You are on page 1of 9

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Electrochemistry

Review Article

Electrochemical detection of ATP release in-vitro and


in-vivo
Andreas Hellmann1, Annika Schundner2, Manfred Frick2 and
Christine Kranz1

Abstract CALHM1 ion channel and P2X7 receptors [1*e3].


ATP is one of the most widely distributed extracellular signaling When released into the extracellular space, ATP binds to
molecules, regulating many physiological and pathological purinergic P2 receptors (metabotropic P2Y or ionotropic
processes via activation of purinergic receptors. Extracellular P2X receptors) [4]. Almost every cell type possesses
ATP may originate from cell-regulated release but also from some kind of purinoceptors, and ATP acts as autocrine
cell membrane injury. To measure ATP release in-vitro or in- and/or paracrine signaling molecule essentially in all
vivo for insight into the mechanisms of transmission requires tissues and organs [5,6]. ATP-mediated purinergic
its direct, sensitive, non-invasive detection, ideally with high signaling contributes to a wide range of physiological
spatial and temporal resolution. This current opinion article processes [7e10]. Malfunction, however, has been
highlights recent research in ATP detection using electro- associated with the development or progression of many
chemical methods and presents examples how in-vitro elec- diseases [11] and targeting purinergic signaling offers
trochemical ATP measurements can contribute to insights in promising therapeutic potential [12]. For example, in
purinergic signaling for pivotal functions like alveolar homeo- the lung, extracellular ATP and its metabolites, maintain
stasis and how ATP acts as major excitatory neurotransmitter mucociliary clearance in the airways [13] and alveolar
in the central nervous system. homeostasis by regulating secretion of pulmonary sur-
factant and alveolar fluid transport [14**e16]. On the
Addresses other hand, extracellular ATP also serves as a danger
1
Institute of Analytical and Bioanalytical Chemistry, Ulm University,
signal in the lungs [17]. It is linked to pulmonary
Ulm, Germany
2
Institute of General Physiology, Ulm University, Ulm, Germany inflammation, development of acute lung injury (ALI/
ARDS) and chronic lung diseases [10,18e20]. Clinical
Corresponding author: Kranz, Christine (Christine.kranz@uni-ulm.de) trials are underway for the use of purinergic receptor
antagonists for the treatment of chronic cough [21] and
inhaled P2Y2 agonists for treatment of cystic fibrosis
Current Opinion in Electrochemistry 2023, 39:101282
(CF) [22].
This review comes from a themed issue on Bioelectrochemistry
(2023)
ATP is detected and quantified by various methods
Edited by Elena Ferapontova and Heinz-Bernhard Kraatz including chromatographic methods [23,24], micro-
For complete overview about the section, refer Bioelectrochemistry scopic methods (colorimetry and fluorescence, FRET-
(2023) based measurements) [25,26], and the well-known
Available online 30 March 2023 luciferin/luciferase-based bioluminescence assay
https://doi.org/10.1016/j.coelec.2023.101282 [27,28]. Given the complexity of purinergic signaling,
2451-9103/© 2023 Elsevier B.V. All rights reserved. the determination of local ATP concentrations is still
challenging. Also, the structural similarity of ATP with
the adenylate homologues, adenosine diphosphate
Keywords
Adenosine triphosphate, Purinergic signaling, Electrochemical bio-
(ADP), adenosine monophosphate (AMP) and cyclic
sensors, Aptamers, In-vitro and in-vivo measurements. (c)-AMP) is challenging and requires highly specific
methods. Specificity can be achieved via separation
Introduction techniques (e.g., reversed-phase HPLC) coupled to
Adenosine triphosphate (ATP) is one of the most e.g., mass spectrometry [24], the luciferin/luciferase-
important extracellular signaling molecules. Extracel- based bioluminescence assay [28], or genetically enco-
lular ATP may be released from cells upon cell mem- ded recognition schemes [29,30]. Detection methods
brane injury or cell lysis. In addition, a multiplicity of requiring a sampling step are usually limited in
regulated ATP release mechanisms has been identified. displaying real-time dynamics of ATP release. Electro-
ATP can be released via exocytosis of ATP containing chemical methods have some advantages as they can be
vesicles, or through various channels, including connexin used in-situ, particularly, if micro-sized electrochemical
or pannexin hemichannels, maxi-anion channels, transducers are used. Since neither ATP nor its

