You are on page 1of 13

Biosensors and Bioelectronics 102 (2018) 540–552

Contents lists available at ScienceDirect

Biosensors and Bioelectronics


journal homepage: www.elsevier.com/locate/bios

Conducting polymer-based electrochemical biosensors for T


neurotransmitters: A review

Jong-Min Moona,1, Neeta Thapliyalb,1, Khalil Khadim Hussaina, Rajendra N. Goyalc, ,

Yoon-Bo Shima,
a
Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea
b
Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
c
Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India

A R T I C L E I N F O A B S T R A C T

Keywords: Neurotransmitters are important biochemical molecules that control behavioral and physiological functions in
Neurotransmitters central and peripheral nervous system. Therefore, the analysis of neurotransmitters in biological samples has a
Conducting polymers great clinical and pharmaceutical importance. To date, various methods have been developed for their assay. Of
Neurotransmitter sensors the various methods, the electrochemical sensors demonstrated the potential of being robust, selective, sensitive,
Conducting polymer composites
and real time measurements. Recently, conducting polymers (CPs) and their composites have been widely
Biosensors
employed in the fabrication of various electrochemical sensors for the determination of neurotransmitters.
Hence, this review presents a brief introduction to the electrochemical biosensors, with the detailed discussion
on recent trends in the development and applications of electrochemical neurotransmitter sensors based on CPs
and their composites. The review covers the sensing principle of prime neurotransmitters, including glutamate,
aspartate, tyrosine, epinephrine, norepinephrine, dopamine, serotonin, histamine, choline, acetylcholine, ni-
trogen monoxide, and hydrogen sulfide. In addition, the combination with other analytical techniques was also
highlighted. Detection challenges and future prospective of the neurotransmitter sensors were discussed for the
development of biomedical and healthcare applications.

1. Introduction to electrochemical biosensors enzymes, cells or tissue slices that identify the target analyte and gen-
erate electroactive molecules. By contrast, affinity sensors depend on a
Biosensors are analytical devices composed of biological sensing specific binding interaction between the analyte and an immobilized
elements that are capable of producing sensitive and selective analy- biological elements such as an antibody or aptamer (Wang, 2006). The
tical signals. The physiochemical changes due to the interactions be- performance evaluation of biosensor is mainly based on its sensitivity,
tween a target and the corresponding biorecognition elements, such as limit of detection (LOD), linear dynamic range, reproducibility, se-
enzymes, proteins, and antibodies, are converted into quantifiable lectivity, response to interferences, and other features.
electronic signals by a transducer, where the amount of signal gener- Electrochemical biosensors are part of the electrochemical cell that
ated is directly related to the analyte concentration. Biosensors are used consists of either three or two electrodes. A typical three electrode
in various applications, such as medical diagnosis, drug discovery, food system consists of a working, a reference, and a counter electrode. The
safety, and environmental monitoring. Various types of biosensors have working electrode consists of a chemically stable solid conductive
been reported, including optical, mechanical, colorimetric, piezo-elec- material, such as platinum, gold, or carbon; the reference electrode
tric, and electrochemical biosensors (Turner et al., 1987). According to usually consists of silver metal coated with a layer of silver chloride
the International Union of Pure and Applied Chemistry (IUPAC), bio- (Ag/AgCl); and a platinum wire is typically used as the auxiliary elec-
sensors can be classified in two ways based on their composition: 1) trode. In contrast, a two-electrode system consists of working and re-
transducers including mass-based, electrochemical, and optical bio- ference electrodes only. Electrochemical techniques can be classified
sensors, or 2) type of bioelements including biocatalytic and affinity into three main categories based on the types of measurements (Bard
sensors. Biocatalytic sensors are fabricated by immobilization of et al., 1980, Wang et al., 2001): (1) current (voltammetric and


Corresponding authors.
E-mail addresses: rngcyfcy@iitr.ac.in (R.N. Goyal), ybshim@pusan.ac.kr (Y.-B. Shim).
1
These authors equally contributed.

https://doi.org/10.1016/j.bios.2017.11.069
Received 1 September 2017; Received in revised form 25 November 2017; Accepted 29 November 2017
0956-5663/ © 2017 Elsevier B.V. All rights reserved.
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

amperometric), (2) potential difference (potentiometry), and (3) im- Table 1


pedance (electrochemical impedance spectroscopy). Of them, bio- Classification of neurotransmitters, associated diseases and chemical structures.
sensors based on current measurements are mostly common in use.
Category Analysts Associated diseases Chemical structure
Current measurement based sensors are characterized by applying a
potential to a working electrode versus a reference electrode and mea- Amino acid Glutamic acid Seizures, neural
suring the current. The current is a result of electrolysis due to the degeneration, lethargy
and cognitive dysfunction
electrochemical reduction or oxidation at the surface of the working
Aspartic acid Stroke, chronic fatigue
electrode. However, this process depends not only on the mass trans-
syndrome, depression,
port rate of molecules to the electrode but also on the electron transfer Huntington’s disease
rate at the electrode surface. There are various types of voltammetric
Tyrosine Parkinson’s disease,
methods such as linear sweep, cyclic, hydrodynamic, differential pulse, behavioral deficit
square-wave, and stripping voltammetry. In voltammetry, a potential is
scanned over a set potential range, and the current response is generally Biogenic Nor- Schizophrenia,
a peak or a plateau that is proportional to the concentration of analyte amines epinephrine depression, ADD
in the measured sample. However, in amperometry, a constant poten- (Attention deficit
disorder)
tial is applied at the working electrode with respect to the reference
Epinephrine Depression, Addison’s
electrode, and the changes in current caused by the electrochemical disease, palpitation, high
reduction or oxidation are directly monitored with respect to time. blood pressure
Overall, amperometric biosensors are more selective and sensitive be- Dopamine Tourette’s disease,
cause the technique relies on a constant specific potential for a given schizophrenia, psychosis,
analyte, therefore, it can minimize the interference effect of other depression, Parkinson’s
electroactive substances. Potentiometric sensors are based on mea- disease, ADD
Serotonin Depression, anxiety
suring the potential of an electrochemical cell while drawing a negli-
disorders, especially
gible current. They function under equilibrium conditions and monitor obsessive-compulsive
the accumulation of charge, at zero current, that is caused by selective disorder
binding at the electrode surface. Such chemical sensors can be turned Histamine Immune system disorder,
into biosensors by coating them with a biological element, like an en- schizophrenia,
zyme, that catalyzes a reaction to form ions which sensed at the mod- convulsion, seizure, and
Parkinson’s disease
ified electrode when the ions bind to suitable ion exchange membrane.
Acetyl Acetyl choline, Depression, Alzheimer’s
The potentiometric sensors are non-invasive, real time, low cost, and choline choline disease, dementia
ease of miniaturization. Electrochemical impedance spectroscopy (EIS)
measures the resistances and capacitances of the materials by a small
amplitude sinusoidal AC excitation signal. The frequency is varied over
Soluble Nitric oxide Huntington’s, Alzheimer’s
a wide range to produces the impedance spectrum. The in-phase and gases Parkinson’s disease,
out-phase AC current responses are then determined to obtain the re- vascular stroke.
sistive and capacitive components of the impedance, respectively. Im- Hydrogen Down syndrome, Chronic
pedance methods enable the monitoring of charge transfer at high sulfide obstructive pulmonary
disease
frequency and mass transfer at low frequency; therefore, impedimetric
detection techniques are primarily used with affinity-type biosensors
(Turner et al., 1987; Skoog et al., 1998). Various aspects of these NTs are summarized in Table 1. Moreover, NTs
play a key role in the functioning of the brain and control various be-
2. Neurotransmitter sensors havioral and physiological conditions that affect daily life, for example,
learning, memory, sleeping, consciousness, mood and the regulation of
Neurotransmitters (NTs) are endogenous chemicals that are in- muscle tone, heart rate and blood pressure (Tomkins and Sellers, 2001;
volved in the transmission of signals from one neuron to another or Dunn and Dishman, 1991; Freed and Yamamoto, 1985). Variations in
between non-neuronal body cells across chemical synapses exchanging the production, secretion, uptake and/or metabolism of these chemicals
information throughout the brain and body (Patestas and Gartner, may lead to various mental and physical disorders, such as Hunting-
2006). Produced by glands such as the pituitary, pineal and adrenal ton’s, Alzheimer’s, Parkinson’s diseases, schizophrenia, epilepsy,
glands, NTs are stored in vesicles clustered at neuronal terminals. An thyroid hormone deficiency, glaucoma, congestive heart failure, and
action potential at a synapse stimulates the release of NTs, which cross different types of cancers. Hence, timely and accurate determination of
the synaptic gap to reach the receptor site of the other neuron or cell, NTs level in various physiological media (such as urine, plasma, and
where they are reabsorbed. A new action potential is then created at the cerebral fluids) is imperative for effective diagnosis, monitoring of the
axon terminal of the next neuron followed by a similar release of NTs disease, therapeutic interventions as well as to understand the role of
subsequently, communicating information to another adjacent neuron. these chemicals in brain functions. Various analytical tools has been
Thus, a complex cascade is initiated via neurons that eventually elicits a reported for the quantification of NTs in biological matrices. These
biological response. Since the discovery of the first neurotransmitter in include mass spectroscopy, fluorimetry, chemiluminescence, chroma-
1921, more than one hundred chemical messengers have been identi- tography, and capillary electrophoresis (Zhang et al., 2007; Santos-
fied, which are involved in synaptic transmission (Yamada et al., 1998). Fandila et al., 2013; De Benedetto et al., 2014; Wang et al., 2012; Li
NTs can be classified according to their 1) molecular structure; 2) mode et al., 2011; Zhao and Suo, 2008; Lapainis et al., 2009). Most of them
of action either direct or as neuromodulator; and 3) physiological are complex, expensive, require tedious procedures, and suffer from
function either excitatory or inhibitory. However, in this review, the poor sensitivity and selectivity. By contrast, electrochemical methods
classification is based on their chemical structure, therefore they are are known to provide low-cost, simple, sensitive, fast response time and
divided into four groups: amino acids primarily (glutamic acid, aspartic selective determination of various biological species. The advent of
acid, and tyrosine), biogenic amines (epinephrine, nor-epinephrine, chemically modified electrodes brought rapid improvements in the field
dopamine, serotonin and histamine), acetyl choline (acetyl-choline and of electroanalysis, meeting the higher demands for sensitivity and
choline), and soluble gases (such as nitric oxide and hydrogen sulfide).

