Next Article in Journal
First Synthesis of Ergotamine-13CD3 and Ergotaminine-13CD3 from Unlabeled Ergotamine
Previous Article in Journal
Effectiveness of Extracorporeal Shock Wave Therapy after Botulinum Toxin Injection for Post-Stroke Upper Extremity Spasticity: A Randomized Controlled Study
Previous Article in Special Issue
Lake Avernus Has Turned Red: Bioindicator Monitoring Unveils the Secrets of “Gates of Hades”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Analytical Methods for Anatoxin-a Determination: A Review

by
Cristina Plata-Calzado
,
Ana I. Prieto
*,
Ana M. Cameán
and
Angeles Jos
Area of Toxicology, Faculty of Pharmacy, Universidad de Sevilla, Profesor García González 2, 41012 Seville, Spain
*
Author to whom correspondence should be addressed.
Toxins 2024, 16(4), 198; https://doi.org/10.3390/toxins16040198
Submission received: 7 February 2024 / Revised: 17 April 2024 / Accepted: 18 April 2024 / Published: 19 April 2024
(This article belongs to the Special Issue Toxic Cyano Blooms around the World and Related Molecules)

Abstract

:
Anatoxin-a (ATX-a) is a potent neurotoxin produced by several species of cyanobacteria whose exposure can have direct consequences, including neurological disorders and death. The increasing prevalence of harmful cyanobacterial blooms makes the detection and reliable assessment of ATX-a levels essential to prevent the risk associated with public health. Therefore, the aim of this review is to compile the analytical methods developed to date for the detection and quantification of ATX-a levels alone and in mixtures with other cyanotoxins and their suitability. A classification of the analytical methods available is fundamental to make an appropriate choice according to the type of sample, the equipment available, and the required sensitivity and specificity for each specific purpose. The most widely used detection technique for the quantification of this toxin is liquid chromatography–tandem mass spectrometry (LC-MS/MS). The analytical methods reviewed herein focus mainly on water and cyanobacterial samples, so the need for validated analytical methods in more complex matrices (vegetables and fish) for the determination of ATX-a to assess dietary exposure to this toxin is evidenced. There is currently a trend towards the validation of multitoxin methods as opposed to single-ATX-a determination methods, which corresponds to the real situation of cyanotoxins’ confluence in nature.
Key Contribution: The methods with the best validation parameters are those based on LC-MS/MS detection. More validated methods in complex matrices such as plants and fish are needed to determine ATX-a alone or in combination with other cyanotoxins.

1. Introduction

Anatoxin-a (ATX-a) belongs to the group of cyanobacterial toxins, which includes other well-known members such as microcystins (MCs) and cylindrospermopsin (CYN). Chemically, it is a bicyclic amine alkaloid whose molecular formula is C10H15NO. Its molecular weight is 165.23 g/mol, and it is very soluble in water. ATX-a, its analog homoanatoxin-a (HATX-a), and its dihydro derivatives (Figure 1) are produced by different cyanobacterial genera, including Anabaena, Dolichospermum, Aphanizomenon, and Cuspidothrix, among others, with a worldwide distribution. However, ATX-a has generally been detected less frequently than MCs and CYN [1]. This could be related to its degradation through photolysis and non-photochemical reactions under natural conditions [2,3]. In any case, ATX-a also has a significant occurrence, and, for example, Bouma-Gregson et al. [4] found ATX-a in 58.9% of the benthic mat samples they analyzed, while Moreira et al. [5] found that 33% of the samples from seven freshwater ecosystems in Portugal were above the guideline value established for ATX-a in Portuguese freshwater systems. Moreover, this toxin has been detected jointly with other cyanotoxins such as MCs [6,7] or CYN [8]. In certain cases, ATX-a has even been detected at higher levels than MCs, as is the case for samples collected from the Eel River (California), alerting us to the importance of this toxin [4].
The growing occurrence and bioaccumulation of cyanotoxins [9,10] raises concerns about their potential risks. These risks are based on two main factors: hazards and exposure. Regarding the hazards, ATX-a is a well-known neurotoxin. It is a nicotinic agonist and has been shown to bind to acetylcholine receptors and cause the inhibition of the enzyme acetylcholinesterase. Consequently, the neurons continuously propagates neuronal impulses that lead to nerve depolarization through the movement of positively charged ions across the receptor, in addition to the desensitivity of the receptor, blocking neurotransmission [11,12,13]. However, toxicity data for ATX-a are comparatively scarcer than for other cyanotoxins and are a topic worthy of research. Recently, Plata-Calzado et al. [14] reviewed the toxic effects produced by ATX-a under laboratory conditions and evidenced that, in addition to neurotoxic effects, the scientific literature shows that ATX-a can also have immunotoxic effects [15,16] and that oxidative stress and apoptosis are the mechanisms involved. Moreover, ATX-a was shown to be genotoxic in vitro by the micronucleus test [17].
With regard to field studies and case reports, ATX-a has been involved in plenty of animal intoxication cases, which has led to great concern [3,18]. In most of the detected cases, ATX-a caused significant behavioral changes, vomiting, ataxia, locomotor deficiency, respiratory distress, and even death in different animal species, such as ducks, cows, flamingos, etc., with dogs being the animal in which most cases of poisoning have been detected [3]. Concentrations of up to 8700 μg/L ATX-a have been detected in their stomach contents [19], and concentrations up to 357 mg/kg have been detected in dogs’ vomit [20].
In contrast, human data poisonings specifically attributed to ATX-a are not so evident in the literature, as co-exposure with other additional cyanotoxins has been described [1]. However, Biré et al. [21] reported human poisoning in 26 patients due to the consumption of sea figs in France between January 2011 and March 2018. These patients presented with diarrhea, nausea, vomiting, paresthesia, dizziness, and headache, among other symptoms that evoke a cerebellar syndrome. This study reported ATX-a concentrations ranging from 193.7 to 1240.2 µg/kg in sea figs (Microcosmus), lower than those reported to cause toxic effects in animals [1]. This emphasizes the toxicological relevance of ATX-a in humans. Nevertheless, the database of ATX-a toxicity is still limited, and that is the reason why the World Health Organization (WHO) has not yet established a long-term provisional health-based reference value [1].
With respect to the exposure data for ATX-a, it has already been stated that it has a worldwide distribution. The levels reported in the waters are very variable. In open water, the concentrations rarely exceed 10 of µg/L, but they can exceed 1000 µg/L in surface blooms, and in drinking water concentrations, they vary from the low-µg/L range to 8.5 µg/L [1]. Also, in relation to water, recreational activities can be another form of exposure, either by ingestion or inhalation, though this has a lower impact. Regarding oral exposure, the ingestion of contaminated food is another possibility to take into account. Currently, data on the occurrence of ATX-a in food are still limited. However, concentrations of up to 33.00 μg/g of this toxin have been detected in dietary supplements containing cyanobacteria [22]. In relation to seafood, in addition to the study by Biré et al. [21] mentioned above, Amzil et al. [23] detected, for the first time, the presence of this neurotoxin in mussels in France. Moreover, interestingly, different studies have investigated the potential bioaccumulation of ATX-a in fish. Osswald et al. [24] calculated a bioaccumulation factor of 2.65 in juvenile Cyprinus carpio exposed to cells of ATX-a producing strains of Anabaena sp. Osswald et al. [25] determined a bioconcentration factor ranging from 30 to 47 based on fresh weight in juvenile Oncorhynchus mykiss exposed to ATX-a. Thus, they concluded that it was necessary to consider potential human exposure through the ingestion of contaminated fish. Similarly, Pawlik-Skowrońska et al. [26] reported the simultaneous accumulation of ATX-a (up to 30 ng/g fresh weight, muscle) and MCs in three species of fish. On the contrary, Colas et al. [27] did not observe ATX-a accumulation in medaka fish.
For all of this, the exposure assessment must rely on analytical results obtained by techniques and validated methods that allow for the unequivocal determination and quantification of ATX-a. In this sense, there are plenty of analytical methods for ATX-a detection that have been noted in the scientific literature. Thus, the aim of this work was to review the research papers focused on analyzing ATX-a, both alone and in combination with other cyanotoxins, and its classification to facilitate the selection of an appropriate method based on the type of sample (matrix), the available equipment, and the required sensitivity and specificity for each specific purpose. Previously, other reviews have been published looking at methods for the determination of cyanotoxins, including ATX-a [28,29]. However, these works mainly focus on extraction procedures prior to sample analysis and do not consider the validation parameters of the methods, which are important when selecting the most appropriate technique according to the requirements of each specific scenario (type of matrix, toxin recoveries, LOQ, LOD, etc.). Furthermore, they do not focus on ATX-a analytical methods, so these studies do not cover all of the existing literature on ATX-a analytical methods. For all this, the novelty of the present review lies in the fact that it summarizes and classifies all the validation parameters of each method according to the type of sample and detection system used and provides a complete overview and update of all available information up to 2023, which allows one to obtain a picture of the benefits and drawbacks of each method. This review aims to support those who need to work with ATX-a for research, surveillance, or monitoring purposes in their decision-making process to select the best analytical option available currently. Also, it will contribute to improving ATX-a exposure assessments in the framework of risk evaluations.

2. Analytical Methods for ATX-a Determination

The first studies focused on the validation of methods for the determination of ATX- date back to the 1980s, focusing only on ATX-a (Figure 2). However, the trend in recent years has shifted towards the development of methods that allow for the detection of several cyanotoxins simultaneously, as this is a more common scenario in nature. In this case, with regard to multitoxin methods, only those methods for the determination of cyanotoxin mixtures containing ATX-a will be discussed. In addition, the need for the development of rapid and reliable methods considering both scenarios (ATX-a alone or in combination) has led to further research into new methodologies.
The methods developed for ATX-a analysis employ a variety of detection systems. One of the first techniques used for the isolation and purification of ATX-a was thin-layer chromatography (TLC), which is, in addition, a low-cost and swift screening technique [30,31]. However, it is a very basic technique. Therefore, more complex techniques have been developed, such as gas or liquid chromatography coupled with spectroscopy or mass spectrometry (MS) detection. Additionally, immunochemical assays or biosensors can also be employed for its detection (Figure 3). However, more than half of the methods have been performed with MS detection systems, highlighting the tandem mass system (MS/MS) mainly for the analysis of cyanotoxin mixtures.
Based on the type of sample analyzed in the revised articles, most of the methods developed for the determination of ATX-a are in water (54.2%) and cyanobacterial cultures (41.7%), compared to methods in which the presence of ATX-a is analyzed in more complex matrices such as aquatic animals (11.1%), vegetables (2.8%), or algae-based supplements (6.9%) (Figure 4).
To our knowledge, all the methods for the determination of ATX-a alone are listed in Table 1 and Table S1 (Supplementary Materials). In addition, multitoxin methods that include ATX-a determination are summarized in Table 2 and Table S2 (Supplementary Materials).

2.1. Enzyme-Linked Immunosorbent Assay (ELISA)

Cyanotoxins can be determined and quantified by their interactions with specialized antibodies, commonly used to determine other cyanotoxins such as MCs, saxitoxins (STXs), and nodularins (NODs). In fact, in 2016, the US Environmental Protection Agency (US EPA) validated an immunoassay for the determination of MCs and NODs [84]. On the contrary, specific antibodies against ATX-a were not available until recently. For the first time, Cevallos-Cedeño et al. [54] developed a sensitive immunoassay for ATX-a after the same research group generated immunoreagents (bioconjugates and antibodies) following the synthesis of three different functionalized derivatives of ATX-a [85], allowing ATX-a to be controlled at lower part-per-billion levels. The authors also used these immunoreagents to optimize a dipstick assay that provides rapid screening in water samples with levels higher than 2 ng/mL ATX-a [54]. This advancement has made the determination of this cyanotoxin possible through commercial kits (Abraxis, Warminster, PA, USA). In this regard, the kit based on the recognition of toxins by a monoclonal antibody shows a limit of detection (LOD) of around 0.1 µg/L, with recoveries between 98 and 104.4%. However, it is important to keep in mind the limitations of this rapid screening method. This test may experience interferences in more complex matrices; the presence of solvents should be reduced to avoid the matrix effect (methanol concentration should be <2.5%.), and in those cases where positive results require regulatory measures, it is also necessary to perform an analysis using an alternative method.

2.2. DNA Aptamers/Biosensors

Aptamers are single-stranded DNA or RNA sequences that take the form of unique three-dimensional structures, enabling them to recognize a specific target with high affinity. These have multiple applications, including biosensors, which are analytical instruments that allow for the detection of an analyte of interest by means of biorecognition and result in a measurable signal. In this respect, biosensors using ATX-a specific DNA aptamers have been published using different detection modes, such as impedimetric [45], electrical conductance [49], colorimetric [51], and electrochemiluminescence [52] aptasensors. The DNA aptamer characterized by Elshafey et al. [45] was one of the first to be developed. The technique revealed an LOD of 0.5 nM, a linear range for ATX-a concentrations ranging from 1 to 100 nM, and recoveries between 94.8 and 108.6%. In general, these methods have lower LODs than other techniques, even detecting ATX-a at picomolar levels [51]. In addition, aptamers exhibit many advantages because they are simple, specific, and cost-effective, as they do not require complex instrumentation, which makes them a useful tool for field studies and allows for the analysis of a large number of samples in a short time, which is a suitable screening method [86]. However, only Li et al. [73] have developed a fluorescence sensor-based adaptor method for the detection of cyanotoxin mixtures, including ATX-a, CYN, NOD, and MC-LR, in water samples. This method shows very good recoveries (in the range of 97.0–104.6%), proving to be a simple, sensitive, and robust method for the determination of four of the most common cyanotoxins in field samples.

