Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter January 13, 2011

The selection of aptamers specific for membrane molecular targets

  • Teresa Janas EMAIL logo and Tadeusz Janas

Abstract

A growing number of RNA aptamers have been selected experimentally using the SELEX combinatorial approach, and these aptamers have several advantages over monoclonal protein antibodies or peptides with respect to their applications in medicine and nanobiotechnology. Relatively few successful selections have been reported for membrane molecular targets, in contrast to the situation with non-membrane molecular targets. This review compares the procedures and techniques used in selections against membrane proteins and membrane lipids. In the case of membrane proteins, the selections were performed against soluble protein fragments, detergent-membrane protein mixed micelles, whole cells, vesicles derived from cellular membranes, and enveloped viruses. Liposomes were used as an experimental system for the selection of aptamers against membrane lipids. RNA structure-dependent aptamer binding for rafts in lipid vesicles was reported. Based on the selected aptamers against DOPC and the amino acid tryptophan, a specific passive membrane transporter composed of RNA was constructed. The determination of the selectivity of aptamers appears to be a crucial step in a selection, but has rarely been fully investigated. The selections, which use whole cells or vesicles derived from membranes, can yield aptamers not only against proteins but also against membrane lipids.

[1] Yarus, M. Life from an RNA World: the ancestor within. Harvard University Press, New York, 2010. Search in Google Scholar

[2] Connell, G.J., Illangsekare, M., Yarus, M. Three small ribooligonucleotides with specific arginine sites. Biochemistry 32 (1993) 5497–5502. http://dx.doi.org/10.1021/bi00072a00210.1021/bi00072a002Search in Google Scholar PubMed

[3] Khvorova, A., Kwak, Y.-G., Tamkun, M., Majerfeld, I. and Yarus, M. RNAs that bind and change the permeability of phospholipid membranes. Proc. Natl. Acad. Sci. USA 96 (1999) 10649–10654. http://dx.doi.org/10.1073/pnas.96.19.1064910.1073/pnas.96.19.10649Search in Google Scholar PubMed PubMed Central

[4] Yarus, M. A specific amino acid binding site composed of RNA. Science 240 (1988) 1751–1758. http://dx.doi.org/10.1126/science.338109910.1126/science.3381099Search in Google Scholar PubMed

[5] Roth, A., Winkler, W.C., Regulski, E.E., Lee, B.W.K., Lim, J., Jona, I., Barrick, J.E., Ritwik, A., Kim, J.N., Welz, R., Iwata-Reuyl, D. and Breaker, R.R. A riboswitch selective for the queuosine precursor preQ1 contains an unusually small aptamer domain. Nat. Struct. Mol. Biol. 14 (2007) 308–317. http://dx.doi.org/10.1038/nsmb122410.1038/nsmb1224Search in Google Scholar PubMed

[6] Spitale, R.C., Terelli, A.T., Krucinska, J., Bandarlan, V., Wedekind, J.E. The structural basis for recognition of the preQ0 metabolite by an unusually small riboswitch aptamer domain. J. Biol. Chem. 284 (2009) 11012–11016. http://dx.doi.org/10.1074/jbc.C90002420010.1074/jbc.C900024200Search in Google Scholar PubMed PubMed Central

[7] Ellington, A.D. and Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 346 (1990) 818–822. http://dx.doi.org/10.1038/346818a010.1038/346818a0Search in Google Scholar PubMed

[8] Tuerk, C. and Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA-polymerase. Science 249 (1990) 505–510. http://dx.doi.org/10.1126/science.220012110.1126/science.2200121Search in Google Scholar PubMed

[9] Janas, T., Widmann, J.J., Knight, R. and Yarus, M. Simple, recurrent RNA binding sites for L-arginine. RNA (2010) 805–816. 10.1261/rna.1979410Search in Google Scholar PubMed PubMed Central

[10] Bock, L.C., Griffin, L.C., Latham, J.A., Vermaas, E.H. and Toole, J.J. Selection of single-stranded DNA molecules that bind and inhibit human thrombin. Nature 355 (1992) 564–566. http://dx.doi.org/10.1038/355564a010.1038/355564a0Search in Google Scholar PubMed

