CMV promoter). These light-activated plasmids might be used to remotely control cellular activity and reduce off-target toxicity for future medical use. Our simple approach to plasmid modification might also be used to introduce novel chemical moieties for advanced function.
Collapse
Affiliation(s)
- Khoa Chung
- Department of Chemistry, University of Oxford Mansfield Road OX1 3TA Oxford UK
| | - Michael J Booth
- Department of Chemistry, University of Oxford Mansfield Road OX1 3TA Oxford UK
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
13
|
Farkas S, Cioca D, Murányi J, Hornyák P, Brunyánszki A, Szekér P, Boros E, Horváth P, Hujber Z, Rácz GZ, Nagy N, Tóth R, Nyitray L, Péterfi Z. Chlorotoxin binds to both matrix metalloproteinase 2 and neuropilin 1. J Biol Chem 2023; 299:104998. [PMID: 37394009 PMCID: PMC10477481 DOI: 10.1016/j.jbc.2023.104998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/03/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023] Open
Abstract
Chlorotoxin (CTX), a scorpion venom-derived 36-residue miniprotein, binds to and is taken up selectively by glioblastoma cells. Previous studies provided controversial results concerning target protein(s) of CTX. These included CLC3 chloride channel, matrix metalloproteinase 2 (MMP-2), regulators of MMP-2, annexin A2, and neuropilin 1 (NRP1). The present study aimed at clarifying which of the proposed binding partners can really interact with CTX using biochemical methods and recombinant proteins. For this purpose, we established two new binding assays based on anchoring the tested proteins to microbeads and quantifying the binding of CTX by flow cytometry. Screening of His-tagged proteins anchored to cobalt-coated beads indicated strong interaction of CTX with MMP-2 and NRP1, whereas binding to annexin A2 was not confirmed. Similar results were obtained with fluorophore-labeled CTX and CTX-displaying phages. Affinity of CTX to MMP-2 and NRP1 was assessed by the "immunoglobulin-coated bead" test, in which the proteins were anchored to beads by specific antibodies. This assay yielded highly reproducible data using both direct titration and displacement approach. The affinities of labeled and unlabeled CTX appeared to be similar for both MMP-2 and NRP1 with estimated KD values of 0.5 to 0.7 μM. Contrary to previous reports, we found that CTX does not inhibit the activity of MMP-2 and that CTX not only with free carboxyl end but also with carboxamide terminal end binds to NRP1. We conclude that the presented robust assays could also be applied for affinity-improving studies of CTX to its genuine targets using phage display libraries.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eszter Boros
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Patrik Horváth
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | | | | | | | | | - László Nyitray
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | | |
Collapse
|
14
|
Chew KS, Wells RC, Moshkforoush A, Chan D, Lechtenberg KJ, Tran HL, Chow J, Kim DJ, Robles-Colmenares Y, Srivastava DB, Tong RK, Tong M, Xa K, Yang A, Zhou Y, Akkapeddi P, Annamalai L, Bajc K, Blanchette M, Cherf GM, Earr TK, Gill A, Huynh D, Joy D, Knight KN, Lac D, Leung AWS, Lexa KW, Liau NPD, Becerra I, Malfavon M, McInnes J, Nguyen HN, Lozano EI, Pizzo ME, Roche E, Sacayon P, Calvert MEK, Daneman R, Dennis MS, Duque J, Gadkar K, Lewcock JW, Mahon CS, Meisner R, Solanoy H, Thorne RG, Watts RJ, Zuchero YJY, Kariolis MS. CD98hc is a target for brain delivery of biotherapeutics. Nat Commun 2023; 14:5053. [PMID: 37598178 PMCID: PMC10439950 DOI: 10.1038/s41467-023-40681-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Brain exposure of systemically administered biotherapeutics is highly restricted by the blood-brain barrier (BBB). Here, we report the engineering and characterization of a BBB transport vehicle targeting the CD98 heavy chain (CD98hc or SLC3A2) of heterodimeric amino acid transporters (TVCD98hc). The pharmacokinetic and biodistribution properties of a CD98hc antibody transport vehicle (ATVCD98hc) are assessed in humanized CD98hc knock-in mice and cynomolgus monkeys. Compared to most existing BBB platforms targeting the transferrin receptor, peripherally administered ATVCD98hc demonstrates differentiated brain delivery with markedly slower and more prolonged kinetic properties. Specific biodistribution profiles within the brain parenchyma can be modulated by introducing Fc mutations on ATVCD98hc that impact FcγR engagement, changing the valency of CD98hc binding, and by altering the extent of target engagement with Fabs. Our study establishes TVCD98hc as a modular brain delivery platform with favorable kinetic, biodistribution, and safety properties distinct from previously reported BBB platforms.
Collapse
Affiliation(s)
- Kylie S Chew
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Robert C Wells
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Arash Moshkforoush
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Darren Chan
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kendra J Lechtenberg
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hai L Tran
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Johann Chow
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Do Jin Kim
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Devendra B Srivastava
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Raymond K Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mabel Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaitlin Xa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Alexander Yang
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Yinhan Zhou
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Padma Akkapeddi
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Lakshman Annamalai
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaja Bajc
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Marie Blanchette
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Gerald Maxwell Cherf
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Timothy K Earr
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Audrey Gill
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Huynh
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Joy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kristen N Knight
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Diana Lac
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Amy Wing-Sze Leung
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Katrina W Lexa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Nicholas P D Liau
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Isabel Becerra
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mario Malfavon
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Joseph McInnes
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hoang N Nguyen
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Edwin I Lozano
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Michelle E Pizzo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Elysia Roche
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Patricia Sacayon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Meredith E K Calvert
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Richard Daneman
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Mark S Dennis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph Duque
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kapil Gadkar
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph W Lewcock
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Cathal S Mahon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - René Meisner
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hilda Solanoy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Robert G Thorne
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Ryan J Watts
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Y Joy Yu Zuchero
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Mihalis S Kariolis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| |
Collapse
|
15
|
Contreras MA, Serrano-Rivero Y, González-Pose A, Salazar-Uribe J, Rubio-Carrasquilla M, Soares-Alves M, Parra NC, Camacho-Casanova F, Sánchez-Ramos O, Moreno E. Design and Construction of a Synthetic Nanobody Library: Testing Its Potential with a Single Selection Round Strategy. Molecules 2023; 28:molecules28093708. [PMID: 37175117 PMCID: PMC10180287 DOI: 10.3390/molecules28093708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Nanobodies (Nbs) are single domain antibody fragments derived from heavy-chain antibodies found in members of the Camelidae family. They have become a relevant class of biomolecules for many different applications because of several important advantages such as their small size, high solubility and stability, and low production costs. On the other hand, synthetic Nb libraries are emerging as an attractive alternative to animal immunization for the selection of antigen-specific Nbs. Here, we present the design and construction of a new synthetic nanobody library using the phage display technology, following a structure-based approach in which the three hypervariable loops were subjected to position-specific randomization schemes. The constructed library has a clonal diversity of 108 and an amino acid variability that matches the codon distribution set by design at each randomized position. We have explored the capabilities of the new library by selecting nanobodies specific for three antigens: vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF) and the glycoprotein complex (GnGc) of Andes virus. To test the potential of the library to yield a variety of antigen-specific Nbs, we introduced a biopanning strategy consisting of a single selection round using stringent conditions. Using this approach, we obtained several binders for each of the target antigens. The constructed library represents a promising nanobody source for different applications.
Collapse
Affiliation(s)
- María Angélica Contreras
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | | | - Alaín González-Pose
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| | | | | | - Matheus Soares-Alves
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Natalie C Parra
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Frank Camacho-Casanova
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Oliberto Sánchez-Ramos
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Ernesto Moreno
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| |
Collapse
|
16
|
Thakur AK, Miller SE, Liau NPD, Hwang S, Hansen S, de Sousa E Melo F, Sudhamsu J, Hannoush RN. Synthetic Multivalent Disulfide-Constrained Peptide Agonists Potentiate Wnt1/β-Catenin Signaling via LRP6 Coreceptor Clustering. ACS Chem Biol 2023; 18:772-784. [PMID: 36893429 DOI: 10.1021/acschembio.2c00753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Wnt ligands are critical for tissue homeostasis and form a complex with LRP6 and frizzled coreceptors to initiate Wnt/β-catenin signaling. Yet, how different Wnts achieve various levels of signaling activation through distinct domains on LRP6 remains elusive. Developing tool ligands that target individual LRP6 domains could help elucidate the mechanism of Wnt signaling regulation and uncover pharmacological approaches for pathway modulation. We employed directed evolution of a disulfide constrained peptide (DCP) to identify molecules that bind to the third β-propeller domain of LRP6. The DCPs antagonize Wnt3a while sparing Wnt1 signaling. Using PEG linkers with different geometries, we converted the Wnt3a antagonist DCPs to multivalent molecules that potentiated Wnt1 signaling by clustering the LRP6 coreceptor. The mechanism of potentiation is unique as it occurred only in the presence of extracellular secreted Wnt1 ligand. While all DCPs recognized a similar binding interface on LRP6, they displayed different spatial orientations that influenced their cellular activities. Moreover, structural analyses revealed that the DCPs exhibited new folds that were distinct from the parent DCP framework they were evolved from. The multivalent ligand design principles highlighted in this study provide a path for developing peptide agonists that modulate different branches of cellular Wnt signaling.
