Facile supermolecular aptamer inhibitors of L-selectin

PLoS One. 2015 Mar 31;10(3):e0123034. doi: 10.1371/journal.pone.0123034. eCollection 2015.

Abstract

Multivalent interactions occur frequently in nature, where they mediate high-affinity interactions between cells, proteins, or molecules. Here, we report on a method to generate multivalent aptamers (Multi-Aptamers) that target L-selectin function using rolling circle amplification (RCA). We find that the L-selectin Multi-Aptamers have increased affinity compared to the monovalent aptamer, are specific to L-selectin, and are capable of inhibiting interactions with endogenous ligands. In addition, the Multi-Aptamers efficiently inhibit L-selectin mediated dynamic adhesion in vitro and homing to secondary lymphoid tissues in vivo. Importantly, our method of generating multivalent materials using RCA avoids many of the challenges associated with current multivalent materials in that the Multi-Aptamers are high affinity, easily produced and modified, and biocompatible. We anticipate that the Multi-Aptamers can serve as a platform technology to modulate diverse cellular processes.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Aptamers, Nucleotide / pharmacology*
  • Humans
  • Jurkat Cells
  • L-Selectin / drug effects*
  • L-Selectin / metabolism
  • Protein Binding

Substances

  • Aptamers, Nucleotide
  • L-Selectin

Grants and funding

This work was supported by the start-up fund from the Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center and the Chao Family Comprehensive Cancer Center at University of California Irvine, and University of California, Cancer Research Coordinating Committee. MAE is supported by a California Institute for Regenerative Medicine (CIRM) Training Grant (TG2-01152). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.