www.sciencedirect.com Current Opinion in Electrochemistry 2023, 39:101282


2 Bioelectrochemistry (2023)

metabolic products are reduced or oxidized at suitable demonstrated using amino (NH2)-functionalized
potentials (>1 V [31]), selective detection schemes carbon-fiber electrodes [41]. The amino functionaliza-
using bio-recognition elements such as aptamers [32,33] tion via N-(3- dimethylaminopropyl)-N0 -ethyl-
or enzymes [34,35] are mostly employed. Enzyme-based carbodiimide (EDC) and ethylenediamine (EDA)
biosensors for ATP were pioneered by Scheller and enhances electrostatic interaction and adsorption of the
Pfeiffer [34]. analyte and increases the sensitivity of the measure-
ments. Besides amino functionalization, physical treat-
In the following, we highlight electrochemical ATP ment using N2, O2 or Ar plasma resulted in enhanced
measurements suitable for in-vitro and in-vivo measure- adsorption properties and sensitivity of the carbon-fiber
ments, e.g., stimulated chemically and/or mechanically. electrodes towards ATP [42,43]. Recently, the modifi-
cation of carbon fiber electrodes with metal nano-
Direct electrochemical detection particles (NPs) (e.g., Au NPs or Pt NPs) has been
Fast-scan cyclic voltammetry (FSCV) using carbon-fiber reported, which resulted in higher sensitivities in ATP
microelectrodes (CFMEs), which is well-established for detection using FSCV [44,45*]. Spontaneous and
in-vivo measurements of catecholamines like dopamine mechanosensitive adenosine release was investigated
[36e38], has also been used to monitor purinergic using FSCV in hippocampal (CA1) brain slices [46**].
transmitters, including ATP and adenosine [39] The authors could show that Pannexin1 channels seem
(Figure 1a and 1b). In FSCV, the applied voltage is not to be involved in the spontaneous release of aden-
alternated in a triangular wave fashion with high scan osine, as in wildtype (WT) and global deletion pannexin
rates (up to 2000 V/s), enabling high temporal resolution knockout mice (1Panex1KO), there was neither a sig-
[40]. Real-time detection of exogenously delivered ATP nificant difference in frequency nor concentration. In
to mimic a transient event in brain slices was case of mechanically induced release, lower amounts of

Figure 1

(a) Direct electrochemical oxidation of ATP and (b) FSCV of ATP and adenosine using a triangular and optimized sawhorse waveform (CFE; −0.4 to
1.45 V; scan rate: 400 V/s). Mechanically stimulated adenosine release was detected in rat brain slices and evaluated using an in-slice training set.
Adapted with permission from Ross et al. [39], copyright 2014. (c) Conformational change of an immobilized and redox labeled aptamer due to binding of
ATP, which results in an enhanced electron transfer. (d) Elevated ATP levels were observed with an electrochemical RNA-aptamer sensor upon chemical
stimulation with Ca2+ of astrocytes in cell culture. Adapted with permission from Santos-Cancel et al. [54**], copyright 2019.

Current Opinion in Electrochemistry 2023, 39:101282 www.sciencedirect.com


Electrochemical ATP detection Hellmann et al. 3

adenosine were found in Panex1KO, which was similar to in solutions of high ionic strength like buffered solutions
WT mice treated with inhibitor. Despite the advantages using FET-based sensing is challenging due to shielding
such as sub-second temporal resolution in FSCV, usually by the electrical double layer, however this issue can be
selectivity remains an issue, as this method relies on overcome by using specific stem-loop aptamers as
direct oxidation at the electrode surface at relatively recently demonstrated [55]. Besides issues concerning
high potentials, and thus may be prone to interferences ionic strength of the media, aptamers, and in particular
from other electroactive species present in solution RNA-based ones, are prone to degradation in biological
(e.g., catecholamines, H2O2 etc.). samples. In addition, limited temporal resolution and
sensor size restrict further applications, e.g., implanta-
Aptamer-based biosensors tion in tissue and displaying real-time dynamics of ATP.
Aptamersdsingle- or double-stranded oligonucleotides
are used for selective ATP detection due their high Enzyme-based biosensors
binding affinity and specificity [47e49]. Aptamers are Potentiometric ATP detection
produced in-vitro by a systematic evolution of ligands by Mainly, the ATPases catalyzed reaction of ATP to ADP is
exponential enrichment (SELEX) process [50]. Immo- used in potentiometric ATP measurements. Protons
bilized on the electrode surface, aptamers are designed to (Hþ) are generated and potentiometrically detected.
fold their flexible chains into well-defined three-dimen- For example, ATPases immobilized at the transducer
sional (3D) structures upon binding to their target surface of a FET, generate Hþ by ATP hydrolysis
molecule as schematically shown in Figure 1c. The (Figure 2a), which are recorded as change of charge
conformational change leads to enhanced electron distribution at the transducer, leading to a change in
transfer based on a redox-active moiety tethered to the overall device conductance [56]. A potentiometric
end of the chain. One of the first aptamer-based elec- sensor for ATP using a semiconductor charge-coupled
trochemical ATP sensor was introduced by Fan and co- device (CCD) for label-free imaging of pH has been
workers using target-induced structural switching of a developed using apyrase, which is immobilized via 3-
ferrocence-tagged anti-ATP aptamer [33]. Via self- aminopropyltriethoxysilane (3-APTES) or carboxyethyl
assembly, the 27-base anti-ATP aptamer containing one silanetriol (CEST) at the pH sensitive layer; whereby
thiol moiety at the 30 and one ferrocene moiety at the 50 CEST gave a homogeneous distribution of the enzyme
terminus was immobilized onto gold electrodes. Binding (Figure 2b and 2c) [57]. In a follow-up, the distribution
of ATP leads to a change in distance of the ferrocene of local ATP (ca. 0.5 mM) at a mouse hippocampal slice
moiety to the electrode surface enabling “electron upon electrical stimulation could be obtained without
transfer (eT) ON” state, whereas in absence of ATP re- labeling and further processing of the tissue [58*]. In a
flects the “eT OFF” state. Cyclic voltammetry (CV) and collaborative effort, an ATP-sensing FET has been
square wave voltammetry (SWV) were used for ATP realized at the tip of a dual carbon nanoelectrode [59*].
determination. The aptamer sensor is selective towards Immobilized hexokinase (HEX) produces Hþ in pres-
ATP in presence of e.g., guanosine triphosphate (GTP) ence of ATP and glucose, leading to changes in
and cytidine triphosphate (CTP). However, its metabo- drainesource currents or to shifts in gate voltage.
lites were not investigated, despite their presence in Placing the FET at the orifice of a double barrel nano-
biological matrices. The binding affinity of ATP-aptamers pipette enabled the detection of local ATP secretion
towards ADP, AMP and adenosine remains an issue [51]. from cardiac cell ensembles as well as release of ATP
from single cells upon mechanical and osmotic stress.
Aptamers can be used as recognition elements in field- There is a limited number of reports describing the
effect-transistors (FET), which provide high sensi- potentiometric detection of ATP, as they suffer from
tivity, low-cost fabrication, miniaturization and the drifts and are prone to non-specific interferences e.g.,
advantage that only a small amount of sample volume is pH changes. In addition, if apyrase is used as the
required [52]. For example, bivalent split aptamers can recognition element, the sensor is responding to ATP
significantly enhance the sensitivity towards ATP by a and ADP [60], thus selectivity remains an issue.
simple hybridization of two identical monovalent split
aptamers as demonstrated recently [53]. As shown in Amperometric detection of purinergic signaling
Figure 1d, real-time, selective detection of ATP release molecules
from an astrocyte containing hydrogel-based cell-culture Amperometric ATP biosensors capitalize on the enzy-
was achieved using RNA aptamer-modified electrodes matically catalyzed conversion of ATP to generate
interfaced with the cell culture (Figure 1d top) [54**]. hydrogen peroxide (H2O2) via a consecutive or
ATP release was investigated in dependence of various competitive enzymatic assay (Figure 3a); H2O2 is
stimuli, including Ca2þ, glutamate, and ionomycin and reduced or oxidized at the electrode with the resulting
could be detected with a temporal resolution of seconds. Faraday current being proportional to the ATP amount.
The authors also employed the luciferin/luciferase assay While the consecutive approach uses mainly glycerol
to validate the results. The detection of small molecules kinase (GK) and glycerol-3-phosphate oxidase (G3POx)