Collapse
Affiliation(s)
- Avinash K Thakur
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Stephen E Miller
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Nicholas P D Liau
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Simon Hansen
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Felipe de Sousa E Melo
- Department of Molecular Oncology, Genentech, South San Francisco, California 94080, United States
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
17
|
Yun J, Hansen S, Morris O, Madden DT, Libeu CP, Kumar AJ, Wehrfritz C, Nile AH, Zhang Y, Zhou L, Liang Y, Modrusan Z, Chen MB, Overall CC, Garfield D, Campisi J, Schilling B, Hannoush RN, Jasper H. Senescent cells perturb intestinal stem cell differentiation through Ptk7 induced noncanonical Wnt and YAP signaling. Nat Commun 2023; 14:156. [PMID: 36631445 PMCID: PMC9834240 DOI: 10.1038/s41467-022-35487-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/06/2022] [Indexed: 01/13/2023] Open
Abstract
Cellular senescence and the senescence-associated secretory phenotype (SASP) are implicated in aging and age-related disease, and SASP-related inflammation is thought to contribute to tissue dysfunction in aging and diseased animals. However, whether and how SASP factors influence the regenerative capacity of tissues remains unclear. Here, using intestinal organoids as a model of tissue regeneration, we show that SASP factors released by senescent fibroblasts deregulate stem cell activity and differentiation and ultimately impair crypt formation. We identify the secreted N-terminal domain of Ptk7 as a key component of the SASP that activates non-canonical Wnt / Ca2+ signaling through FZD7 in intestinal stem cells (ISCs). Changes in cytosolic [Ca2+] elicited by Ptk7 promote nuclear translocation of YAP and induce expression of YAP/TEAD target genes, impairing symmetry breaking and stem cell differentiation. Our study discovers secreted Ptk7 as a factor released by senescent cells and provides insight into the mechanism by which cellular senescence contributes to tissue dysfunction in aging and disease.
Collapse
Affiliation(s)
- Jina Yun
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Simon Hansen
- NBE Therapeutics, Hochbergstrasse 60C, 4057, Basel, Switzerland
| | - Otto Morris
- Exscientia Ltd., The Schrödinger Building Oxford Science Park, Oxford, OX4 4GE, UK
| | - David T Madden
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | - Clare Peters Libeu
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | - Arjun J Kumar
- Fred Hutch/University of Washington, 1100 Fairview Ave. N., Seattle, WA, 98109, USA
| | - Cameron Wehrfritz
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | - Aaron H Nile
- Calico Labs LLC., 1170 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Yingnan Zhang
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Lijuan Zhou
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Yuxin Liang
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Zora Modrusan
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Michelle B Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | - David Garfield
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | - Birgit Schilling
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | - Rami N Hannoush
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Heinrich Jasper
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| |
Collapse
|
18
|
Ge Q, Teng M, Li X, Guo Q, Tao Y. An epitope-directed selection strategy facilitating the identification of Frizzled receptor selective antibodies. Structure 2023; 31:33-43.e5. [PMID: 36513066 DOI: 10.1016/j.str.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/27/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
The lack of incorporating epitope information into the selection process makes the conventional antibody screening method less effective in identifying antibodies with desired functions. Here, we developed an epitope-directed antibody selection method by designing a directed library favoring the target epitope and a precise "counter" antigen for clearing irrelevant binders in the library. With this method, we successfully isolated an antibody, pF7_A5, that targets the less conserved region on the FZD2/7 CRD as designed. Guided by the structure of pF7_A5-FZD2CRD, a further round of evolution was conducted together with the "counter" antigen selection strategy, and ultimately, an FZD2-specific antibody and an FZD7-preferred antibody were obtained. Because of targeting the predefined functional site, all these antibodies exhibited the expected modulatory activity on the Wnt pathway. Together, the method developed here will be useful in antibody drug discovery, and the identified FZD antibodies will have clinical potential in FZD-related cancer therapy.
Collapse
Affiliation(s)
- Qiangqiang Ge
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China
| | - Maikun Teng
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Xu Li
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Qiong Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Yuyong Tao
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China; Joint Laboratory of Innovation in Life Sciences University of Science and Technology of China (USTC) and Changchun Zhuoyi Biological Co. Ltd., 130616 Changchun, P.R. China.
| |
Collapse
|
19
|
Martyn GD, Veggiani G, Sidhu SS. Engineering SH2 Domains with Tailored Specificities and Affinities. Methods Mol Biol 2023; 2705:307-348. [PMID: 37668982 DOI: 10.1007/978-1-0716-3393-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The Src Homology 2 (SH2) domain is an emerging biotechnology with applications in basic science, drug discovery, and even diagnostics. The SH2 domains rapid uptake into different areas of research is a direct result of the wealth of information generated on its biochemical, biological, and biophysical role in mammalian cell biology. Functionally, the SH2 domain binds and recognizes specific phosphotyrosine (pTyr) residues in the cell to mediate protein-protein interactions (PPIs) that govern signal transduction networks. These signal transduction networks are responsible for relaying growth and stress state signals to the cell's nucleus, ultimately effecting a change in cell biology. Protein engineers have been able to increase the affinity of SH2 domains for pTyr while also tailoring the domains' specificity to unique amino acid sequences flanking the pTyr residue. In this way, it has been possible to develop unique SH2 variants for use in affinity-purification coupled to mass spectrometry (AP-MS) experiments, microscopy, or even synthetic biology. This chapter outlines methods to tailor the affinity and specificity of virtually any human SH2 domain using a combination of rational engineering and phage-display approaches.
Collapse
Affiliation(s)
- Gregory D Martyn
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gianluca Veggiani
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Sachdev S Sidhu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
20
|
Targeted degradation via direct 26S proteasome recruitment. Nat Chem Biol 2023; 19:55-63. [PMID: 36577875 PMCID: PMC9797404 DOI: 10.1038/s41589-022-01218-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/25/2022] [Indexed: 12/29/2022]
Abstract
Engineered destruction of target proteins by recruitment to the cell's degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.
Collapse
|
21
|
Sherpa D, Mueller J, Karayel Ö, Xu P, Yao Y, Chrustowicz J, Gottemukkala KV, Baumann C, Gross A, Czarnecki O, Zhang W, Gu J, Nilvebrant J, Sidhu SS, Murray PJ, Mann M, Weiss MJ, Schulman BA, Alpi AF. Modular UBE2H-CTLH E2-E3 complexes regulate erythroid maturation. eLife 2022; 11:e77937. [PMID: 36459484 PMCID: PMC9718529 DOI: 10.7554/elife.77937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
The development of haematopoietic stem cells into mature erythrocytes - erythropoiesis - is a controlled process characterized by cellular reorganization and drastic reshaping of the proteome landscape. Failure of ordered erythropoiesis is associated with anaemias and haematological malignancies. Although the ubiquitin system is a known crucial post-translational regulator in erythropoiesis, how the erythrocyte is reshaped by the ubiquitin system is poorly understood. By measuring the proteomic landscape of in vitro human erythropoiesis models, we found dynamic differential expression of subunits of the CTLH E3 ubiquitin ligase complex that formed maturation stage-dependent assemblies of topologically homologous RANBP9- and RANBP10-CTLH complexes. Moreover, protein abundance of CTLH's cognate E2 ubiquitin conjugating enzyme UBE2H increased during terminal differentiation, and UBE2H expression depended on catalytically active CTLH E3 complexes. CRISPR-Cas9-mediated inactivation of CTLH E3 assemblies or UBE2H in erythroid progenitors revealed defects, including spontaneous and accelerated erythroid maturation as well as inefficient enucleation. Thus, we propose that dynamic maturation stage-specific changes of UBE2H-CTLH E2-E3 modules control the orderly progression of human erythropoiesis.
Collapse
Affiliation(s)
- Dawafuti Sherpa
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Judith Mueller
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Özge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Peng Xu
- Cyrus Tang Medical Institute, National Clinical Research Centre for Hematologic Diseases, Collaborative Innovation Centre of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow UniversitySuzhouChina
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Yu Yao
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Jakub Chrustowicz
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Karthik V Gottemukkala
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Christine Baumann
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Annette Gross
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
- Department of Immunoregulation, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Oliver Czarnecki
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Wei Zhang
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Jun Gu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Johan Nilvebrant
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Sachdev S Sidhu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Peter J Murray
- Department of Immunoregulation, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Arno F Alpi
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
22
|
Berge-Seidl S, Nielsen NV, Rodriguez Alfonso AA, Etscheid M, Kandanur SPS, Haug BE, Stensland M, Thiede B, Karacan M, Preising N, Wiese S, Ständker L, Declerck PJ, Løset GÅ, Kanse SM. Identification of a Phage Display-Derived Peptide Interacting with the N-Terminal Region of Factor VII Activating Protease (FSAP) Enables Characterization of Zymogen Activation. ACS Chem Biol 2022; 17:2631-2642. [PMID: 36070465 PMCID: PMC9486805 DOI: 10.1021/acschembio.2c00538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/25/2022] [Indexed: 01/19/2023]
Abstract
Factor VII Activating protease (FSAP) has a protective effect in diverse disease conditions as inferred from studies in FSAP-/- mice and humans deficient in FSAP activity due to single-nucleotide polymorphism. The zymogen form of FSAP in plasma is activated by extracellular histones that are released during tissue injury or inflammation or by positively charged surfaces. However, it is not clear whether this activation mechanism is specific and amenable to manipulation. Using a phage display approach, we have identified a Cys-constrained 11 amino acid peptide, NNKC9/41, that activates pro-FSAP in plasma. The synthetic linear peptide has a propensity to cyclize through the terminal Cys groups, of which the antiparallel cyclic dimer, but not the monocyclic peptide, is the active component. Other commonly found zymogens in the plasma, related to the hemostasis system, were not activated. Binding studies with FSAP domain deletion mutants indicate that the N-terminus of FSAP is the key interaction site of this peptide. In a monoclonal antibody screen, we identified MA-FSAP-38C7 that prevented the activation of pro-FSAP by the peptide. This antibody bound to the LESLDP sequence (amino acids 30-35) in an intrinsically disordered stretch in the N-terminus of FSAP. The plasma clotting time was shortened by NNKC9/41, and this was reversed by MA-FSAP-38C7, demonstrating the utility of this peptide. Peptide NNKC9/41 will be useful as a tool to delineate the molecular mechanism of activation of pro-FSAP, elucidate its biological role, and provide a starting point for the pharmacological manipulation of FSAP activity.
