578
Views
27
CrossRef citations to date
0
Altmetric
Review

DNA-assisted protein detection technologies

, , , &
Pages 21-32 | Published online: 09 Jan 2014

References

  • Crick FH. On protein synthesis. Symp. Soc. Exp. Biol.12, 138–163 (1958).
  • Picotti P, Bodenmiller B, Mueller LN, Domon B, Aebersold R. Full dynamic range proteome analysis of S. cerevisiae by targeted proteomics. Cell138(4), 795–806 (2009).
  • Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol. Cell Proteomics1(11), 845–867 (2002).
  • Coons AH, Creech HJ, Jones RN, Berliner E. Demonstration of pneumococcal antigen in tissues by use of fluorescent antibody. J. Immunol.45, 159–170 (1942).
  • Yalow RS, Berson SA. Immunoassay of endogenous plasma insulin in man. J. Clin. Invest.39, 1157–1175 (1960).
  • Wide L, Bennich H, Johansson SG. Diagnosis of allergy by an in vitro test for allergen antibodies. Lancet2(7526), 1105–1107 (1967).
  • Addona TA, Abbatiello SE, Schilling B et al. Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat. Biotechnol.27(7), 633–641 (2009).
  • Gullberg M, Fredriksson S, Taussig M, Jarvius J, Gustafsdottir S, Landegren U. A sense of closeness: protein detection by proximity ligation. Curr. Opin. Biotechnol.14(1), 82–86 (2003).
  • Landegren U, Schallmeiner E, Nilsson M et al. Molecular tools for a molecular medicine: analyzing genes, transcripts and proteins using padlock and proximity probes. J. Mol. Recognit.17(3), 194–197 (2004).
  • Gustafsdottir SM, Schallmeiner E, Fredriksson S et al. Proximity ligation assays for sensitive and specific protein analyses. Anal. Biochem.345(1), 2–9 (2005).
  • Niemeyer CM, Adler M, Wacker R. Immuno-PCR: high sensitivity detection of proteins by nucleic acid amplification. Trends Biotechnol.23(4), 208–216 (2005).
  • Tanke HJ, Dirks RW, Raap T. FISH and immunocytochemistry: towards visualising single target molecules in living cells. Curr. Opin. Biotechnol.16(1), 49–54 (2005).
  • Barletta J. Applications of real-time immuno-polymerase chain reaction (rt-IPCR) for the rapid diagnoses of viral antigens and pathologic proteins. Mol. Aspects Med.27(2–3), 224–253 (2006).
  • Zhang H, Zhao Q, Li XF, Le XC. Ultrasensitive assays for proteins. Analyst132(8), 724–737 (2007).
  • Adler M, Wacker R, Niemeyer CM. Sensitivity by combination: immuno-PCR and related technologies. Analyst133(6), 702–718 (2008).
  • Blokzijl A, Friedman M, Ponten F, Landegren U. Profiling protein expression and interactions: proximity ligation as a tool for personalized medicine. J. Intern. Med.268(3), 232–245 (2010).
  • Weibrecht I, Leuchowius KJ, Clausson CM et al. Proximity ligation assays: a recent addition to the proteomics toolbox. Expert Rev. Proteomics7(3), 401–409 (2010).
  • Swartzman E, Shannon M, Lieu P et al. Expanding applications of protein analysis using proximity ligation and qPCR. Methods50(4), S23–26 (2010).
  • Binz HK, Amstutz P, Pluckthun A. Engineering novel binding proteins from nonimmunoglobulin domains. Nat. Biotechnol.23(10), 1257–1268 (2005).
  • Nygren PA, Skerra A. Binding proteins from alternative scaffolds. J. Immunol. Methods290(1–2), 3–28 (2004).
  • Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature346(6287), 818–822 (1990).
  • Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science249(4968), 505–510 (1990).
  • Salas M. Protein-priming of DNA replication. Annu. Rev. Biochem.60, 39–71 (1991).
