Download PDF

Journal Of Biological Chemistry

Publication date: 2017-07-07
Volume: 292 Pages: 11452 - 11465
Publisher: American Society for Biochemistry and Molecular Biology

Author:

Schoonaert, Lies
Rue, Laura ; Roucourt, Bart ; Timmers, Mieke ; Little, Susan ; Chavez-Gutierrez, Lucia ; Dewilde, Maarten ; Joyce, Peter ; Curnock, Adam ; Weber, Peter ; Haustraete, Jurgen ; Hassanzadeh-Ghassabeh, Gholamreza ; De Strooper, Bart ; Van den Bosch, Ludo ; Van Damme, Philip ; Lemmens, Robin ; Robberecht, Wim

Keywords:

Science & Technology, Life Sciences & Biomedicine, Biochemistry & Molecular Biology, SINGLE-DOMAIN ANTIBODIES, BLOOD-BRAIN-BARRIER, CORTICOSPINAL TRACT AXONS, CYCLIC PEPTIDE ANTAGONIST, SPINAL-CORD-INJURY, SMALL MOLECULES, NERVOUS-SYSTEM, EPHRIN-BINDING, FUSION PROTEIN, REGENERATION, EphA4, VHH, inhibition, ligand-binding domain, phosphorylation, protein-protein interaction, receptor-tyrosine kinase, regeneration, single-domain antibody (sdAb,Nanobody), Animals, Antibody Affinity, Cell Line, Humans, Mice, Protein Domains, Receptor, EphA4, Single-Domain Antibodies, 03 Chemical Sciences, 06 Biological Sciences, 11 Medical and Health Sciences, 31 Biological sciences, 32 Biomedical and clinical sciences, 34 Chemical sciences

Abstract:

The ephrin receptor A4 (EphA4) is one of the receptors in the ephrin system that plays a pivotal role in a variety of cell-cell interactions, mostly studied during development. In addition, EphA4 has been found to play a role in cancer biology as well as in the pathogenesis of several neurological disorders such as stroke, spinal cord injury, multiple sclerosis, amyotrophic lateral sclerosis (ALS), and Alzheimer's disease. Pharmacological blocking of EphA4 has been suggested to be a therapeutic strategy for these disorders. Therefore, the aim of our study was to generate potent and selective Nanobodies against the ligand-binding domain of the human EphA4 receptor. We identified two Nanobodies, Nb 39 and Nb 53, that bind EphA4 with affinities in the nanomolar range. These Nanobodies were most selective for EphA4, with residual binding to EphA7 only. Using Alphascreen technology, we found that both Nanobodies displaced all known EphA4-binding ephrins from the receptor. Furthermore, Nb 39 and Nb 53 inhibited ephrin-induced phosphorylation of the EphA4 protein in a cell-based assay. Finally, in a cortical neuron primary culture, both Nanobodies were able to inhibit endogenous EphA4-mediated growth-cone collapse induced by ephrin-B3. Our results demonstrate the potential of Nanobodies to target the ligand-binding domain of EphA4. These Nanobodies may deserve further evaluation as potential therapeutics in disorders in which EphA4-mediated signaling plays a role.