www.sciencedirect.com Current Opinion in Electrochemistry 2023, 39:101282


4 Bioelectrochemistry (2023)

Figure 2

(a) Hydrolysis of ATP and ADP by the enzyme apyrase (Apy) releasing H+. (b) Imaging of ATP distribution via a pH sensitive CCD-based FET using
apyrase; no signal response was observed in absence of the enzyme. (c) Histogram of the potential difference with increasing ATP concentrations.
Adapted with permission from Endo et al. [57], copyright 2018.

Figure 3

(a) Enzyme-based detection schemes using either a consecutive enzyme biosensor based on glycerol kinase (GK) and glycerol-3-phosphate oxidase
(G3POx) or a competitive assay based on glucose oxidase (GOX) and hexokinase (HEX). In both approaches H2O2 is generated and detected as
electroactive by-product. (b) An enzyme-based purine sensor is realized using a consecutive approach with adenosine deaminase, nucleoside phos-
phorylase and xanthine oxidase. (c) The purine sensor was applied in a study of 133 patients including 76 stroke and 9 stroke mimics revealing elevated
purine levels in venous blood caused by the stroke. Adapted with permission from Dale et al. [67], copyright 2019.

Current Opinion in Electrochemistry 2023, 39:101282 www.sciencedirect.com


Electrochemical ATP detection Hellmann et al. 5

Figure 4

(a) ATP is loaded into the lamellar body (LB) by a vesicular nucleotide transporter (VNUT) leading to fusion activated calcium entry (FACE) and ultimately
secretion of surfactant. (b) Fusion events are directly linked with released ATP (GOX/HEX-based ATP microbiosensor) after chemical stimulation. The
total ATP concentration could be determined based on the volume under the electrode. Adapted with permission from Fois et al. [1*], copyright 2018. (c)
For determination of mechanically stimulated ATP release, the microbiosensor was positioned 30 mm above alveolar cells seeded on a flexible membrane.
(d) ATP release correlated with mechanical stretch. A variety of control experiments confirmed that ATP was released from ATI cells and depend on the
expression of caveolin-1 and Ca2+-entry via piezo1 channel. Adapted with permission from Diem et al. .[14**], copyright 2020.

with glycerol as co-substrate [61], in the competitive In the following, some physiologically relevant examples
assay, the enzymes hexokinase (HEX) and glucose oxi- using either the consecutive or competitive biosensor
dase (GOX) compete for the substrate glucose in the are highlighted for in-vitro measurements.
presence of ATP [34,62e64]. Due to the presence of
glucose in many biomedical/biological samples, no In-situ ATP release measurements in complex matrices
additional substrate must be added, but changes of such as brain slices, complex cell assemblies and whole
glucose levels during measurements may influence the blood samples for point of care diagnostics that
sensor response. For the consecutive assay glycerol must contribute to the understanding and early diagnosis of
be artificially added as substrate. More recently, direct diseases remain a significant challenge due to rapid
electron transfer of the enzyme to the electrode surface breakdown of ATP and possibly extensive sample
has been described by coupling cellobiose dehydroge- preparation. Dale and co-workers developed a so-called
nase with HEX [65*]. Co-immobilization of pyruvate SMARTchip (Figure 3b) for rapid and on-site analysis
kinase enables recycling of produced ADP by the of total purine content in blood samples, as depicted in
phosphoenolpyruvate catalyzed reaction. Signal ampli- Figure 3e [67,68]. Elevated purine concentrations were
fication up to 220-fold could be achieved, resulting in a observed in mice after status epilepticus correlating
sub-nanomolar limit of detection (LOD) of ATP. A novel with seizure burden and post-seizure neuro-
integrated sensing system for ATP and lactate using a degeneration in the hippocampus [69*]. This sensing
series of catalytically active enzymes (adenylate kinase, approach may contribute to the early diagnosis of
pyruvate kinase, and pyruvate oxidase) and Prussian neonatal encephalopathy as demonstrated in a clinical
blue as electrocatalytic layer for H2O2 reduction was test of 26 infants [70].
demonstrated by K. Nishiyama et al. [66**]. Reliable
detection of ATP in blood samples with high recovery ATP plays a significant role in surfactant secretion,
rates as well as good selectivity in the presence of regulation of alveolar fluid homeostasis, and maintenance
ascorbate, pyruvate, ADP, and urate was demonstrated. of the gasblood barrier [10,15,16]. P2X4 receptors are