[11] Anderson, P.C. and Mecozzi, S. Unusually short RNA sequences: design of a 13-mer RNA that selectively binds and recognizes theophylline. J. Am. Chem. Soc. 127 (2005) 5290–5291. http://dx.doi.org/10.1021/ja043246310.1021/ja0432463Search in Google Scholar PubMed

[12] Farokhzad, O.C., Cheng, J., Teply, B.A., Sherifi, I., Jon, S., Kantoff, P.W., Richie, J.P. and Langer, R. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl. Acad. Sci. USA 103 (2006) 6315–6320. http://dx.doi.org/10.1073/pnas.060175510310.1073/pnas.0601755103Search in Google Scholar PubMed PubMed Central

[13] Song, S., Wang, L., Li, J., Zhao, J. and Fan, C. Aptamer-based biosensors. Trends Anal. Chem. 27 (2008) 108–117. http://dx.doi.org/10.1016/j.trac.2007.12.00410.1016/j.trac.2007.12.004Search in Google Scholar

[14] Lee, J.O., So, H.M., Jeon, E.K., Chang, H., Won, K. and Kim, Y.H. Aptamers as molecular recognition elements for electrical nanobiosensors. Anal. Bioanal. Chem. 390 (2008) 1023–1032. http://dx.doi.org/10.1007/s00216-007-1643-y10.1007/s00216-007-1643-ySearch in Google Scholar PubMed PubMed Central

[15] Barbas, A.S. and White, R.R. The development and testing for cancer. Curr. Opin. Investig. Drugs 10 (2009) 572–578. Search in Google Scholar

[16] Hicke, B.J., Marion, C., Chang, Y.F., Gould, T., Lynott, C.K., Parma, D., Schmidt, P.G. and Warren, S. Tenascin-C aptamers are generated using tumor cells and purified protein. J. Biol. Chem. 276 (2001) 48644–48654. http://dx.doi.org/10.1074/jbc.M10465120010.1074/jbc.M104651200Search in Google Scholar PubMed

[17] Pestourie, C. Tavitian, B. and Duconge, F. Aptamets against extracellular targets for in vivo applications. Biochimie 87 (2005) 921–930. http://dx.doi.org/10.1016/j.biochi.2005.04.01310.1016/j.biochi.2005.04.013Search in Google Scholar PubMed

[18] Janas, T., Janas, T. and Yarus, M. RNA, lipids and membranes. in: The RNA World III (Gesteland,, R., Cech, T.R. and Atkins, J., Eds.), Cold Spring Harbor Laboratory Press, 2006, 207–225. Search in Google Scholar

[19] Shamah, S.M., Healy, J.M. and Cload, S.T. Complex target SELEX. Acc. Chem. Res. 41 (2008) 130–138. http://dx.doi.org/10.1021/ar700142z10.1021/ar700142zSearch in Google Scholar PubMed

[20] Li, N., Ebright, J.N., Stovall, G.M., Chen, X., Nguyen, H.H., Singh, A., Syrett, A. and Ellington, A.D. Technical and biological issues relevant to cell typing with aptamers. J. Proteome Res. 8 (2009) 2438–2448. http://dx.doi.org/10.1021/pr801048z10.1021/pr801048zSearch in Google Scholar PubMed

[21] Lupold, S.E., Hicke, B.J., Lin, Y. and Coffey, D.S. Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigen. Cancer Res. 62 (2002) 4029–4033. Search in Google Scholar

[22] Chen, C.h.B., Chernis, G.A., Hoang, V.Q. and Landgraf, R. Inhibition of heregulin signaling by an aptamer that preferentially binds to the oligomeric form of human epidermal growth factor receptor-3. Proc. Natl. Acad. Sci. USA 100 (2003) 9226–9231. http://dx.doi.org/10.1073/pnas.133266010010.1073/pnas.1332660100Search in Google Scholar PubMed PubMed Central

[23] Rentmeister, A., Bill, A., Wahle, T., Walter, J. and Famulok, M. RNA aptamers selectively modulate protein recruitment to the cytoplasmic domain of β-secretase BACE1 in vitro. RNA 12 (2006) 1650–1660. http://dx.doi.org/10.1261/rna.12630610.1261/rna.126306Search in Google Scholar PubMed PubMed Central

