14.01.2013 Views

Analytical Chemistry Chemical Cytometry Quantitates Superoxide

Analytical Chemistry Chemical Cytometry Quantitates Superoxide

Analytical Chemistry Chemical Cytometry Quantitates Superoxide

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Scheme 1. Top: Formulas of Purine Nucleobases (A,<br />

G) and Their 7-Deaza Analogues (A*, G*) a<br />

in primer extension (PEX) experiments. 12,13 Nevertheless, many<br />

of the nucleobase conjugates have displayed less facile incorporation<br />

at adjacent positions 7,9,10 and only some of them have been<br />

efficiently used in PCR. 13,14<br />

The 7-deazapurines, 7-deazaguanine (G*), or 7-deazaadenine<br />

(A*) (Scheme 1) are substitutes of standard purine nucleobases<br />

incorporable into DNA by PCR. Although the rate of G* and A*<br />

incorporation by DNA polymerases has been reported 15 to be<br />

lower, compared to the “parent” purines, both of these nucleobase<br />

analogues allow efficient sequence-specific DNA amplification by<br />

PCR and it has been possible to prepare PCR products with a<br />

high density of the corresponding modification. The 7-deazapurines<br />

are able to form Watson-Crick base pairs, maintaining<br />

pairing specificity of the respective natural nucleobases, but cannot<br />

form Hoogsteen pairs due to absence of the N7 atom (which is<br />

substituted by CH group, Scheme 1). Thus, the 7-deazapurines<br />

cannot be involved in triplex and tetraplex DNA structures. 16<br />

Reduced tendency of the “deaza-modified” DNA to adopting<br />

multistranded conformations has been utilized to improve PCR<br />

amplification of G-rich sequences such as (CGG)n repeat, 17 the<br />

length of which is analyzed during molecular diagnostics of<br />

fragile X syndrome. Similarly, 7-deaza-8-azaguanine has been<br />

used to create G-rich DNA probes with reduced propensity to<br />

aggregate and improved specificity. 18 Absence of the nitrogen<br />

atom at the 7-position in G* was also utilized in a study of<br />

sequence-specificity of DNA modification with cisplatin 19 and<br />

in studies of echinomycin-DNA interaction modes. 20 Moreover,<br />

DNA substituted with G* or A* has been shown to resist<br />

cleavage with certain endonucleases. 15<br />

a<br />

Atoms at 7-position are highlighted in red. Bottom: Watson-Crick<br />

and Hoogsteen base pairing. N7 is involved in the Hoogsteen pairing<br />

of natural purines.<br />

(13) Cahova, H.; Pohl, R.; Bednarova, L.; Novakova, K.; Cvacka, J.; Hocek, M.<br />

Org. Biomol. Chem. 2008, 6, 3657–3660.<br />

(14) Raindlova, V.; Pohl, R.; Sanda, M.; Hocek, M. Angew. Chem., Int. Ed. 2010,<br />

49, 1064–1066.<br />

(15) Seela, F.; Roling, A. Nucleic Acids Res. 1992, 20, 55–61.<br />

(16) Palecek, E. Crit. Rev. Biochem. Mol. Biol. 1991, 26, 151–226.<br />

(17) Cao, J.; Tarleton, J.; Barberio, D.; Davidow, L. S. Mol. Cell. Probes 1994,<br />

8, 177–180.<br />

(18) Kutyavin, I. V.; Lokhov, S. G.; Afonina, I. A.; Dempcy, R.; Gall, A. A.; Gorn,<br />

V. V.; Lukhtanov, E.; Metcalf, M.; Mills, A.; Reed, M. W.; Sanders, S.;<br />

Shishkina, I.; Vermeulen, N. M. J. Nucleic Acids Res. 2002, 30, 4952–4959.<br />

(19) Cairns, M. J.; Murray, V. Biochim. Biophys. Acta 1994, 1218, 315–321.<br />

(20) Sayers, E. W.; Waring, M. J. Biochemistry 1993, 32, 9094–9107.<br />

6808 <strong>Analytical</strong> <strong>Chemistry</strong>, Vol. 82, No. 16, August 15, 2010<br />

Table 1. Oligonucleotides Used in This Work a<br />

prim rnd 5′-CATGGGCGGCATGGG-3′<br />

prim noG 5′-TACTCATCATATCAA-3′<br />

temp rnd16 5′-CTAGCATGAGCTCAGTCCCATGCCGCCCATG-3′<br />

temp noG 5′-AATATAAATATATTGATATGATGAGTA-3′<br />

p53-for 5′-GAGGTTGTGAGGCGCTGCCC-3′<br />

p53-rev 5′-TCCTCTGTGCGCCGGTCTCT-3′<br />

a The template strands (temp rnd16 , temp noG ) used in PEX experiments<br />

were 5′end-biotinylated.<br />

Compared to the standard purines, 7-deazapurines exhibit<br />

significantly lower potentials of their oxidation. 9,21,22 G* is thus<br />

easily photooxidized by intercalated ethidium and this photooxidation<br />

has been interrogated as a function of distance, nucleotide<br />

sequence and integrity of π-stacking in studies of DNA-mediated<br />

charge transfer. 21 The G* ability of being selectively oxidized by<br />

a redox mediator with relatively low redox potential, such as<br />

Ru(dmb)3 3+/2+ (dmb )4,4′-dimethyl-2,2′-bipyridine), compared<br />

to A* together with natural G requiring stronger oxidants, such<br />

as Ru(bpy)3 3+/2+ (bpy )2,2′-bipyridine), has been utilized in a<br />

PCR-coupled electrochemical technique proposed for parallel<br />

detection of two genes. 22 Mediated electrooxidation of G* (or<br />

G) was also applied in an indirect electrochemical real time<br />

monitoring of PCR via measuring consumption of the respective<br />

dNTPs. 23 To our best knowledge, direct electrochemical<br />

analysis of DNA with incorporated G* or A* as electrochemically<br />

oxidizable tags has not been reported to date.<br />

In this paper we report on adsorptive transfer stripping<br />

voltammetric analysis of DNA with enzymatically incorporated G*<br />

or A* residues at a carbon electrode. We show that, particularly<br />

G* producing a specific signal separated from those yielded by<br />

natural DNA components, can be utilized as an excellent electroactive<br />

label suitable for monitoring of DNA amplification by PCR.<br />

MATERIALS AND METHODS<br />

Material. Synthetic ODNs (Table 1) were purchased from<br />

VBC genomics (Austria). Templates used in experiments involving<br />

the magnetoseparation procedure were biotinylated at their 5′<br />

ends. Plasmid pT77 bearing wild type p53 cDNA insert 24 (used<br />

as primary template for PCR amplification of the 347-bp fragment)<br />

was isolated from E. coli cells using Qiagen Plasmid Purification<br />

Kit and linearized with EcoR I restrictase (Takara). Streptavidincoated<br />

magnetic beads (MBstv) were purchased from Novagen<br />

(Germany), DyNAzyme II DNA Polymerase from Finnzymes<br />

(Finland), Pfu DNA Polymerase from Promega (U.S.), unmodified<br />

nucleoside triphosphates (dATP, dTTP, dCTP and dGTP),<br />

SYBR Green I, SYBR Gold and Stains-All reagent from Sigma,<br />

7-deaza-dGTP and 7-deaza-dATP from Jena Bioscience. Other<br />

chemicals were of analytical grade.<br />

Primer Extension (PEX). The primer (0.7 µM) was mixed<br />

with corresponding template ODN (0.7 µM), dNTPs (100 µM<br />

each; composition of the dNTP is specified in the text and Figure<br />

legends for individual experiments) and the DyNAzyme II DNA<br />

(21) Kelley, S. O.; Barton, J. K. Chem. Biol. 1998, 5, 413–425.<br />

(22) Yang, I. V.; Ropp, P. A.; Thorp, H. H. Anal. Chem. 2002, 74, 347–354.<br />

(23) Defever, T.; Druet, M.; Rochelet-Dequaire, M.; Joannes, M.; Grossiord, C.;<br />

Limoges, B.; Marchal, D. J. Am. Chem. Soc. 2009, 131, 11433–11441.<br />

(24) Hupp, T. R.; Meek, D. W.; Midgley, C. A.; Lane, D. P. Cell 1992, 71, 875–<br />

886.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!