2.3. Ultraviolet and Fluorescence Detection

The early methods developed based on ATX-a determination mainly employed high-performance liquid chromatography (HPLC) coupled with absorbance detection techniques such as ultraviolet–visible (UV-vis) or fluorescence spectroscopy, providing both qualitative and quantitative data in ATX-a analyses. However, most of these methods contained little detailed data on the validation parameters [30,87,88,89,90,91]. Due to the presence of an α, β-unsaturated ketone, ATX-a has maximum UV absorption at 227 nm, allowing for its discrimination from phenylalanine (257 nm), a compound that frequently produces interferences with ATX-a [92]. For UV detectors, the LOD to determine ATX-a can be up to 0.025 µg/L in water samples [91]. Nevertheless, despite being a fast and simple method, it has limitations, such as its low sensitivity and the numerous interferences in more complex matrices, such as environmental samples or animal and plant tissues, causing false positives or negatives. In addition, HPLC-UV does not allow for the detection of ATX-a degradation products, which is of particular relevance in the case of rapidly degrading ATX-a under natural conditions [93]. Such tests on environmental samples can provide useful information in cases of investigating sudden animal deaths after ATX-a has been degraded [94].
Other authors, to increase ATX-a selectivity and LODs, developed different methods using derivatization and fluorescence detection (FLD) [22,36,39,42,90,94,95]. The method developed by James and Sherlock [37] allowed for the detection of ATX-a in fresh water in Ireland and was used to relate this cyanotoxin to cases of neurotoxicosis in dogs [96]. A year later, the method was improved to allow for the simultaneous determination of ATX-a and HATX-a, as well as their degradation products (dihydroanatoxin-a, epoxyanatoxin-a, dihydrohomoanatoxin-a, and epoxyhomoanatoxin-a), with LODs below 10 ng/L and recoveries greater than 80% in water samples and cyanobacterial blooms [94]. In FLD, the excitation and emission wavelengths of ATX-a are frequently set at 470 and 530 nm, respectively [22,39,90]. However, Azevedo et al. [42] investigated the best detection conditions using different excitation and emission wavelengths, obtaining better results with a 480 nm excitation length and a 546 nm emission wavelength. Regarding derivatization, 4-fluoro-7-nitro-2,1,3-benzoxadiazole (NBD-F) is usually used, as it is a simple method that only takes a few minutes and exhibits selective reactivity with primary and secondary amines to form highly fluorescence derivatives [36,90,94,95]. Namera et al. [95] used the derivatization–solid-phase microextraction (SPME) technique coupled to HPLC-FLD to analyze ATX-a in aqueous simples. An improvement to this method was reported by Rellán and Gago-Martínez [39], who showed an LOD of 0.29 ng/mL in river water samples and changed the position of the derivatizing agent at the bottom of the reaction vial to extend the working life of the fiber. Thus, SPME is an effective alternative to conventional solid-phase extraction (SPE) for monitoring samples with a great number of analytes.
HPLC-FLD after derivatization with NBD-F has also been used for ATX-a analysis in more complex samples, such as algae-based dietary supplements [22,36], showing that the method can be robust to the matrix effect. In this case, the method allows for the determination of ATX-a and two of its degradation products (epoxyanatoxin-a and dihydroanatoxin-a) due to the use of o-phthaldialdehyde (OPA) and mercaptoethanol prior to the derivatization reagent [36].
In relation to multitoxin analysis, Vasas et al. [59] demonstrated the applicability of capillary zone electrophoresis (CZE) and micellar electrokinetic chromatography (MEKC) for the spectrophotometric detection (at 230 nm) of ATX-a, CYN, and MC-LR in water bloom samples and culture extracts. Low LODs were obtained for all three toxins (0.89–3.65 µg/mL), including ATX-a (2.63 µg/mL), proving to be a simple and rapid tool for testing the content of these cyanotoxins in environmental samples. However, for more complex samples, the authors recommended using both techniques for the confirmation of the results. Later, other authors applied HPLC-FLD for the analysis of ATX-a, β-methylamino-L-alanine (BMAA), and (2,4-diaminobutyric) DAB using a polymeric cation exchange solid-phase extraction (SPE) method in more complex matrices, as well as water samples (fish and aquatic plants) [63].

2.4. Mass Spectrometry Detection

The MS detection system is mainly based on the comparison of the specific analytical standards of the compound to be determined, being widely used for the determination of cyanotoxins [29]. Both the initial mass and the characteristic molecular ionic fragments are used to ensure the specificity of the assignment. However, in the case of ATX-a, it is important to note that this pattern is similar to that of phenylalanine, so it is necessary to know how this amino acid behaves when developing a method that allows for the detection of ATX-a with MS [97].
Ross et al. [98] were among the first research groups to use MS for ATX-a determination. They performed a comparison of different MS methods, obtaining the best results with the desorption chemical ionization/mass spectrometry tandem (DCI-MS/MS), with an LOD of pure ATX-a of 10 pg/µL. These authors used this method for the analysis of ATX-a in toxin-spiked urine samples, demonstrating that it could also be suitable for these types of matrices. However, it is important to note that, to date, it is not known whether ATX-a can be eliminated in urine, as the metabolism pathways for this toxin are still uncertain [1].
MS coupled to different techniques, such as LC or gas chromatography (GC), has been used for ATX-a determination in different matrices. Nevertheless, most of the methods developed employ LC-MS/MS when using water and cyanobacterial samples. To our knowledge, to date, only one study has focused on the analysis of fish samples using LC-MS/MS, achieving an LOD of 0.2 ng/g of ATX-a after carrying out an extraction by matrix solid-phase dispersion [37]. Moreover, the authors also evaluated the influence of the type of fish on the recovery rate of ATX-a, showing similar recovery values in all cases (around 75%). Dimitrakopoulus et al. [41] developed a LC-ESI-MS/MS method with the lowest reported LOD for the detection of ATX-a in freshwater samples using LC-MS/MS (LOD = 0.65 ng/L). To accomplish this, different types of SPE cartridges were also evaluated to select the most suitable one to carry out ATX-a preconcentration and extraction. Porous graphitized carbon cartridges showed the best recoveries compared to silica-based C18 or polymeric cartridges [41].
On the other hand, few studies employ GC-MS compared to LC-MS [22,34,38,99]. This may be due to the fact that the LODs achieved by this method are higher (µg/L ATX-a levels) than those obtained by LC-MS (ng/L ATX-a levels) and the fact that this method requires more analysis time due to the use of a derivatization process prior to the analysis. Rellán et al. [22] developed a method for the detection of ATX-a in dietary food supplements containing cyanobacteria, using GC-MS as a tool to confirm results obtained by HPLC-FLD. Although this technique has high sensitivity, as mentioned above, it can present errors in more complex matrices.
The use of other techniques coupled to MS has also been explored for ATX-a analysis. For instance, matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) has been performed for the characterization of ATX-a [40]; however, it does not indicate validation parameters for quantitative purposes. Also, laser diode thermal desorption–atmospheric pressure chemical ionization–tandem mass spectrometry (LDTD-APCI-MS/MS) was employed for the first time by Lemoine et al. [43] to identify ATX-a. This technique allows for the elimination of phenylalanine interferences without the need to use LC, thus reducing analysis time. Shortly thereafter, analytical data from this technique were improved using LDTD-APCI coupled with high-resolution mass spectrometry (HRMS) [46]. An LOD and a limit of quantification (LOQ) of up to 0.2 and 0.6 µg/L, respectively, were achieved using less than 15 s of analysis per sample.
More recently, other authors have used HRMS combined with direct analysis in real time (DART) for the analysis of ATX-a in cyanobacterial samples. This technique can be useful for the analysis of a large number of samples in a short time at the expense of the reproducibility of the method (around 30% RSD), with an LOD = 1 ng/mL [50]. Shortly afterwards, an improvement to this method was made using DART-HRMS/MS, which also allowed for the analysis of HATX-a and dihydroanatoxin-a, as well as the reduction of the matrix effect due to the use of an isotopically labeled ATX-a standard [53].
Regarding the methods for the determination of ATX-a in mixtures with other cyanotoxins, most of them have been validated using an MS detection system in different matrix samples. In addition to ATX-a, these methods have been validated to determine MCs (mainly MC-LR) and CYN, followed by other less common toxins such as STX, BMAA, DAB, NOD, okadaic acid (OA), and domoic acid (DA), among others. The first documented methods for the detection of ATX-a and MCs in water and fish muscle were developed by Hormozábal et al. [100,101], who obtained better validation parameters for the less complex matrix (water). Worse results were obtained by Pietsch et al. [102], who reported a very wide range of general toxin recoveries (3.2–96.0%) and a recovery of 50% for ATX-a after performing HPLC-ESI-MS/MS on water samples. They also determined STX and NOD. Subsequently, other authors improved these parameters by reducing the range of toxin recoveries (96–113%) and decreasing the LODs in phytoplankton samples with a higher number of toxins (STX, ATX-a, DA, NOD, six congeners of MCs, OA, and dinophysistoxin-1) [56]. These authors obtained the best overall toxin recovery parameters, with one of the highest ATX-a recoveries (103%) reported in this type of matrix, by LC-ESI-MS. Ortiz et al. [83] obtained similar parameters by LC-QtoF HRMS in cyanobacterial blooms, determining up to 13 congeners of MCs together with ATX-a.
In general, most of these methods have been developed for water samples, obtaining important differences in the validation parameters and showing wide ranges of toxin recoveries (recoveries of 35.5–107.5% were reported by Greer et al. [66]; recoveries of 65–138% were reported by Pekar et al. [67]; and recoveries of 44–113% were reported by Zervou et al. [71]. Among them, Pekar et al. [67] presented the best recoveries for ATX-a (range of 89–138%, compared to 35.5% [66] and 62.3% [71]) using UPLC-MS/MS and LC-MS/MS (see Table 2).
With respect to more complex matrices such as fish tissues, very few works have been published. As mentioned above, the first published report was from Hormozábal et al. [100], who obtained an ATX-a recovery of up to 73% using LC-MS. More recently, Haddad et al. [72] obtained a varied range of recoveries (45–103%) depending on each toxin, presenting a high percentage in the case of ATX-a (103%) using the same technique. Nevertheless, other authors have presented an overall high range of recoveries (83.2–109.8%) for all cyanotoxins, with 97.8% for ATX-a by LC-MS/MS [78]. However, when choosing a multitoxin method, it is very important to consider the recovery parameters of all toxins in general, in addition to those of ATX-a.

Method EPA 545

Because LC-MS is one of the most sensitive techniques, the Environmental Protection Agency (EPA) proposed this analytical method as the official method for the analysis of ATX-a in drinking water samples [103]. This guide indicates all the quality parameters that must be met by laboratories that wish to use this method for the detection of ATX-a and CYN. This method is a liquid chromatography, electrospray ionization, tandem mass spectrometry (LC/ESI-MS/MS) method. Thus, it requires the presence of analysts skilled in the operation of LC/ESI-MS/MS instrumentation and the interpretation of the associated data. Moreover, the minimum reporting levels for the lowest concentration in the laboratory were 0.018 μg/L for ATX-a and 0.063 μg/L for CYN.
The method describes the precision (95–111% for ATX-a) and accuracy (1.0–7.4% for ATX-a) values for ATX-a depending on the type of fortified water tested (water, groundwater, chlorinated surface water), as well as the influence of the contact packaging material (plastic and glass). Moreover, this method allows for the selection of LC columns and their conditions as long as they do not reduce analytical efficiency, and the analyst checks that all acceptance criteria for the quality control of the EPA-545 method are satisfied.

2.5. Gas Chromatography–Electron Capture Detection

GC coupled to electron capture detection (ECD) is also among the techniques used for ATX-a determination [104,105]. GC-ECD analysis of ATX-a requires the use of derivatization to increase the sensitivity of the analysis, resulting in a longer analysis time. To accomplish this, different derivatization processes have been employed, such as ones involving the use of trichloroacetic anhydride [104] or pentafluorobenzylbromide [105]. This technique was able to detect ATX-a levels up to pg levels in both water and cyanobacterial bloom samples [105].