www.sciencedirect.com Current Opinion in Electrochemistry 2023, 39:101282


6 Bioelectrochemistry (2023)

expressed in the membrane of lamellar bodies (LBs), Declaration of competing interest


storage organelles for lung surfactant within alveolar type The authors declare that they have no known competing
II (ATII) cells. Upon exocytosis of LBs, the acidic pH financial interests or personal relationships that could
within the LB is neutralized and binding of ATP to P2X4 have appeared to influence the work reported in
receptors results in local fusion-activated cation entry this paper.
(FACE) (Figure 4a). ATP for P2X4 activation is also
stored in the LB but can only bind to P2X4 receptors
Data availability
after pH neutralization [1*]. FACE is essential for sur- No data was used for the research described in
factant secretion and activation [16]. A key aspect of this the article.
study is the ability to correlate the initial fusion event
with a jump in extracellular ATP with high temporal and
Acknowledgments
spatial resolution. Fusion of LBs could be directly linked The Deutsche Forschungsgemeinschaft (DFG e German Research
to ATP release from individual LBs by combining live Foundation) is acknowledged in the frame of GRK 2203 (Pulmosens).
fluorescence imaging with measurements using the ATP
microbiosensor, which was positioned with the help of a References
Papers of particular interest, published within the period of review,
second working electrode (dual-electrode assembly) have been highlighted as:
close to the cells (Figure 4b). Similar sensors were used
* of special interest
to unravel the effects of mechanical stretch on surfactant * * of outstanding interest
secretion. Mechanical deformation/expansion of the
alveoli acts as a strong stimulus for surfactant secretion 1. Fois G, Winkelmann VE, Bareis L, Staudenmaier L, Hecht E,
[71,72]. ATI cells, that cover > 95% of the alveolar * Ziller C, Ehinger K, Schymeinsky J, Kranz C, Frick M: ATP is
stored in lamellar bodies to activate vesicular P2X4 in an
surface, act as mechanosensors in the alveoli and stim- autocrine fashion upon exocytosis. J Gen Physiol 2018, 150:
ulate surfactant secretion from ATII cells in a paracrine 277–291, https://doi.org/10.1085/jgp.201711870.
In this paper, a GOX/HEX-based ATP biosensor was used alongside
way. Diem et al. used a co-culture model of ATI cells and live fluorescence imaging to investigate fusion of lamellar bodies (LB)
surfactant secreting ATII cells on an elastic PDMS and simultaneous ATP release provoked by chemical stimulation. It is
unambiguously demonstrated that ATP is stored in the LB and involved
substrate to show that mechanical stretch results in se- in fusion-activated cation entry (FACE) which is crucial for surfactant
lective, strain-induced ATP release from ATI cells. They secretion and activation.
positioned an ATP microbiosensor above the cells while 2. Lazarowski ER: Vesicular and conductive mechanisms of
applying mechanical strain in live cell imaging experi- nucleotide release. Purinergic Signal 2012, 8:359–373, https://
doi.org/10.1007/s11302-012-9304-9.
ments (Figure 4c,d) [14**].
3. Taruno A: ATP release channels. Int J Mol Sci 2018, 19:808,
https://doi.org/10.3390/ijms19030808.
Conclusions 4. North RA, Barnard EA: Nucleotide receptors. Curr Opin
Purines and in particular ATP as secondary messenger Neurobiol 1997, 7:346–357, https://doi.org/10.1016/S0959-
4388(97)80062-1.
molecule, regulate numerous cellular processes through
purinergic signaling pathways. For in-vitro or even in-vivo 5. Verkhratsky A: Early evolutionary history (from bacteria to
hemichordata) of the omnipresent purinergic signalling: a
detection of purinergic transmitters, spatial and/or tribute to Geoff Burnstock inquisitive mind. Biochem Phar-
temporal resolution is a prerequisite. Electrochemical macol 2021, 187:114261, https://doi.org/10.1016/
j.bcp.2020.114261.
approaches have several advantages for local measure-
6. Burnstock G, Knight GE: Cellular distribution and functions of
ments, which may contribute to address signaling P2 receptor subtypes in different systems. In Int. Rev. Cytol..
pathways and with that regulation of important cellular Edited by Jeon K, Cambridge: Academic Press; 2004:31–304,
functions. Novel electrode designs of nearly any shape https://doi.org/10.1016/S0074-7696(04)40002-3.
and size as well as modification with bio-recognition 7. Novak I: ATP as a signaling molecule: the exocrine focus.
elements such as enzymes and aptamers enable in-situ Physiology 2003, 18:12–17, https://doi.org/10.1152/
nips.01409.2002.
measurements down to the single-cell level. However,
8. Idzko M, Ferrari D, Eltzschig HK: Nucleotide signalling during
necessary advancements such as improved immobiliza- inflammation. Nature 2014, 509:310–317, https://doi.org/
tion techniques and rational design are required to 10.1038/nature13085.
ensure improved operational stability. For in-vivo mea- 9. Burnstock G: Introduction to purinergic signaling. In Methods
surements, FSCV with recent advances in surface Mol. Biol., Humana, New York. Edited by Pelegrín P; 2020:1–15,
https://doi.org/10.1007/978-1-4939-9717-6_1.
modification of carbon fiber electrodes are promising
given the improved selectivity and sensitivity. 10. Wirsching E, Fauler M, Fois G, Frick M: P2 purinergic signaling
in the distal lung in health and disease. Int J Mol Sci 2020, 21:
Combining such measurements with principal compo- 4973, https://doi.org/10.3390/ijms21144973.
nent analysis can assist in distinguishing different ATP 11. Vultaggio-Poma V, Falzoni S, Salvi G, Giuliani AL, Di Virgilio F:
analogues. In our opinion, electrochemical detection of Signalling by extracellular nucleotides in health and disease.
ATP is and will also in futuredalongside well- Biochim Biophys Acta Mol Cell Res 2022, 1869:119237, https://
doi.org/10.1016/j.bbamcr.2022.119237.
established instrumental techniques and biochemical
assaysdcontribute to further insight into purinergic 12. Burnstock G: The therapeutic potential of purinergic signal-
ling. Biochem Pharmacol 2018, 151:157–165, https://doi.org/
signaling and in early diagnosis of diseases. 10.1016/j.bcp.2017.07.016.