[24] O’Connell, D., Koenig, A., Jennings, S., Hicke, B., Han, H.L., Fitzwater, T., Chang, Y.F., Varki, N., Parma, D. and Varki, A. Calcium-dependent oligonucleotide antagonists specific for L-selectin. Proc. Natl. Acad. Sci. USA 93 (1996) 5883–5887. http://dx.doi.org/10.1073/pnas.93.12.588310.1073/pnas.93.12.5883Search in Google Scholar PubMed PubMed Central

[25] Lee, H.K., Choi, Y.S., Park, Y.A. and Jeong, S. Modulation of oncogenic transcription and alternative splicing by β-catenin and an RNA aptamer in colon cancer cells. Cancer Res. 66 (2006) 10560–10566. http://dx.doi.org/10.1158/0008-5472.CAN-06-252610.1158/0008-5472.CAN-06-2526Search in Google Scholar PubMed

[26] Tanaka, Y., Akagi, K., Nakamura, Y. and Kozu, T. RNA aptamers targeting the carboxyl terminus of KRAS oncoprotein generated by an improved SELEX with isothermal RNA amplification. Oligonucleotides 17 (2007) 12–21. http://dx.doi.org/10.1089/oli.2006.0035R110.1089/oli.2006.0035R1Search in Google Scholar PubMed

[27] Jeon, S.H., Kayhan, B., Ben-Yedidia, T. and Arnon, R. A DNA aptamer prevents influenza infection by blocking the receptor binding region of the viral hemagglutinin. J. Biol. Chem. 279 (2004) 48410–48419. http://dx.doi.org/10.1074/jbc.M40905920010.1074/jbc.M409059200Search in Google Scholar PubMed

[28] Ferreira, C.S.M., Matthews, C.S. and Missailidis, S. DNA aptamers that bind to MUC1 tumour marker: design and characterization of MUC1-binding single-stranded DNA aptamers. Tumor Biol. 27 (2006) 289–301. http://dx.doi.org/10.1159/00009608510.1159/000096085Search in Google Scholar PubMed

[29] Daniels, D.A., Sohal, A.K., Rees, S. and Grisshammer, R. Generation of RNA aptamers to the G-protein-coupled receptor for neurotensin, NTS-1. Anal. Biochem. 305 (2002) 214–226. http://dx.doi.org/10.1006/abio.2002.566310.1006/abio.2002.5663Search in Google Scholar PubMed

[30] Joshi, R., Janagama, H., Dwivedi, H.P., Kumar, T.M.A.S., Jaykus, L.A. Schefers, J. and Sreevatsan, S. Selection, characterization, and application of DNA aptamers for the capture and detection of Salmonella enterica serovars. Mol. Cell. Probes 23 (2009) 20–28. http://dx.doi.org/10.1016/j.mcp.2008.10.00610.1016/j.mcp.2008.10.006Search in Google Scholar PubMed

[31] Ulrich, H., Ippolito, J.E., Pagan, O.R., Eterovic, V.A., Hann, R.M., Shi, H., Lis, J.T., Eldefrawi, M.E. and Hess, G.P. In vitro selection of RNA molecules that displace cocaine from the membrane-bound nicotinic acetylcholine receptor. Proc. Natl. Acad. Sci. USA 95 (1998) 14051–14056. http://dx.doi.org/10.1073/pnas.95.24.1405110.1073/pnas.95.24.14051Search in Google Scholar PubMed PubMed Central

[32] Morris, K.N., Jensen, K.B., Julin, C.M., Weil, M. and Gold, L. High affinity ligands from in vitro selection: complex target. Proc. Natl. Acad. Sci. USA 95 (1998) 2902–2907. http://dx.doi.org/10.1073/pnas.95.6.290210.1073/pnas.95.6.2902Search in Google Scholar PubMed PubMed Central

[33] Shangguan, D., Li, Y., Tang, Z., Cao, Z.C., Chen, H.W., Mallikaratchy, P., Sefah, K., Yang, C.J. and Tan, W. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc. Natl. Acad. Sci. USA 103 (2006) 11838–11843. http://dx.doi.org/10.1073/pnas.060261510310.1073/pnas.0602615103Search in Google Scholar PubMed PubMed Central