Collapse
Affiliation(s)
| | - Nis Valentin Nielsen
- Oslo
University Hospital and Medical Faculty, University of Oslo, 0372 Oslo, Norway
| | | | | | | | - Bengt Erik Haug
- Department
of Chemistry and Center for Pharmacy, University
of Bergen, 5007 Bergen, Norway
| | - Maria Stensland
- Oslo
University Hospital and Medical Faculty, University of Oslo, 0372 Oslo, Norway
| | - Bernd Thiede
- Department
of Biosciences, University of Oslo, 0371 Oslo, Norway
| | | | | | | | | | - Paul J. Declerck
- Department
of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Geir Åge Løset
- Department
of Biosciences, University of Oslo, 0371 Oslo, Norway
- Nextera
AS, 0349 Oslo, Norway
| | - Sandip M. Kanse
- Oslo
University Hospital and Medical Faculty, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
23
|
Zhao D, Liu L, Liu X, Zhang J, Yin Y, Luan L, Jiang D, Yang X, Li L, Xiong H, Xing D, Zheng Q, Xia N, Tao Y, Li S, Huang H. A potent synthetic nanobody with broad-spectrum activity neutralizes SARS-CoV-2 virus and the Omicron variant BA.1 through a unique binding mode. J Nanobiotechnology 2022; 20:411. [PMID: 36109732 PMCID: PMC9479348 DOI: 10.1186/s12951-022-01619-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/05/2022] [Indexed: 12/23/2022] Open
Abstract
The major challenge to controlling the COVID pandemic is the rapid mutation rate of the SARS-CoV-2 virus, leading to the escape of the protection of vaccines and most of the neutralizing antibodies to date. Thus, it is essential to develop neutralizing antibodies with broad-spectrum activity targeting multiple SARS-CoV-2 variants. Here, we report a synthetic nanobody (named C5G2) obtained by phage display and subsequent antibody engineering. C5G2 has a single-digit nanomolar binding affinity to the RBD domain and inhibits its binding to ACE2 with an IC50 of 3.7 nM. Pseudovirus assays indicated that monovalent C5G2 could protect the cells from infection with SARS-CoV-2 wild-type virus and most of the viruses of concern, i.e., Alpha, Beta, Gamma and Omicron variants. Strikingly, C5G2 has the highest potency against Omicron BA.1 among all the variants, with an IC50 of 4.9 ng/mL. The cryo-EM structure of C5G2 in complex with the spike trimer showed that C5G2 binds to RBD mainly through its CDR3 at a conserved region that does not overlap with the ACE2 binding surface. Additionally, C5G2 binds simultaneously to the neighboring NTD domain of the spike trimer through the same CDR3 loop, which may further increase its potency against viral infection. Third, the steric hindrance caused by FR2 of C5G2 could inhibit the binding of ACE2 to RBD as well. Thus, this triple-function nanobody may serve as an effective drug for prophylaxis and therapy against Omicron as well as future variants.
Collapse
|
24
|
Frosi Y, Lin YC, Shimin J, Ramlan SR, Hew K, Engman AH, Pillai A, Yeung K, Cheng YX, Cornvik T, Nordlund P, Goh M, Lama D, Gates ZP, Verma CS, Thean D, Lane DP, Asial I, Brown CJ. Engineering an autonomous VH domain to modulate intracellular pathways and to interrogate the eIF4F complex. Nat Commun 2022; 13:4854. [PMID: 35982046 PMCID: PMC9388512 DOI: 10.1038/s41467-022-32463-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/02/2022] [Indexed: 11/12/2022] Open
Abstract
An attractive approach to target intracellular macromolecular interfaces and to model putative drug interactions is to design small high-affinity proteins. Variable domains of the immunoglobulin heavy chain (VH domains) are ideal miniproteins, but their development has been restricted by poor intracellular stability and expression. Here we show that an autonomous and disufhide-free VH domain is suitable for intracellular studies and use it to construct a high-diversity phage display library. Using this library and affinity maturation techniques we identify VH domains with picomolar affinity against eIF4E, a protein commonly hyper-activated in cancer. We demonstrate that these molecules interact with eIF4E at the eIF4G binding site via a distinct structural pose. Intracellular overexpression of these miniproteins reduce cellular proliferation and expression of malignancy-related proteins in cancer cell lines. The linkage of high-diversity in vitro libraries with an intracellularly expressible miniprotein scaffold will facilitate the discovery of VH domains suitable for intracellular applications.
Collapse
Affiliation(s)
- Yuri Frosi
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Yen-Chu Lin
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Insilico Medicine Taiwan Ltd., Taipei City, 110208, Taiwan
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei City, 112304, Taiwan
| | - Jiang Shimin
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Siti Radhiah Ramlan
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Kelly Hew
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Alf Henrik Engman
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Anil Pillai
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Kit Yeung
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Yue Xiang Cheng
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Tobias Cornvik
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Par Nordlund
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Megan Goh
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - Dilraj Lama
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Biomedicum Quarter 7B-C Solnavägen 9, 17165, Solna, Sweden
| | - Zachary P Gates
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), A*STAR, 8 A Biomedical Grove, #07-01 Neuros Building, 138665, Singapore, Singapore
| | - Chandra S Verma
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, 138671, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore, Singapore
| | - Dawn Thean
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - David P Lane
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - Ignacio Asial
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore.
| | - Christopher J Brown
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore.
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore.
| |
Collapse
|
25
|
Maruthachalam BV, Barreto K, Hogan D, Kusalik A, Geyer CR. Generation of synthetic antibody fragments with optimal complementarity determining region lengths for Notch-1 recognition. Front Microbiol 2022; 13:931307. [PMID: 35992693 PMCID: PMC9381698 DOI: 10.3389/fmicb.2022.931307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Synthetic antibodies have been engineered against a wide variety of antigens with desirable biophysical, biochemical, and pharmacological properties. Here, we describe the generation and characterization of synthetic antigen-binding fragments (Fabs) against Notch-1. Three single-framework synthetic Fab libraries, named S, F, and modified-F, were screened against the recombinant human Notch-1 extracellular domain using phage display. These libraries were built on a modified trastuzumab framework, containing two or four diversified complementarity-determining regions (CDRs) and different CDR diversity designs. In total, 12 Notch-1 Fabs were generated with 10 different CDRH3 lengths. These Fabs possessed a high affinity for Notch-1 (sub-nM to mid-nM KDapp values) and exhibited different binding profiles (mono-, bi-or tri-specific) toward Notch/Jagged receptors. Importantly, we showed that screening focused diversity libraries, implementing next-generation sequencing approaches, and fine-tuning the CDR length diversity provided improved binding solutions for Notch-1 recognition. These findings have implications for antibody library design and antibody phage display.
Collapse
Affiliation(s)
| | - Kris Barreto
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Daniel Hogan
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Anthony Kusalik
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Clarence Ronald Geyer
- Department of Pathology, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Clarence Ronald Geyer,
| |
Collapse
|
26
|
Labriola JM, Miersch S, Chen G, Chen C, Pavlenco A, Saberianfar R, Caccuri F, Zani A, Sharma N, Feng A, Leung DW, Caruso A, Novelli G, Amarasinghe GK, Sidhu SS. Peptide-Antibody Fusions Engineered by Phage Display Exhibit an Ultrapotent and Broad Neutralization of SARS-CoV-2 Variants. ACS Chem Biol 2022; 17:1978-1988. [PMID: 35731947 PMCID: PMC9236212 DOI: 10.1021/acschembio.2c00411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022]
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.