Current Opinion in Electrochemistry 2023, 39:101282 www.sciencedirect.com


Electrochemical ATP detection Hellmann et al. 7

13. Lazarowski ER, Boucher RC: Purinergic receptors in airway transfer-based genetically encoded indicators. Proc Natl Acad
hydration. Biochem Pharmacol 2021, 187:114387, https:// Sci USA 2009, 106:15651–15656, https://doi.org/10.1073/
doi.org/10.1016/j.bcp.2020.114387. pnas.0904764106.
14. Diem K, Fauler M, Fois G, Hellmann A, Winokurow N, 27. McElroy WD: The energy source for bioluminescence in an
* * Schumacher S, Kranz C, Frick M: Mechanical stretch activates isolated system. Proc Natl Acad Sci USA 1947, 33:342–345,
piezo1 in caveolae of alveolar type I cells to trigger ATP https://doi.org/10.1073/pnas.33.11.342.
release and paracrine stimulation of surfactant secretion
from alveolar type II cells. Faseb J 2020, 34:12785–12804, 28. Morciano G, Sarti AC, Marchi S, Missiroli S, Falzoni S,
https://doi.org/10.1096/fj.202000613RRR. Raffaghello L, Pistoia V, Giorgi C, Di Virgilio F, Pinton P: Use of
In this contribution, mechanical deformation and subsequent release of luciferase probes to measure ATP in living cells and animals.
ATP was investigated with an ATP microbiosensor positioned at alve- Nat Protoc 2017, 12:1542–1562, https://doi.org/10.1038/
olar (ATI and ATII) cells seeded on a flexible PDMS substrate. In nprot.2017.052.
combination with fusion assays, it has been demonstrated that
29. White D, Yang Q: Genetically encoded ATP biosensors for
caveolae in ATI cells serve as mechanosensors within the alveoli,
direct monitoring of cellular ATP dynamics. Cells 2022, 11:
controlling surfactant secretion from ATII cells in response to stretching.
1920, https://doi.org/10.3390/cells11121920.
15. Thompson KE, Korbmacher JP, Hecht E, Hobi N, Wittekindt OH,
30. Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di
Dietl P, Kranz C, Frick M: Fusion-activated cation entry (FACE)
Virgilio F: Increased level of extracellular ATP at tumor sites:
via P2X4 couples surfactant secretion and alveolar fluid
in vivo imaging with plasma membrane luciferase. PLoS One
transport. Faseb J 2013, 27:1772–1783, https://doi.org/10.1096/
2008, 3:e2599, https://doi.org/10.1371/journal.pone.0002599.
fj.12-220533.
31. Janik B, Elving PJ: Correlation of electrochemical reduction of
16. Miklavc P, Thompson KE, Frick M: A new role for P2X4 re-
adenine nucleosides and nucleotides with structure and
ceptors as modulators of lung surfactant secretion. Front Cell
orientation in solution. J Am Chem Soc 1970, 92:235–243,
Neurosci 2013, 7:2009–2014, https://doi.org/10.3389/
https://doi.org/10.1021/ja00705a001.
fncel.2013.00171.
32. Jhaveri S, Rajendran M, Ellington AD: In vitro selection of
17. Riteau N, Gasse P, Fauconnier L, Gombault A, Couegnat M,
signaling aptamers. Nat Biotechnol 2000, 18:1293–1297,
Fick L, Kanellopoulos J, Quesniaux VFJ, Marchand-Adam S,
https://doi.org/10.1038/82414.
Crestani B, Ryffel B, Couillin I: Extracellular ATP is a danger
signal activating P2X7 receptor in lung inflammation and 33. Zuo X, Song S, Zhang J, Pan D, Wang L, Fan C: A target-
fibrosis. Am J Respir Crit Care Med 2010, 182:774–783, https:// responsive electrochemical aptamer switch (TREAS) for
doi.org/10.1164/rccm.201003-0359OC. reagentless detection of nanomolar ATP. J Am Chem Soc
2007, 129:1042–1043, https://doi.org/10.1021/ja067024b.
18. van Heusden C, Grubb B, Button B, Lazarowski E: Airway
epithelial nucleotide release contributes to mucociliary 34. Scheller F, Pfeiffer D: Glucose oxidase—hexokinase bienzyme
clearance. Life 2021, 11:430, https://doi.org/10.3390/ electrode sensor for adenosine triphosphate. Anal Chim Acta
life11050430. 1980, 117:383–386, https://doi.org/10.1016/0003-2670(80)
87043-7.
19. Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MAM,
Muskens F, Hoogsteden HC, Luttmann W, Ferrari D, Di Virgilio F, 35. Llaudet E, Botting NP, Crayston JA, Dale N: A three-enzyme
Virchow JC, Lambrecht BN: Extracellular ATP triggers and microelectrode sensor for detecting purine release from
maintains asthmatic airway inflammation by activating den- central nervous system. Biosens Bioelectron 2003, 18:43–52,
dritic cells. Nat Med 2007, 13:913–919, https://doi.org/10.1038/ https://doi.org/10.1016/S0956-5663(02)00106-9.
nm1617.
36. Huffman ML, Venton BJ: Carbon-fiber microelectrodes for
20. Shah D, Romero F, Stafstrom W, Duong M, Summer R: Extra- in vivo applications. Analyst 2009, 134:18–24, https://doi.org/
cellular ATP mediates the late phase of neutrophil recruit- 10.1039/B807563H.
ment to the lung in murine models of acute lung injury. Am. J.
Physiol. Cell. Mol. Physiol. 2014, 306:L152–L161, https://doi.org/ 37. Robinson DL, Venton BJ, V Heien MLA, Wightman RM: Detect-
10.1152/ajplung.00229.2013. ing subsecond dopamine release with fast-scan cyclic
voltammetry in vivo. Clin Chem 2003, 49:1763–1773, https://
21. Dicpinigaitis PV, McGarvey LP, Canning BJ: P2X3-Receptor doi.org/10.1373/49.10.1763.
antagonists as potential antitussives: summary of current
clinical trials in chronic cough. Lung 2020, 198:609–616, 38. Bucher ES, Wightman RM: Electrochemical analysis of neu-
https://doi.org/10.1007/s00408-020-00377-8. rotransmitters. Annu Rev Anal Chem 2015, 8:239–261, https://
doi.org/10.1146/annurev-anchem-071114-040426.
22. Kellerman D, Rossi Mospan A, Engels J, Schaberg A, Gorden J,
Smiley L: Denufosol: a review of studies with inhaled P2Y2 39. Ross AE, Venton BJ: Sawhorse waveform voltammetry for
agonists that led to Phase 3. Pulm Pharmacol Ther 2008, 21: selective detection of adenosine, ATP, and hydrogen
600–607, https://doi.org/10.1016/j.pupt.2007.12.003. peroxide. Anal Chem 2014, 86:7486–7493, https://doi.org/
10.1021/ac501229c.
23. Coolen EJCM, Arts ICW, Swennen ELR, Bast A, Stuart MAC,
Dagnelie PC: Simultaneous determination of adenosine 40. Jacobs CB, Ivanov IN, Nguyen MD, Zestos AG, Venton BJ: High
triphosphate and its metabolites in human whole blood by temporal resolution measurements of dopamine with carbon
RP-HPLC and UV-detection. J Chromatogr B 2008, 864:43–51, nanotube yarn microelectrodes. Anal Chem 2014, 86:
https://doi.org/10.1016/j.jchromb.2008.01.033. 5721–5727, https://doi.org/10.1021/ac404050t.
24. Wang J, Lin T, Lai J, Cai Z, Yang MS: Analysis of adenosine 41. Li Y, Weese ME, Cryan MT, Ross AE: Amine-functionalized
phosphates in HepG-2 cell by a HPLC–ESI-MS system with carbon-fiber microelectrodes for enhanced ATP detection
porous graphitic carbon as stationary phase. J Chromatogr B with fast-scan cyclic voltammetry. Anal Methods 2021, 13:
2009, 877:2019–2024, https://doi.org/10.1016/ 2320–2330, https://doi.org/10.1039/D1AY00089F.
j.jchromb.2009.05.027.
42. Li Y, Ross AE: Plasma-treated carbon-fiber microelectrodes
25. Li C, Numata M, Takeuchi M, Shinkai S: A sensitive colorimetric for improved purine detection with fast-scan cyclic voltam-
and fluorescent probe based on a polythiophene derivative metry. Analyst 2020, 145:805–815, https://doi.org/10.1039/
for the detection of ATP. Angew Chem 2005, 117:6529–6532, C9AN01636H.
https://doi.org/10.1002/ange.200501823.
43. Syeed AJ, Li Y, Ostertag BJ, Brown JW, Ross AE: Nano-
26. Imamura H, Huynh Nhat KP, Togawa H, Saito K, Iino R, Kato- structured carbon-fiber surfaces for improved neurochemical
Yamada Y, Nagai T, Noji H: Visualization of ATP levels inside detection. Faraday Discuss 2022, 233:336–353, https://doi.org/
single living cells with fluorescence resonance energy 10.1039/D1FD00049G.