3. General Discussion

The proliferation of toxin-producing cyanobacteria is increasing worldwide, with more and more variants of cyanotoxins being documented in the literature, as well as more alarming data regarding the concentrations of these toxins in samples. The development and validation of analytical methods is essential to alert the population to the presence of cyanotoxins and reduce potential exposure to cyanotoxins [106]. In recent years, cyanotoxins other than MCs, such as ATX-a, have attracted more attention [8]. Concentrations of up to 1430 µg/L and 8000 µg/g dw of ATX-a have been found in environmental samples [18,92,107]. Furthermore, a high bioaccumulation capacity has been demonstrated in fish [25,26].
On the other hand, different authors have demonstrated the presence of ATX-a or its derivatives in algae-based food supplements [108,109]. Therefore, because of the risk that the consumption of water and food contaminated with this toxin may pose, it is important to have adequate analytical methods to determine ATX-a. Furthermore, these methods should be in constant development to meet new analysis demands: the presence of cyanotoxins in different matrices, the coexistence of several toxins in one sample, different concentration ranges, etc.
Several parameters must be taken into account when selecting an ATX-a analysis method. These include the availability of the analytical method, the type of matrix, the required analysis time, and the main validation parameters, such as the LOD, LOQ, % recovery, and precision. Figure 5 summarizes the main advantages and disadvantages of the analytical methods developed for ATX-a detection.
In general, although there are different methods for the detection of ATX-a alone, most of them use MS/MS detection systems [41,43,78,80]. These methods have shown the best validation parameters compared to other detection systems such as UV or FLD. With respect to the analysis of ATX-a alone in a water matrix, the method of analysis validated by Dimitrakopoulos et al. [41] using LC-ESI-MS/MS presents the best LOD levels (below 1 ng/L), a good range of toxin recoveries (73–97%), and adequate precision. However, methods using an MS/MS detection system require a prior cleaning, extraction, or preconcentration step, which increases the time of analysis, labor demands, and cost of the analytical process. A good alternative to them in this matrix (water) can be biosensors. The colorimetric biosensor validated by Nguyen and Jang [51] shows an LOD of 4.45 pM (equivalent to 0.7 ng/L) and a high recovery (89–112%).
More recently, rapid methods using DART-HRMS have been developed for cyanobloom samples; however, they have not shown good accuracy parameters (RSD% up to 34%) [50,53]. Recently, Cevallos-Cedeño et al. [54] developed an ELISA method for ATX-a. Although ELISA methods are fast and inexpensive methods for cyanotoxin analysis, they are less sensitive than other techniques. Nevertheless, they could be used as an alternative or screening method for an initial ATX-a analysis.
Regarding the methods used to analyze the presence of ATX-a in matrices other than water and cyanobacteria, we can find methods developed for fish [37,42] and algae-based supplements [22,108]. In fish tissue, the method of Bogialli et al. [37] presents a higher sensitivity, allowing for the detection of ATX-a at lower levels using LC-ESI-MS/MS, while the method developed by Azevedo et al. [42] presents a higher recovery rate and an RSD% < 1% when using HPLC-FLD. Considering the bioaccumulation of the above-mentioned toxin, it is essential to be able to develop new validated methods in different matrices (fish, plants, etc.) in order to carry out a correct risk assessment from a food safety point of view.
In recent years, multitoxin determination methods have attracted more interest than ATX-a alone analysis. This is because the presence of different cyanotoxins at the same time in field samples is becoming more and more frequent for different reasons. On the one hand, in nature, it is common to find different strains producing toxins that coexist in the same cyanobacterial bloom, while on the other hand, the same strain can lead to the production of different types of cyanotoxins [110]. Similarly to methods that focus on ATX-a alone, the analysis of ATX-a in combination with other cyanotoxins mainly uses MS/MS as the detection method.
Overall, multitoxin analytical methods show worse validation parameters (recovery rates, LOD, LOQ, and precision) than those focused on individual ATX-a analysis, despite most of them being more novel. This is largely due to the fact that the extraction process must consider the presence of both hydrophilic and hydrophobic toxins. However, we can find some multitoxin methods that show good recoveries of all the toxins analyzed (around 80–110%) [61,62,69,74]. These methods may be a good option for samples containing MCs [69], CYN [61], NOD [62], or even anabaenopeptins [74]. Regarding detection limits, the method of Filatova et al. [75] shows low detection levels for ATX-a, CYN, NOD, and MCs (order of ng/L), but it has the disadvantage of showing low recovery levels for some of the MC variants.
In relation to the type of matrix, water and cyanobacterial samples are also the matrices in which most analytical methods have been developed for cyanotoxin mixtures. However, fewer multitoxin determination methods have been developed in cyanoblooms compared to those for ATX-a. On the other hand, the number of multitoxin methods developed based on aquatic animals and vegetables is higher compared to the analytical methods for ATX-a alone. The method of Skafi et al. [78] can be applied for the analysis of ATX-a, HATX-a, CYN, 12 MC variants, anabaenopeptins A and B, and cyanopeptolin-A in fish tissue by LC-MS/MS, with recoveries between 83.2 and 109.8%. In addition, a validated method for spinach samples has recently been published that reports validation parameters [83]. This method uses capillary electrophoresis with tandem mass spectrometry (CE-MS/MS) to analyze MC-LR, MC-RR, CYN, ATX-a, NOD, DAB, BMAA, and AEG with good precision (%RSD < 12%) and LODs between 0.03 and 0.23 µg/kg. As evidenced by multitoxin methods with other matrices such as water [71,72,75], this method does not show high toxin recoveries (range 65.5–81.0%).
Nevertheless, although better toxin recovery data are obtained with ATX-a-only methods (more than 70%) [36,37,41,42], they may not be as practical for risk assessments. This is because ATX-a-only methods do not consider the possible presence of other cyanotoxins in samples, even those potentially more toxic and more abundant than ATX-a in natural samples, such as MC-LR and CYN.
Another important aspect of the analysis is to consider the possibility of degradation of ATX-a under environmental conditions. Methods that consider the determination of its degradation products allow for more realistic data on the status of this neurotoxin to be obtained. In this regard, there are different methods focused on the determination of ATX-a that can be applied [44,50,53,94]. However, there are few multitoxin methods that consider the detection of the products derived from ATX-a [60], meaning that, in cases in which this toxin is thought to be the main cause of poisoning, it may be advisable to use methods that only determine ATX-a and ATX-a-derived compounds.
Finally, in the determination of ATX-a, it is important to take into account its potential interference with phenylalanine, an amino acid present in foods rich in proteins. In this regard, several methods have been optimized to avoid erroneous results and false positives due to the presence of this amino acid in samples [41,43,46,48].
Taking all this into account, the selection of the most appropriate ATX-a analytical method in each case is multifactorial and must consider all the possibilities described above.

4. Conclusions

The need for rapid and reliable methods for the determination of ATX-a has led to the publication of a large number of articles focused on the analysis of both ATX-a alone and in mixtures with other cyanotoxins, with the multitoxin mode being highlighted in recent years. In general, the methods focused on ATX-a alone present a better sensitivity in determining this toxin. However, realistically, natural samples frequently contain several toxins.
In summary, bearing in mind the methods currently available for ATX-a analysis, to choose an appropriate method, it is necessary to consider the following: (1) the type of matrix; (2) whether it is likely to find more cyanotoxins as well as ATX-a and, if so, the types of these cyanotoxins; (3) the quality of the analytical parameters required; and (4) the cost and speed requirements. Most validated methods for the determination of ATX-a are based on the MS detection system and use simple matrices such as water samples or cyanobacterial blooms. Therefore, it is necessary to develop analytical methods for this toxin, both alone and in combination with other cyanotoxins, in more complex matrices (vegetables, crops, and fish).

5. Materials and Methods

5.1. The Information Sources and Search Strategy

Our search for information was conducted using research databases including Web of Science, Scopus, Science Database, and PubMed up to January 2024. The selected keywords for all search engines were as follows: anatoxin-a, cyanotoxins, detection methods, mass spectrometry. Furthermore, the bibliographies of the retrieved articles were reviewed to enhance the comprehensiveness of the search.

5.2. Inclusion and Exclusion Criteria

The following criteria were considered in the information selection process:
Inclusion criteria: (1) articles based on ATX-a determination methods providing optimization or new analytical validation parameters for ATX-a alone or in cyanotoxin mixtures; (2) articles published prior to January 2024; and (3) articles reporting comprehensive results published in internationally recognized journals.
Exclusion criteria: (1) articles on ATX-a determination that use methods already published by other authors and do not provide any new developments; (2) articles based on ATX-a(s) determination methods; (3) articles based on methods for the determination of cyanotoxin mixtures that do not consider the detection of ATX-a; (4) articles published in a language other than English; and (5) articles for which the abstract is only available.
Based on all the studies that complied with the inclusion criteria, different tables pertaining to the following were prepared: analytical methods focused on the determination of ATX-a published up to 2003 (Table S1) and from 2003 to date (Table 1), and analytical methods for the determination of cyanotoxin mixtures containing ATX-a published up to 2003 (Table S2) and from 2003 to date (Table 2). In addition, Figure 1, Figure 2 and Figure 3 were elaborated with the data contained in the articles of Table 1 and Table 2 and Supplementary Tables S1 and S2 considering all the ATX-a analysis works found.
Regarding the risk of bias due to the quality of the studies used for the analysis (Table S3), most of them showed a low (59.1%) or medium (37.8%) risk of bias, while only a minority (12.1%) demonstrated a high risk of bias, with these studies mainly being older in terms of date of publication and primarily focusing on the determination of AXT-a alone.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/toxins16040198/s1. Table S1: Analytical methods focused on the determination of ATX-a up to 2003 [111]; Table S2: Analytical methods focused on the determination of cyanotoxin mixtures containing ATX-a up to 2003. Table S3: Risk of bias for the methodological quality of studies reporting different analytical methods for ATX-a determination—0: not reported; 1: not appropriately or clearly evaluated; 2: appropriately evaluated. M: medium (4–6); L: low (7–8); H: high (0–3).

Author Contributions

Conceptualization, C.P.-C., A.I.P. and A.J.; methodology, C.P.-C., A.I.P. and A.J.; formal analysis, C.P.-C. and A.I.P.; investigation, C.P.-C., A.I.P., A.M.C. and A.J.; resources, A.J. and A.M.C.; data curation, C.P.-C. and A.I.P.; writing—original draft preparation, C.P.-C. and A.I.P.; writing—review and editing, A.M.C. and A.J.; visualization, C.P.-C., A.I.P., A.M.C. and A.J.; supervision, A.I.P., A.M.C. and A.J.; project administration, A.J. and A.M.C.; funding acquisition, A.J. and A.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Spanish Ministerio de Ciencia e Innovación (Project PID2019-104890RB-I00 funded by MICIU/AEI/10.13039/501100011033). C. Plata-Calzado is grateful for the financial support of the Junta de Andalucía (PREDOC_00447 contract).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article and Supplementary Materials.