541
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

selectivity (Durst, 1997). The distinct property of a chemically modified neurotransmitters sensors including amino acids primarily (glutamic
electrode is that the specific material immobilized on the electrode acid, aspartic acid, and tyrosine), biogenic amines (epinephrine, nor-
surface conveys its chemical, electrochemical, and electrical char- epinephrine, dopamine, serotonin and histamine), acetyl choline
acteristics as well as other desirable properties. Numerous materials, (acetyl- choline and choline), and soluble gases (such as nitric oxide and
such as metal and metal oxide nanoparticles, carbon nanotubes, bio- hydrogen sulfide).
molecules and CPs, have been used for electrode modification (Adams,
1969). Among these, CPs and composites have attracted considerable 3.1. Amino acid neurotransmitters
attention in recent years.
The first electrochemical preparation and characterization of an Amino acids constitute the major group of neurotransmitters pre-
organic π-conjugated polymer (polyaniline) was reported by Letheby in sent in the central nervous system (CNS), providing the majority of
1862 (Letheby, 1862). After that, the conductivity of polyacetylene was inhibitory and excitatory neurotransmission in the body. All amino acid
studied and found to be enhanced 10-folds with the use of iodine vapors neurotransmitters (except γ-aminobutyric acid) are derived from in-
(Shirakawa et al., 1977; Basescu et al., 1987; Chiang et al., 1977). Since termediary metabolism. The three main amino acid neurotransmitters
then, various types of CPs have been synthesized and applied in various in the human system are glutamate, aspartate, and tyrosine.
fields. Due to the groundbreaking work of Heeger, MacDiarmid, and
Shirakawa on CPs, these researchers were jointly awarded with the 3.1.1. Glutamate
Nobel Prize in Chemistry in 2000. Early research on CPs was reported Glutamate is the key excitatory neurotransmitter distributed
by (Diaz et al., 1981) who proposed the original mechanism of poly- throughout all parts of the mammalian CNS. Glutamate is actively in-
pyrrole formation. Similarly, Park’s group extensively studied the volved in normal brain functions, such as memory, learning, and cog-
electrochemistry of CPs; he reported the autocatalytic growth me- nition (Nedergaard et al., 2002; Niswender and Conn, 2010). It med-
chanism of polyaniline along with its growth kinetics on the electrode iates information that determines the survival, differentiation and
surface (Shim and Park, 1989; Shim et al., 1990; Stilwell and Park, elimination of body cells as well as the formation and elimination of
1988, 1989). CPs have received considerable attention in recent years synapses. Glutamate is present in the extracellular fluids of the brain at
due to their extensive potential applications in the fields of batteries, the micro molar level. However, its concentration is thousand times
sensors, solar cells, electrochromic devices, etc. (Park, 1997; Skotheim higher inside the brain neurons. Even a slight increase or decrease in
et al., 2006; Rahman et al., 2008a; Kim et al., 2012; Beaujuge and the concentration of glutamate is harmful, and such discrepancies are
Reynolds, 2010). One of the most practical applications of CPs is in the implicated in several glial associated neurological diseases, such as
fabrication of modified electrodes for developing chemical sensors and schizophrenia, amyotrophic lateral sclerosis, epilepsy, ischemia, Par-
biosensors. CPs are highly sensitive with even slight modulations at kinson’s and Alzheimer’s diseases. Overstimulation of glutamate re-
their surface leading to changes in their electrochemical activity. CPs ceptors may lead to neuronal death through a process of excitotoxicity.
are commonly used to modify electrode surfaces; however, polypyrrole Glutamate functions by binding to specific receptors present on cell
(PPy) and polyaniline (PANI) are not highly stable in air (Shim et al., surface. These receptors possess glutamate-binding sites, where gluta-
1990; Park et al., 1993) than polythiophene (Arbizzani et al., 1997) and mate binds to the receptor and subsequently excites the cell by causing
polyterthiophene derivatives (Blanchared et al., 2006; Heffries-Ml and an influx of positive ions into the cell. This increases the electrical
McCullough, 2006, Lee and Shim, 2001). The electrical conductivity of charge of the cell, which causes neuronal changes resulting in the re-
these polymers can be controlled over a wide range by proper doping/ lease of many neurotransmitters.
de-doping with suitable dopants (Heeger et al., 1988; Lee et al., 1992). In-vivo amperometric glutamate biosensors have been developed
Additionally, CP-modified electrodes display wide potential windows through the covalent immobilization of glutamate oxidase (GluOx) on
thus, they show prospect to catalyze electrochemical reactions that poly (5,2′:5′,2″-terthiophene-3′-carboxylic acid) (pTTCA) (Rahman
have poor selectivity and high over-potential. Despite these unique et al., 2005; Lee et al., 2008). The steps of sensor fabrication are shown
electrical properties, biosensors prepared with pure CPs are somewhat in Fig. 2 (A). A clear oxidation peak was observed at +0.45 V versus
limited of low sensitivity and selectivity as well as poor electron Ag/AgCl, when the CV was recorded in a PBS solution containing
transfer between a target molecule and the electrode surface. Thus, glutamate. The anodic peak was related to the catalytic oxidation of
functionalized CPs and their nanocomposites with metal nanoparticles hydrogen peroxide (H2O2), which was generated by the GluOx. The
(Rahman et al., 2008b; Kim et al., 2009; Naveen et al., 2016; Noh and proposed sensor enabled monitoring of real-time changes in glutamate
Shim, 2016), carbon (Shrivastava et al., 2016), and various organic concentration with respect to cocaine exposure. Among the interference
materials (Kwon et al., 2006), which offer high electrical conductivity, agents ascorbate and dopamine were found to interfere with the elec-
sensitivity, effective surface area, and facile electron transfer. In addi- trochemical response to glutamate. However, the observed interference
tion, CPs and their composites are considered as biocompatible mate- was minimized by co-immobilizing ascorbate oxidase and poly-
rials in biological system due to their less/nontoxic effect, which make ethyleneimine (a cationic polymer) at the sensor surface. The method
them an ideal choice for biosensor fabrication. Various types of poly- was successfully used for the in-vivo monitoring of the extracellular
mers and composites are summarized in Fig. 1. Over the past few glutamate released by cocaine stimulation. Similarly, approaches have
decades, several electrochemical biosensors based on CP nanocompo- been reported, such as an array of platinum microelectrodes modified
sites has been published in numerous journals with major contribution with thin overoxidized PPy and Nafion film (Tseng and Monbouquette,
in electroanalytical and analytical chemistry aiming towards the de- 2012) and microneedle biosensor modified with a GluOx-functionalized
termination of a wide range of compounds. Considering the abundant poly-(o-phenylenediamine) film (Windmiller et al., 2011).
and highly scattered information in the literature, it has become chal- A glutamate biosensor was constructed by sequential modification
lenging to a broad overview of this research area with a focus on the of polyethyleneimine (PEI), GluOx, and poly-ortho-pheylenediamine
electroanalysis of NTs. The present review attempts to outline state-of- (PPD) onto Pt wire (McMahon et al., 2007). The incorporation of PEI
the-art methodologies and recent advancements made in the last few into the PPD/GluOx-based sensor exhibited a significant improvement
years regarding the use of CPs and their composites for the detection in sensing performance. A highly sensitive and selective glutamate
and determination of various important neurotransmitters (Table 2). micro biosensor was developed using PPy, multi-walled carbon nano-
tubes (MWCNTs), and GluOx on a Pt electrode (Ammam and Fransaer,
3. Electrochemical neurotransmitter sensors 2010), where polyurethane at the outer membrane was exploited to
improve the linearity and stability of the sensor. A meta-phenylene-
The following section describes in detail various types of diamine polymer-modified amperometric biosensor was developed

542
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Fig. 1. Schematic representation of various


types of conducting polymers and composite
materials.

using GluOx for the detection of glutamate in liquid samples of isolated acetic acid) electropolymerized onto MWCNTs coated pencil graphite
rat brain nerve terminals (synaptosomes) (Soldatkin et al., 2015). An electrode (Prasad and Pandey, 2013). The developed sensor enabled the
organic electrochemical transistor was reported for the determination successful analysis of ultra-trace amount of aspartate in real samples
of glutamate (Kergoat et al., 2014). The sensor was coated with a (human blood serum and pharmaceutical samples) without any matrix
composite film consisting of a polymeric mixture of poly(3,4-ethyle- complications or interference.
nedioxythiophene) polystyrene sulfonate (PEDOT:PSS) and platinum
nanoparticles (PtNPs) for the enzymatic detection of glutamate, as 3.1.3. Tyrosine
shown in Fig. 2 (B). Tyrosine is a nonessential amino acid that is produced from phe-
In addition, various in-vivo micro biosensors have also been reported nylalanine. It is the precursor to three important catecholamine neu-
for the detection of glutamate, based on poly(amidoamine)-en- rotransmitters (norepinephrine, epinephrine, and dopamine) that affect
capsulated Pt nanoparticles composites (Yu et al., 2011), Pt wire with a focus, mood, memory and alertness. Tyrosine is important for the
permselective film of electropolymerized, overoxidized polypyrrole functioning of organs responsible for producing and regulating hor-
(OPP) (Hamdi et al., 2006), silica-based platinum electrode, modified mones, including the adrenal, thyroid, and pituitary glands. It also in-
with various types of polymers, such as Nafion, polyphenylenediamine, volved in the production of melanin (Slominski et al., 1988). Abnormal
PPy, PANI, and polynaphthol. These common permselective mem- tyrosine levels are closely related to various human diseases, such as
branes were compared in term of sensitivity for hydrogen peroxide that dementia, Parkinson’s disease, hypochondria, depression, albinism, and
generated from the GluOx reaction (Wahono et al., 2012). Of them, alkaptonuria (Tashkhourian et al., 2016; Xu and Wang, 2005).
polyphenylenediamine based sensor showed superior results compared A differential pulse voltammetric (DPV) method was employed for
to other polymers. Further, thermoelectric label-free glutamate bio- the simultaneous analysis of tyrosine, piroxicam, ascorbic acid (AA) and
sensor combined with microfluidic device was reported (Kopparthy uric acid (UA) using a rGO decorated 3′-(2-aminopyrimidyl)-2,2′:5′,2”-
et al., 2015). The thermoelectric sensor measured the heat, which is terthiophene polymer modified glassy carbon electrode (GCE) (Noh
yielded from biochemical reactions (oxidative deamination of gluta- et al., 2014) without any interference from other biological species. The
mate). sensor probe was validated by detecting tyrosine in human urine
samples. An in situ copper oxide modified molecular imprinted poly-
3.1.2. Aspartate pyrrole (MIPPy) sensor was designed for the determination of tyrosine
Aspartate, the conjugate base of aspartic acid, is an excitatory in aqueous media (Saumya et al., 2011). The proposed sensor exhibited
neurotransmitter present in brain with high concentrations. Generally, a significant enhancement of the analytical signal in response to tyr-
a small portion of aspartate (stored intracellularly up to 100 mM), and osine oxidation by incorporation of copper oxide onto the MIPPy film.
the level of aspartate in the extracellular fluid of CNS range from 1 to Moreover, the addition of copper (II) resulted in higher response, which
3 µM. The mechanism of aspartate as a neurotransmitter is still unclear. is attributed by the formation of copper tyrosine phosphate complex in
However, it is claimed that the neurochemical is released from certain the solution. The applicability was examined in human urine samples.
synapses in a Ca2+-dependent manner following the depolarization of Because the electroactive tyrosine plays an important receptor of
nerve endings with K+ ions and is taken up into neurons by the same phosphorylation in proteins, few label-free tyrosine sensors were also
carriers as for glutamate. Aspartate is reported to be involved in fabricated using electron transfer mediator (Qu et al., 2008), DNA-
learning, memory, drug addiction, movement disorders, and other AuNPs (Xu et al., 2009), and graphene (Li et al., 2013).
physiological behaviors and processes (Riedel et al., 2003).
Recently, few methods have been reported for the electroanalysis of 3.2. Biogenic amine neurotransmitters
aspartate. A molecular imprinted polymer (MIP) was fabricated by
electropolymerization of PPy in the presence of L-aspartic acid (Syritski The biogenic amine neurotransmitters are simple molecules that
et al., 2008). The developed MIP sensor was based on an electro- play critical roles in the regulation of the central and peripheral nervous
chemical quartz crystal microbalance that exhibited 20-fold higher systems. There are five well-known naturally occurring biologically
sensitivity to L-aspartic acid than to D-aspartic acid in a strongly acidic active amine neurotransmitters; the three catecholamines (dopamine,
medium (pH 1.6), because L-aspartic acid dominantly exists as a cation epinephrine and norepinephrine), histamine and serotonin.
in acidic solution. Also, negative potential assisted the uptake of L-as-
partate cations into the MIP layer. Another MIP sensor for the en- 3.2.1. Epinephrine
antioselective analysis of L-aspartic acid proposed with poly (indole-3- Epinephrine (EP) is an important inhibitory catecholamine

543
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Table 2
A comparison of the validation parameters of conducting polymer-based sensors for the detection of various neurotransmitters.

Neurotransmitter Electrode Method Linear Range LOD (in µM) Matrix Ref
(in µM)