www.sciencedirect.com Current Opinion in Electrochemistry 2023, 39:101282


8 Bioelectrochemistry (2023)

44. Keller AL, Quarin SM, Strobbia P, Ross AE: Platinum of chemically immobilized apyrase. Bull Chem Soc Jpn 2018,
nanoparticle size and density impacts purine electro- 91:304–310, https://doi.org/10.1246/bcsj.20170304.
chemistry with fast-scan cyclic voltammetry. J Electrochem
Soc 2022, 169, 046514, https://doi.org/10.1149/1945-7111/ 58. Doi H, Parajuli B, Horio T, Shigetomi E, Shinozaki Y, Noda T,
ac65bc. * Takahashi K, Hattori T, Koizumi S, Sawada K: Development of a
label-free ATP image sensor for analyzing spatiotemporal
45. Li Y, Keller AL, Cryan MT, Ross AE: Metal nanoparticle modi- patterns of ATP release from biological tissues. Sensor
* fied carbon-fiber microelectrodes enhance adenosine Actuator B Chem 2021, 335:129686, https://doi.org/10.1016/
triphosphate surface interactions with fast-scan cyclic j.snb.2021.129686.
voltammetry. ACS Meas Sci Au 2022, 2:96–105, https://doi.org/ This paper describes a potentiometric sensor for imaging of ATP based
10.1021/acsmeasuresciau.1c00026. on immobilized apyrase. Visualization of ATP distribution of a mouse
This paper reports the modification of carbon fiber microelectrodes with hippocampal slice was achieved upon electrical stimulation.
gold or platinum nanoparticles to improve the sensitivity of direct ATP
detection using fast-scan cyclic voltammetry. 59. Zhang Y, Clausmeyer J, Babakinejad B, López Córdoba A, Ali T,
* Shevchuk A, Takahashi Y, Novak P, Edwards C, Lab M, Gopal S,
46. Lee ST, Chang Y, Venton BJ: Pannexin1 channels regulate Chiappini C, Anand U, Magnani L, Coombes RC, Gorelik J,
* * mechanically stimulated but not spontaneous adenosine Matsue T, Schuhmann W, Klenerman D, Sviderskaya EV,
release. Anal Bioanal Chem 2022, 414:3781–3789, https:// Korchev Y: Spearhead nanometric field-effect transistor sen-
doi.org/10.1007/s00216-022-04047-x. sors for single-cell analysis. ACS Nano 2016, 10:3214–3221,
In this paper, FSCV is used to study the effects of pannexin1 channels https://doi.org/10.1021/acsnano.5b05211.
in spontaneous or mechanically induced adenosine release in live In this contribution, a hexokinase/polypyrrole-modified nanometric
brain slices from wildtype and 1Panex1KO mice. Adenosine release is field-effect transistor-based sensor is realized at the tip of a dual-barrel
predominantly through mechanical stimulation and is mainly regulated carbon nanoelectrode for real-time detection of ATP at the single cell
by pannexin1 channels as demonstrated by control experiments level upon mechanical and osmotic stress.
including inhibition with carbenoxolone and studies at wildtype and
Panex1KO mice. 60. Smith TM, Hicks-Berger CA, Kim S, Kirley TL: Cloning, expres-
sion, and characterization of a soluble calcium-activated
47. Liu Y, Kong L, Li H, Yuan R, Chai Y: Electrochemical aptamer nucleotidase, a human enzyme belonging to a new family of
biosensor based on ATP-induced 2D DNA structure switch- extracellular nucleotidases. Arch Biochem Biophys 2002, 406:
ing for rapid and ultrasensitive detection of ATP. Anal Chem 105–115, https://doi.org/10.1016/S0003-9861(02)00420-4.
2022, 94:6819–6826, https://doi.org/10.1021/
acs.analchem.2c00613. 61. Llaudet E, Hatz S, Droniou M, Dale N: Microelectrode biosensor
for real-time measurement of ATP in biological tissue. Anal
48. Zheng J, Li X, Wang K, Song J, Qi H: Electrochemical nano- Chem 2005, 77:3267–3273, https://doi.org/10.1021/ac048106q.
aptasensor for continuous monitoring of ATP fluctuation at
subcellular level. Anal Chem 2020, 92:10940–10945, https:// 62. Kueng A, Kranz C, Mizaikoff B: Amperometric ATP biosensor
doi.org/10.1021/acs.analchem.0c00569. based on polymer entrapped enzymes. Biosens Bioelectron
2004, 19:1301–1307, https://doi.org/10.1016/j.bios.2003.11.023.
49. Li Y, Liu J: Aptamer-based strategies for recognizing adenine,
adenosine, ATP and related compounds. Analyst 2020, 145: 63. Kueng A, Kranz C, Mizaikoff B: Imaging of ATP membrane