  • Solomon MJ, Varshavsky A. Formaldehyde-mediated DNA–protein crosslinking: a probe for in vivo chromatin structures. Proc. Natl Acad. Sci. USA82(19), 6470–6474 (1985).
  • Sano T, Smith CL, Cantor CR. Immuno-PCR: very sensitive antigen detection by means of specific antibody–DNA conjugates. Science258(5079), 120–122 (1992).
  • Gullberg M, Gustafsdottir SM, Schallmeiner E et al. Cytokine detection by antibody-based proximity ligation. Proc. Natl Acad. Sci. USA101(22), 8420–8424 (2004).
  • Kozlov IA, Melnyk PC, Stromsborg KE, Chee MS, Barker DL, Zhao C. Efficient strategies for the conjugation of oligonucleotides to antibodies enabling highly sensitive protein detection. Biopolymers73(5), 621–630 (2004).
  • Soderberg O, Gullberg M, Jarvius M et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods3(12), 995–1000 (2006).
  • Nam JM, Thaxton CS, Mirkin CA. Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science301(5641), 1884–1886 (2003).
  • Lee J, Icoz K, Roberts A, Ellington AD, Savran CA. Diffractometric detection of proteins using microbead-based rolling circle amplification. Anal. Chem.82(1), 197–202 (2010).
  • Gold L, Ayers D, Bertino J et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE5(12), e15004 (2010).
  • Niemeyer CM, Wacker R, Adler M. Combination of DNA-directed immobilization and immuno-PCR: very sensitive antigen detection by means of self-assembled DNA–protein conjugates. Nucleic Acids Res.31(16), e90 (2003).
  • Fan R, Vermesh O, Srivastava A et al. Integrated barcode chips for rapid, multiplexed analysis of proteins in microliter quantities of blood. Nat. Biotechnol.26(12), 1373–1378 (2008).
  • Bailey RC, Kwong GA, Radu CG, Witte ON, Heath JR. DNA-encoded antibody libraries: a unified platform for multiplexed cell sorting and detection of genes and proteins. J. Am. Chem. Soc.129(7), 1959–1967 (2007).
  • Thaxton CS, Elghanian R, Thomas AD et al. Nanoparticle-based bio-barcode assay redefines ‘undetectable’ PSA and biochemical recurrence after radical prostatectomy. Proc. Natl Acad. Sci. USA106(44), 18437–18442 (2009).
  • Kim EY, Stanton J, Korber BT et al. Detection of HIV-1 p24 Gag in plasma by a nanoparticle-based bio-barcode-amplification method. Nanomedicine3(3), 293–303 (2008).
  • Chu BC, Kramer FR, Orgel LE. Synthesis of an amplifiable reporter RNA for bioassays. Nucleic Acids Res.14(14), 5591–5603 (1986).
  • Kattah MG, Coller J, Cheung RK, Oshidary N, Utz PJ. HIT: a versatile proteomics platform for multianalyte phenotyping of cytokines, intracellular proteins and surface molecules. Nat. Med.14(11), 1284–1289 (2008).
  • Saiki RK, Scharf S, Faloona F et al. Enzymatic amplification of beta-globin genomic sequences and restriction site analysis for diagnosis of sickle cell anemia. Science230(4732), 1350–1354 (1985).
  • Engvall E, Jonsson K, Perlmann P. Enzyme-linked immunosorbent assay. II. Quantitative assay of protein antigen, immunoglobulin G, by means of enzyme-labelled antigen and antibody-coated tubes. Biochim. Biophys. Acta251(3), 427–434 (1971).
  • Engvall E, Perlmann P. Enzyme-linked immunosorbent assay (ELISA). Quantitative assay of immunoglobulin G. Immunochemistry8(9), 871–874 (1971).
  • Engvall E, Perlmann P. Enzyme-linked immunosorbent assay, ELISA. 3. Quantitation of specific antibodies by enzyme-labeled anti-immunoglobulin in antigen-coated tubes. J. Immunol.109(1), 129–135 (1972).