Collapse
Affiliation(s)
- Jonathan M. Labriola
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Shane Miersch
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Gang Chen
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Chao Chen
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Alevtina Pavlenco
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Reza Saberianfar
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and
Translational Medicine, University of Brescia Medical School,
25123 Brescia, Italy
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and
Translational Medicine, University of Brescia Medical School,
25123 Brescia, Italy
| | - Nitin Sharma
- Department of Pathology and Immunology,
Washington University School of Medicine in St Louis, St
Louis, Missouri 63110, United States
| | - Annie Feng
- Department of Pathology and Immunology,
Washington University School of Medicine in St Louis, St
Louis, Missouri 63110, United States
- Division of Infectious Diseases, John T. Milliken
Department of Medicine, Washington University School of
Medicine, St. Louis, Missouri 63110, United
States
| | - Daisy W. Leung
- Division of Infectious Diseases, John T. Milliken
Department of Medicine, Washington University School of
Medicine, St. Louis, Missouri 63110, United
States
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and
Translational Medicine, University of Brescia Medical School,
25123 Brescia, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention,
University of Rome Tor Vergata, Via Montpellier 1, 00133
Rome, Italy
- Italy 6 IRCCS Neuromed, Pozzilli
(IS) 86077, Italy
- Department of Pharmacology, School of Medicine,
University of Nevada, Reno, Nevada 89557, United
States
| | - Gaya K. Amarasinghe
- Department of Pathology and Immunology,
Washington University School of Medicine in St Louis, St
Louis, Missouri 63110, United States
| | - Sachdev S. Sidhu
- Department of Molecular Genetics, The
Donnelly Centre, University of Toronto, 160 College St., M5S 3E1 Toronto,
Ontario, Canada
| |
Collapse
|
27
|
Liang R, Zhang Y, Ma G, Wang S. Charge-Sensitive Optical Detection of Binding Kinetics between Phage-Displayed Peptide Ligands and Protein Targets. BIOSENSORS 2022; 12:394. [PMID: 35735542 PMCID: PMC9221260 DOI: 10.3390/bios12060394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
Phage display technology has been a powerful tool in peptide drug development. However, the supremacy of phage display-based peptide drug discovery is plagued by the follow-up process of peptides synthesis, which is costly and time consuming, but is necessary for the accurate measurement of binding kinetics in order to properly triage the best peptide leads during the affinity maturation stages. A sensitive technology is needed for directly measuring the binding kinetics of peptides on phages to reduce the time and cost of the entire process. Here, we show the capability of a charge-sensitive optical detection (CSOD) method for the direct quantification of binding kinetics of phage-displayed peptides to their target protein, using whole phages. We anticipate CSOD will contribute to streamline the process of phage display-based drug discovery.
Collapse
Affiliation(s)
- Runli Liang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ 85287, USA; (R.L.); (G.M.)
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA;
| | - Guangzhong Ma
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ 85287, USA; (R.L.); (G.M.)
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ 85287, USA; (R.L.); (G.M.)
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
28
|
Castillo F, Corbi-Verge C, Murciano-Calles J, Candel AM, Han Z, Iglesias-Bexiga M, Ruiz-Sanz J, Kim PM, Harty RN, Martinez JC, Luque I. Phage display identification of nanomolar ligands for human NEDD4-WW3: Energetic and dynamic implications for the development of broad-spectrum antivirals. Int J Biol Macromol 2022; 207:308-323. [PMID: 35257734 DOI: 10.1016/j.ijbiomac.2022.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022]
Abstract
The recognition of PPxY viral Late domains by the third WW domain of the human HECT-E3 ubiquitin ligase NEDD4 (NEDD4-WW3) is essential for the budding of many viruses. Blocking these interactions is a promising strategy to develop broad-spectrum antivirals. As all WW domains, NEDD4-WW3 is a challenging therapeutic target due to the low binding affinity of its natural interactions, its high conformational plasticity, and its complex thermodynamic behavior. In this work, we set out to investigate whether high affinity can be achieved for monovalent ligands binding to the isolated NEDD4-WW3 domain. We show that a competitive phage-display set-up allows for the identification of high-affinity peptides showing inhibitory activity of viral budding. A detailed biophysical study combining calorimetry, nuclear magnetic resonance, and molecular dynamic simulations reveals that the improvement in binding affinity does not arise from the establishment of new interactions with the domain, but is associated to conformational restrictions imposed by a novel C-terminal -LFP motif in the ligand, unprecedented in the PPxY interactome. These results, which highlight the complexity of WW domain interactions, provide valuable insight into the key elements for high binding affinity, of interest to guide virtual screening campaigns for the identification of novel therapeutics targeting NEDD4-WW3 interactions.
Collapse
Affiliation(s)
- Francisco Castillo
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Carles Corbi-Verge
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain; Donnelly Centre for Cellular and Biomolecular Research, Department of Molecular Genetics & Department of Computer Science, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Javier Murciano-Calles
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Adela M Candel
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Ziying Han
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce St., Philadelphia, PA 19104, USA
| | - Manuel Iglesias-Bexiga
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Javier Ruiz-Sanz
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Philip M Kim
- Donnelly Centre for Cellular and Biomolecular Research, Department of Molecular Genetics & Department of Computer Science, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce St., Philadelphia, PA 19104, USA
| | - Jose C Martinez
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain
| | - Irene Luque
- Department of Physical Chemistry, Institute of Biotechnology and Excelence Unit in Chemistry Applied to Biomedicine and Environment, School of Sciences, University of Granada, Campus Fuentenueva s/n 18071, Granada, Spain.
| |
Collapse
|
29
|
Ma C, Jiang C, Zhao D, Li S, Li R, Li L. Development in Detection Methods for the Expression of Surface-Displayed Proteins. Front Microbiol 2022; 13:899578. [PMID: 35558116 PMCID: PMC9085562 DOI: 10.3389/fmicb.2022.899578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Directed evolution is a widely-used engineering strategy for improving the stabilities or biochemical functions of proteins by repeated rounds of mutation and selection. A protein of interest is selected as the template and expressed on a molecular display platform such as a bacteriophage for engineering. Initially, the surface-displayed protein template needs to be checked against the desired target via ELISA to examine whether the functions of the displayed template remain intact. The ELISA signal is subject to the protein-target binding affinity. A low-affinity results in a weak ELISA signal which makes it difficult to determine whether the weak signal is because of low affinity or because of poor expression of the protein. Using a methyllysine-binding chromodomain protein Cbx1 that weakly binds to the histone H3K9me3 peptide, we developed and compared three different approaches to increase the signal-to-background ratio of ELISA measurements. We observed that the specific peptide-binding signal was enhanced by increasing the Cbx1 phage concentration on the ELISA plate. The introduction of previously known gain-of-function mutations to the Cbx1 protein significantly increased the ELISA signals. Moreover, we demonstrated that the H3K9me3-specific binding signal was enhanced by fusing Cbx1 with a high-affinity phosphotyrosine-binding protein and by coating the ELISA plate with a mixture of H3K9me3 and phosphotyrosine peptides. This approach also worked with binding to a lower affinity momomethyllysine peptide H3K9me1. These approaches may help improve ELISA experiments when dealing with low-affinity ligand-protein interactions.
Collapse
Affiliation(s)
- Chenglong Ma
- College of Life Sciences, Qingdao University, Qingdao, China.,School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Chunyang Jiang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Dongping Zhao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shuhao Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Ronggui Li
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Lei Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
30
|
Frick R, Høydahl LS, Hodnebrug I, Vik ES, Dalhus B, Sollid LM, Gray JJ, Sandlie I, Løset GÅ. Affinity maturation of TCR-like antibodies using phage display guided by structural modeling. Protein Eng Des Sel 2022; 35:gzac005. [PMID: 35871543 PMCID: PMC9536190 DOI: 10.1093/protein/gzac005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 12/01/2022] Open
Abstract
TCR-like antibodies represent a unique type of engineered antibodies with specificity toward pHLA, a ligand normally restricted to the sensitive recognition by T cells. Here, we report a phage display-based sequential development path of such antibodies. The strategy goes from initial lead identification through in silico informed CDR engineering in combination with framework engineering for affinity and thermostability optimization, respectively. The strategy allowed the identification of HLA-DQ2.5 gluten peptide-specific TCR-like antibodies with low picomolar affinity. Our method outlines an efficient and general method for development of this promising class of antibodies, which should facilitate their utility including translation to human therapy.
Collapse
Affiliation(s)
- Rahel Frick
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Lene S Høydahl
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Ina Hodnebrug
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Erik S Vik
- Nextera AS, Gaustadalléen 21, 0349 Oslo, Norway
| | - Bjørn Dalhus
- Department for Medical Biochemistry, Institute for Clinical Medicine, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
- Department for Microbiology, Clinic for Laboratory Medicine, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Ludvig M Sollid
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Jeffrey J Gray
- Program in Molecular Biophysics, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering and Institute of NanoBioTechnology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Inger Sandlie
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Geir Åge Løset
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
- Nextera AS, Gaustadalléen 21, 0349 Oslo, Norway
| |
Collapse
|
31
|
Martinez JC, Castillo F, Ruiz-Sanz J, Murciano-Calles J, Camara-Artigas A, Luque I. Understanding binding affinity and specificity of modular protein domains: A focus in ligand design for the polyproline-binding families. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:161-188. [PMID: 35534107 DOI: 10.1016/bs.apcsb.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Within the modular protein domains there are five families that recognize proline-rich sequences: SH3, WW, EVH1, GYF and UEV domains. This chapter reviews the main strategies developed for the design of ligands for these families, including peptides, peptidomimetics and drugs. We also describe some studies aimed to understand the molecular reasons responsible for the intrinsic affinity and specificity of these domains.