Acknowledgments

The authors want to thank the Spanish Ministerio de Ciencia e Innovación (Project PID2019-104890RB-I00 funded by MICIU/AEI/10.13039/501100011033) and the Junta de Andalucía (PREDOC_00447 contract).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. World Health Organization (WHO). Cyanobacterial Toxins: Anatoxin-a and Analogues; Background Document for Development of WHO Guidelines for Drinking-Water Quality and Guidelines for safe Recreational Water Environments; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  2. Stevens, D.K.; Krieger, R.I. Stability studies on the cyanobacterial nicotinic alkaloid anatoxin-a. Toxicon 1991, 29, 167–179. [Google Scholar] [CrossRef]
  3. Colas, S.; Marie, B.; Lance, E.; Quiblier, C.; Tricoire-Leignel, H.; Mattei, C. Anatoxin-a: Overview on a harmful cyanobacterial neurotoxin from the environmental scale to the molecular target. Environ. Res. 2021, 193, 110590. [Google Scholar] [CrossRef]
  4. Bouma-Gregson, K.; Kudela, R.M.; Power, M.E. Widespread anatoxin-a detection in benthic cyanobacterial mats throughout a river network. PLoS ONE 2018, 13, e0197669. [Google Scholar] [CrossRef]
  5. Moreira, C.; Gomes, C.; Vasconcelos, V.; Antunes, A. Cyanotoxins Occurrence in Portugal: A New Report on Their Recent Multiplication. Toxins 2020, 12, 154. [Google Scholar] [CrossRef]
  6. Sabart, M.; Crenn, K.; Perrière, F.; Abila, A.; Leremboure, M.; Colombet, J.; Jousse, C.; Latour, D. Co-occurrence of microcystin and anatoxin-a in the freshwater lake Aydat (France): Analytical and molecular approaches during a three-year survey. Harmful Algae 2015, 48, 12–20. [Google Scholar] [CrossRef]
  7. Cerasino, L.; Salmaso, N. Co-occurrence of anatoxin-a and microcystins in Lake Garda and other deep perialpine lakes. Adv. Oceanogr. Limnol. 2020, 11, 11–21. [Google Scholar] [CrossRef]
  8. Blahova, L.; Sehnal, L.; Lepsova-Skacelova, O.; Szmucova, V.; Babica, P.; Hilscherova, K.; Teikari, J.; Sivonen, K.; Blaha, L. Occurrence of cylindrospermopsin, anatoxin-a and their homologs in the southern Czech Republic—Taxonomical, analytical, and molecular approaches. Harmful Algae 2021, 108, 102101. [Google Scholar] [CrossRef]
  9. Ríos, V.; Moreno, I.; Prieto, A.I.; Puerto, M.; Gutiérrez-Praena, D.; Soria-Díaz, M.E.; Cameán, A.M. Analysis of MC-LR and MC-RR in tissue from freshwater fish (Tinca tinca) and crayfish (Procambarus clarkii) in tench ponds (Cáceres, Spain) by liquid chromatography-mass spectrometry (LC-MS). Food Chem. Toxicol. 2013, 57, 170–178. [Google Scholar] [CrossRef]
  10. Guzmán-Guillén, R.; Moreno, I.; Prieto Ortega, A.I.; Soria-Díaz, M.E.; Vasconcelos, V.; Cameán, A.M. CYN determination in tissues from freshwater fish by LC-MS/MS: Validation and application in tissues from subchronically exposed tilapia (Oreochromis niloticus). Talanta 2015, 131, 452–459. [Google Scholar] [CrossRef]
  11. Spivak, C.E.; Witkop, B.; Albuquerque, E.X. Anatoxin-a: A novel, potent agonist at the nicotinic receptor. Mol. Pharmacol. 1980, 18, 384–394. [Google Scholar]
  12. Aráoz, R.; Molgó, J.; Tandeau de Marsac, N. Neurotoxic cyanobacterial toxins. Toxicon 2010, 56, 813–828. [Google Scholar] [CrossRef]
  13. Bruno, M.; Ploux, O.; Metcalf, J.S.; Mejean, A.; Pawlik-Skowronska, B.; Furey, A. Anatoxin-a, homoanatoxin-a, and natural analogues. In Handbook of Cyanobacterial Monitoring and Cyanotoxin analysis, 1st ed.; Meriluoto, J., Spoof, L., Codd, G.A., Eds.; John Wiley & Sons, Ltd.: New York, NY, USA, 2017; pp. 138–147. [Google Scholar]
  14. Plata-Calzado, C.; Prieto, A.I.; Cameán, A.M.; Jos, A. Toxic effects produced by anatoxin-a under laboratoty conditions: A review. Toxins 2022, 14, 861. [Google Scholar] [CrossRef]
  15. Rao Lakshmana, P.; Bhattacharya, R.; Gupta, N.; Parida, M.; Bhaskar, A.; Dubey, R. Involvement of caspase and reactive oxygen species in cyanobacterial toxin anatoxin-a-induced cytotoxicity and apoptosis in rat thymocytes and Vero cells. Arch. Toxicol. 2002, 76, 227–235. [Google Scholar] [CrossRef]
  16. Zhong, Y.; Shen, L.; Ye, X.; Zhou, D.; He, Y.; Li, Y.; Ding, Y.; Zhu, W.; Ding, J.; Zhang, H. Neurotoxic anatoxin-a can also exert immunotoxicity by the induction of apoptosis on Carassius auratus lymphocytes in vitro when exposed to environmentally relevant concentrations. Front. Physiol. 2020, 11, 316. [Google Scholar] [CrossRef]
  17. Plata-Calzado, C.; Diez-Quijada, L.; Medrano-Padial, C.; Prieto, A.I.; Cameán, A.M.; Jos, A. In vitro mutagenic and genotoxic assessment of anatoxin-a alone and in combination with cylindrospermopsin. Toxins 2023, 15, 458. [Google Scholar] [CrossRef]
  18. Testai, E. Anatoxin-a and analogues. In Toxic Cyanobacteria in Water. A Guide to Their Public Health Consequences, Monitoring and Management, 2nd ed.; Chorus, I., Welker, M., Eds.; CRC Press: Boca Raton, FL, USA, 2021; pp. 72–94. [Google Scholar]
  19. Fastner, J.; Beulker, C.; Geiser, B.; Hoffmann, A.; Kröger, R.; Teske, K.; Hoppe, J.; Mundhenk, L.; Neurath, H.; Sagebiel, D.; et al. Fatal neurotoxicosis in dogs associated with tychoplanktic, anatoxin-a producing Tychonema sp. in mesotrophic Lake Tegel, Berlin. Toxins 2018, 10, 60. [Google Scholar] [CrossRef]
  20. McCarron, P.; Rafuse, C.; Scott, S.; Lawrence, J.; Bruce, M.R.; Doothwright, E.; Murphy, C.; Reith, M.; Beach, D.G. Anatoxins from benthic cyanobacteria responsible for dog motalities in New Brunswick, Canada. Toxicon 2023, 227, 107086. [Google Scholar] [CrossRef]
  21. Biré, R.; Bertin, T.; Dom, I.; Hort, V.; Schmitt, C.; Diogène, J.; Lemée, R.; De Haro, L.; Nicolas, M. First Evidence of the presence of anatoxin-a in sea figs associated with human food poisonings in France. Mar. Drugs 2020, 18, 285. [Google Scholar] [CrossRef]
  22. Rellán, S.; Osswald, J.; Saker, M.; Gago-Martinez, A.; Vasconcelos, V. First detection of anatoxin-a in human and animal dietary supplements containing cyanobacteria. Food Chem. Toxicol. 2009, 47, 2189–2195. [Google Scholar] [CrossRef]
  23. Amzil, Z.; Derrien, A.; Terre Terrillon, A.; Savar, V.; Bertin, T.; Peyrat, M.; Duval, A.; Lhaute, K.; Arnich, N.; Hort, V.; et al. Five years monitoring the emergence of unregulated toxins in shellfish in France (EMERGTOX 2018-2022). Mar. Drugs 2023, 21, 435. [Google Scholar] [CrossRef]
  24. Osswald, J.; Rellan, S.; Carvalho, A.P.; Gago, A.; Vasconcelos, V. Acute effects of an anatoxin-a producing cyanobacterium on juvenile fish—Cyprinus carpio L. Toxicon 2007, 49, 693–698. [Google Scholar] [CrossRef]
  25. Osswald, J.; Azevedo, J.; Vasconcelos, V.; Guilhermino, L. Experimental determination of the bioconcentration factors for anatoxin-a in juvenile rainbow trout (Oncorhynchus mykiss). Proc. Int. Acad. Ecol. Environ. Sci. 2011, 1, 77–86. [Google Scholar]
  26. Pawlik-Skowrońska, B.; Toporowska, M.; Rechulicz, J. Simultaneous accumulation of anatoxin-a and microcystins in three fish species indigenous to lakes affected by cyanobacterial blooms. Oceanol. Hydrobiol. Stud. 2012, 41, 53–65. [Google Scholar] [CrossRef]
  27. Colas, S.; Duval, C.; Marie, B. Toxicity, transfer and depuration of anatoxin-a (cyanobacterial neurotoxin) in medaka fish exposed by single-dose gavage. Aquat. Toxicol. 2020, 222, 105422. [Google Scholar] [CrossRef]
  28. Picardo, M.; Filatova, D.; Nuñez, O.; Farré, M. Recent advances in the detection of natural toxins in freshwater environments. Trend Anal. Chem. 2019, 112, 75–86. [Google Scholar] [CrossRef]
  29. Sundaravadivelu, D.; Sanan, T.T.; Venkatapathy, R.; Mash, H.; Tettenhorst, D.; Danglada, L.; Frey, S.; Tatters, A.O.; Lazorchak, J. Determination of cyanotoxins and prymnesins in water, fish tissue, and other matrices: A review. Toxins 2022, 14, 213. [Google Scholar] [CrossRef]
  30. Harada, K.; Kimura, Y.; Ogawa, K.; Suzuki, M.; Dahlem, A.M.; Beasley, V.R.; Carmichael, W.W. A new procedure for the analysis and purification of naturally occurring anatoxin-a from the blue-green alga Anabaena flos-aquae. Toxicon 1989, 27, 1289–1296. [Google Scholar] [CrossRef]
  31. Ojanperä, I.; Vuori, E.; Himberg, K.; Waris, M.; Niinivaara, K. Facile detection of anatoxin-a in algal material by thin-layer chromatography with fast black K salt. Analyst 1991, 116, 265–267. [Google Scholar] [CrossRef]
  32. Furey, A.; Crowley, J.; Lehane, M.; James, K.J. Liquid chromatography with electrospray ion-trap mass spectrometry for the determination of anatoxins in cyanobacteria and drinking water. Rapid Commun. Mass Spectrom. 2003, 17, 583–588. [Google Scholar] [CrossRef]
  33. Dagnino, D.; Schripsema, J. 1H NMR quantification in very dilute toxin solutions: Application to anatoxin-a analysis. Toxicon 2005, 46, 236–240. [Google Scholar] [CrossRef]
  34. Ghassempour, A.; Mashkouri Najafi, N.; Mehdiania, A.; Hosseiny Davarani, S.S.; Fallahi, M.; Nakhshab, M. Analysis of anatoxin-a using polyaniline as a sorbent in solid-phase microextraction coupled to gas chromatography-mass spectrometry. J. Chromatogr. A 2005, 1078, 120–127. [Google Scholar] [CrossRef] [PubMed]
  35. James, K.J.; Crowley, J.; Hamilton, B.; Lehane, M.; Skulberg, O.; Furey, A. Anatoxins and degradation products, determined using hybrid quadrupole time-of-flight and quadrupole ion-trap mass spectrometry: Forensic investigations of cyanobacterial neurotoxin poisoning. Rapid Commun. Mass Spectrom. 2005, 19, 1167–1175. [Google Scholar] [CrossRef] [PubMed]
  36. Rawn, D.F.K.; Lau, B.; Niedzwiadek, B.; Lawrence, J.F. Improved method for the determination of anatoxin-a and two of its metabolites in blue-green algae using liquid chromatography with fluorescence detection. J. AOAC Int. 2005, 88, 1741–1747. [Google Scholar] [CrossRef] [PubMed]
  37. Bogialli, S.; Bruno, M.; Curini, R.; Di Corcia, A.; Laganà, A. Simple and rapid determination of anatoxin-a in lake water and fish muscle tissue by liquid-chromatography-tandem mass spectrometry. J. Chromatogr. A 2006, 1122, 180–185. [Google Scholar] [CrossRef] [PubMed]
  38. Rodríguez, V.; Yonamine, M.; Pinto, E. Determination of anatoxin-a in environmental water samples by solid-phase microextraction and gas chromatography-mass spectrometry. J. Sep. Sci. 2006, 29, 2085–2090. [Google Scholar] [CrossRef] [PubMed]
  39. Rellán, S.; Gago-Martínez, A. Improved conditions for the application of solid phase microextraction prior to HPLC-FLD analysis of anatoxin-a. J. Sep. Sci. 2007, 30, 2522–2528. [Google Scholar] [CrossRef]
  40. Aráoz, R.; Guérineau, V.; Rippka, R.; Palibroda, N.; Herdman, M.; Laprevote, O.; Döhren, H.; Tandeau de Marsac, N.; Erhard, M. MALDI-TOF-MS detection of the low molecular weight neurotoxins anatoxin-a and homoanatoxin-a on lyophilized and fresh filaments of axenic Oscillatoria strains. Toxicon 2008, 51, 1308–1315. [Google Scholar] [CrossRef]
  41. Dimitrakopoulus, I.K.; Kaloudis, T.S.; Hiskia, A.E.; Thomaidis, N.S.; Koupparis, M.A. Development of a fast and selective method for the sensitive determination of anatoxin-a in lake waters using liquid chromatography-tandem mass spectrometry and phenylalanine-d5 as internal standard. Anal. Bioanal. Chem. 2010, 397, 2245–2252. [Google Scholar] [CrossRef]
  42. Azevedo, J.; Osswald, J.; Guilhermino, L.; Vasconcelos, V. Development and validation of an SPE-HPLC-FL method for the determination of anatoxin-a in water and trout (Oncorhincus mykiss). Anal. Lett. 2011, 44, 1431–1441. [Google Scholar] [CrossRef]