  • Sims PW, Vasser M, Wong WL, Williams PM, Meng YG. Immunopolymerase chain reaction using real-time polymerase chain reaction for detection. Anal. Biochem.281(2), 230–232 (2000).
  • Ye Q, Zhuang H, Zhou C. Detection of naphthalene by real-time immuno-PCR using molecular beacon. Mol. Cell Probes23(1), 29–34 (2009).
  • Ruzicka V, Marz W, Russ A, Gross W. Immuno-PCR with a commercially available avidin system. Science260(5108), 698–699 (1993).
  • Zhou H, Fisher RJ, Papas TS. Universal immuno-PCR for ultra-sensitive target protein detection. Nucleic Acids Res.21(25), 6038–6039 (1993).
  • Sperl J, Paliwal V, Ramabhadran R, Nowak B, Askenase PW. Soluble T cell receptors: detection and quantitative assay in fluid phase via ELISA or immuno-PCR. J. Immunol. Methods186(2), 181–194 (1995).
  • Hendrickson ER, Truby TM, Joerger RD, Majarian WR, Ebersole RC. High sensitivity multianalyte immunoassay using covalent DNA-labeled antibodies and polymerase chain reaction. Nucleic Acids Res.23(3), 522–529 (1995).
  • Fischer A, Von Eiff C, Kuczius T, Omoe K, Peters G, Becker K. A quantitative real-time immuno-PCR approach for detection of staphylococcal enterotoxins. J. Mol. Med.85(5), 461–469 (2007).
  • Burbulis I, Yamaguchi K, Gordon A, Carlson R, Brent R. Using protein–DNA chimeras to detect and count small numbers of molecules. Nat. Methods2(1), 31–37 (2005).
  • Guo YC, Zhou YF, Zhang XE et al. Phage display mediated immuno-PCR. Nucleic Acids Res.34(8), e62 (2006).
  • Yu X, Burgoon MP, Shearer AJ, Gilden DH. Characterization of phage peptide interaction with antibody using phage mediated immuno-PCR. J. Immunol. Methods326(1–2), 33–40 (2007).
  • Yang L, Ellington AD. Real-time PCR detection of protein analytes with conformation-switching aptamers. Anal. Biochem.380(2), 164–173 (2008).
  • Yoshida Y, Horii K, Sakai N, Masuda H, Furuichi M, Waga I. Antibody-specific aptamer-based PCR analysis for sensitive protein detection. Anal. Bioanal. Chem.395(4), 1089–1096 (2009).
  • Wacker R, Ceyhan B, Alhorn P, Schueler D, Lang C, Niemeyer CM. Magneto immuno-PCR: a novel immunoassay based on biogenic magnetosome nanoparticles. Biochem. Biophys. Res. Commun.357(2), 391–396 (2007).
  • Wang TW, Lu HY, Lou PJ, Lin FH. Application of highly sensitive, modified glass substrate-based immuno-PCR on the early detection of nasopharyngeal carcinoma. Biomaterials29(33), 4447–4454 (2008).
  • Tian P, Mandrell R. Detection of norovirus capsid proteins in faecal and food samples by a real time immuno-PCR method. J. Appl. Microbiol.100(3), 564–574 (2006).
  • Perez JW, Vargis EA, Russ PK, Haselton FR, Wright DW. Detection of respiratory syncytial virus using nanoparticle amplified immuno-polymerase chain reaction. Anal. Biochem.410(1), 141–148 (2011).
  • Deng MJ, Xiao XZ, Zhang YM et al. A highly sensitive immuno-PCR assay for detection of H5N1 avian influenza virus. Mol. Biol. Rep.38(3), 1941–1948 (2011).
  • Chen L, Wei H, Guo Y, Cui Z, Zhang Z, Zhang XE. Gold nanoparticle enhanced immuno-PCR for ultrasensitive detection of Hantaan virus nucleocapsid protein. J. Immunol. Methods346(1–2), 64–70 (2009).
  • Barletta JM, Edelman DC, Highsmith WE, Constantine NT. Detection of ultra-low levels of pathologic prion protein in scrapie infected hamster brain homogenates using real-time immuno-PCR. J. Virol. Methods127(2), 154–164 (2005).
  • Barletta J, Bartolome A, Constantine NT. Immunomagnetic quantitative immuno-PCR for detection of less than one HIV-1 virion. J. Virol. Methods157(2), 122–132 (2009).
  • Kuczius T, Becker K, Karch H, Zhang W. High sensitivity detection of the glial fibrillary acidic protein as indicator for TSE risk material in meat products using an immuno-PCR. Mol. Nutr. Food Res.53(10), 1329–1335 (2009).
  • Zhang L, Ren J, Pan K et al. Detection of gastric carcinoma-associated MG7-Ag by serum immuno-PCR assay in a high-risk Chinese population, with implication for screening. Int. J. Cancer126(2), 469–473 (2010).
  • Zhang W, Bielaszewska M, Pulz M et al. New immuno-PCR assay for detection of low concentrations of shiga toxin 2 and its variants. J. Clin. Microbiol.46(4), 1292–1297 (2008).
  • Zhou C, Zhuang H. Determination of fluoranthene by antigen-coated indirect competitive real-time immuno-PCR assay. J. Environ. Monit.11(2), 400–405 (2009).
  • Zhuang HS, Zhou C. Determination of anthracene by real-time immuno-polymerase chain reaction assay. Anal. Chim. Acta633(2), 278–282 (2009).
  • Chen HY, Zhuang HS. Real-time immuno-PCR assay for detecting PCBs in soil samples. Anal. Bioanal. Chem.394(4), 1205–1211 (2009).
  • Singer D, Soininen H, Alafuzoff I, Hoffmann R. Immuno-PCR-based quantification of multiple phosphorylated tau-epitopes linked to Alzheimer’s disease. Anal. Bioanal. Chem.395(7), 2263–2267 (2009).
  • Shibasaki F, Morizane Y, Ishikawa Y et al. [Clinical application of supersensitive and multiplex assay, MUSTag technology]. Rinsho Byori56(9), 802–810 (2008).
  • Shibasaki F, Morizane Y, Makisaka N. [Protein biomarker measurement and simple/rapid diagnostics with supersensitive and multiplex assay, MUSTag technology]. Rinsho Byori57(11), 1104–1112 (2009).
  • Xie MJ, Fukui K, Horie M et al. A novel sensitive immunoassay method based on the Invader technique. Anal. Biochem.374(2), 278–284 (2008).
  • Schweitzer B, Wiltshire S, Lambert J et al. Inaugural article: immunoassays with rolling circle DNA amplification: a versatile platform for ultrasensitive antigen detection. Proc. Natl Acad. Sci. USA97(18), 10113–10119 (2000).
  • Baner J, Nilsson M, Mendel-Hartvig M, Landegren U. Signal amplification of padlock probes by rolling circle replication. Nucleic Acids Res.26(22), 5073–5078 (1998).
  • Ericsson O, Jarvius J, Schallmeiner E et al. A dual-tag microarray platform for high-performance nucleic acid and protein analyses. Nucleic Acids Res.36(8), e45 (2008).
  • Schweitzer B, Roberts S, Grimwade B et al. Multiplexed protein profiling on microarrays by rolling-circle amplification. Nat. Biotechnol.20(4), 359–365 (2002).
  • Bacarese-Hamilton T, Mezzasoma L, Ingham C et al. Detection of allergen-specific IgE on microarrays by use of signal amplification techniques. Clin. Chem.48(8), 1367–1370 (2002).
  • Zhou H, Bouwman K, Schotanus M et al. Two-color, rolling-circle amplification on antibody microarrays for sensitive, multiplexed serum-protein measurements. Genome Biol.5(4), R28 (2004).
  • Van Dessel W, Vandenbussche F, Staes M, Goris N, De Clercq K. Assessment of the diagnostic potential of immuno-RCA in 96-well ELISA plates for foot-and-mouth disease virus. J. Virol. Methods147(1), 151–156 (2008).
  • Kobori T, Matsumoto A, Takahashi H, Sugiyama S. Rolling circle amplification for signal enhancement in ovalbumin detection. Anal. Sci.25(12), 1381–1383 (2009).
  • Zhou L, Ou LJ, Chu X, Shen GL, Yu RQ. Aptamer-based rolling circle amplification: a platform for electrochemical detection of protein. Anal. Chem.79(19), 7492–7500 (2007).
  • Yan J, Song S, Li B et al. An on-nanoparticle rolling-circle amplification platform for ultrasensitive protein detection in biological fluids. Small6(22), 2520–2525 (2010).
  • Cao Y, Kopplow K, Liu GY. In situ immuno-PCR to detect antigens. Lancet356(9234), 1002–1003 (2000).
  • Gusev Y, Sparkowski J, Raghunathan A et al. Rolling circle amplification: a new approach to increase sensitivity for immunohistochemistry and flow cytometry. Am. J. Pathol.159(1), 63–69 (2001).
  • Fredriksson S, Gullberg M, Jarvius J et al. Protein detection using proximity-dependent DNA ligation assays. Nat. Biotechnol.20(5), 473–477 (2002).
  • Rebeski DE, Winger EM, Shin YK et al. Identification of unacceptable background caused by non-specific protein adsorption to the plastic surface of 96-well immunoassay plates using a standardized enzyme-linked immunosorbent assay procedure. J. Immunol. Methods226(1–2), 85–92 (1999).
  • Fredriksson S, Dixon W, Ji H, Koong AC, Mindrinos M, Davis RW. Multiplexed protein detection by proximity ligation for cancer biomarker validation. Nat. Methods4(4), 327–329 (2007).
  • Kim J, Hu J, Sollie RS, Easley CJ. Improvement of sensitivity and dynamic range in proximity ligation assays by asymmetric connector hybridization. Anal. Chem.82(16), 6976–6982 (2010).
  • Schallmeiner E, Oksanen E, Ericsson O et al. Sensitive protein detection via triple-binder proximity ligation assays. Nat. Methods4(2), 135–137 (2007).
  • Gustafsdottir SM, Schlingemann J, Rada-Iglesias A et al.In vitro analysis of DNA–protein interactions by proximity ligation. Proc. Natl Acad. Sci. USA104(9), 3067–3072 (2007).
  • Gustafsdottir SM, Nordengrahn A, Fredriksson S et al. Detection of individual microbial pathogens by proximity ligation. Clin. Chem.52(6), 1152–1160 (2006).
  • Darmanis S, Nong RY, Hammond M et al. Sensitive plasma protein analysis by microparticle-based proximity ligation assays. Mol. Cell Proteomics9(2), 327–335 (2010).
  • Kamali-Moghaddam M, Pettersson FE, Wu D et al. Sensitive detection of Abeta protofibrils by proximity ligation – relevance for Alzheimer’s disease. BMC Neurosci.11, 124 (2010).
  • Schlingemann J, Leijon M, Yacoub A et al. Novel means of viral antigen identification: improved detection of avian influenza viruses by proximity ligation. J. Virol. Methods163(1), 116–122 (2010).
  • Pai SS, Ellington AD. Using RNA aptamers and the proximity ligation assay for the detection of cell surface antigens. Methods Mol. Biol.504, 385–398 (2009).
  • Tavoosidana G, Ronquist G, Darmanis S et al. Multiple recognition assay reveals prostasomes as promising plasma biomarkers for prostate cancer. Proc. Natl Acad. Sci. USA108(21), 8809–8814 (2011).
  • Zhu C, Deng X, Shi F. Rapid detection of Brucella abortus by a novel proximity ligation-based loop-mediated isothermal amplification method. J. Rapid Methods Automation Microbiol.17(2), 154–163 (2009).
  • Fredriksson S, Horecka J, Brustugun OT et al. Multiplexed proximity ligation assays to profile putative plasma biomarkers relevant to pancreatic and ovarian cancer. Clin. Chem.54(3), 582–589 (2008).
  • Lundberg M, Thorsen SB, Assarsson E et al. Multiplexed homogeneous proximity ligation assays for high throughput protein biomarker research in serological material. Mol. Cell Proteomics10(4), M110.004978 (2011).
  • Darmanis S, Nong RY, Vänelid J et al. ProteinSeq: high-performance proteomic analyses by proximity ligation and next generation sequencing. PLoS ONE6(9), e25583 (2011).
  • Conze T, Carvalho AS, Landegren U et al. MUC2 mucin is a major carrier of the cancer-associated sialyl-Tn antigen in intestinal metaplasia and gastric carcinomas. Glycobiology20(2), 199–206 (2010).
  • Jarvius M, Paulsson J, Weibrecht I et al.In situ detection of phosphorylated platelet-derived growth factor receptor beta using a generalized proximity ligation method. Mol. Cell Proteomics6(9), 1500–1509 (2007).
  • Pinidiyaarachchi A, Zieba A, Allalou A, Pardali K, Wahlby C. A detailed analysis of 3D subcellular signal localization. Cytometry A75(4), 319–328 (2009).
  • Allalou A, Wahlby C. BlobFinder, a tool for fluorescence microscopy image cytometry. Comput. Methods Programs Biomed.94(1), 58–65 (2009).
  • Leuchowius KJ, Weibrecht I, Landegren U, Gedda L, Soderberg O. Flow cytometric in situ proximity ligation analyses of protein interactions and post-translational modification of the epidermal growth factor receptor family. Cytometry A75(10), 833–839 (2009).
  • Leuchowius KJ, Jarvius M, Wickstrom M et al. High content screening for inhibitors of protein interactions and post-translational modifications in primary cells by proximity ligation. Mol. Cell Proteomics9(1), 178–183 (2010).
  • Liu Y, Gu J, Hagner-Mcwhirter A et al. Western blotting via proximity ligation for high performance protein analysis. Mol. Cell Proteomics10(11), O111.011031 (2011).
  • Zieba A, Wahlby C, Hjelm F et al. Bright-field microscopy visualization of proteins and protein complexes by in situ proximity ligation with peroxidase detection. Clin. Chem.56(1), 99–110 (2010).
  • Clausson CM, Allalou A, Weibrecht I et al. Increasing the dynamic range of in situ PLA. Nat. Methods8(11), 892–893 (2011).
  • Aubele M, Spears M, Ludyga N et al.In situ quantification of HER2-protein tyrosine kinase 6 (PTK6) protein–protein complexes in paraffin sections from breast cancer tissues. Br. J. Cancer103(5), 663–667 (2010).
  • Di Giusto DA, Wlassoff WA, Gooding JJ, Messerle BA, King GC. Proximity extension of circular DNA aptamers with real-time protein detection. Nucleic Acids Res.33(6), e64 (2005).
  • Mcgregor LM, Gorin DJ, Dumelin CE, Liu DR. Interaction-dependent PCR: identification of ligand–target pairs from libraries of ligands and libraries of targets in a single solution-phase experiment. J. Am. Chem. Soc.132(44), 15522–15524 (2010).
  • Lundberg M, Eriksson A, Tran B, Assarsson E, Fredriksson S. Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood. Nucleic Acids Res.39(15), e102 (2011).
  • Rissin DM, Kan CW, Campbell TG et al. Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol.28(6), 595–599 (2010).
  • Todd J, Freese B, Lu A et al. Ultrasensitive flow-based immunoassays using single-molecule counting. Clin. Chem.53(11), 1990–1995 (2007).
  • Tessler LA, Reifenberger JG, Mitra RD. Protein quantification in complex mixtures by solid phase single-molecule counting. Anal. Chem.81(17), 7141–7148 (2009).
  • Uhlen M, Svahn HA. Affinity reagents for lab on chips. Lab. Chip11(8), 1417–1419 (2011).
  • Dubel S, Stoevesandt O, Taussig MJ, Hust M. Generating recombinant antibodies to the complete human proteome. Trends Biotechnol.28(7), 333–339 (2010).

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.