Collapse
Affiliation(s)
- Jose C Martinez
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain.
| | - Francisco Castillo
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Javier Ruiz-Sanz
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Javier Murciano-Calles
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Ana Camara-Artigas
- Departamento de Química Física, Universidad de Almería, Campus de Excelencia Internacional Agroalimentario ceiA3 y CIAMBITAL, Almeria, Spain
| | - Irene Luque
- Departamento de Química Física e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| |
Collapse
|
32
|
Klaus M, Rossini E, Linden A, Paithankar KS, Zeug M, Ignatova Z, Urlaub H, Khosla C, Köfinger J, Hummer G, Grininger M. Solution Structure and Conformational Flexibility of a Polyketide Synthase Module. JACS AU 2021; 1:2162-2171. [PMID: 34977887 PMCID: PMC8717363 DOI: 10.1021/jacsau.1c00043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 05/28/2023]
Abstract
Polyketide synthases (PKSs) are versatile C-C bond-forming enzymes that are broadly distributed in bacteria and fungi. The polyketide compound family includes many clinically useful drugs such as the antibiotic erythromycin, the antineoplastic epothilone, and the cholesterol-lowering lovastatin. Harnessing PKSs for custom compound synthesis remains an open challenge, largely because of the lack of knowledge about key structural properties. Particularly, the domains-well characterized on their own-are poorly understood in their arrangement, conformational dynamics, and interplay in the intricate quaternary structure of modular PKSs. Here, we characterize module 2 from the 6-deoxyerythronolide B synthase by small-angle X-ray scattering and cross-linking mass spectrometry with coarse-grained structural modeling. The results of this hybrid approach shed light on the solution structure of a cis-AT type PKS module as well as its inherent conformational dynamics. Supported by a directed evolution approach, we also find that acyl carrier protein (ACP)-mediated substrate shuttling appears to be steered by a nonspecific electrostatic interaction network.
Collapse
Affiliation(s)
- Maja Klaus
- Institute
of Organic Chemistry and Chemical Biology, Buchmann Institute for
Molecular Life Sciences, Goethe University
Frankfurt, Max-von-Laue Strasse 15, Frankfurt am Main 60438, Germany
| | - Emanuele Rossini
- Department
of Theoretical Biophysics, Max Planck Institute
of Biophysics, Max-von-Laue
Strasse 3, Frankfurt am Main 60438, Germany
| | - Andreas Linden
- Max
Planck Institute for Biophysical Chemistry, Am Fassberg 11, Goettingen 37077, Germany
- Institute
for Clinical Chemistry, University Medical
Center Göttingen, Robert Koch Strasse 40, Goettingen 37075, Germany
| | - Karthik S. Paithankar
- Institute
of Organic Chemistry and Chemical Biology, Buchmann Institute for
Molecular Life Sciences, Goethe University
Frankfurt, Max-von-Laue Strasse 15, Frankfurt am Main 60438, Germany
| | - Matthias Zeug
- Institute
of Organic Chemistry and Chemical Biology, Buchmann Institute for
Molecular Life Sciences, Goethe University
Frankfurt, Max-von-Laue Strasse 15, Frankfurt am Main 60438, Germany
| | - Zoya Ignatova
- Institute
for Biochemistry and Molecular Biology, University of Hamburg, Notkestrasse 85, Hamburg 22607, Germany
| | - Henning Urlaub
- Max
Planck Institute for Biophysical Chemistry, Am Fassberg 11, Goettingen 37077, Germany
- Institute
for Clinical Chemistry, University Medical
Center Göttingen, Robert Koch Strasse 40, Goettingen 37075, Germany
| | - Chaitan Khosla
- Department
of Chemistry, Stanford ChEM-H, Department of Chemical Engineering Stanford University, Stanford, California 94305, United States
| | - Jürgen Köfinger
- Department
of Theoretical Biophysics, Max Planck Institute
of Biophysics, Max-von-Laue
Strasse 3, Frankfurt am Main 60438, Germany
| | - Gerhard Hummer
- Department
of Theoretical Biophysics, Max Planck Institute
of Biophysics, Max-von-Laue
Strasse 3, Frankfurt am Main 60438, Germany
- Institute
of Biophysics, Goethe University Frankfurt, Max-von-Laue Strasse 1, Frankfurt am Main 60438, Germany
| | - Martin Grininger
- Institute
of Organic Chemistry and Chemical Biology, Buchmann Institute for
Molecular Life Sciences, Goethe University
Frankfurt, Max-von-Laue Strasse 15, Frankfurt am Main 60438, Germany
| |
Collapse
|
33
|
Ch'ng ACW, Lam P, Alassiri M, Lim TS. Application of phage display for T-cell receptor discovery. Biotechnol Adv 2021; 54:107870. [PMID: 34801662 DOI: 10.1016/j.biotechadv.2021.107870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022]
Abstract
The immune system is tasked to keep our body unharmed and healthy. In the immune system, B- and T-lymphocytes are the two main components working together to stop and eliminate invading threats like virus particles, bacteria, fungi and parasite from attacking our healthy cells. The function of antibodies is relatively more direct in target recognition as compared to T-cell receptors (TCR) which recognizes antigenic peptides being presented on the major histocompatibility complex (MHC). Although phage display has been widely applied for antibody presentation, this is the opposite in the case of TCR. The cell surface TCR is a relatively large and complex molecule, making presentation on phage surfaces challenging. Even so, recombinant versions and modifications have been introduced to allow the growing development of TCR in phage display. In addition, the increasing application of TCR for immunotherapy has made it an important binding motif to be developed by phage display. This review will emphasize on the application of phage display for TCR discovery as well as the engineering aspect of TCR for improved characteristics.
Collapse
Affiliation(s)
- Angela Chiew Wen Ch'ng
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Paula Lam
- CellVec Private Limited, 118518, Singapore; National University of Singapore, Department of Physiology, 117597, Singapore; Duke-NUS Graduate Medical School, Cancer and Stem Cells Biology Program, 169857, Singapore
| | - Mohammed Alassiri
- Department of Basic Sciences, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
34
|
Chrustowicz J, Sherpa D, Teyra J, Loke MS, Popowicz GM, Basquin J, Sattler M, Prabu JR, Sidhu SS, Schulman BA. Multifaceted N-Degron Recognition and Ubiquitylation by GID/CTLH E3 Ligases. J Mol Biol 2021; 434:167347. [PMID: 34767800 DOI: 10.1016/j.jmb.2021.167347] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
N-degron E3 ubiquitin ligases recognize specific residues at the N-termini of substrates. Although molecular details of N-degron recognition are known for several E3 ligases, the range of N-terminal motifs that can bind a given E3 substrate binding domain remains unclear. Here, we discovered capacity of Gid4 and Gid10 substrate receptor subunits of yeast "GID"/human "CTLH" multiprotein E3 ligases to tightly bind a wide range of N-terminal residues whose recognition is determined in part by the downstream sequence context. Screening of phage displaying peptide libraries with exposed N-termini identified novel consensus motifs with non-Pro N-terminal residues binding Gid4 or Gid10 with high affinity. Structural data reveal that conformations of flexible loops in Gid4 and Gid10 complement sequences and folds of interacting peptides. Together with analysis of endogenous substrate degrons, the data show that degron identity, substrate domains harboring targeted lysines, and varying E3 ligase higher-order assemblies combinatorially determine efficiency of ubiquitylation and degradation.
Collapse
Affiliation(s)
- Jakub Chrustowicz
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. https://twitter.com/chrustowicz_j
| | - Dawafuti Sherpa
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. https://twitter.com/dawafutisherpa
| | - Joan Teyra
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Mun Siong Loke
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Grzegorz M Popowicz
- Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany; Bavarian NMR Center, Department of Chemistry, Technical University of Munich, Germany
| | - Jerome Basquin
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Michael Sattler
- Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany; Bavarian NMR Center, Department of Chemistry, Technical University of Munich, Germany
| | - J Rajan Prabu
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. https://twitter.com/rajanprabu
| | - Sachdev S Sidhu
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany.
| |
Collapse
|
35
|
Lam KN, Spanogiannopoulos P, Soto-Perez P, Alexander M, Nalley MJ, Bisanz JE, Nayak RR, Weakley AM, Yu FB, Turnbaugh PJ. Phage-delivered CRISPR-Cas9 for strain-specific depletion and genomic deletions in the gut microbiome. Cell Rep 2021; 37:109930. [PMID: 34731631 PMCID: PMC8591988 DOI: 10.1016/j.celrep.2021.109930] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/13/2021] [Accepted: 10/12/2021] [Indexed: 01/04/2023] Open
Abstract
Mechanistic insights into the role of the human microbiome in the predisposition to and treatment of disease are limited by the lack of methods to precisely add or remove microbial strains or genes from complex communities. Here, we demonstrate that engineered bacteriophage M13 can be used to deliver DNA to Escherichia coli within the mouse gastrointestinal (GI) tract. Delivery of a programmable exogenous CRISPR-Cas9 system enables the strain-specific depletion of fluorescently marked isogenic strains during competitive colonization and genomic deletions that encompass the target gene in mice colonized with a single strain. Multiple mechanisms allow E. coli to escape targeting, including loss of the CRISPR array or even the entire CRISPR-Cas9 system. These results provide a robust and experimentally tractable platform for microbiome editing, a foundation for the refinement of this approach to increase targeting efficiency, and a proof of concept for the extension to other phage-bacterial pairs of interest.
