Skip to main content
Log in

CRISPR-Mediated Isogenic Cell-SELEX Approach for Generating Highly Specific Aptamers Against Native Membrane Proteins

  • 2020 CMBE Young Innovators Issue
  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Introduction

The generation of affinity reagents that bind native membrane proteins with high specificity remains challenging. Most in vitro selection paradigms utilize different cell types for positive and negative rounds of selection (where the positive selection is against a cell that expresses the desired membrane protein and the negative selection is against a cell that lacks the protein). However, this strategy can yield affinity reagents that bind unintended membrane proteins on the target cells. To address this issue, we developed a systematic evolution of ligands by exponential enrichment (SELEX) scheme that utilizes isogenic pairs of cells generated via CRISPR techniques.

Methods

Using a Caco-2 epithelial cell line with constitutive Cas9 expression, we knocked out the SLC2A1 gene (encoding the GLUT1 glucose transporter) via lipofection with synthetic gRNAs. Cell-SELEX rounds were carried out against wild-type and GLUT1-null cells using a single-strand DNA (ssDNA) library. Next-generation sequencing (NGS) was used to quantify enrichment of prospective binders to the wild-type cells.

Results

10 rounds of cell-SELEX were conducted via simultaneous exposure of ssDNA pools to wild-type and GLUT1-null Caco-2 cells under continuous perfusion. The top binders identified from NGS were validated by flow cytometry and immunostaining for their specificity to the GLUT1 receptor.

Conclusions

Our data indicate that highly specific aptamers can be isolated with a SELEX strategy that utilizes isogenic cell lines. This approach may be broadly useful for generating affinity reagents that selectively bind to membrane proteins in their native conformations on the cell surface.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8

Similar content being viewed by others

Abbreviations

SELEX:

Systematic evolution of ligands through exponential enrichment

CRISPR:

Clustered regularly interspaced short palindromic repeats

GLUT:

Glucose transporter

TIDE:

Tracking of indels by decomposition

NGS:

Next-generation sequencing

iPSC:

Induced pluripotent stem cell

BMEC:

Brain microvascular endothelial cells

References

  1. Askoxylakis, V., et al. Peptide-based targeting of the platelet-derived growth factor receptor beta. Mol. Imaging Biol. 15(2):212–221, 2013. https://doi.org/10.1007/s11307-012-0578-7.

    Article  Google Scholar 

  2. Bradbury, A. R. M., S. Sidhu, S. Dübel, and J. McCafferty. Beyond natural antibodies: the power of in vitro display technologies. Nat. Biotechnol. 29(3):245–254, 2011. https://doi.org/10.1038/nbt.1791.

    Article  Google Scholar 

  3. Brinkman, E. K., T. Chen, M. Amendola, and B. van Steensel. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42(22):e168, 2014. https://doi.org/10.1093/nar/gku936.

    Article  Google Scholar 

  4. Cruz-Toledo, J., et al. Aptamer base: a collaborative knowledge base to describe aptamers and SELEX experiments. Database J. Biol. Databases Curation 2012. https://doi.org/10.1093/database/bas006.

    Article  Google Scholar 

  5. Dangaj, D., et al. Novel recombinant human B7-H4 antibodies overcome tumoral immune escape to potentiate T cell anti-tumor responses. Cancer Res. 73(15):4820–4829, 2013. https://doi.org/10.1158/0008-5472.CAN-12-3457.

    Article  Google Scholar 

  6. Duan, M., et al. Selection and characterization of DNA aptamer for metastatic prostate cancer recognition and tissue imaging. Oncotarget 7(24):36436–36446, 2016. https://doi.org/10.18632/oncotarget.9262.

    Article  Google Scholar 

  7. Groff, K., J. Brown, and A. J. Clippinger. Modern affinity reagents: recombinant antibodies and aptamers. Biotechnol. Adv. 33(8):1787–1798, 2015. https://doi.org/10.1016/j.biotechadv.2015.10.004.

    Article  Google Scholar 

  8. Hauptmann, S., et al. Glucose transporter GLUT1 in colorectal adenocarcinoma cell lines is inversely correlated with tumour cell proliferation. Anticancer Res. 25(5):3431–3436, 2005.

    Google Scholar 

  9. Heitner, T., A. Moor, J. L. Garrison, C. Marks, T. Hasan, and J. D. Marks. Selection of cell binding and internalizing epidermal growth factor receptor antibodies from a phage display library. J. Immunol. Methods 248(1–2):17–30, 2001. https://doi.org/10.1016/s0022-1759(00)00340-9.

    Article  Google Scholar 

  10. Hoinka, J., R. Backofen, and T. M. Przytycka. AptaSUITE: a full-featured bioinformatics framework for the comprehensive analysis of aptamers from HT-SELEX experiments. Mol. Ther. Nucleic Acids 11:515–517, 2018. https://doi.org/10.1016/j.omtn.2018.04.006.