  43. Lemoine, P.; Roy-Lachapelle, A.; Prévost, M.; Tremblay, P.; Solliec, M.; Sauvé, S. Ultra-fast analysis of anatoxin-a using laser diode thermal desorption-atmospheric pressure chemical ionization-tandem mass spectrometry: Validation and resolution from phenylalanine. Toxicon 2013, 61, 165–174. [Google Scholar] [CrossRef]
  44. Sanchez, J.A.; Otero, P.; Alfonso, A.; Ramos, V.; Vasconcelos, V.; Aráoz, R.; Molgó, J.; Vieytes, M.R.; Botana, L.M. Detection of anatoxin-a and three analogs in Anabaena spp. Cultures: New fluorescence polarization assay and toxin profile by LC-MS/MS. Toxins 2014, 6, 402–415. [Google Scholar] [CrossRef] [PubMed]
  45. Elshafey, R.; Siaj, M.; Zourob, M. DNA aptamers selection and characterization for development of label-free impedimetric aptasensor for neurotoxin anatoxin-a. Biosens. Bioelectron. 2015, 68, 295–302. [Google Scholar] [CrossRef]
  46. Roy-Lachapelle, A.; Solliec, M.; Sinotte, M.; Deblois, C.; Sauvé, S. High resolution/accurate mass (HRMS) detection of anatoxin-a in lake water using LDTD-APCI coupled to a Q-Exactive mass spectrometer. Talanta 2015, 132, 836–844. [Google Scholar] [CrossRef] [PubMed]
  47. Beach, D.G.; Hollingdale, C.; Quilliam, M.A. Isotope-labelling derivatization: A broadly applicable approach to quantitation of algal toxins by isotope dilution LC-MS/MS. Anal. Methods 2016, 8, 2872–2879. [Google Scholar] [CrossRef]
  48. Le, T.; Esteve-Turrillas, F.A.; Armenta, S.; de la Guardia, M.; Quiñones-Reyes, G.; Abad-Fuentes, A.; Abad-Somovilla, A. Dispersive magnetic immunoaffinity extraction. Anatoxin-a determination. J. Chromatogr. A 2017, 1529, 57–62. [Google Scholar] [CrossRef] [PubMed]
  49. Lin, Z.; Gu, J.; Li, C.; Lee, R.; Xie, L.; Chen, S.; Cao, P.; Jiang, S.; Yuan, Y.; Hong, X.; et al. A nanoparticle-decorated biomolecule-responsive polymer enables robust signaling cascade for biosensing. Adv. Mater. 2017, 29, 1702090. [Google Scholar] [CrossRef] [PubMed]
  50. Beach, D.G.; Rafuse, C.; Melanson, J.E.; McCarron, P. Rapid quantitative screening of cyanobacteria for production of anatoxins using direct analysis in real time high-resolution mass spectrometry. Rapid Commun. Mass Spectrom. 2021, 35, e8940. [Google Scholar] [CrossRef] [PubMed]
  51. Nguyen, D.; Jang, C. A simple and ultrasensitive colorimetric biosensor for anatoxin-a based on aptamer and gold nanoparticles. Micromachines 2021, 12, 1526. [Google Scholar] [CrossRef] [PubMed]
  52. Xia, M.; Zhou, F.; Feng, X.; Sun, J.; Wang, L.; Li, N.; Wang, X.; Wang, G. A DNAzyme-Based Dual-Stimuli responsive electrochemiluminescence resonance energy transfer platform for ultrasensitive anatoxin-a detection. Anal. Chem. 2021, 93, 11284–11290. [Google Scholar] [CrossRef] [PubMed]
  53. Beach, D.G.; Bruce, M.; Lawrence, J.; McCarron, P. Rapid quantification of anatoxins in benthic cyanobacterial mats using direct analysis in real-time-high resolution tandem mass spectrometry. Environ. Sci. Technol. 2022, 56, 13837–13844. [Google Scholar] [CrossRef]
  54. Cevallos-Cedeño, R.E.; Quiñones-Reyes, G.; Agulló, C.; Abad-Somovilla, A.; Abad-Fuentes, A.; Mercader, J.V. Rapid immunochemical methods for anatoxin-a monitoring in environmental water samples. Anal. Chem. 2022, 94, 10857–10864. [Google Scholar] [CrossRef] [PubMed]
  55. Beach, D.; Zamlynny, L.; MacArthur, M.; Miles, C. Liquid chromatography-high-resolution tandem mass spectrometry of anatoxins, including new conjugates and reduction products. Anal. Bioanal. Chem. 2023, 415, 5281–5296. [Google Scholar] [CrossRef]
  56. Dahlmann, J.; Budakowski, W.R.; Luckas, B. Liquid chromatography–electrospray ionisation-mass spectrometry based method for the simultaneous determination of algal and cyanobacterial toxins in phytoplankton from marine waters and lakes followed by tentative structural elucidation of microcystins. J. Chromatogr. A 2003, 994, 45–57. [Google Scholar] [CrossRef] [PubMed]
  57. Dell’Aversano, C.; Eaglesham, G.K.; Quilliam, M.A. Analysis of cyanobacterial toxins by hydrophilic interaction liquid chromatography-mass spectrometry. J. Chromatogr. A 2004, 1028, 155–164. [Google Scholar] [CrossRef] [PubMed]
  58. Maizels, M.; Budde, W.L. A LC/MS method for the determination of cyanobacteria toxins in water. Anal. Chem. 2004, 76, 1342–1351. [Google Scholar] [CrossRef] [PubMed]
  59. Vasas, G.; Gáspar, A.; Páger, C.; Surányi, G.; Mathé, C.; Hamvas, M.M.; Borbely, G. Analysis of cyanobacterial toxins (anatoxin-a, cylindrospermopsin, microcystin-LR) by capillary electrophoresis. Electrophoresis 2004, 25, 108–115. [Google Scholar] [CrossRef] [PubMed]
  60. Hiller, S.; Krock, B.; Cembella, A.; Luckas, B. Rapid detection of cyanobacterial toxins in precursor ion mode by liquid chromatography tandem mass spectrometry. J. Mass Spectrom. 2007, 42, 1238–1250. [Google Scholar] [CrossRef] [PubMed]
  61. Oehrle, S.A.; Southwell, B.; Westrick, J. Detection of various freshwater cyanobacterial toxins using ultra-performance liquid chromatography tandem mass spectrometry. Toxicon 2010, 55, 965–972. [Google Scholar] [CrossRef] [PubMed]
  62. Yen, H.; Lin, T.; Liao, P. Simultaneous detection of nine cyanotoxins in drinking water using dual solid-phase extraction and liquid chromatography–mass spectrometry. Toxicon 2011, 58, 209–218. [Google Scholar] [CrossRef]
  63. Al-Sammak, M.A.; Hoagland, K.D.; Snow, D.D.; Cassada, D. Methods for simultaneous detection of the cyanotoxins BMAA, DABA, and anatoxin-a in environmental samples. Toxicon 2013, 76, 316–325. [Google Scholar] [CrossRef]
  64. Rodríguez, I.; Alfonso, C.; Alfonso, A.; Otero, P.; Meyer, T.; Breitenback, U.; Botana, L.M. Toxin profile in samples collected in fresh and brackish water in Germany. Toxicon 2014, 91, 35–44. [Google Scholar] [CrossRef] [PubMed]
  65. Fayad, P.B.; Roy-Lachapelle, A.; Duy, S.V.; Prévost, M.; Sauvé, S. On-line solid-phase extraction coupled to liquid chromatography tandem mass spectrometry for the analysis of cyanotoxins in algal blooms. Toxicon 2015, 108, 167–175. [Google Scholar] [CrossRef] [PubMed]
  66. Greer, B.; McNamee, S.E.; Boots, B.; Cimarelli, L.; Guillebault, D.; Helmi, K.; Marcheggiani, S.; Panaiotov, S.; Breitenbach, U.; Akçaalan, R.; et al. A validated UPLC–MS/MS method for the surveillance of ten aquatic biotoxins in European brackish and freshwater systems. Harmful Algae 2016, 55, 31–40. [Google Scholar] [CrossRef] [PubMed]
  67. Pekar, H.; Westerberg, E.; Bruno, O.; Lääne, A.; Persson, K.M.; Sundström, L.F.; Thim, A. Fast, rugged and sensitive ultra-high pressure liquid chromatography tandem mass spectrometry method for analysis of cyanotoxins in raw water and drinking water—First findings of anatoxins, cylindrospermopsins and microcystin variants in Swedish source waters and infiltration ponds. J. Chromatogr. A 2016, 1429, 265–276. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, C.C.; Petty, E.E.; Smith, C.M. Rapid and Efficient Analysis of Microcystins, Nodularin, Cylindrospermopsin, and Anatoxin-a in Drinking Water by LC Tandem MS. J. AOAC Int. 2016, 99, 1565–1571. [Google Scholar] [CrossRef] [PubMed]
  69. Ortiz, X.; Korenkova, E.; Jobst, K.; MacPherson, K.A.; Reiner, E.J. A high throughput targeted and non-targeted method for the analysis of microcystins and anatoxin-A using on-line solid phase extraction coupled to liquid chromatography–quadrupole time-of-flight high resolution mass spectrometry. Anal. Bioanal. Chem. 2017, 409, 4959–4969. [Google Scholar] [CrossRef]
  70. Roy-Lachapelle, A.; Solliec, M.; Bouchard, M.F.; Sauvé, S. Detection of cyanotoxins in algae dietary supplements. Toxins 2017, 9, 76. [Google Scholar] [CrossRef] [PubMed]
  71. Zervou, S.; Christoporidis, C.; Kaloudis, T.; Triantis, T.M.; Hiskia, A. New SPE-LC-MS/MS method for simultaneous determination of multi-class cyanobacterial and algal toxins. J. Hazard. Mater. 2017, 323, 56–66. [Google Scholar] [CrossRef] [PubMed]
  72. Haddad, S.P.; Bobbitt, J.M.; Taylor, R.B.; Lovin, L.M.; Conkle, J.L.; Chambliss, C.K.; Brooks, B.W. Determination of microcystins, nodularin, anatoxin-a, cylindrospermopsin, and saxitoxin in water and fish tissue using isotope dilution liquid chromatography tandem mass spectrometry. J. Chromatogr. A 2019, 1599, 66–74. [Google Scholar] [CrossRef]
  73. Li, Z.; Zhang, S.; Yu, T.; Dai, Z.; Wei, Q. Aptamer-based fluorescent sensor array for multiplexed detection of cyanotoxins on a smartphone. Anal. Chem. 2019, 91, 10448–10457. [Google Scholar] [CrossRef]
  74. Roy-Lachapelle, A.; Duy, S.V.; Munoz, G.; Dihn, Q.T.; Bahl, E.; Simon, D.F.; Sauvé, S. Analysis of multiclass cyanotoxins (microcystins, anabaenopeptins, cylindrospermopsin and anatoxins) in lake waters using on-line SPE liquid chromatography high-resolution Orbitrap mass spectrometry. Anal. Methods 2019, 11, 5289. [Google Scholar] [CrossRef]
  75. Filatova, D.; Núñez, O.; Farré, M. Ultra-Trace analysis of cyanotoxins by liquid chromatography coupled to High-Resolution Mass Spectrometry. Toxins 2020, 12, 247. [Google Scholar] [CrossRef] [PubMed]
  76. Tran, N.H.; Li, Y.; Reinhard, M.; Goh, K.C.; Sukarji, N.H.B.; You, L.; He, Y.; Gin, K.Y. Quantification of cylindrospermopsin, anatoxin-a and homoanatoxin-a in cyanobacterial bloom freshwater using direct injection/SPE coupled with UPLC-MS/MS. Sci. Total Environ. 2020, 731, 139014. [Google Scholar] [CrossRef] [PubMed]
  77. Romera-García, E.; Helmus, R.; Ballesteros-Gómez, A.; Visser, P.M. Multi-class determination of intracellular and extracellular cyanotoxins in freshwater samples by ultra-high performance liquid chromatography coupled to high resolution mass spectrometry. Chemosphere 2021, 274, 129770. [Google Scholar] [CrossRef] [PubMed]
  78. Skafi, M.; Duy, S.V.; Munoz, G.; Dinh, Q.T.; Simon, D.F.; Juneau, P.; Sauvé, S. Ocurrence of microcystins, anabaenopeptins and other cyanotoxins in fish from a freshwater wildlife reserve impacted by harmful cyanobacterial blooms. Toxicon 2021, 194, 44–52. [Google Scholar] [CrossRef] [PubMed]
  79. Aparicio-Muriana, M.M.; Carmona-Molero, R.; Lara, F.J.; García-Campaña, A.M.; Olmo-Iruela, M. Multiclass cyanotoxin analysis in reservoir waters: Tandem solid-phase extraction followed by zwitterionic hydrophilic interaction liquid chromatography-mass spectrometry. Talanta 2022, 237, 122929. [Google Scholar] [CrossRef]
  80. Aparicio-Muriana, M.M.; Lara, F.J.; Olmo-Iruela, M.; García-Campaña, A.M. Determination of multiclass cyanotoxins in Blue-Green Algae (BGA) dietary supplements using Hydrophilic Interaction Liquid Chromatography-Tandem Mass Spectrometry. Toxins 2023, 15, 127. [Google Scholar] [CrossRef] [PubMed]
  81. España Amórtegui, J.C.; Pekar, H.; Chico Retrato, M.D.; Persson, M.; Karlson, B.; Bergquist, J.; Zuberovic-Muratovic, A. LC-MS/MS analysis of cyanotoxins in bivalve mullusks-method development, validation and first evidence of occurrence of nodularin in mussels (Mytilus edulis) and Oysters (Magallana gigas) from the West Coast of Sweden. Toxins 2023, 15, 329. [Google Scholar] [CrossRef] [PubMed]
  82. Rios Jacinavicius, F.; Valverde Campos, T.G.; Souza Passos, L.; Pinto, E.; Geraldes, V. A rapid LC-MS/MS method for multi-class identification and quantification of cyanotoxins. Toxicon 2023, 234, 107282. [Google Scholar] [CrossRef]
  83. Carmona-Molero, R.; Aparicio-Muriana, M.M.; Lara, F.J.; García-Campaña, A.M.; del Olmo-Iruela, M. Capillary electrophoresis tandem mass spectrometry to determine multiclass cyanotoxins in reservoir water and spinach samples. J. Chromatogr. A 2024, 1717, 464666. [Google Scholar] [CrossRef]
