Abstract

BackgroundNotch receptors normally play a key role in guiding a variety of cell fate decisions during development and differentiation of metazoan organisms. On the other hand, dysregulation of Notch1 signaling is associated with many different types of cancer as well as tumor angiogenesis, making Notch1 a potential therapeutic target.Principal FindingsHere we report the in vitro activities of inhibitory Notch1 monoclonal antibodies derived from cell-based and solid-phase screening of a phage display library. Two classes of antibodies were found, one directed against the EGF-repeat region that encompasses the ligand-binding domain (LBD), and the second directed against the activation switch of the receptor, the Notch negative regulatory region (NRR). The antibodies are selective for Notch1, inhibiting Jag2-dependent signaling by Notch1 but not by Notch 2 and 3 in reporter gene assays, with EC50 values as low as 5±3 nM and 0.13±0.09 nM for the LBD and NRR antibodies, respectively, and fail to recognize Notch4. While more potent, NRR antibodies are incomplete antagonists of Notch1 signaling. The antagonistic activity of LBD, but not NRR, antibodies is strongly dependent on the activating ligand. Both LBD and NRR antibodies bind to Notch1 on human tumor cell lines and inhibit the expression of sentinel Notch target genes, including HES1, HES5, and DTX1. NRR antibodies also strongly inhibit ligand-independent signaling in heterologous cells transiently expressing Notch1 receptors with diverse NRR “class I” point mutations, the most common type of mutation found in human T-cell acute lymphoblastic leukemia (T-ALL). In contrast, NRR antibodies failed to antagonize Notch1 receptors bearing rare “class II” or “class III” mutations, in which amino acid insertions generate a duplicated or constitutively sensitive metalloprotease cleavage site. Signaling in T-ALL cell lines bearing class I mutations is partially refractory to inhibitory antibodies as compared to cell-penetrating gamma-secretase inhibitors.Conclusions/SignificanceAntibodies that compete with Notch1 ligand binding or that bind to the negative regulatory region can act as potent inhibitors of Notch1 signaling. These antibodies may have clinical utility for conditions in which inhibition of signaling by wild-type Notch1 is desired, but are likely to be of limited value for treatment of T-ALLs associated with aberrant Notch1 activation.

Highlights

  • Notch signals normally participate in a variety of cellular processes, including cell fate specification, differentiation, proliferation, apoptosis, migration, and angiogenesis [1]

  • negative regulatory region (NRR) antibodies strongly inhibit ligand-independent signaling in heterologous cells transiently expressing Notch1 receptors with diverse NRR ‘‘class I’’ point mutations, the most common type of mutation found in human T-cell acute lymphoblastic leukemia (T-ALL)

  • All antibodies were shown by flow cytometry to bind to full-length Notch1 overexpressed in HEK cells (Table 1 and data not shown)

Read more

Summary

Introduction

Notch signals normally participate in a variety of cellular processes, including cell fate specification, differentiation, proliferation, apoptosis, migration, and angiogenesis [1]. The first is catalyzed by a metalloprotease of the ADAM (a disintegrin and metalloprotease) family [14,15] and occurs at a site called S2, which lies within the NRR just external to the transmembrane domain This primes Notch for additional cleavages within the transmembrane domain that are carried out by the multiprotein membrane complex c-secretase [16]. The final cleavage liberates ICN from the membrane, allowing it to enter the nucleus and activate the transcription of Notch-responsive genes (e.g., HES1, HES5, NRARP, Deltex (DTX1), c-MYC) This depends on binding of ICN to the transcription factor CSL [7,17,18] and recruitment of Mastermind-like coactivators [19,20,21,22]. Dysregulation of Notch signaling is associated with many different types of cancer as well as tumor angiogenesis, making Notch a potential therapeutic target

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call