Collapse
Affiliation(s)
- Kathy N Lam
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peter Spanogiannopoulos
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paola Soto-Perez
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret Alexander
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew J Nalley
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jordan E Bisanz
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Renuka R Nayak
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Allison M Weakley
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA; Stanford ChEM-H: Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA 94305, USA
| | - Feiqiao B Yu
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Peter J Turnbaugh
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
36
|
Domain Analysis and Motif Matcher (DAMM): A Program to Predict Selectivity Determinants in Monosiga brevicollis PDZ Domains Using Human PDZ Data. Molecules 2021; 26:molecules26196034. [PMID: 34641578 PMCID: PMC8512817 DOI: 10.3390/molecules26196034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022] Open
Abstract
Choanoflagellates are single-celled eukaryotes with complex signaling pathways. They are considered the closest non-metazoan ancestors to mammals and other metazoans and form multicellular-like states called rosettes. The choanoflagellate Monosiga brevicollis contains over 150 PDZ domains, an important peptide-binding domain in all three domains of life (Archaea, Bacteria, and Eukarya). Therefore, an understanding of PDZ domain signaling pathways in choanoflagellates may provide insight into the origins of multicellularity. PDZ domains recognize the C-terminus of target proteins and regulate signaling and trafficking pathways, as well as cellular adhesion. Here, we developed a computational software suite, Domain Analysis and Motif Matcher (DAMM), that analyzes peptide-binding cleft sequence identity as compared with human PDZ domains and that can be used in combination with literature searches of known human PDZ-interacting sequences to predict target specificity in choanoflagellate PDZ domains. We used this program, protein biochemistry, fluorescence polarization, and structural analyses to characterize the specificity of A9UPE9_MONBE, a M. brevicollis PDZ domain-containing protein with no homology to any metazoan protein, finding that its PDZ domain is most similar to those of the DLG family. We then identified two endogenous sequences that bind A9UPE9 PDZ with <100 μM affinity, a value commonly considered the threshold for cellular PDZ-peptide interactions. Taken together, this approach can be used to predict cellular targets of previously uncharacterized PDZ domains in choanoflagellates and other organisms. Our data contribute to investigations into choanoflagellate signaling and how it informs metazoan evolution.
Collapse
|
37
|
Guo Y, Liu Q, Mallette E, Caba C, Hou F, Fux J, LaPlante G, Dong A, Zhang Q, Zheng H, Tong Y, Zhang W. Structural and functional characterization of ubiquitin variant inhibitors for the JAMM-family deubiquitinases STAMBP and STAMBPL1. J Biol Chem 2021; 297:101107. [PMID: 34425109 PMCID: PMC8449267 DOI: 10.1016/j.jbc.2021.101107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 01/23/2023] Open
Abstract
Ubiquitination is a crucial posttranslational protein modification involved in a myriad of biological pathways. This modification is reversed by deubiquitinases (DUBs) that deconjugate the single ubiquitin (Ub) moiety or poly-Ub chains from substrates. In the past decade, tremendous efforts have been focused on targeting DUBs for drug discovery. However, most chemical compounds with inhibitory activity for DUBs suffer from mild potency and low selectivity. To overcome these obstacles, we developed a phage display-based protein engineering strategy for generating Ub variant (UbV) inhibitors, which was previously successfully applied to the Ub-specific protease (USP) family of cysteine proteases. In this work, we leveraged the UbV platform to selectively target STAMBP, a member of the JAB1/MPN/MOV34 (JAMM) metalloprotease family of DUB enzymes. We identified two UbVs (UbVSP.1 and UbVSP.3) that bind to STAMBP with high affinity but differ in their selectivity for the closely related paralog STAMBPL1. We determined the STAMBPL1-UbVSP.1 complex structure by X-ray crystallography, revealing hotspots of the JAMM-UbV interaction. Finally, we show that UbVSP.1 and UbVSP.3 are potent inhibitors of STAMBP isopeptidase activity, far exceeding the reported small-molecule inhibitor BC-1471. This work demonstrates that UbV technology is suitable to develop molecules as tools to target metalloproteases, which can be used to further understand the cellular function of JAMM family DUBs.
Collapse
Affiliation(s)
- Yusong Guo
- Fisheries College, Guangdong Ocean University, Guangdong, China; Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Qi Liu
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Canada
| | - Evan Mallette
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Canada
| | - Cody Caba
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Canada
| | - Feng Hou
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Julia Fux
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Canada
| | - Gabriel LaPlante
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Canada
| | - Aiping Dong
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Qi Zhang
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Hui Zheng
- Jiangsu Key Laboratory of Infection and Immunity, International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yufeng Tong
- Structural Genomics Consortium, University of Toronto, Toronto, Canada; Department of Chemistry and Biochemistry, University of Windsor, Windsor, Canada.
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Canada; CIFAR Azrieli Global Scholars Program, Canadian Institute for Advanced Research, Toronto, Canada.
| |
Collapse
|
38
|
Short Read-Length Next Generation DNA Sequencing of Antibody CDR Combinations from Phage Selection Outputs. Methods Mol Biol 2021. [PMID: 34478134 DOI: 10.1007/978-1-0716-1450-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Phage display is commonly used to select target-binding antibody fragments from large libraries containing billions of unique antibody clones. In practice, selection outputs are often highly heterogenous, making it desirable to recover sequence information from the selected pool. Next Generation DNA Sequencing (NGS) enables the acquisition of sufficient sequencing reads to cover the pool diversity, however read-lengths are typically too short to capture paired antibody complementarity-determining regions (CDRs), which is needed to reconstruct target-binding antibody fragments. Here, we describe a simple in vitro protocol to bring the DNA encoding the antibody CDRs closer together. The final PCR product referred to as a "CDR strip" is suitable for short read-length NGS. In this method, phagemid ssDNA is recovered from antibody phage display biopanning and used as a template to create a heteroduplex with deletions between CDRs of interest. The shorter strand in the heteroduplex is preferentially PCR amplified to generate a CDR strip that is sequenced using NGS. We have also included a bioinformatics approach to analyze the CDR strip populations so that single antibody clones can be created from paired CDR sequences.
Collapse
|
39
|
Frick R, Høydahl LS, Petersen J, du Pré MF, Kumari S, Berntsen G, Dewan AE, Jeliazkov JR, Gunnarsen KS, Frigstad T, Vik ES, Llerena C, Lundin KEA, Yaqub S, Jahnsen J, Gray JJ, Rossjohn J, Sollid LM, Sandlie I, Løset GÅ. A high-affinity human TCR-like antibody detects celiac disease gluten peptide-MHC complexes and inhibits T cell activation. Sci Immunol 2021; 6:6/62/eabg4925. [PMID: 34417258 DOI: 10.1126/sciimmunol.abg4925] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
Antibodies specific for peptides bound to human leukocyte antigen (HLA) molecules are valuable tools for studies of antigen presentation and may have therapeutic potential. Here, we generated human T cell receptor (TCR)-like antibodies toward the immunodominant signature gluten epitope DQ2.5-glia-α2 in celiac disease (CeD). Phage display selection combined with secondary targeted engineering was used to obtain highly specific antibodies with picomolar affinity. The crystal structure of a Fab fragment of the lead antibody 3.C11 in complex with HLA-DQ2.5:DQ2.5-glia-α2 revealed a binding geometry and interaction mode highly similar to prototypic TCRs specific for the same complex. Assessment of CeD biopsy material confirmed disease specificity and reinforced the notion that abundant plasma cells present antigen in the inflamed CeD gut. Furthermore, 3.C11 specifically inhibited activation and proliferation of gluten-specific CD4+ T cells in vitro and in HLA-DQ2.5 humanized mice, suggesting a potential for targeted intervention without compromising systemic immunity.
Collapse
Affiliation(s)
- Rahel Frick
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Centre for Immune Regulation and Department of Biosciences, University of Oslo, Oslo, Norway
| | - Lene S Høydahl
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Centre for Immune Regulation and Department of Biosciences, University of Oslo, Oslo, Norway.,KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Jan Petersen
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - M Fleur du Pré
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | | | | | - Alisa E Dewan
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | | | - Kristin S Gunnarsen
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Centre for Immune Regulation and Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | - Carmen Llerena
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Knut E A Lundin
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway.,Department of Gastroenterology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Sheraz Yaqub
- Department of Gastrointestinal Surgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Jeffrey J Gray
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering and Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Ludvig M Sollid
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Centre for Immune Regulation and Department of Biosciences, University of Oslo, Oslo, Norway
| | - Geir Åge Løset
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway. .,Centre for Immune Regulation and Department of Biosciences, University of Oslo, Oslo, Norway.,Nextera AS, Oslo, Norway
| |
Collapse
|
40
|
Positive charge in the complementarity-determining regions of synthetic nanobody prevents aggregation. Biochem Biophys Res Commun 2021; 572:1-6. [PMID: 34332323 DOI: 10.1016/j.bbrc.2021.07.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 01/19/2023]
Abstract
In the past, specificity and affinity were the priority for synthetic antibody library. However, therapeutic antibodies need good stability for medical use. Through carefully adjust the chemical diversity in CDRs, one hopes to design a synthetic antibody library with good developability. Here we thoroughly analyzed 296 nanobody sequences and structures, constructed a fully-functional synthetic nanobody library, evaluated the relationship between aggregation and isoelectric point, and found that high-pI nanobodies were more resistant to aggregation than low-pIs. As we used the same framework for constructing the library, CDRs charge played a crucial role in mediating nanobody aggregation. We also analyzed the theoretical pI of 296 nanobodies from PDB, about 75% had basic pI, only 25% were acidic. Those results provided useful guidelines for designing next-generation synthetic nanobody libraries and for identifying potent and safe nanobody therapeutics.