6753–6768, https://doi.org/10.1039/D0AN00886A. transport with dual micro-disk electrodes and scanning
electrochemical microscopy. Biosens Bioelectron 2005, 21:
50. Gold L: SELEX: how it happened and where it will go. J Mol 346–353, https://doi.org/10.1016/j.bios.2004.10.020.
Evol 2015, 81:140–143, https://doi.org/10.1007/s00239-015-
9705-9. 64. Hecht E, Liedert A, Ignatius A, Mizaikoff B, Kranz C: Local
detection of mechanically induced ATP release from bone
51. Ng S, Lim HS, Ma Q, Gao Z: Optical aptasensors for adenosine cells with ATP microbiosensors. Biosens Bioelectron 2013, 44:
triphosphate. Theranostics 2016, 6:1683–1702, https://doi.org/ 27–33, https://doi.org/10.1016/j.bios.2013.01.008.
10.7150/thno.15850.
65. Yarman A, Schulz C, Sygmund C, Ludwig R, Gorton L,
52. Vu C-A, Chen W-Y: Predicting future prospects of aptamers in * Wollenberger U, Scheller FW: Third generation ATP sensor
field-effect transistor biosensors. Molecules 2020, 25:680, with enzymatic analyte recycling. Electroanalysis 2014, 26:
https://doi.org/10.3390/molecules25030680. 2043–2048, https://doi.org/10.1002/elan.201400231.
In this contribution, coupling of hexokinase with a mutated cellobiose
53. Zhang Z, Zhao X, Liu J, Yin J, Cao X: Highly sensitive sandwich dehydrogenase (CDH) and co-immobilized pyruvate kinase enabled
electrochemical sensor based on DNA-scaffolded bivalent sensitive detection of ATP while direct electron transfer from the
split aptamer signal probe. Sensor Actuator B Chem 2020, 311: enzyme CDH to the electrode surface was proposed.
127920, https://doi.org/10.1016/j.snb.2020.127920.
66. Nishiyama K, Mizukami R, Kuki S, Ishida A, Chida J, Kido H,
54. Santos-Cancel M, Simpson LW, Leach JB, White RJ: Direct, real- * * Maeki M, Tani H, Tokeshi M: Electrochemical enzyme-based
* * time detection of adenosine triphosphate release from as- blood ATP and lactate sensor for a rapid and straightforward
trocytes in three-dimensional culture using an integrated evaluation of illness severity. Biosens Bioelectron 2022, 198:
electrochemical aptamer-based sensor. ACS Chem Neurosci 113832, https://doi.org/10.1016/j.bios.2021.113832.
2019, 10:2070–2079, https://doi.org/10.1021/ This paper demonstrates an approach for monitoring ATP and lactate
acschemneuro.9b00033. levels in blood samples based on adenylate kinase, pyruvate kinase
In this study, an electrochemical aptamer sensor is used for detection and pyruvate oxidase in combination with Prussian blue modified
of ATP release in-vitro from astrocytes in a 3D cell culture induced by electrodes for H2O2 reduction.
stimulation with Ca2+, glutamate and ionomycin.
67. Dale N, Tian F, Sagoo R, Phillips N, Imray C, Roffe C: Point-of-
55. Nakatsuka N, Yang K-A, Abendroth JM, Cheung KM, Xu X, care measurements reveal release of purines into venous
Yang H, Zhao C, Zhu B, Rim YS, Yang Y, Weiss PS, blood of stroke patients. Purinergic Signal 2019, 15:237–246,
Stojanovi
c MN, Andrews AM: Aptamer–field-effect transistors https://doi.org/10.1007/s11302-019-09647-4.
overcome Debye length limitations for small-molecule
sensing. Science 2018, 362:319–324, https://doi.org/10.1126/ 68. Tian F, Bibi F, Dale N, Imray CHE: Blood purine measurements
science.aao6750. as a rapid real-time indicator of reversible brain ischaemia.
Purinergic Signal 2017, 13:521–528, https://doi.org/10.1007/
56. Gotoh M, Tamiya E, Karube I, Kagawa Y: A microsensor for s11302-017-9578-z.
adenosine-50 -triphosphate pH-sensitive field effect transis-
tors. Anal Chim Acta 1986, 187:287–291, https://doi.org/10.1016/ 69. Beamer E, Lacey A, Alves M, Conte G, Tian F, Diego-Garcia L,
S0003-2670(00)82920-7. * Khalil M, Rosenow F, Delanty N, Dale N, El-Naggar H,
Henshall DC, Engel T: Elevated blood purine levels as a
57. Endo S, Kato R, Sawada K, Hattori T: Two-dimensional array biomarker of seizures and epilepsy. Epilepsia 2021, 62:
ATP/ADP sensitive image sensor with a uniform distribution 817–828, https://doi.org/10.1111/epi.16839.