[34] Homann, M. and Göringer, H.U. Combinatorial selection of high affinity RNA ligands to live African trypanosomes. Nucleic Acids Res. 27 (1999) 2006–2014. http://dx.doi.org/10.1093/nar/27.9.200610.1093/nar/27.9.2006Search in Google Scholar PubMed PubMed Central

[35] Blank, M., Weinschenk, T., Priemer, M. and Schluesener, H. Systematic evolution of DNA aptamer binding to rat brain tumor microvessels. J. Biol. Chem. 276 (2001) 16464–16468. http://dx.doi.org/10.1074/jbc.M10034720010.1074/jbc.M100347200Search in Google Scholar PubMed

[36] Mallikaratchy, P., Tang, Z., Kwame, S., Meng, L., Shangguan, D. and Tan, W. Aptamer directly evolved from live cells recognizes membrane bound immunoglobin heavy mu chain in Burkitt’s lymphoma cells. Mol. Cell. Proteomics 6 (2007) 2230–2238. http://dx.doi.org/10.1074/mcp.M700026-MCP20010.1074/mcp.M700026-MCP200Search in Google Scholar PubMed

[37] Shangguan, D., Cao, Z., Meng, L., Mallikaratchy, P., Sefah, K., Wang, H., Li, Y. and Tan, W. Cell-specific aptamer probes for membrane protein elucidation in cancer cells. J. Proteome Res. 7 (2008) 2133–2139. http://dx.doi.org/10.1021/pr700894d10.1021/pr700894dSearch in Google Scholar PubMed PubMed Central

[38] Berezovski, M.V., Lechmann, M., Musheev, M.U., Mak, T.W. and Krylov, S.N. Aptamer-facilitated biomarker discovery (AptaBiD). J. Am. Chem. Soc. 130 (2008) 9137–9143. http://dx.doi.org/10.1021/ja801951p10.1021/ja801951pSearch in Google Scholar PubMed

[39] Ulrich, H., Magdesian, M.H., Alves, M.J.M. and Colli, W. In vitro selection of RNA aptamers that bind to cell adhesion receptors of Trypanosoma cruzi and inhibit cell invasion. J. Biol. Chem. 277 (2002) 20756–20762. http://dx.doi.org/10.1074/jbc.M11185920010.1074/jbc.M111859200Search in Google Scholar PubMed

[40] Cerchia, L., Duconge, F., Pestourie, C., Boulay, J., Aissouni, Y., Gombert, K., Tavitian, B., de Franciscis, V. and Libri, D. Neutralizing aptamers from whole-cell SELEX inhibit the RET receptor tyrosine kinase. PLOS Biol. 3 (2005) 697–704. http://dx.doi.org/10.1371/journal.pbio.003012310.1371/journal.pbio.0030123Search in Google Scholar PubMed PubMed Central

[41] Gopinath, S.C.B., Misono, T.S., Kawasaki, K., Mizuno, T., Imai, M., Odegiri, T. and Kumar, P.K.R. An RNA aptamer that distinguishes between closely related human influenza viruses and inhibits haemagglutinin-mediated membrane fusion. J. Gen. Virol. 87 (2006) 479–487. http://dx.doi.org/10.1099/vir.0.81508-010.1099/vir.0.81508-0Search in Google Scholar PubMed

[42] Ohuchi, S.P., Ohtsu, T. and Nakamura, Y. Selection of RNA aptamers against recombinant transforming growth factor-β type III receptor displayed on cell surface. Biochimie 88 (2006) 897–904. http://dx.doi.org/10.1016/j.biochi.2006.02.00410.1016/j.biochi.2006.02.004Search in Google Scholar PubMed

[43] Sazani, P.L., Larraide, R. and Szostak, J.W. A small aptamer with strong and specific recognition of the triphosphate of ATP. J. Am. Chem Soc. 126 (2004) 8370–8371. http://dx.doi.org/10.1021/ja049171k10.1021/ja049171kSearch in Google Scholar PubMed PubMed Central