Collapse
|
41
|
Comprehensive Assessment of the Relationship Between Site -2 Specificity and Helix α2 in the Erbin PDZ Domain. J Mol Biol 2021; 433:167115. [PMID: 34171344 DOI: 10.1016/j.jmb.2021.167115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/27/2021] [Accepted: 06/16/2021] [Indexed: 11/20/2022]
Abstract
PDZ domains are key players in signalling pathways. These modular domains generally recognize short linear C-terminal stretches of sequences in proteins that organize the formation of complex multi-component assemblies. The development of new methodologies for the characterization of the molecular principles governing these interactions is critical to fully understand the functional diversity of the family and to elucidate biological functions for family members. Here, we applied an in vitro evolution strategy to explore comprehensively the capacity of PDZ domains for specific recognition of different amino acids at a key position in C-terminal peptide ligands. We constructed a phage-displayed library of the Erbin PDZ domain by randomizing the binding site-2 and adjacent residues, which are all contained in helix α2, and we selected for variants binding to a panel of peptides representing all possible position-2 residues. This approach generated insights into the basis for the common natural class I and II specificities, demonstrated an alternative basis for a rare natural class III specificity for Asp-2, and revealed a novel specificity for Arg-2 that has not been reported in natural PDZ domains. A structure of a PDZ-peptide complex explained the minimum requirement for switching specificity from class I ligands containing Thr/Ser-2 to class II ligands containing hydrophobic residues at position-2. A second structure explained the molecular basis for the specificity for ligands containing Arg-2. Overall, the evolved PDZ variants greatly expand our understanding of site-2 specificities and the variants themselves may prove useful as building blocks for synthetic biology.
Collapse
|
42
|
Discovery of a caspase cleavage motif antibody reveals insights into noncanonical inflammasome function. Proc Natl Acad Sci U S A 2021; 118:2018024118. [PMID: 33723046 DOI: 10.1073/pnas.2018024118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammasomes sense a number of pathogen and host damage signals to initiate a signaling cascade that triggers inflammatory cell death, termed pyroptosis. The inflammatory caspases (1/4/5/11) are the key effectors of this process through cleavage and activation of the pore-forming protein gasdermin D. Caspase-1 also activates proinflammatory interleukins, IL-1β and IL-18, via proteolysis. However, compared to the well-studied apoptotic caspases, the identity of substrates and therefore biological functions of the inflammatory caspases remain limited. Here, we construct, validate, and apply an antibody toolset for direct detection of neo-C termini generated by inflammatory caspase proteolysis. By combining rabbit immune phage display with a set of degenerate and defined target peptides, we discovered two monoclonal antibodies that bind peptides with a similar degenerate recognition motif as the inflammatory caspases without recognizing the canonical apoptotic caspase recognition motif. Crystal structure analyses revealed the molecular basis of this strong yet paradoxical degenerate mode of peptide recognition. One antibody selectively immunoprecipitated cleaved forms of known and unknown inflammatory caspase substrates, allowing the identification of over 300 putative substrates of the caspase-4 noncanonical inflammasome, including caspase-7. This dataset will provide a path toward developing blood-based biomarkers of inflammasome activation. Overall, our study establishes tools to discover and detect inflammatory caspase substrates and functions, provides a workflow for designing antibody reagents to study cell signaling, and extends the growing evidence of biological cross talk between the apoptotic and inflammatory caspases.
Collapse
|
43
|
Liang CT, Roscow OMA, Zhang W. Recent developments in engineering protein-protein interactions using phage display. Protein Eng Des Sel 2021; 34:6297171. [PMID: 34117768 DOI: 10.1093/protein/gzab014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/09/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
Targeted inhibition of misregulated protein-protein interactions (PPIs) has been a promising area of investigation in drug discovery and development for human diseases. However, many constraints remain, including shallow binding surfaces and dynamic conformation changes upon interaction. A particularly challenging aspect is the undesirable off-target effects caused by inherent structural similarity among the protein families. To tackle this problem, phage display has been used to engineer PPIs for high-specificity binders with improved binding affinity and greatly reduced undesirable interactions with closely related proteins. Although general steps of phage display are standardized, library design is highly variable depending on experimental contexts. Here in this review, we examined recent advances in the structure-based combinatorial library design and the advantages and limitations of different approaches. The strategies described here can be explored for other protein-protein interactions and aid in designing new libraries or improving on previous libraries.
Collapse
Affiliation(s)
- Chen T Liang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G2W1, Canada
| | - Olivia M A Roscow
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G2W1, Canada
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G2W1, Canada.,CIFAR Azrieli Global Scholars Program, Canadian Institute for Advanced Research, MaRS Centre West Tower, 661 University Avenue, Toronto, Ontario M5G1M1, Canada
| |
Collapse
|
44
|
Tombling BJ, Lammi C, Lawrence N, Gilding EK, Grazioso G, Craik DJ, Wang CK. Bioactive Cyclization Optimizes the Affinity of a Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Peptide Inhibitor. J Med Chem 2020; 64:2523-2533. [PMID: 33356222 DOI: 10.1021/acs.jmedchem.0c01766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Peptides are regarded as promising next-generation therapeutics. However, an analysis of over 1000 bioactive peptide candidates suggests that many have underdeveloped affinities and could benefit from cyclization using a bridging linker sequence. Until now, the primary focus has been on the use of inert peptide linkers. Here, we show that affinity can be significantly improved by enriching the linker with functional amino acids. We engineered a peptide inhibitor of PCSK9, a target for clinical management of hypercholesterolemia, to demonstrate this concept. Cyclization linker optimization from library screening produced a cyclic peptide with ∼100-fold improved activity over the parent peptide and efficiently restored low-density lipoprotein (LDL) receptor levels and cleared extracellular LDL. The linker forms favorable interactions with PCSK9 as evidenced by thermodynamics, structure-activity relationship (SAR), NMR, and molecular dynamics (MD) studies. This PCSK9 inhibitor is one of many peptides that could benefit from bioactive cyclization, a strategy that is amenable to broad application in pharmaceutical design.
Collapse
Affiliation(s)
- Benjamin J Tombling
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Carmen Lammi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Nicole Lawrence
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Edward K Gilding
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Giovanni Grazioso
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Conan K Wang
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia
| |
Collapse
|
45
|
Dong Y, Meng F, Wang Z, Yu T, Chen A, Xu S, Wang J, Yin M, Tang L, Hu C, Wang H, Cai J. Construction and application of a human scFv phage display library based on Cre‑LoxP recombination for anti‑PCSK9 antibody selection. Int J Mol Med 2020; 47:708-718. [PMID: 33416098 PMCID: PMC7797424 DOI: 10.3892/ijmm.2020.4822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/10/2020] [Indexed: 11/08/2022] Open
Abstract
A large human natural single-chain fragment variable (scFv) phage library was constructed based on Cre-LoxP recombination, and used to successfully identify antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9). The library was derived from 400 blood samples, 30 bone marrow samples, and 10 cord blood samples from healthy donors. Lymphocytes were isolated from each sample and cDNA was synthesized using reverse transcription-quantitative PCR. Two-step overlap PCR was then used for scFv synthesis using a LoxP peptide as the linker. The scFv gene was inserted into the phagemid vector pDF by enzymatic digestion and ligation, and then transformed into Escherichia coli (E. coli) SS320 to establish a primary antibody library in the form of scFvs. A primary antibody library consisting of 5×107 peripheral blood and umbilical cord blood sources, as well as a primary antibody library of 5×107 bone marrow samples were obtained. By optimizing the recombination conditions, the primary phage library was used to infect E. coliBS1365 strain (which expresses the Cre enzyme), and a human scFv recombinant library with a size of 1×1011 was obtained through Cre-LoxP enzyme-mediated heavy and light chain replacement and recombination. This constructed recombinant library was employed to screen for antibodies against recombinant PCSK9. After four rounds of selection, a fully human antibody (3D2) was identified with a binding affinity of 1.96±1.56ⅹ10−10 M towards PCSK9. In vitro, the PCSK9/low-density lipoprotein receptor (LDLR) pathway of Hep-G2 cells was inhibited by 3D2 treatment, thereby increasing LDL uptake in these cells. In addition, combination treatment with 3D2 and statin was more effective at increasing LDLR levels than treatment with 3D2 or statin alone. Furthermore, 3D2 resulted in a 3-fold increase in hepatic LDLR levels, and lowered total serum cholesterol by up to 61.5% in vivo. Taken together, these results suggest that the constructed human Cre-LoxP scFv phage display library can be used to screen fully human scFv, and that 3D2 may serve as a candidate hypolipidemic therapy.
Collapse
Affiliation(s)
- Yuan Dong
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - Fanwei Meng
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - Zhiheng Wang
- Clinical Laboratory, 944th Hospital of People's Liberation Army, Jiuquan, Gansu 735000, P.R. China
| | - Tianyi Yu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - An Chen
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, P.R. China
| | - Song Xu
- Research and Development Department, Chongqing Biomean Technology Co., Ltd., Chongqing 400715, P.R. China
| | - Jianming Wang
- Research and Development Department, Chongqing Biomean Technology Co., Ltd., Chongqing 400715, P.R. China
| | - Moli Yin
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - Lu Tang
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - Chuanmin Hu
- Research and Development Department, Chongqing Biomean Technology Co., Ltd., Chongqing 400715, P.R. China
| | - Huiyan Wang
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| | - Jianhui Cai
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, Jilin 132013, P.R. China
| |
Collapse
|
46
|
Teyra J, Kelil A, Jain S, Helmy M, Jajodia R, Hooda Y, Gu J, D’Cruz AA, Nicholson SE, Min J, Sudol M, Kim PM, Bader GD, Sidhu SS. Large-scale survey and database of high affinity ligands for peptide recognition modules. Mol Syst Biol 2020; 16:e9310. [PMID: 33438817 PMCID: PMC7724964 DOI: 10.15252/msb.20199310] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
Many proteins involved in signal transduction contain peptide recognition modules (PRMs) that recognize short linear motifs (SLiMs) within their interaction partners. Here, we used large-scale peptide-phage display methods to derive optimal ligands for 163 unique PRMs representing 79 distinct structural families. We combined the new data with previous data that we collected for the large SH3, PDZ, and WW domain families to assemble a database containing 7,984 unique peptide ligands for 500 PRMs representing 82 structural families. For 74 PRMs, we acquired enough new data to map the specificity profiles in detail and derived position weight matrices and binding specificity logos based on multiple peptide ligands. These analyses showed that optimal peptide ligands resembled peptides observed in existing structures of PRM-ligand complexes, indicating that a large majority of the phage-derived peptides are likely to target natural peptide-binding sites and could thus act as inhibitors of natural protein-protein interactions. The complete dataset has been assembled in an online database (http://www.prm-db.org) that will enable many structural, functional, and biological studies of PRMs and SLiMs.