  84. U.S. EPA. Method 546: Determination of Total Microcystins and Nodularins in Drinking Water and Ambient Water by Adda Enzyme-Linked Immunosorbent Assay. Available online: https://www.epa.gov/esam/method-546-determination-total-microcystins-and-nodularins-drinking-water-and-ambient-water (accessed on 15 October 2023).
  85. Quiñones-Reyes, G.; Agulló, C.; Mercader, J.V.; Abad-Somovilla, A.; Abad-Fuentes, A. Synthetic haptens and monoclonal antibodies to the cyanotoxin anatoxin-a. Angew. Chem. Int. Ed. 2019, 58, 9134–9139. [Google Scholar] [CrossRef] [PubMed]
  86. Song, S.; Wang, L.; Li, J.; Zhao, J.; Fan, C. Aptamer-based biosensors. Trends Anal. Chem. 2008, 27, 108–117. [Google Scholar] [CrossRef]
  87. Wong, S.H.; Hindin, E. Detecting an algal toxin by high-pressure liquid chromatography. J. Am. Water Works Assoc. 1982, 74, 528–529. [Google Scholar] [CrossRef]
  88. Zotou, A.; Jefferies, T.M.; Brough, P.A.; Gallagher, T. Determination of anatoxin-a and homoanatoxin in blue-green algal extracts by high-performance liquid chromatography and gas chromatography-mass spectrometry. Analyst 1993, 118, 753–758. [Google Scholar] [CrossRef]
  89. Jefferies, T.M.; Brammer, G.; Zotou, A.; Brough, P.A.; Gallagher, T. Determination of Anatoxin-a, Homoanatoxin and Propylanatoxin in Cyanobacterial Extracts by HPLC, GC-Mass Spectrometry and Capillary Electrophoresis. In Detection Methods for Cyanobacterial Toxins, 1st ed.; Codd, G.A., Jefferies, T.M., Keevil, C.W., Potter, E., Eds.; Royal Society of Chemistry: Cambridge, UK, 1994; pp. 34–39. [Google Scholar]
  90. James, K.; Sherlock, I.R. Determination of the cyanobacterial neurotoxin, anatoxin-a, by derivatisation using 7-fluoro-4-nitro-2,1,3-benzoxadiazole (NBD-F) and HPLC analysis with fluorometric detection. Biomed. Chromatogr. 1996, 10, 46–47. [Google Scholar] [CrossRef]
  91. Powell, M.W. Analysis of anatoxin-a in aqueous samples. Chromatographia 1997, 45, 25–28. [Google Scholar] [CrossRef]
  92. Gugger, M.; Lenoir, S.; Berger, C.; Ledreux, A.; Druart, J.; Humbert, J.; Guette, C.; Bernard, C. First report in a river in France of the benthic cyanobacterium Phormidium favosum producing anatoxin-a associated with dog neurotoxicosis. Toxicon 2005, 45, 919–928. [Google Scholar] [CrossRef] [PubMed]
  93. Osswald, J.; Rellán, S.; Gago, A.; Vasconcelos, V. Toxicology and detection methods of the alkaloid neurotoxin produced by cyanobacteria, anatoxin-a. Environ. Int. 2007, 33, 1070–1089. [Google Scholar] [CrossRef] [PubMed]
  94. James, K.; Furey, A.; Sherlock, I.R.; Stack, M.A.; Twohig, M.; Caudwell, F.B.; Skulberg, O.M. Sensitive determination of anatoxin-a, homoanatoxin-a and their degradation products by liquid chromatography with fluorometric detection. J. Chromatogr. A 1998, 798, 147–157. [Google Scholar] [CrossRef]
  95. Namera, A.; So, A.; Pawliszyn, J. Analysis of anatoxin-a in aqueous samples by solid-phase microextraction coupled to high-performance liquid chromatography with fluorescence detection and on-fiber derivatization. J. Chromatogr. A 2002, 963, 295–302. [Google Scholar] [CrossRef]
  96. James, K.; Sherlock, I.R.; Stack, M.A. Anatoxin-a in Irish freshwater and cyanobacteria, determined using a new fluorimetric liquid chromatographic method. Toxicon 1997, 35, 963–971. [Google Scholar] [CrossRef] [PubMed]
  97. Furey, A.; Crowley, J.; Hamilton, B.; Lehane, M.; James, K.J. Strategies to avoid the mis-identification of anatoxin-a using mass spectrometry in the forensic investigation of acute neurotoxic poisoning. J. Chromatogr. A 2005, 1082, 91–97. [Google Scholar] [CrossRef] [PubMed]
  98. Ross, M.M.; Kidwell, D.A.; Callahan, J.H. Mass spectrometric analysis of anatoxin-a. J. Anal. Toxicol. 1989, 13, 317–321. [Google Scholar] [CrossRef] [PubMed]
  99. Himberg, K. Determination of anatoxin-a, the neurotoxin of Anabaena flos-aquae cyanobacterium, in algae and water by gas chromatography-mass spectrometry. J. Chromatogr. A 1989, 481, 358–362. [Google Scholar] [CrossRef] [PubMed]
  100. Hormazábal, V.; Østensvik, Ø.; Underval, B.; Skulberg, O.M. Simultaneous determination of the cyanotoxins anatoxin-a, microcystin desmethyl-3, LR, RR, and YR in fish muscle using liquid chromatography-mass spectrometry. J. Liq. Chrom. Rel. Technol. 2000, 23, 185–196. [Google Scholar] [CrossRef]
  101. Hormazábal, V.; Østensvik, Ø.; Underval, B.; Skulberg, O.M. Simultaneous determination of the cyanotoxins anatoxin-a, microcystin desmethyl-3, LR, RR, and YR in water using liquid chromatography-mass spectrometry. J. Liq. Chrom. Rel. Technol. 2000, 23, 3155–3164. [Google Scholar] [CrossRef]
  102. Pietsch, J.; Fichtner, S.; Imhof, L.; Schmidt, W.; Brauch, H.J. Simultaneous determination of cyanobacterial hepato- and neurotoxins in water samples by ion-pair supported enrichment and HPLC-ESI-MS-MS. Chromatographia 2001, 54, 339–344. [Google Scholar] [CrossRef]
  103. U.S. EPA. Method 545: Determination of Cylindrospermopsin and Anatoxin-a in Ambient Freshwaters by Liquid Chromatography/Tandem Mass Spectrometry (LC/MS/MS), EPA 815-R-15-009. Available online: https://www.epa.gov/esam/method-545-determination-cylindrospermopsin-and-anatoxin-drinking-water-liquid-chromatography (accessed on 15 October 2023).
  104. Stevens, D.K.; Krieger, R.I. Analysis of anatoxin-a by GC/ECD. J. Anal. Toxicol. 1988, 12, 126–131. [Google Scholar] [CrossRef] [PubMed]
  105. Bumke-Vogt, C.; Mailhan, W.; Rotard, W.; Chorus, I. A highly sensitive analytical method for the neurotoxin anatoxin-a, using GC-ECD, and first application to laboratory cultures. Phycologia 1996, 35, 51–61. [Google Scholar] [CrossRef]
  106. Guzmán-Guillén, R.; Prieto Ortega, A.I.; Moreno, I.; González, G.; Soria-Díaz, M.E.; Vasconcelos, V.; Cameán, A.M. Development and optimization of a method for the determination of Cylindrospermopsin from strains of Aphanizomenon cultures: Intra-laboratory assessment of its accuracy by using validation standards. Talanta 2012, 100, 356–363. [Google Scholar] [CrossRef]
  107. Wood, S.; Rasmussen, J.P.; Holland, P.; Campbell, R.; Crowe, A.L. First report of the cyanotoxin anatoxin-a from Aphanizomenon issatschenkoi (cyanobacteria). J. Phycol. 2007, 43, 356–365. [Google Scholar] [CrossRef]
  108. Draisci, R.; Ferretti, E.; Palleschi, L.; Marchiafava, C. Identification of anatoxins in blue-green algae food supplements using liquid chromatography-tandem mass spectrometry. Food Addit. Contam. 2001, 18, 525–531. [Google Scholar] [CrossRef] [PubMed]
  109. Sánchez-Parra, E.; Boutarfa, S.; Aboal, M. Are cyanotoxins the only toxic compound potentially present in microalgae supplements? Results from a study of ecological and non-ecological products. Toxins 2020, 12, 552. [Google Scholar] [CrossRef] [PubMed]
  110. Buratti, F.M.; Manganelli, M.; Vichi, S.; Stefanelli, M.; Scardala, S.; Testai, E.; Funari, E. Cyanotoxins: Producing organisms, occurrence, toxicity, mechanism of action and human health toxicological risk evaluation. Arch. Toxicol. 2017, 91, 1049–1130. [Google Scholar] [CrossRef]
  111. Takino, M.; Daishima, S.; Yamaguchi, K. Analysis of anatoxin-a in freshwater by automated on-line derivatization-liquid chromatography-electrospray mass spectrometry. J. Chromatogr. A 1999, 862, 191–197. [Google Scholar] [CrossRef]
Figure 1. Structures of (A) ATX-a and (B) HATX-a and their respective derivatives.
Figure 1. Structures of (A) ATX-a and (B) HATX-a and their respective derivatives.
Toxins 16 00198 g001
Figure 2. % of publications focused on determination of ATX-a alone or in combination with other cyanotoxins in relation to years of publication.
Figure 2. % of publications focused on determination of ATX-a alone or in combination with other cyanotoxins in relation to years of publication.
Toxins 16 00198 g002
Figure 3. Techniques employed for determination of ATX-a alone or in combination with other cyanotoxins. ELISA: enzyme-linked immunosorbent assay; TLC: thin-layer chromatography; UV: ultraviolet; FLD: fluorescence detection; MS: mass spectrometry; ECD: electron capture detector; MS/MS: tandem mass spectrometry; HRMS: high-resolution mass spectrometry: HRMS/MS: high-resolution mass spectrometry tandem mass spectrometry.
Figure 3. Techniques employed for determination of ATX-a alone or in combination with other cyanotoxins. ELISA: enzyme-linked immunosorbent assay; TLC: thin-layer chromatography; UV: ultraviolet; FLD: fluorescence detection; MS: mass spectrometry; ECD: electron capture detector; MS/MS: tandem mass spectrometry; HRMS: high-resolution mass spectrometry: HRMS/MS: high-resolution mass spectrometry tandem mass spectrometry.
Toxins 16 00198 g003
Figure 4. Matrices of the samples employed for the determination of ATX-a alone or in combination with other cyanotoxins.
Figure 4. Matrices of the samples employed for the determination of ATX-a alone or in combination with other cyanotoxins.
Toxins 16 00198 g004
Figure 5. Advantages (green) and disadvantages (red) of the methods developed for ATX-a analysis. ELISA: enzyme-linked immunosorbent assay; HPLC-FLD: high-performance liquid chromatography coupled to fluorescence detection; LC-MS/MS: liquid chromatography–tandem mass spectrometry.
Figure 5. Advantages (green) and disadvantages (red) of the methods developed for ATX-a analysis. ELISA: enzyme-linked immunosorbent assay; HPLC-FLD: high-performance liquid chromatography coupled to fluorescence detection; LC-MS/MS: liquid chromatography–tandem mass spectrometry.
Toxins 16 00198 g005
Table 1. Analytical methods focused on the determination of ATX-a published from 2003 to date.
Table 1. Analytical methods focused on the determination of ATX-a published from 2003 to date.
Type of SampleAnalytical MethodLinear Concentration RangeValidation ParametersMore InformationReferences
Cyanobacteria and water samplesLC-MSn-LOD < 0.6 µg/L
Reproducibility (RSD%): ≤7% (LC-MS/MS and LC-MS3)
LC-MS4 did not produce reproductible quantitative data.[32]
Cyanobacteria culturesGC-MS
1H NMR
2–400 ngLOD: 0.5 ngDerivatization is not necessary.
To improve the detection limits, SIM is necessary.
[33]
Water samples and cyanobacterial bloomGC-MS50–10,000 ng/LLOD: 11.2 ng/mL
LOQ: 200.1 ng/mL
Intra-day (RSD%): 5.15%
Inter-day (RSD%): 2.7%
Three forms of PANI films and a single form of PPY film were used, showing that the leucoemeraldine form of PANI displayed a better selectivity to ATX-a.[34]
Cyanobacterial samplesQqTOF-MS
QIT-MS
--The investigated compounds were ATX-a, HATX-a, and their degradation products.[35]
Food supplements (BGA and spirulina tablets, powders, and capsules)LC-FLD0.1–2.0 µg/gLOD: 50 µg/kg (BGA) and 10 µg/kg (spirulina)
Recovery: 74–108%
Derivatization reagent: NBD-F.
Two transformation products of ATX-a were also analyzed: epoxyanatoxin-a (LOD: 10–55 µg/kg) and dihydroanatoxin-a (LOD: 10–65 µg/kg).
[36]
Water and fish muscle tissueLC-MS/MS-LOD: 8 ng/L (water) and 0.2 ng/g (fish)
LOQ: 13 ng/L (water) and 0.5 ng/g (fish)
Recovery: 71–79%
Water samples were directly injected after filtration, whereas fish tissue required the matrix SPD technique. Provides sensitivity for analyzing ATX-a in fish at levels < 1 ng/g.[37]
Water samplesGC-MS2.5–200 ng/mLLOD: 2.0 ng/mL
LOQ: 2.5 ng/mL
Repeatability (%RSD): 6.8–10.9%
Direct derivatization of the ATX-a by adding hexylchloroformate in a sample with pH = 9.[38]
Water samples (deionized water and river water) and cyanobacterial samplesLC-FLD6.25–1250 ng/mLLOD: 0.18 ng/mL (deionized water) and 0.29 ng/mL (river water)