Glutamate GluOx/pTTCA/Pt Amperometry 0.2-100 0.1 Rat brain Rahman et al., 2005, Lee
et al., 2008
GluOx-BSA/Nafion/OPPy/Pt micro electrode Amperometry upto 630 2.1 – Tseng and Monbouquette,
2012
GluOx/functionalized PPD/microneedle array Amperometry upto140 3.0 Human serum Windmiller et al., 2011
electrode
PPD/GluOx/PEI/Pt electrode Amperometry 5.0-150 0.02 – Mcmahon et al., 2007
PU/GluOx/MWCNT/PPy/Pt electrode Amperometry 0.3-500 0.3 – Ammam and Fransaer, 2010
GluOx/PPD/Pt electrode Amperometry 2.0-800 0.5 Rat brain nerve Soldatkin et al., 2015
terminals
GluOx/PEDOT:PSS/Pt NPs/OECD Amperometry 0.9-14 0.5 – Kergoat et al., 2014
Aspartate MIP/oPPy/Au electrode QCM – – – Syritski et al., 2008
MI-PI3AA/ MWCNTs-PG electrode DPASV 0.15-7.4 0.025 Human serum Prasad and Pandey, 2013
0.15-8.64 0.016 Pharmaceutical
formulations
Tyrosine MSNs/CPE DPV 0.5-600 0.15 Artificial urine Tashkhourian et al., 2016
MWCNT/GC SWV 2.0-500 0.4 – Xu and Wang, 2005
p-APT/rGO/GC DPV 50-600 5.97 Human urine Noh et al., 2014
MIPPy /GC DPV 0.01-1 and 2-8 0.004 Human urine Saumya et al., 2011
Epinephrine PPy/AuNPs/SWCNTs/Au DPV 0.004-0.10 0.002 Injections Lu et al., 2011
Electrode Human serum, plasma,
and urine
SDBS-doped polypyrrole /MWCNT DPV 0.1-8.0 and 0.04 Injection Shahrokhian and Saberi,
10-100 2011
2+
Cu -PANI-Nano-ZSM-5/GC DPV 0.01-600 0.004 Injection Kaur and Srivastava, 2015a
Au-MWCNT-PANI-TiO2 and Au-MWCNT- CV 4.9-76.9 0.16 and 0.18 Pharmaceutical samples Tsele et al., 2017
PANI-RuO2
Polythionine/AuNPs/GC DPV 5.5-218 1.6 Serum Huang et al., 2016
Norepinephrine pAcrylic acid/PAA Amperometry – – – Alvarez et al., 2012
microchannel
RGD peptide on inkjet-printed PANi Amperometry 0.002 0.002-1 PC12 cells Oh et al., 2013
p-AMT/GC Amperometry 0.01-100 0.00002 Human plasma Revin and John, 2012
PDAN/Pt electrode DPV 9.90-90.9 1.82 Pharmaceutical Guedes et al., 2011
Formulation
Dopamine F3GA/PDAN/GC LSV 5.0-100 0.1 Human urine Abdelwahab et al., 2009
GO/AuNPs/pDAN-EDTA Amperometry 0.01-1 0.005 PC12 cells Mir et al., 2015
polyDAN-RB4/GC DPV 0.1-1500 0.061 Human serum and urine Chandra et al., 2013
PEDOT:PSS/ITO DPV 1-50 6.84 – Pal et al., 2017
Nf/Cu-MPS/GC SWV 0.08-5.0 0.05 – Won et al., 2005
GR/pAHWSA/SPCE SWV 0.05-100 0.002 Human urine and blood Raj et al., 2017
PEDOT/nafion/microelectrode CV – – Rat brain Vreeland et al., 2015
PEDOT/RGO/GC Amperometry 0.1 to 175 0.039 – Wang et al., 2014
PEDOT/CNT/CPE DPV 0.1 to 20 0.02 – Xu et al., 2013
PEDOT-Aunano/Au in presence of SDS LSV 0.5 to 20 0.0004 and Human urine Atta et al., 2012
25 to 140 0.002
PNMPy, PNCPy, PEDOT/GC CV – – – Fabregat et al., 2014a,
2014b
PEDOT:PSS/ITO Amperometry 10-900 1 – Scavetta et al., 2014
PEDOT/ssDNA/CPE Amperometry 0.25-66.5 0.074 – Xu et al., 2015
PEDOT/rGO/aptamer DPV 0.000001 0.000000078 Human serum Wang et al., 2015a, 2015b
-0.160
GO-PEDOT/GC CV 1.0-40 0.083 – Weaver et al., 2014
PEDOT/DES/GC DPV 5-180 1.3 Prathish et al., 2016
PEDOT/Pt electrode CV 0.5-25 0.061 Human urine Atta et al., 2011a
in presence of SDS 30-100 0.086
PEDOT:tosylate microelectrode CV – – PC12 Larsen and Taboryski, 2012
PEDOT/IL/GC Amperometry 0.2-312 0.051 – Sheng et al., 2015
EDOT/PNMPy/PEDOT/GC CV 500-2000 – – Fabregat et al., 2014a,
2014b
(Au/PEDOT–Pt–Ag/AgCl) DPV 0.2–300 0.1 – Belaidi et al., 2015
Pt-CPPy microelectrode array FET 0.0000001- – – Lee et al., 2015
0.001
PNMPy,PNCPy/AuNP/GC CV 100-10000 – – Fabregat et al., 2011
PPy/CNTs-MIPs/GC DPV 0.0000005-5.0 0.00001 – Qian et al., 2014a
MIPs/MWNTs/GC DPV 0.63-100 0.06 – Kan et al., 2012
OPPy–MSA–MWCNTs/Au electrode DPV 0.001-2.87 0.0004 Human serum Su et al., 2012
PPy/graphene/MEA Amperometry 0.8-10 0.004 PC12 Wang et al., 2015a, 2015b
Py/PSS electrode Amperometry – – PC12 Sasso et al., 2013
Au NPs/OPPy NT arrays electrode SWV 0.025-2.5 0.01 – Lin, 2015
nano-Cu/PPy/GC DPV 0.001-0.1 0.00085 Injection, human urine Ulubay and Dursun, 2010
Ppy/FCN/CPE DPV 100-1200 15.1 – Raoof et al., 2005
tyrosinase–SWNTs–Ppy Amperometry 5-50 5 – Min and Yoo, 2009
Au@PPy/GS DPV 0.0001-5 0.00001829 – Qian et al., 2014b
(continued on next page)

544
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Table 2 (continued)

Neurotransmitter Electrode Method Linear Range LOD (in µM) Matrix Ref
(in µM)

ET-SDBS-NPPy/ERGO SWV 0.1-100 0.02 Human serum Arulraj et al., 2016


Lac/PPy/MWCNT/Pt DPV 0.50-4.75 0.14 Human urine Cesarino et al., 2013
aptamer/GR–PANI/GC SWV 0.000007-0.09 0.000002 Human serum Liu et al., 2012
Ag/PANI Composite CV – – – Gao et al., 2009
AuNPs@PANI DPV 10-1700 5 Human serum Yang et al., 2012
PANI/Au composite hollow spheres CV 1000-10000 – – Feng et al.,2006
LbLdeposited PANI–AuN DPV 7-148 3 – Stoyanova et al., 2011
PANI/PDDMAC/AuNps CV – 50 – Prakash et al., 2009
TS-PANI/GC Amperometry 10-300 0.7 – Jin et al., 2010
P3MT/γ-CD SWV 0.5-50 0.2 – Bouchta et al., 2005
Nafion/SWNTs/poly(3-methylthiophene) DPV 0.020–0.10, 0.005 Human blood serum, Wang et al., 2006
0.10–1.0, injection
1.0–6.0
Pt/PMT(BE)/Pd DPV 0.05–1 0.008 Urine, Atta and El-Kady, 2010
human blood serum
PIn5COOH/TYR Amperometry 0.5–20 – – Maciejewska et al., 2011
MBIP DPV 0.02-7 0.006 Urine and plasma Rezaei et al., 2015
AuNPs-PTAP/GC DPV 0.15–1.5 0.017 Human serum, Khudaish et al., 2016
pharmaceutical drug
Pl-LEU/DNA composite DPV 0.1–100.0 0.04 Urine Zheng et al., 2013
AuNP/PAN Amperometry 1-100 0.91 – Chu et al., 2015
Nafion-CNT- ABTS/ITO DPV 1.87–20.00 1.75 Human serum Chih and Yang, 2013
Pty/GC LSSV 1-7 0.142 Serum Khudaish et al.,2012
PILs/PPy/GO DPV 4–18 0.073 – Mao et al.,2015
PPyox-PTSA/Ag-NP/Pt DPV 0.001-0.12 0.00058 Serum Saha et al., 2014
PMR/CPE CV 0.01-0.1, 0.005 Pharmaceutical samples Zhou et al.,2011
0.1-1000
PNMPy/PS CV 10000-20000 1.5 – Marti et al., 2010
PPy/eRGO DPV 0.1–150 0.023 Human serum Si et al.,2011
PEDOT/GO/CFE CV 0.5-10 0.22 Rat dorsal striatum Taylor et al., 2017
hDRD1-MCPEDOT NF/FET Amperometry 0.0001-0.001 – – Park et al., 2016
Serotonin CD/PNAANI/CNT DPV 4-200 0.2 Bovine assayed multi- Abbaspour and Noori,2011
sera
polymelamine/EPPGS SWV 0.1-100 0.03 Serum and urine Gupta and Goyal, 2014
AuNPs@MIES DPV 0.2-10 0.0113 Human blood serum Xue et al., 2014
PEDOT: PSS/TPyP-3IP/FTO CV 0-224 0.23 – Song et al., 2014
PEDOT/Pt in SDS LSV 0.05-10 0.048 Human urine Atta et al., 2011b
20-100 0.071
AuNPs@PPyNPs/GSPE SWV 0.1-15 0.03 Human serum Tertiș et al., 2017
PEDOTNTs/rGO/AgNPs/GC DPV 0.001-500 0.0001 Bovine assayed multi- Sadanandhan et al., 2017
sera
Histamine MIP on B : NCD :O electrode EIS – – – Ratautaita et al., 2014
MWCNTs/p-(AHNSA)/GC DPV 0.1-100 0.0762 Fish muscle Geto et al., 2014
lignin modified GC SWV 5-200 0.28 Wine and human urine Degefu et al., 2014
Acetylcholine pAu/pTTBA/ChOx-Hydrazine Amperometry 0.0007-1500 0.0006 Leukemic T-cells Akhtar et al., 2017
AchE-ChO/Fe2O3/rGO/PEDOT/FTO Amperometry 0.004-800 0.004 Human serum Chauhan et al., 2017
PEDOT/PSS/gold wire Amperometry 100-1000000 5.69 Rat brain He et al., 2016
poly(SNS-NH2)/AChE–ChO Amperometry 120-10000 111 Pesticide Kanik et al., 2013
Naf–FCNTs/AChE–ChO (10:1)/PoPD/CFElip Amperometry – 0.045 – Khan and Ab Ghani,2012
ChO/poly(TBT6–NH2)-graphite Amperometry 100-10000 16.8 Pesticide Kanik et al.,2012
AChE/PANI-Nano-ZSM-5/GC Amperometry 1-1000 0.03 Pesticide Kaur and Srivastava, 2015b
Pt/PPYox/P2NAP/ChO–AChE Amperometry – 0.1 – Guerrieri et al., 2006
Pt/PPYox/P2NAP/ChO
Polypyrrole- Amperometry 0.01-0.1 0.005 Synthetic blood Aynaci et al., 2014
polyvinylsulphonate
film 0.1-1
MIP(PANI/MWCNT) Potentiometry – 34.5 Synthetic serum Sacramento et al., 2017
Nitric oxide Cytc /poly-TTCA Amperometry 2.4–55.0 0.013 Rat brain Koh et al., 2008
Nafion/Cytc /poly-TTCA/Pt microelectrode Amperometry 0 – 55.0 0.013 Rat brain Lee et al., 2010
CAS/SOD/MP/MWCNT-PTTCA/AuNP Amperometry 1.0-40 0.0043 Liver and cultured cells Abdelwahab et al., 2010
anti-iNOS/AuNP(TTBA) Amperometry 0.001-0.02 0.0002 Cultured cells Koh et al., 2011
p-EDA- MWNTs/GC Amperometry 0.095-11 0.095 Drug Wang et al., 2013
GC/AuPs/PPBZ Amperometry 20-140 0.0037 Goat and chicken Liver Mohan et al., 2014
Hydrogen sulfide SPE-Pt electrode Amperometry 0-100 ppm 1 ppm – Yourong et al., 2001
NASICON and Pr6O11- Potentiometric 5-50 ppm – – Liang et al.,2007
doped SnO2
PANI nanofibers dopes with – – 10 ppm – Virji et al., 175
Zn, Cd, Cu
PANI nanowires-AuNPS Chemiresistive 0.1-500 ppb 0.1 ppb – Shirsat et al.,2009
sensor
PANI-CuCl2-Carbon IDE Chemiresistive 10-100 ppmv 2.5 ppmv – Crowley et al., 2010
sensor
HRP–SAM–Au electrode Amperometry 0.5–12.7 0.3 Mountain stream water Yang et al., 2004