[44] Zimmerman, G.R, Jenison, R.D., Wick, C.L., Simorre, J.P. and Pardi, A. Interlocking structural motifs mediate molecular discrimination by a theophylline-binding RNA. Nat. Struct. Biol. 4 (1997) 644–649. http://dx.doi.org/10.1038/nsb0897-64410.1038/nsb0897-644Search in Google Scholar PubMed

[45] Majerfeld, I. and Yarus, M. An RNA pocker for an aliphatic hydrophobe. Nature Struct. Biol. 1 (1994) 287–292. http://dx.doi.org/10.1038/nsb0594-28710.1038/nsb0594-287Search in Google Scholar

[46] Majerfeld, I. and Yarus, M. Isoleucine:RNA sites with associated coding sequences. RNA 4 (1998) 471–478. Search in Google Scholar

[47] Gilbert, B.A., Sha, M., Wathen, S.T. and Rando, R.R. RNA aptamers that specifically bind to a K Ras-derived farnesylated peptide. Bioorg. Med. Chem. 5 (1997) 1115–1122. http://dx.doi.org/10.1016/S0968-0896(97)00047-310.1016/S0968-0896(97)00047-3Search in Google Scholar

[48] Sussman, D., Nix, J.C. and Wilson, C. The structural basis for molecular recognition by the vitamin B12 RNA aptamer. Nat. Struct. Biol. 7 (2000) 53–57. http://dx.doi.org/10.1038/7125310.1038/71253Search in Google Scholar PubMed

[49] Illangasekare, M. and Yarus, M. Phenylalanine-binding RNAs and genetic code evolution. J. Mol. Evol. 54 (2002) 298–311. Search in Google Scholar

[50] Betat, H., Vogel, S., Struhalla, M., Förster, H.H., Famulok, M., Welzel, P. and Hahn, U. Aptamers that recognize the lipid moiety of the antibiotic moenomycin A. Biol. Chem. 384 (2003) 1497–1500. http://dx.doi.org/10.1515/BC.2003.16510.1515/BC.2003.165Search in Google Scholar PubMed

[51] Brockstedt, U., Uzarowska, A., Montpetit, A., Pfau, W. and Labuda, D. In vitro evolution of RNA aptamers recognizing carcinogenic aromatic amines. Biochem. Biophys. Res Commun. 313 (2004) 1004–1008. http://dx.doi.org/10.1016/j.bbrc.2003.12.03010.1016/j.bbrc.2003.12.030Search in Google Scholar PubMed

[52] Janas, T., Janas, T. and Yarus, M. Specific RNA binding to ordered phospholipid bilayers. Nucleic Acids Res. 34 (2006) 2128–2136. http://dx.doi.org/10.1093/nar/gkl22010.1093/nar/gkl220Search in Google Scholar PubMed PubMed Central

[53] Vlassov, A., Khvorova, A., and Yarus, M. Binding and disruption of phosholipid bilayers by supramolecular RNA complexes. Proc. Natl. Acad. Sci. USA 98 (2001) 7706–7711. http://dx.doi.org/10.1073/pnas.14104109810.1073/pnas.141041098Search in Google Scholar PubMed PubMed Central

[54] Janas, T. and Yarus, M. Visualization of membrane RNAs. RNA 9 (2003) 1353–1361. http://dx.doi.org/10.1261/rna.512980310.1261/rna.5129803Search in Google Scholar PubMed PubMed Central

[55] Janas, T., Janas, T. and Yarus, M. A membrane transporter for tryptophan composed of RNA. RNA 10 (2004) 1541–1549. http://dx.doi.org/10.1261/rna.711270410.1261/rna.7112704Search in Google Scholar PubMed PubMed Central

[56] Janas, T. and Janas, T. A Search for membrane RNAs that can inhibit formation of toxic amyloid aggregates. Sie Foundation Symposium, Aurora, 2006, 13. Search in Google Scholar

Published Online: 2011-1-13
Published in Print: 2011-3-1

© 2011 University of Wrocław, Poland

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.

Downloaded on 25.5.2024 from https://www.degruyter.com/document/doi/10.2478/s11658-010-0023-3/html
Scroll to top button