Collapse
Affiliation(s)
- Joan Teyra
- The Donnelly CentreUniversity of TorontoTorontoONCanada
| | | | - Shobhit Jain
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Computer ScienceUniversity of TorontoTorontoONCanada
| | - Mohamed Helmy
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Present address:
Singapore Institute of Food and Biotechnology Innovation (SIFBI)Agency for ScienceTechnology and Research (A*STAR)Singapore CitySingapore
| | - Raghav Jajodia
- Indian Institute of Engineering Science and TechnologyShibpurIndia
| | - Yogesh Hooda
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Present address:
MRC Laboratory of Molecular BiologyCambridgeUK
| | - Jun Gu
- The Donnelly CentreUniversity of TorontoTorontoONCanada
| | - Akshay A D’Cruz
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia
| | - Jinrong Min
- Structural Genomics ConsortiumUniversity of TorontoTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
| | - Marius Sudol
- Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Philip M Kim
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Computer ScienceUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Gary D Bader
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Computer ScienceUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Sachdev S Sidhu
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| |
Collapse
|
47
|
Deep profiling of protease substrate specificity enabled by dual random and scanned human proteome substrate phage libraries. Proc Natl Acad Sci U S A 2020; 117:25464-25475. [PMID: 32973096 DOI: 10.1073/pnas.2009279117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proteolysis is a major posttranslational regulator of biology inside and outside of cells. Broad identification of optimal cleavage sites and natural substrates of proteases is critical for drug discovery and to understand protease biology. Here, we present a method that employs two genetically encoded substrate phage display libraries coupled with next generation sequencing (SPD-NGS) that allows up to 10,000-fold deeper sequence coverage of the typical six- to eight-residue protease cleavage sites compared to state-of-the-art synthetic peptide libraries or proteomics. We applied SPD-NGS to two classes of proteases, the intracellular caspases, and the ectodomains of the sheddases, ADAMs 10 and 17. The first library (Lib 10AA) allowed us to identify 104 to 105 unique cleavage sites over a 1,000-fold dynamic range of NGS counts and produced consensus and optimal cleavage motifs based position-specific scoring matrices. A second SPD-NGS library (Lib hP), which displayed virtually the entire human proteome tiled in contiguous 49 amino acid sequences with 25 amino acid overlaps, enabled us to identify candidate human proteome sequences. We identified up to 104 natural linear cut sites, depending on the protease, and captured most of the examples previously identified by proteomics and predicted 10- to 100-fold more. Structural bioinformatics was used to facilitate the identification of candidate natural protein substrates. SPD-NGS is rapid, reproducible, simple to perform and analyze, inexpensive, and renewable, with unprecedented depth of coverage for substrate sequences, and is an important tool for protease biologists interested in protease specificity for specific assays and inhibitors and to facilitate identification of natural protein substrates.
Collapse
|
48
|
Hanna R, Cardarelli L, Patel N, Blazer LL, Adams JJ, Sidhu SS. A phage-displayed single-chain Fab library optimized for rapid production of single-chain IgGs. Protein Sci 2020; 29:2075-2084. [PMID: 32803886 DOI: 10.1002/pro.3931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/25/2022]
Abstract
Phage-displayed synthetic antibody (Ab) repertoires have become a major source of affinity reagents for basic and clinical research. Specific Abs identified from such libraries are often screened as fragments antigen binding (Fabs) produced in bacteria, and those with desired biochemical characteristics are reformatted for production as full-length immunoglobulin G (IgG) in mammalian cells. The conversion of Fabs to IgGs is a cumbersome and often rate-limiting step in the development of Abs. Moreover, biochemical properties required for lead IgG development are not always shared by the Fabs, and these issues are not uncovered until a significant effort has been spent on Abs that ultimately will not be useful. Thus, there is a need for simple and rapid techniques to convert phage-displayed Fabs to IgGs at an early stage of the Ab screening process. We report the generation of a highly diverse phage-displayed synthetic single-chain Fab (scFab) library, in which the light and heavy chains were tethered with an optimized linker. Following selection, pools of scFabs were converted to single-chain IgGs (scIgGs) en masse, enabling facile screening of hundreds of phage-derived scIgGs. We show that this approach can be used to rapidly screen for and select scIgGs that target cell-surface receptors, and scIgGs behave the same as conventional IgGs.
Collapse
Affiliation(s)
- Rachel Hanna
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Lia Cardarelli
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Nish Patel
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Levi L Blazer
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jarrett J Adams
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sachdev S Sidhu
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Kara E, Nielsen NV, Eggertsdottir B, Thiede B, Kanse SM, Løset GÅ. Design and Characterization of a New pVII Combinatorial Phage Display Peptide Library for Protease Substrate Mining Using Factor VII Activating Protease (FSAP) as Model. Chembiochem 2020; 21:1875-1884. [PMID: 32180321 PMCID: PMC7383712 DOI: 10.1002/cbic.201900705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/07/2020] [Indexed: 12/18/2022]
Abstract
We describe a novel, easy and efficient combinatorial phage display peptide substrate-mining method to map the substrate specificity of proteases. The peptide library is displayed on the pVII capsid of the M13 bacteriophage, which renders pIII necessary for infectivity and efficient retrieval, in an unmodified state. As capture module, the 3XFLAG was chosen due to its very high binding efficiency to anti-FLAG mAbs and its independency of any post-translational modification. This library was tested with Factor-VII activating protease (WT-FSAP) and its single-nucleotide polymorphism variant Marburg-I (MI)-FSAP. The WT-FSAP results confirmed the previously reported Arg/Lys centered FSAP cleavage site consensus as dominant, as well as reinforcing MI-FSAP as a loss-of-function mutant. Surprisingly, rare substrate clones devoid of basic amino acids were also identified. Indeed one of these peptides was cleaved as free peptide, thus suggesting a broader range of WT-FSAP substrates than previously anticipated.
Collapse
Affiliation(s)
- Emrah Kara
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | - Nis Valentin Nielsen
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | | | - Bernd Thiede
- Department of BiosciencesUniversity of Oslo0316OsloNorway
| | - Sandip M. Kanse
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | - Geir Åge Løset
- Department of BiosciencesUniversity of Oslo0316OsloNorway
- Nextera ASOsloNorway
| |
Collapse
|
50
|
Landgraf KE, Williams SR, Steiger D, Gebhart D, Lok S, Martin DW, Roybal KT, Kim KC. convertibleCARs: A chimeric antigen receptor system for flexible control of activity and antigen targeting. Commun Biol 2020; 3:296. [PMID: 32518350 PMCID: PMC7283332 DOI: 10.1038/s42003-020-1021-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
We have developed a chimeric antigen receptor (CAR) platform that functions as a modular system to address limitations of traditional CAR therapies. An inert form of the human NKG2D extracellular domain (iNKG2D) was engineered as the ectodomain of the CAR to generate convertibleCARTM-T cells. These cells were specifically directed to kill antigen-expressing target cells only in the presence of an activating bispecific adapter comprised of an iNKG2D-exclusive ULBP2-based ligand fused to an antigen-targeting antibody (MicAbodyTM). Efficacy against Raji tumors in NSG mice was dependent upon doses of both a rituximab-based MicAbody and convertibleCAR-T cells. We have also demonstrated that the exclusive ligand-receptor partnering enabled the targeted delivery of a mutant form of IL-2 to selectively promote the expansion of convertibleCAR-T cells in vitro and in vivo. By altering the Fv domains of the MicAbody or the payload fused to the orthogonal ligand, convertibleCAR-T cells can be readily targeted or regulated.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigen Presentation/immunology
- Apoptosis
- Cell Proliferation
- Female
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunotherapy, Adoptive/methods
- Interleukin-2/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mutation
- NK Cell Lectin-Like Receptor Subfamily K/genetics
- NK Cell Lectin-Like Receptor Subfamily K/immunology
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Sequence Homology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kyle E Landgraf
- Reflexion Pharmaceuticals, 937 Tahoe Blvd, Suite 150, Incline Village, NV, 89451, USA
| | - Steven R Williams
- Xyphos Biosciences, an Astellas Company, 100 Kimball Way, South San Francisco, CA, 94080, USA
| | - Daniel Steiger
- Freenome, 279 E Grand Ave 5th Floor, South San Francisco, CA, 94080, USA
| | - Dana Gebhart
- Xyphos Biosciences, an Astellas Company, 100 Kimball Way, South San Francisco, CA, 94080, USA
| | - Stephen Lok
- Zymergen, 5980 Horton St #105, Emeryville, CA, 94608, USA
| | - David W Martin
- Xyphos Biosciences, an Astellas Company, 100 Kimball Way, South San Francisco, CA, 94080, USA
| | - Kole T Roybal
- University of California, San Francisco, 513 Parnassus Avenue HSE-301, San Francisco, CA, 94143, USA
| | - Kaman Chan Kim
- Xyphos Biosciences, an Astellas Company, 100 Kimball Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|