545
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

neurotransmitter primarily used for the transfer of information in the employing microchip electrophoresis with electrochemical detection.
mammalian CNS (Eisenhofer et al., 2004). Also referred to as the flight The negative charges from carboxylates improved the separation effi-
hormone, it boosts the supply of oxygen and glucose to the brain and ciency through the interaction with cationic analytes. A real-time
muscles in emergency situations. Changes in EP concentration are re- neurotransmitters (dopamine, EP, and NE) detection system was de-
lated to Parkinson’s disease (Spencer et al., 1998). EP is also prescribed veloped based on flexible inkjet-printed and arginine-glycine-aspartate
in the treatment of asthma, sepsis, severe allergic reactions, cardiac (RGD) peptide-immobilized PANI patterns on polyethylene ter-
arrest, and anaphylaxis (Cicchetti and Cohen, 2006; Kemp et al., 2008). ephthalate film (Oh et al., 2013). The interaction between the neuro-
Most of EP detections were performed by direct oxidation of EP at transmitters and the PANI were reported to affect the charge-carrier
the sensor surface modified with various catalytic materials. A nano- density of the conjugated PANI patterns, enabling the recognition of
composite CP film comprising single-walled carbon nanotubes neuronal neurotransmitter secretions in live cells. A modified electrode
(SWCNTs), PPy, and AuNPs was employed to modify a gold electrode, was fabricated by depositing 3-amino-5-mercapto-1,2,4-triazole onto a
which was then used for the electrochemical analysis of EP (Lu et al., GCE and was used for the simultaneous determination of NE and 5-HT
2011). The proposed method was evaluated for the determination of EP (Revin and John, 2012). The practical applicability of the developed
in pharmaceutical formulations and human biological fluids (serum, electrode was successfully validated by quantifying NE and 5-HT in
plasma, and urine) with satisfactory results. An over-oxidized compo- spiked human blood plasma samples, with good recovery results. A poly
site film consisting of carboxylated MWCNTs and sodium dode- (1,5-diaminonaphthalene) (PDAN) film was electropolymerized onto a
cylbenzene sulfonate-doped PPy was electropolymerized on GCE sur- Pt electrode followed by its cathodic pretreatment to obtain the final
face (Shahrokhian and Saberi, 2011). The proposed sensor exhibited a electrode surface (CPPDAN/Pt) which was used for the determination
low detection limit, high reproducibility and stability, and was vali- of NE in pharmaceutical formulations (Guedes da silva et al., 2011).
dated for the determination of EP in pharmaceutical preparations and Short time cathodic pretreatment allowed better electrochemical re-
human synthetic serum samples. A PANI-transition metal ion ex- sponse as compared to non-treated electrode. However, sodium bisul-
changed zeolite based electrochemical sensor was developed for the phite was found to interfere in NE determination at equal concentra-
simultaneous analysis of EP, paracetamol (PCM) and folic acid (FA) tions. Further, fast-scan cyclic voltammetry at carbon-fiber
(Kaur and Srivastava, 2015a). Metal ion-exchanged PANI-Nano-ZSM-5 microelectrode was also reported for in-vivo detection of NE (Park et al.,
nanocomposites were synthesized, where the metal ions were Co2+, 2011).
Fe2+, Mn2+, Cu2+ and Ni2+, and the nanocomposites were coated onto
the electrode surface. Among them, Cu2+-PANI-Nano-ZSM-5 modified
3.2.3. Dopamine
GCE demonstrated excellent electrocatalytic activity for the simulta-
Dopamine (DA) is an inhibitory neurotransmitter discovered in the
neous determination of EP, PCM, and FA in pharmaceutical prepara-
1950s by Arvid Carlsson (Carlsson et al., 1957) and play a crucial role
tions. PANI nanocomposite films doped with TiO2 and RuO2 nano-
in physical and mental health, such as cardiovascular, hormonal, renal
particles on MWCNTs was reported for EP detection (Tsele et al., 2017).
and central nervous systems (Gowrishankar et al., 2014). DA in the
The prepared sensor was tested for the detection of EP in pharmaceu-
frontal lobe is known to control the flow of information from other
tical injections. Similarly, polythionine/AuNPs composites (PTh/
areas of the brain which is an important for attention, problem solving
AuNPs) based sensor was prepared for the determination of EP (Huang
and memory. DA is also termed as the “reward chemical” since it is
et al., 2016) and it was evaluated to detect EP in serum samples.
released in the course of rewarding experiences such as food, sex and
other stimulating experiences. Abnormal DA level is associated with
3.2.2. Norepinephrine sleeping and eating disorders, addictive behaviors due to drug abuse,
Norepinephrine (NE), a catecholamine neurotransmitter present in schizophrenia, attention deficit hyperactivity disorder/attention deficit
the CNS which secreted by the adrenal medulla (Glowinski and disorder, social anxiety and Parkinson's disease (Dawson and Dawson,
Baldessarini, 1966). Abnormal NE concentrations are associated with 2003; Camardese et al., 2014).
various diseases, such as neuroblastoma, coronary heart disease, mul- An in-vivo amperometric DA sensor was developed by incorporating
tiple sclerosis, psychosocial stress, anxiety, depression, Alzheimer’s and Cibacron Blue (F3GA) into poly (1,5-DAN) (Abdelwahab et al., 2009).
Parkinson’s diseases (Klimek et al., 1997; Friedman et al., 1999). The sensor probe was prepared by electropolymerization of the DAN in
Poly-(acrylic acid) (PAA) was immobilized on soda-lime glass mi- the presence of F3GA and was applied for the determination of DA in a
crochips. The immobilization was based on the reaction of the car- human urine sample. The sulfonate group containing F3GA composite
boxylate groups of PAA with an amine-functionalized surface obtained film provided a cation exchange site which can act as a charge-selective
using the bifunctional reagent 3-aminopropyl triethoxysilane (Álvarez- compound. Furthermore, it was also employed for the real-time de-
Martos et al., 2012). The modified microchips (PAA-MEs) were then tection of the DA concentration in a rat’s brain (Oh et al., 2015; Lee
applied for the separation of catecholamines (dopamine, EP, and NE) et al., 2013a, 2013b). An amperometric nanobiosensor was fabricated

Fig. 2. Schematic representations of glutamate sensors based on (A) a GluOx/pTTCA/Pt microelectrode (Rahman et al., 2005) and (B) a GluOx/PEDOT: PSS/Pt NPs/OECD (Kergoat et al.,
2014).

546
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

for the determination of K+-induced DA released from living cells mediator for the electrooxidation of DA. Similarly, a single-stranded
(PC12) (Mir et al., 2015); as shown in Fig. 3(A). Ethylenediaminete- DNA doped PEDOT-modified carbon paste electrode (PEDOT/DNA/
traacetic acid (EDTA) was immobilized onto poly (1,5-DAN)/GO/ CPE) and aptamer sensor was constructed on a nanocomposite layer
AuNPs layer. Interaction between DA and the EDTA-modified probe consisting of rGO and PEDOT were constructed for the analysis of DA
through hydrophilic and charge interactions allowed its detection without interference (Xu et al., 2015; Wang et al., 2015a, 2015b) re-
hence it was applied for the monitoring of DA released from PC12 upon spectively. In addition, several other PEDOT-based electrodes were
extracellular stimulation with K+ ions. Similarly, DA was analyzed in prepared and employed for the determination of DA with high sensi-
the presence of acetaminophen using reactive blue-4 dye entrapped in a tivity and selectivity (Weaver et al., 2014; Prathish et al., 2016; Atta
poly (1,5-DAN) composite layer (polyDAN-RB4) (Chandra et al., 2013). et al., 2011a; Larsen and Taboryski, 2012a; Larsen et al., 2012b; Sheng
The polyDAN-RB4-modified electrode showed a significantly enhanced et al., 2015; Fabregat et al., 2014a, 2014b; Belaidi et al., 2015; Pal
analytical signal for DA due to the electrostatic interaction between the et al., 2017; Taylor et al., 2017).
negatively charged sulfonic acid group of the RB4 and the positively A non-enzyme field effect transistor (FET) sensor based on im-
charged DA. The sensor was evaluated to detect DA in biological fluids mobilized platinum particle-decorated carboxylated polypyrrole nano-
(human blood serum and urine samples). A copper-(3-mercaptopropyl) particles (Pt-CPPy) conjugated onto an amine-functionalized inter-
trimethoxy silane (Cu-MPS) complex modified electrode was developed digitated microelectrode array was fabricated for the detection of DA
for DA detection (Won et al., 2005). The modified electrode exhibited (Lee et al., 2015). The proposed sensors exhibited high sensitivity to-
excellent electrocatalytic activity in response to the oxidation of DA and wards DA at remarkably low concentrations. Composites of AuNPs with
avoided interference from other biological compounds by Nafion ultra-thin films of poly[N-(2-cyanoethyl) pyrrole] and poly(N-methyl-
coated onto Cu-MPS layer. Similarly, graphene and poly 4-amino-3- pyrrole) were coated onto the electrode surface and used for the elec-
hydroxyl-naphtalenesulfonic acid electrode was developed for the si- trochemical detection of DA (Fabregat et al., 2011) where the response
multaneous detection of DA and 5-HT in human urine, plasma, and of the poly[N(2cyanoethyl) pyrrole] was better than poly(N-methyl-
pharmaceutical samples (Raj et al., 2017). pyrrole) electrode. An MIP oxygen-containing PPy decorated CNTs
A selective detection of DA for in-vivo experiment was demonstrated composite sensor was reported for DA detection (Qian et al., 2014a).
using PEDOT:Nafion coated carbon-fiber microelectrode (Vreeland The π-π stacking between aromatic rings and hydrogen bonds between
et al., 2015). The proposed microelectrode was implanted in the rat amino groups of DA and oxygen-containing PPy are assigned for the
nucleus accumbens for 6 h. A nanocomposite composed of PEDOT high electrocalaytic activity observed at the modified electrode. An
doped with reduced graphene oxide (rGO) (PEDOT/rGO) was achieved electrochemical sensor combined with a molecular imprinted technique
by a simple electrochemical process (Wang et al., 2014). The modified was developed, where the sensor was fabricated by electro-
electrode showed enhanced electrocatalytic activity due to the large polymerization of pyrrole in the presence of DA onto carboxyl func-
surface area, which provided many active sites to accelerate the elec- tionalized MWCNTs (Kim et al., 2012). A thiolated polymeric nano-
tron transfer process of DA. Similarly, PEDOT doped with carbon na- composite film incorporating overoxidized PPy, a carboxylated thiol
notubes (PEDOT/CNTs) on carbon paste electrodes was developed for (mercaptosuccinic acid), and MWCNTs based sensor was developed for
DA detection (Xu et al., 2013) without common biological inter- efficient enrichment and sensitive electroanalysis of DA (Su et al.,
ferences. An AuNPs-coated PEDOT-modified gold electrode (PEDOT- 2012). A microelectrode array electrodeposited directionally with PPy
Aunano/Au) was developed for the determination of DA in the presence graphene nanocomposites was fabricated as depicted in Fig. 3 (B). The
of sodium dodecyl sulfate (SDS) (Atta et al., 2012). Higher catalytic proposed sensor was applied for the detection of DA release and the
activity was achieved due to the rich electron cloud in PEDOT, and coordinating neurotransmission in the nerve system (Wang et al.,
electrocatalytic properties of the AuNPs and SDS. The modified elec- 2015a, 2015b). In addition, various types of PPy composite films with
trode was validated for the determination of DA in human urine. Poly metal and carbon materials have been reported for determination of DA
(N-methylpyrrole) (PNMPy), poly (N-cyanoethylpyrrole) (PNCPy), (Ulubay and Dursun, 2010; Raoof et al., 2005; Min and Yoo, 2009; Qian
PEDOT, and poly (hydroxymethyl-3,4-ethylenedioxythiophene) et al., 2014b; Arulraj et al., 2016; Cesarino et al., 2013; Sasso et al.,
(PHMeDOT) films were prepared and composited with AuNPs for the 2013; Lin et al., 2015).
selective determination of DA, UA, and AA (Fabregat et al., 2014a, A label-free electrochemical aptasensor was developed based on a
2014b). PEDOT composites showed better results compared with other graphene-PANI nanocomposite film (Liu et al., 2012). The nano-
composites. However, AuNPs were ineffective due to the outstanding composite was prepared using multistep procedure, where the DA ap-
electrical and electrochemical properties of PEDOT. A ferrocene-func- tamer was immobilized onto the graphene-PANI nanocomposite layer.
tionalized (PEDOT-Fc:PSS) layer was prepared on an indium tin oxide The proposed sensor was highly selective due to the aptamer selectivity
(ITO) electrode for the amperometric detection of DA (Scavetta et al., however, the sensor required additional fabrication steps. Further, it
2014). The peak current of PEDOT-Fc:PSS was increased with an in- was evaluated in human blood serum samples for the detection of DA. A
creasing DA concentration, indicating that the ferrocene was acting as a self-assemble assisted Ag/PANI composite nanotubes decorated ITO

Fig. 3. Schematic representations of dopamine sensors based on (A) GO/AuNPs/pDAN-EDTA (Mir et al., 2015) and (B) a PPy/graphene/MEA (Wang et al., 2015a, 2015b).