Repeatability (RSD%): <15%
Reproducibility (RSD%): <15%
Derivatization reagent: NBD-F
SPME coupled to HPLC was used.
[39]
Cyanobacterial culturesMALDI-TOF-MS--Derivatization reagent: N-methyl-N(tert-butyldimethylsilyl) trifluoroacetamide.
Useful method for small sample volumes (1 µL) and has an analysis time of 1 min/sample.
[40]
Dietary food supplements containing cyanobacteriaLC-FLD0.03–0.59 µg/mLRecovery: 84%
LOD: 3 ng/g
LOQ: 10 ng/g
Repeatability (%RSD): 6.3%
No significant effect on response of ATX-a in the matrix sample.[22]
GC-MS5.87–23.50 µg/mLLOD: 24 ng/g
LOQ: 70 ng/g
Repeatability (%RSD): 10.9%
13% of suppression of ATX-a response in sample matrix extract compared with solvent.
Water samplesLC-MS/MS0.5–2000 µg/LRecovery: 73–97%
LOD: 0.65 ng/L
LOQ: 1.96 ng/L
Intra-day (RSD%): 4.2–5.9%
Inter-day (RSD%): 4.2–9.1%
Three different SPE cartridges were assessed—hypersep PGC SPE cartridges gave better recoveries.
PHE-d5 was used as internal standard.
[41]
Water and lyophilized troutLC-FLD0.1–23.0 µg/mLLOD: 0.17 µg/mL (water) and 80 ng/g (d.w. trout)
LOQ: 0.58 µg/mL (water) and 170 ng/g (d.w. trout)
Repeatability (RSD%): 0.4–0.9%
Recovery: 84–94%
Intermediate precision <2%
Accuracy (RSD%): 6% (water) and <0.5% (trout)
Methanol is used as the mobile phase.[42]
Cyanobacterial bloomLDTD-APCI-MS/MS3–250 µg/LLOD: 1 µg/L
LOQ: 3 µg/L
Accuracy: 108%
Inter-day (RSD%): 8%
Remove interference from PHE.[43]
Cyanobacterial culturesFP0.1–200 µmLOD: 33.3 nM
LOQ: 100.0 nM
No effects of the solvent (methanol) in fluorescence intensity.[44]
LC-MS/MS-LOD: 1.5 ng/mL (5.33 nM)
LOQ: 5.0 ng/mL (17.77 nM)
The sample was analyzed in positive MRM mode, searching for the transitions of the ATX-a and the most common ATX-a analogues: HATX-a, dihydroanatoxin-a, dihydrohomoanatoxin-a, epoxyanatoxin-a, and epoxyhomoanatoxin-a.
Water samplesAptasensor (DNA aptamers)1–100 nMLOD: 0.5 nM
Recovery: 94.8–108.6%
The aptasensor exhibited high stability and selectivity of ATX-a against CYN and MC-LR.[45]
Water samplesLDTD-APCI-HRMS0.5–1000 µg/LRecovery: 96–108%
LOD: 0.2 µg/L
LOQ: 0.6 µg/L
Accuracy and precision values (RSD%) < 15%
Resolve the misidentification of ATX-a and PHE. Two scan modes were assessed, showing that targeted-MS/MS increased the selectivity of compound detection.[46]
Cyanobacteria samplesLC-MS0.005–1.25 µMLOD: 0.02 mg/kg
LOQ: 0.06 mg/kg
Precision (RSD%): 1–12%
A novel mixed reverse-phase/weak anion exchange solid phase clean-up was employed.
Also determines HATX-a (LOD: 0.04 mg/kg and LOQ: 0.12 mg/kg).
[47]
Water samplesIMS20–150 µg/LRecovery: 91–115%
LOD: 0.02 µg/L
LOQ: 0.08 µg/L
Repeatability (RSD%): 3–9%
Interferences with PHE avoided using the d-MagIA extraction methodology. [48]
Water samplesUPSS sensor10−15–10−10 MLOD: 10−14 MSelectivity of ATX-a against CYN and brevetoxin-2.[49]
Cyanobacterial samplesDART-HRMS0.283–206 ng/mLLOD: 1 ng/mL
Intra-day (RSD%): 10–40%
Inter-day (RSD%): 33%
<2 min of analysis per sample for triplicate analysis. Also determines HATX-a and dihydroanatoxin-a.[50]
Water samplesColorimetric biosensor (DNA aptamer)10 pM–200 nMLOD: 4.45 pM
Repeatability (RSD%): 3.6%
Recoveries: 89.72–112.43% (RSD: 4.12–10.91%)
Gold nanoparticles are used as probes. Excellent specificity toward ATX-a.[51]
Water samplesECL-RET aptasensor0.001–1 mg/mLLOD: 0.00034 mg/mLThis sensor showed high assay performance for ATX-a determination. [52]
Cyanobacterial samplesDART-HRMS/MS0.14–86 ng/mLLOD: 4.8 ng/g cyanobacteria
Recovery: 82–84%
Precision (RSD%): 12–34%
Also determines HATX-a and dihydroanatoxin-a.[53]
Water samplesELISA (direct and indirect) 0.5–500 ng/mLDirect and indirect ELISA:
LOD: 0.1 ng/mL
Recovery: 82–117.4%
CV < 20%
Intra-day (RSD%) < 10%
Inter-day (RSD%) < 20%
Detected (+)-ATX-a.
Robust to pH variations.
It is advisable to minimize the concentration of organic solvents, and PBS proved to be the most suitable buffer for the ELISA.
[54]
LFICA-LOD: 2 ng/mLNo influence of pH.
Cyanobacterial samplesLC-HRMS/MS--Allows for the detection of new ATX-a derivatives.[55]
APCI: atmospheric pressure chemical ionization; ATX-a: anatoxin-a; BGA: blue–green algae; CV: coefficients of variation; DART: direct analysis in real time; CYN: cylindrospermopsin; DNA: deoxyribonucleic acid; d-MagIA: dispersive magnetic immunoaffinity; d.w.: dry weight; ECL-RET: electrochemiluminescence resonance energy transfer; ELISA: enzyme-linked immunosorbent assay; FLD: fluorescence detection; FP: fluorescent polarization; GC: gas chromatography; HATX-a: homoanatoxin-a; 1H NMR: proton nuclear magnetic resonance; HPLC: high-performance liquid chromatography; HRMS: high-resolution mass spectrometry; IMS: ion mobility spectrometry; LC: liquid chromatography; LDTD: laser diode thermal desorption; LFICA: lateral flow immunochromatography assay; LOD: limit of detection; LOQ: limit of quantification; MALDI-TOF-MS: matrix-assisted laser desorption ionization time of flight mass spectrometry; MC-LR: microcystin-LR; MRM: Multiple Reaction Monitoring; MS/MS: tandem mass spectrometry; MSn: multiple tandem mass spectrometry; NBD-F: 7-Fluoro-4-nitro-2,1,3-benzoxadiazole; PANI: polyaniline; PBS: phosphate-buffered saline; PGC: porous graphitic carbon; PHE: phenylalanine; PPY: polypyrrole; QIT: quadrupole ion-trap; QqTOF: hybrid quadrupole time-of-flight; RSD: relative standard deviation; SIM: selected ion monitoring; SPD: solid-phase dispersion; SPE: solid-phase extraction; SPME: solid-phase microextraction; UPSS: ultrasensitive polymeric sensing system.
Table 2. Analytical methods for the determination of cyanotoxin mixtures containing ATX-a published from 2003 to date.
Table 2. Analytical methods for the determination of cyanotoxin mixtures containing ATX-a published from 2003 to date.
Type of SampleCyanotoxinsAnalytical MethodLinear Concentration RangeGlobal Validation Parameters of Multitoxin MethodsMore Information and Specific ATX-a DataReferences
Phytoplankton and aqueous phytoplanktonSTX, ATX-a, DA, NOD, MCs (-RR, -YR, -LR, LA, -LW, and -LF), OA, and DTX-1 LC-MS1 to 50 ng on columnLOD: 0.5 to 1.0 ng
RSD%: 0.6–7.1%
Recoveries: 96–113%
Direct analysis after the simple preparation of the sample.
The applied chromatographic conditions allow for the isolation and identification of substances suspected to be “new” microcystins.
For ATX-a: LOD: 0.5 ng, RSD%: 3.2%, and recovery: 103%.
[56]
Cyanobacterial culturesSTX and its various analogues, ATX-a, CYN, doCYN, MCs (-LR and -RR)LC-MS-LOD: 1.4–3.2 pmol injected on column in SIM modeMS detection was carried out in the SIM and SRM modes for the positive ions, with SRM being the best performing.
In samples, a simple extraction method was used, and no clean-up was performed on the crude extracts in order to demonstrate rapid
analysis.
For ATX-a: LOD: 1.4 pmol injected on column in SIM mode.
[57]
Water samplesATX-a, MCs (-LR, -RR, and -YR), and NODLC-MS5–50 µg/mLLOD: less than 1 µg/L
RSD%: 5–19%
Recoveries: 68–98%
Toxins are partitioned from water samples with extraction disks.
It allows for sample analysis, including processing in 1 h.
Does not provide multipoint linearity data due to the lack of sufficient quantities of analyte.
For ATX-a: LOD: less than 1 µg/L, and recovery: 68%.
[58]
Water bloom samples and crude cyanobacterial extractsATX-a, MC-LR, and CYN CZE-UV -LOD: 0.73–3.77 µg/mL
RSD% peak area: 1.48–2.93%
RSD% migration time: 0.27–0.71%
For ATX-a: LOD: 1.12 µg/mL; RSD% peak area: 2.93%; and RSD% migration time: 0.27%.[59]
MEKC-UV-LOD: 0.89–3.65 µg/mL
RSD% peak area: 1.87–2.98%
RSD% migration time: 0.58–0.74%
It is recommended to use both CZE and MEKC for the analysis of the same sample in order to confirm the results.
For ATX-a: LOD: 2.63 µg/mL; RSD% peak area: 2.98%; and RSD% migration time: 0.66%.
Cyanobacteria-containing samplesPSP toxins, ATXs, CYNs, MCs (-RR, -LR, -YR, -LA, -LW, and -LF), and NODs LC-MS/MS-LOD: 6–700 pg
LOQ: 10–1150 pg
It is necessary to clean the sample to avoid false positives.
This method allows one to detect potential unknown variants of cyanobacterial toxins.
For ATX-a: LOD: 700 pg and LOQ: 1150 pg
[60]
Water samplesATX-a, MCs (-RR, -LR, and -LF), and CYNLC-MS/MS0.5–100 ppb (µg/L)LOD: 0.10–0.21 ppb (µg/L)
Recoveries: 88–110%
This method can be used to either screen or quantify the main cyanotoxins in a few minutes.
For ATX-a: LOD: 0.13 µg/L and recoveries: 88–102%.
[61]
Water samplesMCs (-LR, -RR, -YR, -LW, -LF, and -LA), NOD, ATX-a, and CYNLC-MS8–1000 ng/L (MCs and NOD)
40–2000 ng/L (ATX-a)
40–2000 ng/L (CYN)
LOD: 2–100 ng/L (pure water)
Recoveries: 83–104%
Dual-cartridge SPE extraction is necessary.
Toxin recoveries were lower in reservoir water than in pure water (83–90% vs. 94–104%).
Differences of less than 10% were obtained in the values of MC-LR and CYN concentrations measured by this method (SPE-LC-MS) compared to the ELISA test.
For ATX-a (pure water): LOD: 46 ng/L and recovery: 96%.
[62]
Water, fish, and plant samplesBMAA, DAB, and ATX-aLC-FLD 0.01–0.70 mg/L (BMAA and
ATX-a)
0.01–1 mg/L (DAB)
LOD: 5–7 µg/LDerivatization reagent: 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate.
An extraction step with SPE cartridges is required. The sensitivity provided by LC-MS/MS was improved over HPLC-FLD.
For ATX-a: LOD: 6 µg/L in HPLC-FLD and 3.2 µg/L in LC-MS/MS.
[63]
LC-MS/MS0.01–0.70 mg/L (BMAA and
ATX-a)
0.01–1 mg/L (DAB)
LOD: 0.8–3.2 µg/L
Water samplesCYN, MCs (-LR, -RR, and -YR), and ATX-aLC-MS/MS-LOD: 1.9–3.9 ng/mL
LOQ: 5–10 ng/mL
The method separates the amino acid PHE, so there is no interference with ATX-a.
The most common cyanotoxins can be simultaneously identified within 12.5 min, including several ATX-a analogues.
For ATX-a: LOD: 1.9 ng/mL and LOQ: 5 ng/mL.
[64]
Algal bloom water samplesMCs (-RR, -YR, -LR, -LY, -LW, and -LF), ATX-a, and CYNLC-MS/MS0.1–10 µg/LLOD: 0.01–0.02 µg/L
LOQ: 0.03–0.06 µg/L
Intra-day (RSD%): 3–9%
Inter-day (RSD%): 7–13%
Bias (%): 7–12%
Recoveries: 72–102%
SPE extraction is required.
The analysis time was 7 min per sample.
The method permitted the chromatographic separation of ATX-a and PHE.
For ATX-a: LOD: 0.01 µg/L, LOQ: 0.03, bias: 8–9%, intra-day (RSD%): 4–6%, inter-day (RSD%): 9–13%, and recovery: 72%.
[65]
Aquatic samplesMCs (-LR, -YR,
-RR, -LA, -LY, and -LF), NOD, CYN, ATX-a, and DA
LC-MS/MS0.01–2 ng/mLLOD: 0.3 and 5.6 ng/L
LOQ: 0.8 and 18.5 ng/L
Intra-day (RSD%): 1.2–9.6%
Inter-day (RSD%): 1.3–12.0%
Recoveries: 35.5–107.5%
SPE extraction is required.
Very low recoveries are obtained for ATX-a and DA (35.5 and 65.5%, respectively). The rest of the toxins show good recoveries (93.8–107.5%).
For ATX-a: LOD: 5.6 ng/L, LOQ: 18.5, ng/L: 1.3–12.0%, and recoveries: 35.5%.
[66]
Water samplesATX-a, HATX-a, CYN, doCYN, NOD, and MCs (-LR, -RR, -YR, HtyR, -LA, -LF, -LW, -LY, -WR, -[Dha7]-LR, -[d-Asp3]-LR, -[d-Asp3]-RR, -HilR, -(N-methyl-l)-R, -[d-Asp3,
(E)-Dhb7]-HphR, -[d-Asp3,
(E)-Dhb7]-HtyR, -[d-Asp3,
(E)-Dhb7]-RR, -[Dha7]-LR)
LC-MS/MS0.1–10 µg/LLOQ: 0.1–0.5 µg/L
RSD%: 0.9–5.4%
Recoveries: 60–111% for drinking water
Recoveries: 65–138% for raw water
The time of analysis, including the lysis of cell-bound toxins, is less than three hours.
For ATX-a: LOQ: 0.5 µg/L and recoveries: 89–138% (raw water).
[67]
Drinking water samplesMCs (-RR, -YR, -LR, -WR, -LA, -LY, -LW, and -LF), NOD, ATX-a, and CYN LC-MS0.1–10 µg/L (ATX-a, MC-RR, -WR, -LA, -LF, and NOD)
0.2–20 µg/L (MC-YR, and -LW)