Current Opinion in Electrochemistry 2023, 39:101282 www.sciencedirect.com


Electrochemical ATP detection Hellmann et al. 9

The authors present proof of concept studies of purine measurements 71. Dietl P, Frick M, Mair N, Bertocchi C, Haller T: Pulmonary con-
in blood samples towards an approach for supporting early diagnosis of sequences of a deep breath revisited. Neonatology 2004, 85:
seizures and epilepsy. 299–304, https://doi.org/10.1159/000078176.
70. Beamer E, O’Dea MI, Garvey AA, Smith J, Menéndez-Méndez A, 72. Edwards YS: Stretch stimulation: its effects on alveolar type II
Kelly L, Pavel A, Quinlan S, Alves M, Jimenez-Mateos EM, cell function in the lung. Comp Biochem Physiol Part A Mol
Tian F, Dempsey E, Dale N, Murray DM, Boylan GB, Molloy EJ, Integr Physiol 2001, 129:245–260, https://doi.org/10.1016/
Engel T: Novel point-of-care diagnostic method for neonatal S1095-6433(01)00321-X.
encephalopathy using purine nucleosides. Front Mol Neurosci
2021, 14:1–9, https://doi.org/10.3389/fnmol.2021.732199.

www.sciencedirect.com Current Opinion in Electrochemistry 2023, 39:101282

You might also like