547
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

electrode was used for the detection of DA (Gao et al., 2009) where, the 3.2.4. Serotonin
prepared nanocomposites showed an improved electrocatalytic oxida- Serotonin (5-hydroxytryptamine; 5-HT) is a monoamine neuro-
tion of DA compared with the pure PANI which could be due to the transmitter discovered by Vittorio Erspamer. 5-HT plays an important
enhancement in charge migration to the PANI. AuNPs@PANI core-shell role in the regulation of various physiological functions, especially in
nanocomposites were synthesized and deposited onto GCE surface human emotions (Whitaker-Azmitia, 1999). Deficiency of 5-HT leads to
(Yang et al., 2012) for the simultaneous determination of DA and AA mental disorders, such as Alzheimer’s disease, infantile autism, mental
using DPV. The π-rich nature of PANI, the π–π interaction between the retardation, sleep disorders and depression, whereas high level of ser-
phenyl ring of DA and the PANI promoted the influx of DA molecules to otonin in the body may lead to carcinoid syndrome (Meltzer et al.,
the electrode surface. Further, π–π interaction between PANI and 1998; Robiolio et al., 1995).
penta-heterocycle of AA is not strong, positively charged chitosan ex- A polymelamine-modified edge plane pyrolytic graphite sensor
erted a significant electrostatic attraction to negatively charged AA in (EPPGE) was developed for the determination of 5-HT (Gupta and
PBS (pH 6.0). The combined effect of the above two aspects is attrib- Goyal, 2014), where polymelamine was used to avoid superficial poi-
uted to the simultaneous determination of DA and AA in human serum. soning of the EPPGE. The proposed sensor was evaluated to detect 5-HT
Similar works have been done by other groups using PANI/Au com- in spiked human serum and urine samples. A β-cyclodextrin (β-CD)/
posite hollow spheres (Feng et al., 2006), AuNPs-PANI nanocomposite poly(N-acetylaniline)/MWCNTs composite modified carbon paste
multilayer (Stoyanova et al., 2011), PANI-polyelectrolyte-Au ternary electrode was developed for simultaneous quantification of 5-HT and
nanocomposites (Prakash et al., 2009), and tetragonal star-like struc- DA (Abbaspour and Noori, 2011). The β-CD layer comprising nano-
ture of aspartic acid doped PANI (Jin et al., 2010). cavity offered the encapsulation site of 5-HT and DA, resulting in ne-
A functionalized stable film of poly-3-methylthiophene combined gative potential shift and improvement of the oxidation current. The
with γ-cyclodextrin (P3MT/γ-CD) was employed for the detection of proposed sensor was employed to detect 5-HT and DA in spiked syn-
chlorpromazine, DA, and L-dopa (Bouchta et al., 2005). The γ-CD in the thetic bovine serum samples with acceptable recoveries. Similarly,
P3MT/γ-CD film possessed a hydrophobic interior cavity, which may carbon-based materials were also used for the sensor fabrication, such
have played a key role in the selectivity; thus, the response was not as rGO/PANI nanocomposites (Xue et al., 2014) and PEDOT-rGO-silver
affected by AA. A Nafion/single-walled carbon nanotubes/poly (3-me- hybrid nanocomposite (PEDOTNTs/rGO/AgNPs) (Sadanandhan et al.,
thylthiophene) hybrid film was fabricated and exploited for the de- 2017). A PEDOT:PSS modified fluorine doped tin oxide (FTO) electrode
termination of DA (Wang et al., 2006). The proposed sensor was suc- surface was fabricated using 3-iodopropionate (3IP) and 5,10,15,20-
cessfully applied for the quantitation of DA in dopamine hydrochloride tetrakis(4-pyridyl)-21H,23H-porphyrin to generate a binary self-as-
injections and human blood serum. Similarly, Pt or Pd nanoparticles sembled monolayer for the detection of 5-HT. However, the developed
were introduced into a poly (3-methylthiophene) matrix to develop a sensor was not applied in real samples. Similarly, PEDOT modified Pt
composite layer for the simultaneous detection of DA and AA (Atta and electrode was reported for the detection of 5-HT in the presence of SDS
El-Kady, 2010). Polyindole-camphor sulfonic acid composite coated (Atta et al., 2011b). A nanocomposite based on PPyNPs and AuNPs was
over Pt disk electrode was reported for DA detection without inter- also employed for the quantification of serotonin (Tertiș et al., 2017).
ference (Pandey et al., 2011). A composite poly (indole-5-carboxylic The sensor probe was applied in human serum sample showing ex-
acid)/tyrosinase modified electrode system was employed for the de- cellent recoveries. In addition, various in-vitro microelectrode sensors
tection of DA in the presence of two common biological interferences, have been reported for 5-HT, such as boron-doped diamond thin film,
AA and UA (Maciejewska et al., 2011). The method was applied to which was deposited on Pt wire (Zhao et al., 2010), cylindrical carbon
urine and blood serum samples. A molecularly bioimprinted polymer fiber microelectrodes (Yang et.al, 2007, Abdalla et al., 2017), and Ni
sensor was prepared for the detection of DA, which prepared through (II)–phthalocyanine/Nafion modified carbon fiber microelectrode (De
the electrochemical entrapment of double stranded-DNA and AuNPs in Irazu et al., 2001).
o-phenylenediamine by one-step electropolymerization (Rezaei et al.,
2015). The applicability of the developed sensor was conducted for the 3.2.5. Histamine
detection of DA in real samples, such as urine and plasma. A liquid ion- Histamine is a vasoactive amine produced from the decarboxylation
gated FET system was reported (Park et al., 2016). The sensor was of the amino acid, histidine (Shan et al., 2015). A major portion of the
fabricated using a DA receptor-conjugated multidimensional CP nano- histamine in the human body is generated by granules in mast cells and
fiber membrane that demonstrated ultrasensitive detection of DA in white blood cells. Non-mast cell histamine is released in the brain,
PBS. where it functions as a neurotransmitter with the histamine neurons
In addition, various DA sensors has been reported based on polymer affecting sleep-wake cycle (Chikahisa et al., 2013; Atsushi et al., 1982).
composites modified electrodes, such as modified with poly(2,4,6- Histamine in the brain is known to be involved in several physiological
triaminopyrimidine) decorated with AuNPs (Khudaish et al., 2016), and neurological diseases, such as multiple sclerosis and Alzheimer's
poly(4-aminobutyric acid) (Zheng et al., 2013), AuNPs incorporated disease (Haas et al., 2008).
into PANI (Chu et al., 2015), 2,2′-azino-bis(3-ethylbenzthiazoline-6- An electrochemical sensor was fabricated based on histamine-im-
sulfonic acid)-immobilized CNTs (Chih and Yang, 2013), polytyramine printed PPy deposited onto boron-doped oxygen-terminated nanocrys-
film (Khudaish et al., 2012), poly(ionic liquids)-functionalized PPy/GO talline diamond (B:NCD) (Ratautaite et al., 2014). However, no attempt
nanosheets (Mao et al., 2015), chitosan-stabilized silver nanoparticles was made towards quantification of the analyte. A MWCNT 4-amino-3-
and p-toluene sulfonic acid-doped ultrathin PPy film (Saha et al., 2014), hydroxynaphthalene sulfonic acid-modified GCE was fabricated for the
poly(methyl red) film (Zhou et al., 2011), AuNPs-doped poly(N-me- determination of histamine (Geto et al., 2014) and was applied to detect
thylpyrrole) (Martí et al., 2010), and PPy/graphene film (Si et al., histamine in fish muscle extract. A lignin modified GCE was constructed
2011). Recently, ultra-microelectrodes (UMEs) have been also in- for the determination of histamine in spiked human urine and red wine
troduced for in-vivo and in-vitro detection of DA, where carbon based samples (Degefu et al., 2014). The lignin modified electrode showed the
material was used in the fabrication of UME sensors because of its catalytic activity towards the oxidation of histamine in PBS. Though the
biocompatible nature. Among the carbon-based sensors, CNT and gra- method was reliable, however, the effect of interference agents was not
phene have attracted much attention, however, graphene showed better investigated. A potentiometric sensor was also developed for the de-
results compared to that of CNT modified electrodes in selective de- tection of histamine based on molecularly imprinted nanoparticles
tection of DA, this is due to the π–π stacking interactions between DA (Basozabal et al., 2014). The sensor showed high affinity and specifi-
and the graphene surface (Jackowska et al., 2013). city, which allowed label-free quantification of histamine in real sam-
ples with fast response time. Further, organic ion-sensitive

548
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

microelectrode was also reported for the sensitive detection of hista- 3.4. Soluble gases
mine (Bi, 1989).
Certain soluble gases have recently been identified to act as neu-
rotransmitters. These soluble gas neurotransmitters are synthesized and
3.3. Acetyl-choline and choline released in response to Ca2+ binding (Nathan and Xie, 1994). They are
produced in the neural cytoplasm from where they move immediately
Acetylcholine (ACh) is the first known neurotransmitter (Kandel through the cell membrane into the extracellular fluid and penetrate by
and Squire, 2000), Isolated in 1921 by Otto Loewi. ACh synthesized in diffusion into the membrane of the receiving neuron (Calabrese et al.,
nerve terminals from acetyl coenzyme A and choline in the presence of 2007). They act very fast and are instantly broken down, existing for
choline acetyltransferase (Krnjević, 1974). ACh is found in both the only a few seconds, thus making them difficult to investigate. The most
central and peripheral nervous systems and mainly acts at the neuro- important members of this category are nitrogen monoxide (NO) and
muscular junctions, allowing motor neurons to activate muscle action. hydrogen sulfide (H2S). NO is involved in synaptic plasticity, apoptosis
Moreover, it carries signals from motor neurons to the skeletal muscles as part of neurodegenerative disease (Kim et al., 1999; Yamada et al.,
(Sine, 2012). ACh modulates communication between various neurons 1995), and variety of neurological functions, including sleep and ap-
in different brain parts that control motivation, arousal, attention, petite regulation, neurotransmission, learning and memory (Vincent,
formation of memories, verbal functions and logical reasoning and is 2010). In contrast, H2S is a toxic gas that acts as an active physiological
associated with the onset of dementia and Alzheimer's disease messenger regulating the release of corticotropin-releasing hormone
(Danielmeier et al., 2015; Lombardo and Maskos, 2015). Choline and has a leading role in smooth muscle relaxation.
(ChO), an essential nutrient for humans, is the precursor molecule for A cytochrome c modified pTTCA coated Pt microelectrode was
ACh (Cooper et al., 1997). ChO deficiency is associated with liver dis- fabricated for the in-vivo measurement of NO released upon stimulation
ease, atherosclerosis, and possibly neurological disorders (Coyle et al., by cocaine (Koh et al., 2008). Cytochrome c has been known to have a
1983). binding affinity for small ligands and was introduced as a catalyst for
An enzyme-based ACh sensor based on a microfluidic structure was the NO detection. A Nafion film was used to protect the sensor from
reported (Akhtar et al., 2017). The sensor probe consisted of two screen fouling effect and interferences, such as oxygen (O2), superoxide (O2-),
printed carbon electrodes (SPCEs), one of them utilized for enzyme and H2O2. Administration of cocaine increased the extracellular NO
reaction and the other for detection assembled in microfluidic archi- level in the rat dorsal striatum, which was effectively detected by the
tecture and employed to detect Ca2+-induced ACh released from cancer micro biosensor (Lee et al., 2010). Similarly, multienzymes-im-
cells. The sequential enzymatic oxidation of Ach yielded H2O2 in the mobilized MWCNT-pTTCA nanocomposite was also reported for NO
presence of acetylcholinesterase/choline oxidase (AChE/ChOx) bioca- detection (Abdelwahab et al., 2010). Step-by-step modification of the
talyst cascade in the microfluidic channel structure. Subsequently, proposed sensor is shown in Fig. 5. The sensor probe was evaluated to
H2O2 was catalytically reduced by hydrazine to generate the analytical detect NO in rat liver as well as human stomach (AGS) and intestinal
signal. Similarly, AChE and ChOx were immobilized onto the surface of (HT-29) cancer cell samples. In addition, other NO sensors were fabri-
iron oxide nanoparticles (Fe2O3NPs), PEDOT-rGO nanocomposite cated based on poly (ethylenediamine) layer on MWCNTs (Wang et al.,
modified FTO (Chauhan et al., 2017). The proposed sensor was applied 2013), AuNPs dispersed in poly (2-(2-pyridyl) benzimidazole) (Mohan
to determine ACh in serum samples from healthy and Alzheimer's pa- et al., 2014), and Nafion and o-phenylenediamine (Özel et al., 2015).
tients. A new CP, poly (6-(4,7-di(thiophen-2-yl)-2H-benzo[d] [1,2,3] An amperometric sensor was reported for H2S sensing based on a
triazol-2-yl)-hexan-1-amine) was synthesized and deposited on a gra- solid-polymer electrolyte (SPE)-Pt electrode in a H2S + N2 mixture
phite electrode followed by the immobilization of ChOx, stepwise fab- (Yourong et al., 2001). The detection mechanism was based on the
rication of the biosensor is illustrated in Fig. 4 (A). (Kanik et al., 2012). electrochemical oxidation of H2S at the SPE-Pt electrode. However, it
Similarly, AChE, ChOx sensor was developed using a PPy-poly- was not applied in real sample analysis. In contrast, various potentio-
vinylsulphonate conducting layer (Aynaci et al., 2014). Enzymes were metric sensors have also been reported for the determination of H2S. Of
immobilized by crosslinking with glutaraldehyde, and physical en- those sensors, a compact tubular sensor based on a sodium super-ionic
trapment as shown in Fig. 4 (B). Crosslinked method was superior and conductor (NASICON) and a Pr6O11-doped SnO2 electrode showed
was applied for the detection of ACh in synthetic blood samples. In ultra-sensitive detection of H2S oxidation (Liang et al., 2007), similarly
addition, various enzymatic biosensors were studied using different these sensors were not investigated in biological matrix. Moreover,
types of CPs, such as PEDOT:PSS and PtNPs (Kergoat et al., 2014), poly various composite materials have been synthesized to enhance the
(4-(2,5-di(thiophen-2-yl)-1H-pyrrol-1-yl)benzenamine) modified gra- sensitivity, such as metal salts with PANI nanofibers (Virji et al., 2005),
phite electrode (Kanik et al., 2013), poly(o-phenylenediamine) on a AuNPs-PANI nanowire (Shirsat et al., 2009), and PANI-Cu(II)-chloride
carbon fiber (Khan and Ab Ghani, 2012), PANI-zeolite (Kaur and (Crowley et al., 2010).
Srivastava, 2015b), and overoxidized PPy and poly(2-naphthol) bilayer An amperometric biosensor was constructed by the immobilization
membrane (Guerrieri et al., 2006). of horseradish peroxidase (HRP) on a thiolate self-assembled monolayer
In contrast, a potentiometric ion-selective nonenzymatic ACh sensor (SAM) (Yang et al., 2004). The Au-modified HRP-SAM probe inhibited
was developed based on gold wire (He et al., 2016), where the in- HRP activity in the presence of H2S. However, it suffered from the in-
troduction of a selective membrane containing heptakis (2,3,6-tri-Ο- terference of common cations. Recently, H2S oxidation was reported
methyl)-β-cyclodextrin as an ionophore enhanced the selectivity. The using ferric hemoglobin (Bostelaar et al., 2016).
proposed sensor was applied to detect ACh in rat brain. A molecular
imprinting technique was employed to construct an ACh potentiometric 4. Electrochemical methods coupled with other techniques
sensor (Sacramento et al., 2017). The MIP layer was constructed by
polymerization of composite matrix, which composed of ACh, Electrochemical analysis of neurotransmitters combined with other
MWCNTs, and aniline and subsequently, yielded a polymeric structure techniques, such as liquid chromatography (LC), capillary electro-
of PANI. The sensor probe was applied in synthetic serum samples, phoresis (CE), and mass spectrometry (MS). These techniques allow
where the detection limit was far from the physiological levels. Fur- multi-analyte detection, but such approaches lack spatial and temporal
thermore, various microelectrode sensors have been reported for the resolution due to the lag time between sample collection and simulta-
determination of ACh and ChO, such as carbon fiber/filament sensors neous detection. Further, these methods must be coupled with micro-
based on the redox polymer gel (Kulagina et. al, 2003), and Pt-based dyalsis for the in-vivo detection. To date, limited literatures have re-
microelectrode (Bruno et al., 2004). ported on the use of CPs in such combinations. An amperometric

549
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Fig. 4. Schematic representations of (A) choline and (B) acetylcholine sensors based on.
ChO/poly(TBT6–NH2)-graphite (Kanik et al., 2012) and PPy-polyvinyl sulfonate film (Aynaci et al., 2014).

Fig. 5. Schematic representation of a NO sensor


based on CAS/SOD/MP/MWCNT-PTTCA/AuNPs
(Abdelwahab et al., 2010).

biosensor- was developed based on the flow through a microdetector nanoparticles and CPs prominently exhibited an improved performance
for the determination of glutamate and ChO (Gáspár et al., 2004). A of the sensors. In addition, the direct electron transfer process of elec-
melanin-type polymer-modified graphite carbon electrode acting as an troactive neurotransmitters has captured more attention compared to
amperometric detector combined with CE was reported for the quan- other detection methods, including enzymatic or MIP ones. Despite the
tification of DA, EP and norepinephrine (Chicharro et al., 2004), where advancements in the sensing of neurotransmitters, the measurements
CE was employed to enhance the separation efficiency. A system for were performed mostly under standard conditions with few reports
electrophoresis coupled with in-channel electrochemical detection was using in-vitro systems. The slow translation of this technology from the
fabricated and applied to detect DA, 5-HT, and EP (Wang et al., 2007). standard condition to clinical applications can be resolved by addres-
A film was obtained via the sequential immobilization of poly (dia- sing several challenges before implanted in point-of-care devices, these
llyldimethyl ammonium chloride) (PDDA) and glucose oxidase (GOx) challenges include, nonspecific adsorption, interferences, and bio-
on the microfluidic channel surface via layer-by-layer (LBL) assembly. A compatibility. Such challenges can be addressed by the synthesis of
similar microchip was designed using PDMS/glass CE microfluidic de- highly selective and sensitive interface materials. In addition, the future
vice for the detection of DA (Dawoud et al., 2007). The modified perspectives of such platforms are to develop portable, non-invasive,
electrochemical detector provided well-resolved separation of DA and cheap, wearable, and point-of-care oriented devices that are capable to
catechol in less than 60 s with enhanced sensitivity. reduce time and frequency of sampling. Further, the development of
ultra-small, multiplexed, array of nanoelectrode sensors, and label-free
5. Conclusion and future perspective biosensors is a vibrant research area that can contribute in the devel-
opment of clinical and pharmaceutical applications.
Electrochemical sensors are a promising technique for the detection
of neurotransmitters. In the last couple of years, most of the work was Acknowledgement
devoted to synthesize new sensing probe materials to improve the
sensor performance, such as sensitivity, selectivity, and biocompat- This work was supported by the National Research Foundation of
ibility. Of those synthesized nanomaterials, CPs and their composites Korea (NRF) grant funded by the Korea government (MSIP) (No.
have been widely used in the construction of sensor surfaces. These 2015R1A2A1A13027762).
nano composites offer great enhancement in sensitivity, selectivity, and
stability of the sensors. Several CPs like polythiophene, functionalized References
polyterthiophene, PPy, PANI, PEDOT, and poly(o-phenylenediamine)
display advantages due to their charge transport properties and elec- Abbaspour, A., Noori, A., 2011. Biosens. Bioelectron. 26, 4674–4680.
Abdalla, A., Atcherley, C.W., Pathirathna, P., Samaranayake, S., Qiang, B., Peña, E.,
trochemical redox efficiency, which are attributed to the delocalization
Morgan, S.L., Heien, M.L., Hashemi, P., 2017. Anal. Chem. 89, 9703–9711.
of π-electrons over the polymeric back bone. The synergistic effect of

550
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Abdelwahab, A.A., Koh, W.C.A., Noh, H.-B., Shim, Y.-B., 2010. Biosens. Bioelectron. 26, Friedman, J.I., Adler, D.N., Davis, K.L., 1999. Biol. Psychiatry 46, 1243–1252.
1080–1086. Gao, Y., Shan, D., Cao, F., Gong, J., Li, X., Ma, H.Y., Su, Z.M., Qu, L.Y., 2009. J. Phys.
Abdelwahab, A.A., Lee, H.M., Shim, Y.-B., 2009. Anal. Chim. Acta 650, 247–253. Chem. C 113, 15175–15181.
Adams, R.N., 1969. Electrochemistry at Solid Electrodes. Marcel Dekker, New York. Gáspár, S., Wang, X., Suzuki, H., Csöregi, E., 2004. Anal. Chim. Acta 525, 75–82.
Akhtar, M.H., Hussain, K.K., Gurudatt, N.G., Shim, Y.-B., 2017. Biosens. Bioelectron. 98, Geto, A., Tessema, M., Admassie, S., 2014. Synth. Met. 191, 135–140.
364–370. Glowinski, J., Baldessarini, R.J., 1966. Pharmacol. Rev. 18, 1201–1238.
Álvarez-Martos, I., Fernández-Abedul, M.T., Anillo, A., Fierro, J.L.G., Alonso, F.J.G., Gowrishankar, R., Hahn, M.K., Blakely, R.D., 2014. Neurochem. Int. 73, 42–48.
Costa-García, A., 2012. Anal. Chim. Acta 724, 136–143. Guedes da Silva, Q., Vieira Barbosa, N., de Pieri Troiani, E., Censi Faria, R., 2011.
Ammam, M., Fransaer, J., 2010. Biosens. Bioelectron. 25, 1597–1602. Electroanalysis 23, 1359–1364.
Arbizzani, C., Mastragostino, M., Scrosati, B., 1997. In: Nalwa, H.S. (Ed.), Handbook of Guerrieri, A., Lattanzio, V., Palmisano, F., Zambonin, P.G., 2006. Biosens. Bioelectron. 21,
Organic Conductive Molecules and Polymers. Wiley, Chichester. 1710–1718.
Arulraj, A.D., Arunkumar, A., Vijayan, M., Viswanath, K.B., Vasantha, V.S., 2016. Gupta, P., Goyal, R.N., 2014. Talanta 120, 17–22.
Electrochim. Acta 206, 77–85. Haas, H.L., Sergeeva, O.A., Selbach, O., 2008. Physiol. Rev. 88, 1183–1241.
Atsushi, Y., Kazutaka, M., Takehiko, W., Hiroshi, W., Yukihiko, K., 1982. Biochem. Hamdi, N., Wang, J., Walker, E., Maidment, N.T., Monbouquette, H.G., 2006. J.
Pharmacol. 31, 305–309. Electroanal. Chem. 591, 33–40.
Atta, N.F., El-Kady, M.F., 2010. Sens. Actuators B 145, 299–310. He, C., Wang, Z., Wang, Y., Hu, R., Li, G., 2016. Biosens. Bioelectron. 85, 679–683.
Atta, N.F., Galal, A., Ahmed, R.A., 2011a. Bioelectrochemistry 80, 132–141. Heeger, A.J., Kivelson, S., Schrieffer, J.R., Su, W.-P., 1988. Rev. Mod. Phys. 60, 781–850.
Atta, N.F., Galal, A., Ahmed, R.A., 2011b. J. Electrochem. Soc. 158, F52–F60. Heffries-Ml, M., Mccullough, D., 2006. Regioregular polythiophene, J. In: Handbook of
Atta, N.F., Galal, A., Ekram, H., 2012. Electrochim. Acta 69, 102–111. Conducting Polymers, Conjugated Polymer, 3rd Ed., CRS press, Boca Raton, FL.
Aynaci, E., Yaşar, A., Arslan, F., 2014. Sens. Actuators B 202, 1028–1036. Huang, J., Xu, W., Gong, Y., Weng, S., Lin, X., 2016. Int. J. Electrochem. Sci. 11,
Basescu, N., Liu, Z.X., Heeger, A.J., Naarmann, H., Theophilou, N., 1987. Nature 327, 8193–8203.
403–405. Jackowska, K., Krysinski, P., 2013. Anal. Bioanal. Chem. 405, 3753–3771.
Basozabal, I., Guerreiro, A., Gomez-Caballero, A., Goicolea, M.A., Barrio, R.J., 2014. Jin, E., Lu, X., Bian, X., Kong, L., Zhang, W., Wang, C., 2010. J. Mater. Chem. 20,
Biosens. Bioelectron. 58, 138–144. 3079–3083.
Bard, A.J., Faulkner, L.R., Leddy, J., Zoski, C.G., 1980. Electrochemical Methods: Kan, X., Zhou, H., Li, C., Zhu, A., Xing, Z., Zhao, Z., 2012. Electrochim. Acta 63, 69–75.
Fundamentals and Applications 2 Wiley, New York. Kandel, E.R., Squire, L.R., 2000. Science 290, 1113–1120.
Beaujuge, P.M., Reynolds, J.R., 2010. Chem. Rev. 110, 268–320. Kanik, F.E., Kolb, M., Timur, S., Bahadir, M., Toppare, L., 2013. Int. J. Biol. Macromol. 59,
Belaidi, F.S., Civélas, A., Castagnola, V., Tsopela, A., Mazenq, L., Gros, P., Launay, 2015. 111–118.
Sens. Actuators B 214, 1–9. Kanik, F.E., Rende, E., Timur, S., Toppare, L., 2012. J. Mater. Chem. 22, 22517–22525.
Bi, Y., 1989. Biosensors 4, 373–380. Kaur, B., Srivastava, R., 2015a. Sens. Actuators B 211, 476–488.
Blanchared P., Leriche P., Frere P., Roncali J., 2006. Advanced functional polythiophene Kaur, B., Srivastava, R., 2015b. New J. Chem. 39, 6899–6906.
based on tailored precursors. In: Handbook of Conducting Polymers, Conjugated Kemp, S.F., Lockey, R.F., Simons, F.E.R., 2008. World Allergy Organ. J. 1, S18.
polymer, 3rd ed., CRS Press, Boca Raton, FL. Kergoat, L., Piro, B., Simon, D.T., Pham, M.C., Noël, V., Berggren, M., 2014. Adv. Mater.
Bostelaar, T., Vitvitsky, V., Kumutima, J., Lewis, B.E., Yadav, P.K., Brunold, T.C., 26, 5658–5664.
Filipovic, M., Lehnert, N., Stemmler, T.L., Banerjee, R., 2016. J. Am. Chem. Soc. 138, Khan, A., Ab Ghani, S., 2012. Biosens. Bioelectron. 31, 433–438.
8476–8488. Khudaish, E.A., Al-Ajmi, K.Y., Al-Harthi, S.H., Al-Hinai, A.T., 2012. J. Electroanal. Chem.
Bouchta, D., Izaoumen, N., Zejli, H., El Kaoutit, M., Temsamani, K.R., 2005. Biosens. 676, 27–34.
Bioelectron. 20, 2228–2235. Khudaish, E.A., Al-Nofli, F., Rather, J.A., Al-Hinaai, M., Laxman, K., Kyaw, H.H., Al-
Bruno, J.P., Sarter, M., Gash, C., Parikh, V., 2004. Eur. J. Neurosci. 20, 1545–1554. Harthy, S., 2016. J. Electroanal. Chem. 761, 80–88.
Calabrese, V., Mancuso, C., Calvani, M., Rizzarelli, E., Butterfield, D.A., Stella, A.M.G., Kim, D.-M., Noh, H.-B., Park, D.S., Ryu, S.-H., Koo, J.S., Shim, Y.-B., 2009. Biosens.
2007. Nat. Rev. Neurosci. 8, 766–775. Bioelectron. 25, 456–462.
Camardese, G., Di Giuda, D., Di Nicola, M., Cocciolillo, F., Giordano, A., Janiri, L., Kim, D.-M., Yoon, J.-H., Won, M.-S., Shim, Y.-B., 2012. Electrochim. Acta 67, 201–207.
Guglielmo, R., 2014. J. Psychiatr. Res. 51, 7–18. Kim, Y.M., Bombeck, C.A., Billiar, T.R., 1999. Circ. Res. 84, 253–256.
Carlsson, A., Lindqvist, M., Magnusson, T.O.R., 1957. Nature 180, 1200. Klimek, V., Stockmeier, C., Overholser, J., Meltzer, H.Y., Kalka, S., Dilley, G., Ordway,
Cesarino, I., Galesco, H.V., Moraes, F.C., Lanza, M.R., Machado, S.A., 2013. G.A., 1997. J. Neurosci. 17, 8451–8458.
Electroanalysis 25, 394–400. Koh, W.C.A., Chandra, P., Kim, D.-M., Shim, Y.-B., 2011. Anal. Chem. 83, 6177–6183.
Chandra, P., Son, N.X., Noh, H.-B., Goyal, R.N., Shim, Y.-B., 2013. Biosens. Bioelectron. Koh, W.C.A., Rahman, M.A., Choe, E.S., Lee, D.K., Shim, Y.-B., 2008. Biosens. Bioelectron.
39, 139–144. 23, 1374–1381.
Chauhan, N., Chawla, S., Pundir, C.S., Jain, U., 2017. Biosens. Bioelectron. 89, 377–383. Kopparthy, V.L., Tangutooru, S.M., Guilbeau, E.J., 2015. Bioengineering 2, 2–14.
Chiang, C.K., Fincher Jr., C.R., Park, Y.W., Heeger, A.J., Shirakawa, H., Louis, E.J., Gau, Krnjević, K., 1974. Physiol. Rev. 54, 418–540.
S.C., MacDiarmid, A.G., 1977. Phys. Rev. Lett. 39, 1098–1101. Kulagina, N.V., Michael, A.C., 2003. Anal. Chem. 75, 4875–4881.
Chicharro, M., Sánchez, A., Zapardiel, A., Rubianes, M.D., Rivas, G., 2004. Anal. Chim. Kwon, N.H., Rahman, M.A., Won, M.-S., Shim, Y.-B., 2006. Anal. Chem. 78, 52–60.
Acta 523, 185–191. Lapainis, T., Rubakhin, S.S., Sweedler, J.V., 2009. Anal. Chem. 81, 5858–5864.
Chih, Y.K., Yang, M.C., 2013. Bioelectrochemistry 91, 44–51. Larsen, S.T., Taboryski, R., 2012a. Analyst 137, 5057–5061.
Chikahisa, S., Kodama, T., Soya, A., Sagawa, Y., Ishimaru, Y., Séi, H., Nishino, S., 2013. Larsen, S.T., Vreeland, R.F., Heien, M.L., Taboryski, R., 2012b. Analyst 137, 1831–1836.
PLoS One 8, e78434. Lee, D.K., Bian, S., Rahman, M.A., Shim, Y.-B., Shim, I., Choe, E.S., 2008. Eur. J.
Chu, W., Zhou, Q., Li, S., Zhao, W., Li, N., Zheng, J., 2015. Appl. Surf. Sci. 353, 425–432. Pharmacol. 590, 157–162.
Cicchetti, D., Cohen, D.J., 2006. Developmental Psychopathology. Volume 3: Risk, Lee, D.K., Koh, W.C.A., Shim, Y.-B., Shim, I., Choe, E.S., 2010. Psychopharmacology 208,
Disorder and Adaptation. 3. John Wiley & Sons, Inc, New York, pp. 129–201. 245–256.
Cooper, J.R., Bloom, F.E., Roth, R.H., 1997. Psyccritiques 42, 172. Lee, D.K., Oh, J.H., Shim, Y.-B., Choe, E.S., 2013a. Neurochem. Res. 38, 1424–1433.
Coyle, J.T., Price, D.L., Delong, M.R., 1983. Science 219, 1184–1190. Lee, D.K., Oh, J.H., Yang, J.H., Youn, B., Shim, Y.-B., Shim, I., Wang, J.Q., Choe, E.S.,
Crowley, K., Morrin, A., Shepherd, R.L., in het Panhuis, M., Wallace, G.G., Smyth, M.R., 2013b. Neurosci. Lett. 541, 120–125.
Killard, A.J., 2010. IEEE Sens. J. 10, 1419–1426. Lee, J.S., Oh, J., Kim, S.G., Jang, J., 2015. Small 11, 2399–2406.
Danielmeier, C., Allen, E.A., Jocham, G., Onur, O.A., Eichele, T., Ullsperger, M., 2015. Lee, J.W., Park, D.S., Shim, Y.-B., Park, S.M., 1992. J. Electrochem. Soc. 139, 3507–3514.
Curr. Biol. 25, 1461–1468. Lee, T.-Y., Shim, Y.-B., 2001. Anal. Chem. 73, 5629–5632.
Dawoud, A.A., Kawaguchi, T., Jankowiak, R., 2007. Electrochem. Commun. 9, Letheby, H., 1862. J. Chem. Soc. 15, 161–163.
1536–1541. Li, B., Shi, X., Gu, W., Zhao, K., Chen, N., Xian, Y., 2013. Analyst 138, 7212–7217.
Dawson, T.M., Dawson, V.L., 2003. Science 302, 819–822. Li, L., Liu, H., Shen, Y., Zhang, J., Zhu, J.J., 2011. Anal. Chem. 83, 661–665.
De Benedetto, G.E., Fico, D., Pennetta, A., Malitesta, C., Nicolardi, G., Lofrumento, D.D., Liang, X., He, Y., Liu, F., Wang, B., Zhong, T., Quan, B., Lu, G., 2007. Sens. Actuators B
De Nuccio, F., La Pesa, V., 2014. J. Pharm. Biomed. Anal. 98, 266–270. 125, 544–549.
Degefu, H., Amare, M., Tessema, M., Admassie, S., 2014. Electrochim. Acta 121, 307–314. Lin, M., 2015. RSC Adv. 5, 9848–9851.
De Irazu, S., Unceta, N., Sampedro, M.C., Goicolea, M.A., Barrio, R.J., 2001. Analyst 126, Liu, S., Xing, X., Yu, J., Lian, W., Li, J., Cui, M., Huang, J., 2012. Biosens. Bioelectron. 36,
495–500. 186–191.
Diaz, A.F., Castillo, J.I., Logan, J.A., Lee, W.-Y., 1981. J. Electroanal. Chem. 129, Lombardo, S., Maskos, U., 2015. Neuropharmacology 96, 255–262.
115–132. Lu, X., Li, Y., Du, J., Zhou, X., Xue, Z., Liu, X., Wang, Z., 2011. Electrochim. Acta 56,
Dunn, A.L., Dishman, R.K., 1991. Exerc. Sport Sci. Rev. 19, 41–98. 7261–7266.
Durst, R.A., 1997. Pure Appl. Chem. 69, 1317–1324. Maciejewska, J., Pisarek, K., Bartosiewicz, I., Krysiński, P., Jackowska, K., Bieguński,
Eisenhofer, G., Kopin, I.J., Goldstein, D.S., 2004. Pharmacol. Rev. 56, 331–349. A.T., 2011. Electrochim. Acta 56, 3700–3706.
Fabregat, G., Casanovas, J., Redondo, E., Armelin, E., Alemán, C., 2014a. Phys. Chem. Mao, H., Liang, J., Zhang, H., Pei, Q., Liu, D., Wu, S., Zhang, Y., Song, X.M., 2015.
Chem. Phys. 16, 7850–7861. Biosens. Bioelectron. 70, 289–298.
Fabregat, G., Córdova-Mateo, E., Armelin, E., Bertran, O., Alemán, C., 2011. J. Phys. Martí, M., Fabregat, G., Estrany, F., Alemán, C., Armelin, E., 2010. J. Mater. Chem. 20,
Chem. C 115, 14933–14941. 10652–10660.
Fabregat, G., Estrany, F., Casas, M.T., Alemán, C., Armelin, E., 2014b. J. Phys. Chem. B McMahon, C.P., Rocchitta, G., Kirwan, S.M., Killoran, S.J., Serra, P.A., Lowry, J.P.,
118, 4702–4709. O’Neill, R.D., 2007. Biosens. Bioelectron. 22, 1466–1473.
Feng, X., Mao, C., Yang, G., Hou, W., Zhu, J.J., 2006. Langmuir 22, 4384–4389. Meltzer, C.C., Smith, G., DeKosky, S.T., Pollock, B.G., Mathis, C.A., Moore, R.Y., Kupfer,
Freed, C.R., Yamamoto, B.K., 1985. Science 229, 62–66. D.J., Reynolds, C.F., 1998. Neuropsychopharmacology 18, 407–430.

551
J.-M. Moon et al. Biosensors and Bioelectronics 102 (2018) 540–552

Min, K., Yoo, Y.J., 2009. Talanta 80, 1007–1011. Polymer, Conjugated Polymer: Processing and Applications, 3rd ed. CRS press, Boca
Mir, T.A., Akhtar, M.H., Gurudatt, N.G., Kim, J.I., Choi, C.S., Shim, Y.-B., 2015. Biosens. Raton, FL.
Bioelectron. 68, 421–428. Slominski, A., Moellmann, G., Kuklinska, E., Bomirski, A., Pawelek, J., 1988. J. Cell Sci.
Mohan, A.V., Aswini, K.K., Biju, V.M., 2014. Sens. Actuators B 196, 406–412. 89, 287–296.
Nathan, C., Xie, Q.W., 1994. Cell 78, 915–918. Soldatkin, O., Nazarova, A., Krisanova, N., Borуsov, A., Kucherenko, D., Kucherenko, I.,
Naveen, M.H., Gurudatt, N.G., Noh, H.-B., Shim, Y.-B., 2016. Adv. Funct. Mater. 26, Pozdnyakova, N., Soldatkin, A., Borisova, T., 2015. Talanta 135, 67–74.
1590–1601. Song, M.J., Kim, S., Min, N.K., Jin, J.H., 2014. Biosens. Bioelectron. 52, 411–416.
Nedergaard, M., Takano, T., Hansen, A.J., 2002. Nat. Rev. Neurosci. 3, 748–755. Spencer, J.P., Jenner, P., Daniel, S.E., Lees, A.J., Marsden, D.C., Halliwell, B., 1998. J.
Niswender, C.M., Conn, P.J., 2010. Annu. Rev. Pharmacol. Toxicol. 50, 295–322. Neurochem. 71, 2112–2122.
Noh, H.-B., Shim, Y.-B., 2016. J. Mater. Chem. A 4, 2720–2728. Stilwell, D.E., Park, S.-M., 1988. J. Electrochem. Soc. 135, 2254–2262.
Noh, H.-B., Revin, S.B., Shim, Y.-B., 2014. Electrochim. Acta 139, 315–322. Stilwell, D.E., Park, S.-M., 1989. J. Electrochem. Soc. 136, 427–433.
Oh, J.H., Lee, D.K., Shim, Y.-B., Ryu, I.S., Seo, S.Y., Kim, J., Yang, J.H., Cho, H.W., Choe, Stoyanova, A., Ivanov, S., Tsakova, V., Bund, A., 2011. Electrochim. Acta 56, 3693–3699.
E.S., 2015. Exp. Brain Res. 233, 1511–1518. Su, Z., Liu, Y., Xie, Q., Chen, L., Zhang, Y., Meng, Y., Li, Y., Fu, Y., Ma, M., Yao, S., 2012.
Oh, W.K., Kim, S., Shin, K.H., Jang, Y., Choi, M., Jang, J., 2013. Talanta 105, 333–339. Biosens. Bioelectron. 36, 154–160.
Özel, R.E., Hayat, A., Andreescu, S., 2015. Anal. Lett. 48, 1044–1069. Syritski, V., Reut, J., Menaker, A., Gyurcsányi, R.E., Öpik, A., 2008. Electrochim. Acta 53,
Pal, R.K., Kundu, S.C., Yadavalli, V.K., 2017. Sens. Actuators B 242, 140–147. 2729–2736.
Pandey, P.C., Chauhan, D.S., Kumari, S., 2011. Developments in strategic materials and Tashkhourian, J., Daneshi, M., Nami-Ana, S.F., 2016. Anal. Chim. Acta 902, 89–96.
computational design II. In: In: Gyekenyesi, A.L., Kriven, W.M., Wang, J., Widjaja, S., Taylor, I.M., Robbins, E.M., Cat, K.A., Cody, P.A., Happe, C.L., Cui, X.T., 2017. Biosens.
Singh, D. (Eds.), Ceramic Engineering and Science Proceedings 32(10). Wiley Press, Bioelectron. 89, 400–410.
Florida, pp. 235–243. Tertiș, M., Cernat, A., Lacatiș, D., Florea, A., Bogdan, D., Suciu, M., Săndulescu, R.,
Park, D.-S., Shim, Y.-B., Park, S.-M., 1993. J. Electrochem. Soc. 140, 609–614. Cristea, C., 2017. Electrochem. Commun. 75, 43–47.
Park, J., Takmakov, P., Wightman, R.M., 2011. J. Neurochem. 119, 932–944. Tomkins, D.M., Sellers, E.M., 2001. Can. Med. Assoc. J. 164, 817–821.
Park, S.J., Lee, S.H., Yang, H., Park, C.S., Lee, C.-S., Kwon, O.S., Park, T.H., Jang, J., 2016. Tsele, T.P., Adekunle, A.S., Fayemi, O.E., Ebenso, E.E., 2017. Electrochim. Acta 243,
ACS Appl. Mater. Interfaces 8, 28897–28903. 331–348.
Park, S.-M., 1997. Electrochemistry of π-conjugated polymer. In: Nalwa, H.S., E. (Ed.), Tseng, T.T.C., Monbouquette, H.G., 2012. J. Electroanal. Chem. 682, 141–146.
Handbook of Organic Conductive Molecules and Polymer. Wiley, Chichestrer. Turner, A., Karube, I., Wilson, G.S., 1987. Biosensors: Fundamentals and Applications.
Patestas, M.A., Gartner, L.P., 2006. A Textbook of Neuroanatomy. Blackwell publishing, Oxford University Press.
Oxford. Ulubay, Ş., Dursun, Z., 2010. Talanta 80, 1461–1466.
Prakash, S., Rao, C.R., Vijayan, M., 2009. Electrochim. Acta 54, 5919–5927. Vincent, S.R., 2010. Prog. Neurobiol. 90, 246–255.
Prasad, B.B., Pandey, I., 2013. Electrochim. Acta 88, 24–34. Virji, S., Fowler, J.D., Baker, C.O., Huang, J., Kaner, R.B., Weiller, B.H., 2005. Small 1,
Prathish, K.P., Carvalho, R.C., Brett, C.M., 2016. Electrochim. Acta 187, 704–713. 624–627.
Qian, T., Yu, C., Zhou, X., Ma, P., Wu, S., Xu, L., Shen, J., 2014a. Biosens. Bioelectron. 58, Vreeland, R.F., Atcherley, C.W., Russell, W.S., Xie, J.Y., Lu, D., Laude, N.D., Porreca, F.,
237–241. Heien, M.L., 2015. Anal. Chem. 87, 2600–2607.
Qian, T., Yu, C., Zhou, X., Wu, S., Shen, J., 2014b. Sens. Actuators B 193, 759–763. Wahono, N., Qin, S., Oomen, P., Cremers, T.I.F., de Vries, M.G., Westerink, B.H.C., 2012.
Qu, N., Wan, B., Guo, L.-H., 2008. Analyst 133, 1246–1249. Biosens. Bioelectron. 33, 260–266.
Rahman, M.A., Kwon, N.H., Won, M.-S., Choe, E.S., Shim, Y.-B., 2005. Anal. Chem. 77, Wang, A.J., Xu, J.J., Chen, H.Y., 2007. Electroanalysis 19, 674–680.
4854–4860. Wang, H., Walaszczyk, E.J., Li, K., Chung-Davidson, Y.W., Li, W., 2012. Anal. Chim. Acta
Rahman, M.A., Kumar, P., Park, D.-S., Shim, Y.-B., 2008a. Sensors 8, 118–141. 721, 147–153.
Rahman, M.A., Noh, H.-B., Shim, Y.-B., 2008b. Anal. Chem. 80, 8020–8027. Wang, H.S., Li, T.H., Jia, W.L., Xu, H.Y., 2006. Biosens. Bioelectron. 22, 664–669.
Raj, M., Gupta, P., Goyal, R.N., Shim, Y.-B., 2017. Sens. Actuators B 239, 993–1002. Wang, J., 2001. Electroanalysis 13, 983–988.
Raoof, J.B., Ojani, R., Rashid-Nadimi, S., 2005. Electrochim. Acta 50, 4694–4698. Wang, J., 2006. Analytical Electrochemistry. John Wiley & Sons.
Ratautaite, V., Nesladek, M., Ramanaviciene, A., Baleviciute, I., Ramanavicius, A., 2014. Wang, L., Hu, P., Deng, X., Wang, F., Chen, Z., 2013. Biosens. Bioelectron. 50, 57–61.
Electroanalysis 26, 2458–2464. Wang, L., Xu, H., Song, Y., Luo, J., Wei, W., Xu, S., Cai, X., 2015a. ACS Appl. Mater.
Revin, S.B., John, S.A., 2012. Analyst 137, 209–215. Interfaces 7, 7619–7626.
Rezaei, B., Boroujeni, M.K., Ensafi, A.A., 2015. Biosens. Bioelectron. 66, 490–496. Wang, W., Wang, W., Davis, J.J., Luo, X., 2015b. Microchim. Acta 182, 1123–1129.
Riedel, G., Platt, B., Micheau, J., 2003. Behav. Brain Res. 140, 1–47. Wang, W., Xu, G., Cui, X.T., Sheng, G., Luo, X., 2014. Biosens. Bioelectron. 58, 153–156.
Robiolio, P.A., Rigolin, V.H., Wilson, J.S., Harrison, J.K., Sanders, L.L., Bashore, T.M., Weaver, C.L., Li, H., Luo, X., Cui, X.T., 2014. J. Mater. Chem. B 2, 5209–5219.
Feldman, J.M., 1995. Circulation 92, 790–795. Whitaker-Azmitia, P.M., 1999. Neuropsychopharmacology 21, 2S–8S.
Sacramento, A.S., Moreira, F.T.C., Guerreiro, J.L., Tavares, A.P., Sales, M.G.F., 2017. Windmiller, J.R., Valdés-Ramírez, G., Zhou, N., Zhou, M., Miller, P.R., Jin, C., Brozik,
Mater. Sci. Eng. C 79, 541–549. S.M., Polsky, R., Katz, E., Narayan, R., Wang, J., 2011. Electroanalysis 23,
Sadanandhan, N.K., Cheriyathuchenaaramvalli, M., Devaki, S.J., Menon, A.R.R., 2017. J. 2302–2309.
Electroanal. Chem. 794, 244–253. Won, M.-S., Rahman, M., Kwon, N.H., Shankaran, D.R., Shim, Y.-B., 2005. Electroanalysis
Saha, S., Sarkar, P., Turner, A.P., 2014. Electroanalysis 26, 2197–2206. 17, 2231–2238.
Santos-Fandila, A., Zafra-Gómez, A., Barranco, A., Navalón, A., Rueda, R., Ramírez, M., Xu, G., Li, B., Cui, X.T., Ling, L., Luo, X., 2013. Sens. Actuators B 188, 405–410.
2013. Talanta 114, 79–89. Xu, G., Wang, W., Li, B., Luo, Z., Luo, X., 2015. Microchim. Acta 182, 679–685.
Sasso, L., Heiskanen, A., Diazzi, F., Dimaki, M., Castillo-León, J., Vergani, M., Landini, E., Xu, Q., Wang, S.F., 2005. Microchim. Acta 151, 47–52.
Raiteri, R., Ferrari, G., Carminati, M., Sampietro, M., 2013. Analyst 138, 3651–3659. Xu, X., Nie, Z., Chen, J., Fu, Y., Li, W., Shena, Q., Yao, S., 2009. Chem. Commun.
Saumya, V., Prathish, K.P., Rao, T.P., 2011. Talanta 85, 1056–1062. 6946–6948.
Scavetta, E., Mazzoni, R., Mariani, F., Margutta, R.G., Bonfiglio, A., Demelas, M., Fiorilli, Xue, C., Wang, X., Zhu, W., Han, Q., Zhu, C., Hong, J., Zhou, X., Jiang, H., 2014. Sens.
S., Marzocchi, M., Fraboni, B., 2014. J. Mater. Chem. B 2, 2861–2867. Actuators B 196, 57–63.
Shahrokhian, S., Saberi, R.S., 2011. Electrochim. Acta 57, 132–138. Yamada, K., Noda, Y., Nakayama, S., Komori, Y., Sugihara, H., Hasegawa, T., Nabeshima,
Shan, L., Dauvilliers, Y., Siegel, J.M., 2015. Nat. Rev. Neurol. 11, 401–413. T., 1995. Br. J. Pharmacol. 115, 852–858.
Sheng, G., Xu, G., Xu, S., Wang, S., Luo, X., 2015. RSC Adv. 5, 20741–20746. Yamada, M., Inanobe, A., Kurachi, Y., 1998. Pharmacol. Rev. 50, 723–757.
Shim, Y.-B., Park, S.-M., 1989. Synth. Met. 29, 169–174. Yang, L., Liu, S., Zhang, Q., Li, F., 2012. Talanta 89, 136–141.
Shim, Y.-B., Won, M.-S., Park, S.-M., 1990. J. Electrochem. Soc. 137, 538–544. Yang, Y., Yang, M., Wang, H., Jiang, J., Shen, G., Yu, R., 2004. Sens. Actuators B 102,
Shirakawa, H., Louis, E.J., MacDiarmid, A.G., Chiang, C.K., Heeger, A.J., 1977. J. Chem. 162–168.
Soc. Chem. Commun. 578–580. Yourong, W., Heqing, Y., E'feng, W., 2001. J. Electroanal. Chem. 497, 163–167.
Shirsat, M.D., Bangar, M.A., Deshusses, M.A., Myung, N.V., Mulchandani, A., 2009. Appl. Yu, Y., Sun, Q., Zhou, T., Zhu, M., Jin, L., Shi, G., 2011. Bioelectrochemistry 81, 53–57.
Phys. Lett. 94, 083502. Zhang, X., Rauch, A., Lee, H., Xiao, H., Rainer, G., Logothetis, N.K., 2007. Rapid Commun.
Shrivastava, S., Jadon, N., Jain, R., 2016. Trac-Trends Anal. Chem. 82, 55–67. Mass Spectrom. 21, 3621–3628.
Si, P., Chen, H., Kannan, P., Kim, D.H., 2011. Analyst 136, 5134–5138. Zhao, H., Bian, X., Galligan, J.J., Swain, G.M., 2010. Diam. Relat. Mater. 19, 182–185.
Sine, S.M., 2012. Physiol. Rev. 92, 1189–1234. Zhao, X.E., Suo, Y.R., 2008. Talanta 76, 690–697.
Skoog, D.A., Holler, F.J. Nieman, T.A., 1998. Principles of Instrumental Analysis. Zheng, X., Zhou, X., Ji, X., Lin, R., Lin, W., 2013. Sens. Actuators B 178, 359–365.
Skotheim, T.A., Lelsenbaumer, R., Reynolds, J.R., 2006. Handbook of Conducting Zhou, Y., He, M., Dong, S., Zheng, J., 2011. J. Electrochem. Soc. 159, F17–F22.

552

You might also like