0.3–30 µg/L (CYN, MC-LR, and -LY)
LOD < 100 ng/L (range of 30–90 ng/L)
LOQ: 0.1–0.3 µg/L
RSD < 20% (intra-day (RSD%): 5.1–16.6% and inter-day (RSD%): 5.8–13.7 for 1 µg/L; intra-day (RSD%): 2.5–16.0% and inter-day (RSD%) 4.6–12.3 for 200 ng/L)
Accuracy: intra-day 93–107% and inter-day: 84–99% for 1 µg/L; accuracy intra-day: 89–106% and inter-day 91–105 for 200 pg/mL
Adequate separation of toxins in 12 min.
The matrix effects between calibrators and samples (≤30%) were negligible.
For ATX-a: LOD: 30 ng/L and RSD < 20% (intra-day (RSD%): 8.6% and inter-day (RSD%) 8.1% for 1 µg/L; intra-day (RSD%): 5.1% and inter-day (RSD%) 6.3 for 200 ng/L; accuracy: intra-day 101% and inter-day: 99% for 1 µg/L; and accuracy intra-day: 106% and inter-day 105% for 200 pg/mL
[68]
Cyanobacterial blooms samplesMCs (-LR, -YR, -RR, -HtyR, -HilR, -WR, -LW, -LA, -LF, -LY, -Dha7, -LR, and -Dha7-RR) and ATX-aLC-HRMS0.05–4.80 µg/LLOD: 0.004 and 0.01 μg/L
Intra-day (RSD%): 1.71–4.68%
Inter-day (RSD%): 1.95–3.63%
Recoveries: 93.5–105.4%
Automated preconcentration of the sample (on-line SPE) was used.
This method presents quantitative results in a period less than 3 h.
Uncertainty of method: 4 and 14%.
For ATX-a: LOD: 0.0044 µg/L, intra-day (RSD%): 3.66%, inter-day (RSD%): 3.42%,
recovery: 98.9%.
[69]
Algal dietary supplements (Spirulina and Aphanizomenon flos-aquae)MCs (-RR, -YR, -LR, -LA, -LY, -LW, and -LF), ATX-a, dihydroanatoxin-a, epoxyanatoxin-a, CYN, STX, and BMAALDTD-APCI-HRMS and LC-HRMS0.03–20 µg/gLODs: 0.01–0.1 µg/gLOQs: 0.03–0.1 µg/gIntra-day (RSD%): 1–9% and 5–12% for high and low concentrations
Inter-day (RSD%): 7–13% and 7–15% for high and low concentrations
Recoveries: 79–97%
The ATX-a recoveries obtained using LDTD-APCI-HRMS analysis were better (92–95%) than applying UHPLC-HESI-HRMS analysis (89–90%). However, a better linearity range and better LOD and LOQ data were obtained with UHPLC-HESI-HRMS compared to LDTD-APCI-HRMS.
For ATX-a: LOD: 0.04 µg/g, LOQ: 0.1 µg/g, intra-day (RSD%): 5%, inter-day (RSD%): 8–9%, and recovery: 89–90%.
[70]
Water samplesCYN, ATX-a, NOD, MCs ([D-Asp3]-RR, -RR, -YR, -HtyR, [D-Asp3]-LR, -LR, -HilR, -WR, -LA, -LY, -LW, and -LF), OA, and DALC-MS/MS1–250 µg/LLODs: 1–10 ng/L
RSD%: 5.5–45.8%
Recoveries: 44–113%
In general, the mean recoveries and precision parameters are in agreement with guidelines for all common toxins.
The worst analytical validation data (recovery and precision) were found for MC-WR, MC-LF, and MC-LW. Nevertheless, the decreased efficiency of the method, observed only for these analytes, is compensated for by the advantage of achieving the simultaneous determination of numerous toxins.
For ATX-a: LODs: 1 ng/L, RSD%: 25.38%, and recovery: 62.3%.
[71]
Water samples and fish tissueMCs (-LA, -LR, -LY, -RR, and -YR), NOD, ATX-a, CYN, and STXLC-MS0.1–100 ng/mL
0.1–20 ng/mL (ATX-a)
LOD: 0.004–0.080 ng/mL
LOQ: 0.01–0.28 ng/mL
RSD% water: 0.6–7.2%
RSD% fish tissue: 1.2–8.2%
Recoveries water: 53–98%
Recoveries fish tissue: 45–103%
SPE extraction was used for water, and liquid–liquid extraction was used for fish tissue.
A zwitterionic HILIC was evaluated to separate toxins.
For ATX-a: LOD: 0.004 ng/mL, LOQ: 0.01 ng/mL, RSD% water: 1.9%, RSD% fish tissue: 3.4%, recovery water: 97%, and recovery fish tissue: 103%.
[72]
Water samplesATX-a, CYN, NOD, and MC-LRDifferential fluorescent sensor array (DNA aptamer)8–100,000 nMLOD of fluorimeter: 0.54–1.8 nM
LOD of smartphone: 1.2–2.8 nM
LOQ: 8–23 nM
Recoveries: 97–104.6%
RSD%: 0.9–3%
The smartphone-based sensor platform showed remarkable chemical specificity against potential interfering agents in water. This assay chip runs to completion after the addition of a drop of environmental water, making it suitable for field applications. Robust to various interferences (minerals, organics, and biomacromolecules).
For ATX-a: LOD of fluorimeter: 0.54 nM and LOD of smartphone: 1.2 nM.
[73]
Water samplesCYN, ATX-a, HATX-a, AP-A and AP-B, and MCs (-RR, [Asp3]-RR, -YR, -HtyR, -LR, [Asp3]-LR, -HilR, -WR, -LA, -LY, -LW, and -LF)LC-HRMS-LOD: 8 and 53 ng/L
LOQ: 36 and 176 ng/L
Intra-day (RSD%): 1.2–17%
Inter-day (RSD%): 3.1–19%
RSD% < 20%
Recoveries: 81–113%
On-line SPE extraction was used.
Extraction and separation of toxins were achieved in 8 min.
Low relative matrix effects (<29%).
For ATX-a: LOD: 15 ng/L and LOQ: 49 ng/L.
[74]
Water reservoirsCYN, ATX-a, NOD, and MCs (-LR, -RR, -YR, -LA, -LY, -LW, -LF).LC-HRMS0.025–50 µg/LLOD: 4 and 150 pg/L
LOQ: 12 and 450 pg/L
Intra-day (RSD%): 1.5–8.8%
Inter-day (RSD%): 2–23.2%
Recoveries: 9.2–84.3% at 2 ng/L
Recoveries: 32.3–70.3% at 10 ng/L
Recoveries: 48.7–87.8% at 20 ng/L
Two-step SPE extraction was used.
The best recoveries of 70.2–87.8% were obtained for ATX-a, and the worst data recoveries (only 9.2–48.7%) were obtained with MC-LW.
For ATX-a: LOD: 20 pg/L, LOQ: 60 pg/L, intra-day (RSD%): 2.1%, inter-day (RSD%): 22.6%, and recoveries: 70.2–87.8%.
[75]
Cyanobacterial bloom freshwaterCYN, ATX-a, and HATX-aLC-MS/MS0.03–100 ng/mLLOD by DI: 15–70 ng/L
LOD by SPE: 0.6–1.3 ng/L
LOQ by DI: 50–240 ng/L
LOQ by SPE: 2–4 ng/L
RSD%: 1.4–3.3%
ATX-a and HATX-a in freshwater samples could be performed by both DI and SPE coupled with UPLC-ESI-MS/MS; the best values were obtained by SPE. Acetaminophen-d4 (an isotopically labeled acetaminophen) is a suitable internal standard for correcting the matrix effects on the signal intensity of ATX-a and HATX-a.
For ATX-a: LOD by DI: 70 ng/L, LOD by SPE: 1.3 ng/L, LOQ by DI: 240 ng/L, LOQ by SPE: 4 ng/L, and RSD%: 2.2%.
[76]
Water samplesATX-a, HATX-a, CYN, and MCs (-LF, -LR, -RR, -YR, [D-Asp3]-LR, [D-Asp3]-RR)LC-HRMS0.02–100 µg/LLOD extracellular: 10–129 ng/L
LOD intracellular: 3–45 ng/L
LOQ extracellular: 25–129 ng/L
LOQ intracellular: 8–45 ng/L
Intra-day (RSD%): 2–7%
Inter-day (RSD%): 2–7%
Recoveries:84–119%
Matrix effects for the intracellular fraction were similar and acceptable for all analytes.
For ATX-a: LOD extracellular: 10 ng/L, LOD intracellular: 3 ng/L, LOQ extracellular: 25 ng/L, LOQ intracellular: 8 ng/L, intra-day (RSD%): 2–4%, inter-day (RSD%): 4%, and recoveries: 84–100%
[77]
Fish muscleATX-a, HATX-a, CYN, 12 MCs, AP-A and AP-B, and cyanopeptolin-ALC-MS/MS0.05–250 µg/kgLOD: 0.3–10 µg/kg
LOQ: 0.3–33 µg/kg
Intra-day (RSD%): 1.8–11.4%
Inter-day (RSD%): 7.2–25.5%
Recoveries: 83.2–109.8%
For the ATX-a, the parameters were as follows: LOD: 10 µg/kg, LOQ: 33 µg/kg, intra-day (RSD%): 1.8%, inter-day (RSD%): 19%, and recovery: 97.8%.[78]
Water reservoirsMCs (-LR and -RR), NOD, CYN, ATX-a, BMAA, DAB, and AEG LC-MS/MS0.004–2.00 µg/LLOD: 0.0012–0.03 µg/L
LOQ: 0.004–0.1 µg/L
Intra-day (RSD%): 3.3–14.1%
Inter-day (RSD%): 5.1–14%
Recoveries: 70.6–101.0%
Matrix effects: 11.2–253%.
Different types of stationary phases were tested. The results showed that the Oasis MCX cartridge provided satisfactory recoveries for all cyanotoxins (above 55%), with the exception of ATX-a.
Moreover, different membrane filters (CA, GF, PTFE, Nylon, and PVDF) were studied, with the hydrophilic PTFE filter membrane being the most reliable filter material.
Separation and detection of toxins were achieved in less than 12 min.
For ATX-a: LOD: 0.03 µg/L, LOQ: 0.1 µg/L, intra-day (RSD%): 3.6–4.5%, inter-day (RSD%): 6.4–14%, recoveries: 82.3–92.8%, and matrix effect: 11.4–16.8%.
[79]
Algal dietary supplements (Spirulina)MCs (-LR and -RR), NOD, ATX-a, BMAA, DAB, and AEGLC-MS/MS60–2500 µg/kgLOD: 15–90 µg/kg
LOQ: 50–300 µg/kg
Intra-day (RSD%): 3.7–19.5%
Inter-day (RSD%): 5.6–25.1%
Recoveries: 64.2–102.9%
Matrix effects: 9.1–467.8%. For ATX-a, matrix effects: 26–33%.
For ATX-a: LOD: 45 µg/kg, LOQ: 150 µg/kg, intra-day (RSD%): 7.1–9.1%, inter-day (RSD%): 8.2–8.6%, and recoveries: 78.7–87.8%.
[80]
ShellfishSTXs, ATX-a, CYN, NOD doCYN, DTXs, DA, MCs, BMAA, DAB, AEG, azaspiracids, yessotoxins, spirolides, pectenotoxins, pinnatoxins, ovatoxins, gymnodimines, and brevetoxins LC-MS/MS-LOD: 1.2–150 µg/kg
LOQ: 3.5–450 µg/kg
Recoveries: 48–107.5%
For ATX-a: LOD: 8 µg/kg, LOQ: 17.5 µg/kg, and recovery: 94%.[23]
Bivalve mollusks and phytoplanktonATX-a, HATX-a, CYN, MCs (-RR[D-Asp3], -RR[D-Asp3, (E)-Dhb7], -LA, -LR-[Dha7], -LR-[Asp3], -LF, -LR, -LY, -HilR, -LW, -YR, -HtyR, and -WR), and NODLC-MS/MS3.12–200 µg/kg LOD: 2.1–4.04 µg/kg
LOQ: 6.29–12.11 µg/kg
Within-batch repeatability (RSD%): 1.3–7.4%; between-batch repeatability (RSD%): 5.6–18%
Recoveries: 18.6–88.8%
ATX-a, HATX-a, MC-LF, MC-LW, and especially CYN presented the worst recovery data.
For ATX-a: LOD: 2.51 µg/kg, LOQ: 7.52 µg/kg, and recoveries: 57.9–60.6%.
[81]
Cyanobacterial samplesATX-a, HATX-a, CYN, doCYN, NOD, GNT, MCs (-RR, [C-Asp3]RR, -LA, -LR, -LY, -LW, and -YR), and STXsLC-MS/MS--First method to detect GNT simultaneously with other cyanotoxins.[82]
Water samplesMCs (-LR and -RR), NOD, CYN, ATX-a, DAB, BMAA, and AEGCE-MS/MS0.03–2.5 µg/L (NOD and CYN)
0.02–0.15 µg/mL (rest of cyanotoxins)
LOD: 0.005–0.102 µg/L
LOQ: 0.016–0.340 µg/L
Recoveries: 53.5–105%
Intra-day (RSD%): <9.8%
Inter-day (RSD%): <13.7%
No significant matrix effect. The lower recoveries were obtained for DAB.
For ATX-a: LOD: 0.005 µg/L and LOQ: 0.016 µg/L.
[83]
Spinach samples0.6–2.4 µg/kg (AEG, DAB, and MC-LR)
0.45–1.80 µg/kg (ATX-a, MC-RR, and BMAA)
1.28–8 µg/kg (NOD and CYN)
LOD: 0.03–0.23 µg/kg
LOQ: 0.10–0.78 µg/kg
Recoveries: 65.5–81.0%
Precision (RSD%): 1.1–11.9%
For ATX-a: LOD: 0.03 µg/Kg and LOQ: 0.10 µg/kg.
AEG: N-(2-aminoethyl)glycine); APs: anabaenopeptins; APCI: atmospheric pressure chemical ionization; ATX-a: anatoxin-a; BMAA: β-methylamino-L-alanine; CA: cellulose acetate; CYN: cylindrospermopsin; CZE: capillary electrophoresis; DA: domoic acid; DAB: 2,4-diaminobutyric acid; DI: direct injection; DNA: deoxyribonucleic acid; doCYN: deoxycylindrospermopsin; DTX-1: dinophysistoxin-1; ELISA: enzyme-linked immunosorbent assay; ESI: electrospray ionization; FLD: fluorescence detection; GNT: guanitoxin; GF: glass microfiber; HATX-a: homoanatoxin-a; HESI: heated electrospray ionization source; HPLC: high-performance liquid chromatography; HRMS: high-resolution mass spectrometry; LC: liquid chromatography; LDTD: laser diode thermal desorption; LOD: limit of detection; LOQ: limit of quantification; MEKC: micellar electrokinetic chromatography; MC: microcystin; MS: mass spectrometry; MS/MS: tandem mass spectrometry; NOD: nodularin; OA: okadaic acid; PHE: phenylalanine; PTFE: polytetrafluoroethylene; PSP: paralytic shellfish poisoning; PVDF: polyvinylidene fluoride; RSD: relative standard deviations (precision, repeatability, and reproducibility); SIM: selected ion monitoring; SPE: solid-phase extraction; SRM: selected reaction monitoring; STX: saxitoxin; UHPLC: ultra-high-performance liquid chromatography; UPLC: ultra-performance liquid chromatography.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Plata-Calzado, C.; Prieto, A.I.; Cameán, A.M.; Jos, A. Analytical Methods for Anatoxin-a Determination: A Review. Toxins 2024, 16, 198. https://doi.org/10.3390/toxins16040198

AMA Style

Plata-Calzado C, Prieto AI, Cameán AM, Jos A. Analytical Methods for Anatoxin-a Determination: A Review. Toxins. 2024; 16(4):198. https://doi.org/10.3390/toxins16040198

Chicago/Turabian Style

Plata-Calzado, Cristina, Ana I. Prieto, Ana M. Cameán, and Angeles Jos. 2024. "Analytical Methods for Anatoxin-a Determination: A Review" Toxins 16, no. 4: 198. https://doi.org/10.3390/toxins16040198

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop