Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

Short Communication

SPECIAL ISSUE
Design of an Amperometric Biosensor for the Determination of
Biogenic Amines Using Screen Printed Carbon Working
Electrodes
Dietlind Telsnig,a Kurt Kalcher,b Andreas Leitner,a Astrid Ortner*a
a Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, Karl-Franzens-University, Humboldtstraße 46, 8010
Graz, Austria
b Institute of Chemistry, Department of Analytical Chemistry, Karl-Franzens-University, Stremayrgasse 16, 8010 Graz, Austria

*e-mail: astrid.ortner@uni-graz.at
Received: July 19, 2012
Accepted: September 5, 2012
Published online: October 16, 2012

Abstract
An amperometric biosensor with pea seedling amine oxidase and screen printed carbon electrode modified with
MnO2 for determining biogenic amines (BAs) was designed. The bio-component was immobilized with Nafion solu-
tion. The enzymatically produced hydrogen peroxide was determined to quantify BAs. All measurements were per-
formed in flow injection analysis system with phosphate buffer (66 mM, pH 7.5) as mobile phase at + 400 mV vs.
Ag/AgCl. A linear correlation was found for cadaverine and putrescine from 1–50 mM, for tyramine and histamine
from 10–300 mM respectively. The limits of detection were determined to be 0.3 mM for cadaverine and putrescine
and 3.0 mM for tyramine and histamine. The sensor was used to quantify BAs in chicken meat samples.

Keywords: Amperometric biosensor, Pea seedling amine oxidase, Screen printed carbon electrode, Biogenic amines

DOI: 10.1002/elan.201200378

BAs are low molecular weight organic bases. They are PSAO is a homodimeric protein, containing a Cu2 + ion
formed of amino acids during bacterial degradation such and a covalently bound 2,4,5-trihydroxyphenylalanine
as ripening, processing or putrefaction. When ingested in quinine (TPQ) at each active site. Due to its sensitivity
high amounts they can have negative health effects, espe- for the irreversible inhibitor semicarbazide PSAO belongs
cially in sensitive people [1]. BAs are thermally stable to the group of semicarbazide sensitive amine oxidases
and are therefore important marker substances for the (SSAOs).
freshness of food. According to the Commission Regula- Hydrogen peroxide is well approved as electrochemical
tion (EC) 2073/2005 the statutory limit of histamine is detecting agent and excellent results have been obtained
fixed to 200 mg/kg in fresh fish. However no limitation is using MnO2 as mediator [8].
given for other BAs. Previous experiments in this group have shown that
Due to their great importance in food industry differ- BAs can be quantified in biological samples using PSAO
ent approaches (HPLC and GC methods) have been as bio-component immobilized with Nafion films without
made to quantify them [2–4]. interfering matrix effects [9]. The enzymatically produced
Bio-sensing systems have proved to be very versatile hydrogen peroxide was then detected with a MnO2 modi-
tools for the determination of BAs since they generate fied carbon paste electrode. In these former investigations
a direct signal, allow fast analysis and do not require these electrodes were produced manually making them
a preceding sample preparation. Several publications are a valuable tool during the development of a new analyti-
dealing with bio-sensing systems to quantify BAs [4–7]. cal method. Their flexibility – which is desirable at this
PSAO is an herbal diamine oxidase extracted from pea point of development – can cause troubles with reprodu-
seedlings. It shows high selectivity for cadaverine and pu- cibility (varying composition, surface characteristics etc.).
trescine and mediocre selectivity for tyramine and hista- To overcome this drawback and to proceed with the opti-
mine. It catalyses the oxidative deamination of BAs ac- mization of this PSAO biosensor, a more stable working
cording to the following reaction equation: electrode was investigated – a SPCE which was produced
semi-automatically.
In the present work, a SPCE modified with 4 % MnO2
RCH2 NH2 þ O2 þ H2 O ! RCHO þ NH3 þ H2 O2
(m:m) and PSAO was optimized for the quantitative de-
termination of BAs. Amperometric measurements with
flow injection analysis (FIA) system were performed to

Electroanalysis 2013, 25, No. 1, 47 – 50  2013 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 47
Short Communication D. Telsnig et al.
SPECIAL ISSUE

Fig. 1. Typical amperogram of cadaverine. Injection of 50, 10, 5, 3, 2, and 1 mM (n = 6–7); mobile phase Sørensen phosphate buffer
(66 mM, pH 7.5), flow rate 1.00 mL/min, working potential + 400 mV vs. Ag/AgCl, injection volume 20 mL.

Table 1. Analytical parameters of the investigated substrates cadaverine, putrescine, tyramine, and histamine.
Analyte Linear range (mM) Slope R2 LOD (mM) LOQ (mM)
Cadaverine 1–50 8.279 0.9999 0.3 1.0
Putrescine 1–50 5.949 0.9999 0.3 1.0
Tyramine 10–300 0.428 0.9998 3.0 10.0
Histamine 10–300 0.455 0.9807 3.0 10.0

obtain data about substrate selectivity, analytical parame- cine, and 3 mM for tyramine and histamine, whereas the
ters, lifespan, and determination in complex, biological LOQs were estimated to be 1 mM for cadaverine and pu-
matrix. trescine and 10 mM for tyramine and histamine.
All measurements were performed with Sørensen phos- The analytical parameters for cadaverine, putrescine,
phate buffer (66 mM, pH 7.5) as mobile phase due to its tyramine, and histamine can be seen in Table 1.
physiological properties. Hydrogen peroxide was analysed The varying sensitivity of the sensor for the respective
at + 400 mV vs. Ag/AgCl. All measurements were per- substrate can be quantified by considering the electrode
formed with a flow rate of 1.00 mL/min. During the anal- surface as well as the concentration of the analyte. The
ysis 20 ml of the respective analyte were injected into the results can be seen in Table 2. The different sensitivities
carrier stream. are due to the selectivity of the enzyme towards the sub-
The obtained hydrogen peroxide signals were well strate.
shaped without any tailing. Due to a very low noise, the Special attention was paid to the repeatability of the
system proved to be very sensitive. results as well as the lifespan of the sensor. When not in
The designed sensor had excellent substrate selectivity use the sensor was stored at room temperature in the de-
for cadaverine and putrescine. Both amines showed tector. The sensor could be used for up to 40 runs (about
a linear dependence of the signal on the concentration 1400 injections) quantifying all four substrates. During
from 1–50 mM with a correlation coefficient of R2 = the first two days a significant decrease in sensitivity
0.9999. The linear correlation was calculated to be I could be observed followed by a long, comparatively
(nA) = 8.28c (mM) + 2.10 for cadaverine and I (nA) = stable period.
5.95c (mM) + 3.25 for putrescine. Tyramine and histamine
showed a linear correlation from 10–300 mM with a corre-
lation coefficient of R2 = 0.9998 for tyramine and 0.9807
Table 2. Sensitivity of the PSAO biosensor for the substrates ca-
for histamine. The linear correlation was calculated to be daverine, putrescine, tyramine, and histamine (n = 6).
I (nA) = 0.43c (mM) + 2.66 for tyramine and I (nA) =
0.46c (mM) + 18.84 for histamine. Figure 1 shows a typical Analyte Sensitivity (nA mM1 cm2) SD [a] (nA mM1 cm2)
amperogram of cadaverine. Cadaverine 12.5 3.0
Limits of detection (LOD) were determined as peak : Putrescine 9.0 2.6
noise ratio of 3 : 1 and limits of quantification (LOQ) Tyramine 0.65 0.2
Histamine 1.0 0.3
were estimated as the three-fold of LOD. The LODs
were determined to be 0.3 mM for cadaverine and putres- [a] Standard deviation

48 www.electroanalysis.wiley-vch.de  2013 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Electroanalysis 2013, 25, No. 1, 47 – 50
Amperometric Biosensor for Determination of Biogenic Amines

SPECIAL ISSUE
Fig. 2. Decrease of the cadaverine sensitivity of the sensor over time; mobile phase Sørensen phosphate buffer (66 mM, pH 7.5),
flow rate 1.00 mL/min, working potential + 400 mV vs. Ag/AgCl, injection volume 20 mL.

Figure 2 shows the decrease of the sensitivity for cadav- Thus the method was calibrated using the standard ad-
erine over a period of 15 days. dition method by adding different amounts of cadaverine
The repeatability and the reproducibility were deter- to the extracts (n = 3).
mined measuring cadaverine 10 mM (medium range). The Figure 3 shows the linear correlation calculated from
standard deviation of the intraday repeatability was calcu- the obtained signals. The correlation coefficient was cal-
lated to be 0.8 % (n = 6), whereas the standard deviation culated to be R2 = 0.9744.
of the interday repeatability was calculated to be 20 % The amount of BAs in the chicken meat sample was
(n = 6). If calculated without the first day of an electro- calculated to be 124 mg/kg ( 5 mg/kg) cadaverine equiv-
des lifespan, the interday repeatability was calculated to alents. This result is comparable to those published for
be 15 %. The reproducibility was determined by measur- food products (fish, pork, cheese) with values ranging
ing cadaverine 10 mM on 4 different electrodes. The stan- from 70–300 mg/kg [4, 10].
dard deviation of the reproducibility was calculated to be The designed PSAO biosensor proved to be a valuable
22 % (n = 4). analytical tool for the determination of BAs. Due to the
The sensor was tested for its applicability in complex optimization of the working electrode and the measure-
matrix by using extracts of fresh chicken meat as sample. ment system, the linear range was broadened and the sen-
Biological samples are known to contain different ratios sitivity was improved significantly compared to earlier
of BAs, such as cadaverine, putrescine, tyramine, and his- studies [9]. Extensive studies of the electrodes lifespan
tamine. The developed sensor determines the sum of the showed that one electrode could be used for up to
BAs present in the investigated extracts and displays 15 days and 1400 sample injections. The PSAO biosensor
them as cadaverine equivalents.

Fig. 3. Determination of BAs in chicken meat via standard addition method (n = 3); mobile phase Sørensen phosphate buffer
(66 mM, pH 7.5), flow rate 1.00 mL/min, working potential + 400 mV vs. Ag/AgCl, injection volume 20 mL.

Electroanalysis 2013, 25, No. 1, 47 – 50  2013 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.electroanalysis.wiley-vch.de 49
Short Communication D. Telsnig et al.

showed excellent results in complex matrix and is well The PSAO modified biosensor was prepared by drop-
SPECIAL ISSUE

suitable for the determination of BAs in food. coating 10 mL PSAO solution on the electrode surface
and allowing it to dry at room temperature. After that
the surface was covered with 10 mL Nafion solution in
Experimental
two steps by drop-coating with 5 mL respectively and al-
lowing it to dry.
All used chemicals and reagents were of analytical grade. The determination of BAs in chicken meat samples was
PSAO EC 1.4.3.6 was a generous gift from Bio-Research performed according to Carelli et al. [10]. Briefly, chicken
Product, Inc. Carbon ink (type C2030519P4) was pur- meat was homogenized using a common household blen-
chased from Gwent Electronic Materials Ltd. Manganese der shaft. 2.0 g meat was weighed in a centrifuge tube
dioxide (85–90 %), KH2PO4 (99 %), Na2HPO4·2H2O and 5.0 mL Sørensen phosphate buffer were added. The
(99.5 %), and ethanol (96 %) were obtained from Merck. sample was homogenized by vortexing for 1 min. The
Nafion 5 % solution (m:m) (perfluorinated, ion exchange sample was extracted by ultrasonication for 10 min. at
resin), cadaverine dihydrochloride, tyramine hydrochlo- room temperature using a Transsonic 660/H; Elma ultra-
ride, and histamine dihydrochloride (> 99 %) were pur- sonication bath. After that the tube was centrifuged at
chased from Sigma Aldrich. Glycerolum purum (> 98 %) 3500 rpm for 10 min at room temperature using a Haereus
was purchased from Fluka Chemie AG. Putrescine, was Instruments Labofuge 400 (d = 110 mm) to separate the
obtained from Acros Organics. Ceramic support plates residue from the liquid. The supernatant was transferred
(sintered aluminum oxide) for the production of the to a 20 mL volumetric flask and the extraction was re-
SPCEs were purchased from CoorsTek. The chicken meat peated two more times. The volumetric flask was filled up
was purchased at a local supermarket. to the mark with phosphate buffer and an aliquot was fil-
All aqueous solutions were prepared with highly pure tered through a 0.22 mm SCFA filter.
water, freshly prepared in the lab by a cartridge system
(Milli-Q).
Acknowledgement
Sørensen phosphate buffer (66 mM, pH 7.5) was pre-
pared by mixing a KH2PO4 solution (66 mM) and
The authors would like to thank Bio-research Product,
a Na2HPO4·2H2O solution (66 mM) until the required pH
Inc. for providing the PSAO.
was achieved. PSAO was dissolved in Sørensen phosphate
buffer with a concentration of 10 mg/mL aliquoted and
stored at 20 8C to avoid frequent freeze-thaw cycles. All
substrate solutions were prepared by dissolving the re-
References
agent in Sørensen phosphate buffer, stored at 4 8C and [1] A. Halsz, . Barth, L. Simon-Sarkadi, W. Holzapfel, Tr.
prepared freshly every two days. Nafion was neutralized Food Sci. Tech. 1994, 5, 42.
with tetrabutylammonium-hydroxid 40 % (m:m) before [2] C. Almeida, J. Fernandes, S. Cunha, Food Control 2012, 25,
use. 380.
All electrochemical measurements were carried out [3] S. Jia, Y. P. Kang, J. H. Park, J. Lee, S. W. Kwon, J. Chroma-
togr. A 2011, 1218, 9174.
with a Perkin Elmer/BAS Amperometric detector LC 4B [4] B. Bka, N. Adnyi, D. Virg, M. Sebela, A. Kiss, Electroa-
using a National Instruments 6120 AD converter and nalysis 2012, 24, 181.
a Gilson Minipuls 3 peristaltic pump with a Tygon R3607 [5] M. Fusco, R. Federico, A. Boffi, A. Macone, G. Favero, F.
tube (ID 1.3 mm, wall 0.86 mm). Data were analyzed Mazzei, Anal. Bioanal. Chem. 2011, 401, 707.
with LabView SignalExpress as well as PeakSimple soft- [6] M. A. Alonso-Lomillo, O. Domnguez-Renedo, P. Matos,
ware. The interpretation of the collected data was per- M. J. Arcos-Martnez, Anal. Chim. Acta 2010, 665, 26.
[7] L. Mureşan, R. Ronda Valera, I. Fr bort, I. C. Popescu, E.
formed with MS Excel.
Csçregi, M. Nistor, Microchim. Acta 2008, 163, 219.
The printing of the working electrodes was performed [8] K. Schachl, H. Alemu, K. Kalcher, J. Jezkova, I. Svancara,
using a semi-automatic screen printer (MLP USA). An K. Vytras, Anal. Lett. 1997, 30, 2655.
Ag/AgCl electrode and a stainless steel electrode were [9] D. Telsnig, A. Terzic, T. Krenn, V. Kassarnig, K. Kalcher, A.
used as reference and auxiliary electrode respectively. A Ortner, Int. J. Electrochem. Sci. 2012, 7, 6893.
spacer with a size of 3  22  0.254 mm was used to gener- [10] D. Carelli, D. Centonze, C. Palermo, M. Quinto, T. Rotun-
no, Biosens. Bioelectron. 2007, 23, 640.
ate a measuring chamber. The electrochemical cell was
put into a Faraday cage to prevent parasitic signals.

50 www.electroanalysis.wiley-vch.de  2013 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Electroanalysis 2013, 25, No. 1, 47 – 50

You might also like