    Article  Google Scholar 

  11. Kim, J. W., et al. Identification of DNA aptamers toward epithelial cell adhesion molecule via cell-SELEX. Mol. Cells 37(10):742–746, 2014. https://doi.org/10.14348/molcells.2014.0208.

    Article  Google Scholar 

  12. Kinet, S., et al. Isolated receptor binding domains of HTLV-1 and HTLV-2 envelopes bind Glut-1 on activated CD4+ and CD8 + T cells. Retrovirology 4:31, 2007. https://doi.org/10.1186/1742-4690-4-31.

    Article  Google Scholar 

  13. Lippmann, E. S., et al. Human blood-brain barrier endothelial cells derived from pluripotent stem cells. Nat. Biotechnol. 30(8):783–791, 2012. https://doi.org/10.1038/nbt.2247.

    Article  Google Scholar 

  14. Mahraoui, L., et al. Presence and differential expression of SGLT1, GLUT1, GLUT2, GLUT3 and GLUT5 hexose-transporter mRNAs in Caco-2 cell clones in relation to cell growth and glucose consumption. Biochem. J. 298(Pt 3):629–633, 1994.

    Article  Google Scholar 

  15. Mercier, M.-C., M. Dontenwill, and L. Choulier. Selection of nucleic acid aptamers targeting tumor cell-surface protein biomarkers. Cancers 2017. https://doi.org/10.3390/cancers9060069.

    Article  Google Scholar 

  16. Meyer, S., et al. Development of an Efficient Targeted Cell-SELEX Procedure for DNA Aptamer Reagents. PLoS ONE 8(8):e71798, 2013. https://doi.org/10.1371/journal.pone.0071798.

    Article  Google Scholar 

  17. Mueckler, M., and C. Makepeace. Model of the exofacial substrate-binding site and helical folding of the human Glut1 glucose transporter based on scanning mutagenesis. Biochemistry (Mosc.) 48(25):5934–5942, 2009. https://doi.org/10.1021/bi900521n.

    Article  Google Scholar 

  18. Mueckler, M., and B. Thorens. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 34:121–138, 2013. https://doi.org/10.1016/j.mam.2012.07.001.

    Article  Google Scholar 

  19. Neal, E. H., et al. A simplified, fully defined differentiation scheme for producing blood-brain barrier endothelial cells from human iPSCs. Stem Cell Rep. 12(6):1380–1388, 2019. https://doi.org/10.1016/j.stemcr.2019.05.008.

    Article  Google Scholar 

  20. Newton, J., and S. L. Deutscher. Phage peptide display. Handb. Exp. Pharmacol. 185(2):145–163, 2008. https://doi.org/10.1007/978-3-540-77496-9_7.

    Article  Google Scholar 

  21. Ohuchi, S. P., T. Ohtsu, and Y. Nakamura. Selection of RNA aptamers against recombinant transforming growth factor-β type III receptor displayed on cell surface. Biochimie 88(7):897–904, 2006. https://doi.org/10.1016/j.biochi.2006.02.004.

    Article  Google Scholar 

  22. Palmer, C. S., C. L. Cherry, I. Sada-Ovalle, A. Singh, and S. M. Crowe. Glucose metabolism in T cells and monocytes: new perspectives in HIV pathogenesis. EBioMedicine 6:31–41, 2016. https://doi.org/10.1016/j.ebiom.2016.02.012.

    Article  Google Scholar 

  23. Pardridge, W. M., R. J. Boado, and C. R. Farrell. Brain-type glucose transporter (GLUT-1) is selectively localized to the blood-brain barrier. Studies with quantitative western blotting and in situ hybridization. J. Biol. Chem. 265(29):18035–18040, 1990.

    Google Scholar 

  24. Pleiko, K., L. Saulite, V. Parfejevs, K. Miculis, E. Vjaters, and U. Riekstina. Differential binding cell-SELEX method to identify cell-specific aptamers using high-throughput sequencing. Sci. Rep. 9(1):1–12, 2019. https://doi.org/10.1038/s41598-019-44654-w.

    Article  Google Scholar 

  25. Rosch, J. C., D. A. Balikov, F. Gong, and E. S. Lippmann. A systematic evolution of ligands by exponential enrichment workflow with consolidated counterselection to efficiently isolate high-affinity aptamers. Eng. Rep. 2020. https://doi.org/10.1002/eng2.12089.

    Article  Google Scholar 

  26. Sefah, K., D. Shangguan, X. Xiong, M. B. O’Donoghue, and W. Tan. Development of DNA aptamers using Cell-SELEX. Nat. Protoc. 5(6):1169–1185, 2010. https://doi.org/10.1038/nprot.2010.66.

    Article  Google Scholar 

  27. Stern, L. A., et al. Geometry and expression enhance enrichment of functional yeast-displayed ligands via cell panning. Biotechnol. Bioeng. 113(11):2328–2341, 2016. https://doi.org/10.1002/bit.26001.

    Article  Google Scholar 

  28. Szablewski, L. Glucose transporters in brain. In health and in Alzheimer’s disease. J. Alzheimers Dis. JAD 55(4):1307–1320, 2017. https://doi.org/10.3233/JAD-160841.

    Article  Google Scholar 

  29. Tang, M., et al. Brain microvasculature defects and Glut1 deficiency syndrome averted by early repletion of the glucose transporter-1 protein. Nat. Commun. 8(1):1–15, 2017. https://doi.org/10.1038/ncomms14152.

    Article  Google Scholar 

  30. Thul, P. J., et al. A subcellular map of the human proteome. Science 2017. https://doi.org/10.1126/science.aal3321.

    Article  Google Scholar 

  31. Uchański, T., et al. An improved yeast surface display platform for the screening of nanobody immune libraries. Sci. Rep. 9(1):1–12, 2019. https://doi.org/10.1038/s41598-018-37212-3.

    Article  Google Scholar 

  32. Wang, G., et al. Selection and characterization of DNA aptamer against glucagon receptor by cell-SELEX. Sci. Rep. 7(1):1–10, 2017. https://doi.org/10.1038/s41598-017-05840-w.

    Article  Google Scholar 

  33. Wang, D., J. M. Pascual, and D. De Vivo. Glucose transporter type 1 deficiency syndrome. In: GeneReviews®, edited by M. P. Adam, H. H. Ardinger, R. A. Pagon, S. E. Wallace, L. J. Bean, K. Stephens, and A. Amemiya. Seattle, WA: University of Washington, 1993.

    Google Scholar 

  34. Wang, X. X., and E. V. Shusta. The use of scFv-displaying yeast in mammalian cell surface selections. J. Immunol. Methods 304(1–2):30–42, 2005. https://doi.org/10.1016/j.jim.2005.05.006.

    Article  Google Scholar 

  35. Wu, X., et al. Cell-SELEX aptamer for highly specific radionuclide molecular imaging of glioblastoma in vivo. PloS ONE 9(6):e90752, 2014. https://doi.org/10.1371/journal.pone.0090752.

    Article  Google Scholar 

  36. Zadeh, J. N., et al. NUPACK: analysis and design of nucleic acid systems. J. Comput. Chem. 32(1):170–173, 2011. https://doi.org/10.1002/jcc.21596.

    Article  Google Scholar 

Download references

Acknowledgments

We gratefully acknowledge Allison Bosworth for providing iPSC-derived BMECs, Ella Hoogenboezem for providing MDA-MB-231 cells, and Everett Allchin for culturing HEK-293 cells. This research was supported by a Ben Barres Early Career Acceleration Award from the Chan Zuckerberg Initiative (Grant 2018-191850 to ESL), Grant A20170945 from the BrightFocus Foundation (ESL), Grant IRG-58-009-56 from the American Cancer Society (ESL), and an Engineering Immunity Pilot Grant from Vanderbilt University (ESL). This work was further supported by facilities at Vanderbilt University, including the Vanderbilt University Medical Center Flow Cytometry shared resource (supported by the Vanderbilt Ingram Cancer Center NIH Grant P30 CA68485 and the Vanderbilt Digestive Disease Research Center NIH Grant DK058404) and the Vanderbilt Technologies for Advanced Genomics core facility (supported by the Vanderbilt Ingram Cancer Center NIH Grant P30 CA68485, the Vanderbilt Vision Center NIH Grant P30 EY08126, and NIH/NCRR Grant G20 RR030956). EHN is supported by a Graduate Research Fellowship from the National Science Foundation (DGE-1445197). DAB was supported by the Vanderbilt University Medical Scientist Training Program (T32 GM007347). We thank the Cooperative Human Tissue Network, an NIH/NCI sponsored resource, for providing human brain tissue.

Conflict of interest

The authors declare no conflicts of interest.

Ethical Approval

All human tissue used in this study was de-identified and did not require IRB approval.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ethan S. Lippmann.

Additional information

Associate Editor Scott Simon oversaw the review of this article.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 19978 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rosch, J.C., Neal, E.H., Balikov, D.A. et al. CRISPR-Mediated Isogenic Cell-SELEX Approach for Generating Highly Specific Aptamers Against Native Membrane Proteins. Cel. Mol. Bioeng. 13, 559–574 (2020). https://doi.org/10.1007/s12195-020-00651-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-020-00651-y

Keywords

Navigation