Next Article in Journal
Effects of Phytotoxic Nonenolides, Stagonolide A and Herbarumin I, on Physiological and Biochemical Processes in Leaves and Roots of Sensitive Plants
Next Article in Special Issue
Gut Microbiota of Freshwater Gastropod (Bellamya aeruginosa) Assist the Adaptation of Host to Toxic Cyanobacterial Stress
Previous Article in Journal
Astaxanthin Alleviates Aflatoxin B1-Induced Oxidative Stress and Apoptosis in IPEC-J2 Cells via the Nrf2 Signaling Pathway
Previous Article in Special Issue
Integration of Transcriptomics and Microbiomics Reveals the Responses of Bellamya aeruginosa to Toxic Cyanobacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Freshwater Cyanobacterial Toxins, Cyanopeptides and Neurodegenerative Diseases

by
Galina Nugumanova
1,
Eugene D. Ponomarev
1,
Sholpan Askarova
2,
Elizaveta Fasler-Kan
3 and
Natasha S. Barteneva
1,4,*
1
Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan
2
Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
3
Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, 3010 Bern, Switzerland
4
The Environment & Resource Efficiency Cluster (EREC), Nazarbayev University, Astana 010000, Kazakhstan
*
Author to whom correspondence should be addressed.
Toxins 2023, 15(3), 233; https://doi.org/10.3390/toxins15030233
Submission received: 31 December 2022 / Revised: 13 March 2023 / Accepted: 19 March 2023 / Published: 21 March 2023
(This article belongs to the Special Issue Ecology and Toxicology of Cyanobacteria and Cyanotoxins)

Abstract

:
Cyanobacteria produce a wide range of structurally diverse cyanotoxins and bioactive cyanopeptides in freshwater, marine, and terrestrial ecosystems. The health significance of these metabolites, which include genotoxic- and neurotoxic agents, is confirmed by continued associations between the occurrence of animal and human acute toxic events and, in the long term, by associations between cyanobacteria and neurodegenerative diseases. Major mechanisms related to the neurotoxicity of cyanobacteria compounds include (1) blocking of key proteins and channels; (2) inhibition of essential enzymes in mammalian cells such as protein phosphatases and phosphoprotein phosphatases as well as new molecular targets such as toll-like receptors 4 and 8. One of the widely discussed implicated mechanisms includes a misincorporation of cyanobacterial non-proteogenic amino acids. Recent research provides evidence that non-proteinogenic amino acid BMAA produced by cyanobacteria have multiple effects on translation process and bypasses the proof-reading ability of the aminoacyl-tRNA-synthetase. Aberrant proteins generated by non-canonical translation may be a factor in neuronal death and neurodegeneration. We hypothesize that the production of cyanopeptides and non-canonical amino acids is a more general mechanism, leading to mistranslation, affecting protein homeostasis, and targeting mitochondria in eukaryotic cells. It can be evolutionarily ancient and initially developed to control phytoplankton communities during algal blooms. Outcompeting gut symbiotic microorganisms may lead to dysbiosis, increased gut permeability, a shift in blood-brain-barrier functionality, and eventually, mitochondrial dysfunction in high-energy demanding neurons. A better understanding of the interaction between cyanopeptides metabolism and the nervous system will be crucial to target or to prevent neurodegenerative diseases.
Key Contribution: This review summarizes what is known concerning cyanopeptides that have neurotoxic potential and are associated with neurodegenerative diseases; detection methods of cyanotoxins are also discussed shortly. We propose a novel hypothesis regarding the role of non-proteinogenic amino acids and related cyanopeptides. The mistranslation mechanism can be evolutionarily ancient, initially serving to outcompete neighboring microorganisms during algal blooms.

1. Introduction

Cyanobacteria are a group of photosynthetic microbes inhabiting diverse aquatic freshwater, marine, and terrestrial environments. Cyanotoxin production is considered to be an ancient trade exceeding 2.5 billion years of age [1].
The ability of cyanobacteria to produce cyanotoxins and their ubiquity in freshwater ecosystems with increasing demands upon water resources require better detection and a more comprehensive understanding of cyanobacterial distribution and its impact on animals and humans. Although cyanobacterial cell numbers change seasonally, the toxins can persist in water for several months, extending low-dose exposure [2]. Aquatic organisms (e.g., grazers/herbivores, fish, mammals) may consume aquatic plants with high concentrations of cyanotoxins, and trophic transfer may happen to higher-order organisms, although, for some toxins, biodilution, and not biomagnification may be a predominant process in the food webs [3]. The detectable presence of cyanobacterial toxins in animal tissues has been found to be associated with mass mortalities of animals, including cows, dogs, and sea mammals [4,5,6,7,8,9,10,11,12], birds [13], and some human cases [14,15]. The terrestrial vertebrates affected by cyanotoxins are more diverse than was thought before [16,17]. Moreover, cyanotoxins demonstrate sublethal effects, including growth inhibition in zooplankton and aquatic plants, macroinvertebrates, and aquatic plants [18].
The predicted climate changes, such as increased water temperatures, together with anthropogenic nutrient loadings, escalate freshwater cyanobacterial frequency, duration, and size of algal blooms [19,20,21]. The cyanobacterial abundance has increased disproportionally relative to other phytoplankton since 1945 [22]. Although our knowledge about cyanotoxins mostly comes from temperate and tropic areas, Arctic regions undergo the most pronounced and rapid climate changes, and high-latitude lakes support cyanobacteria blooming and cyanotoxins production [23]. Moreover, the occurrence and intensity of near-surface phytoplankton harmful algal blooms (HABs) have been increasing across the world [24,25] due to the eutrophication or nutrient enrichment of water bodies [26,27]. The potential presence of low doses of cyanobacterial toxins in drinking water is likely to be a continuing problem.

2. Cyanobacterial Toxins

Cyanobacteria produce a variety of toxins. Traditionally, they are divided based on functional properties into main groups: hepatotoxins, neurotoxins, dermatotoxins, and cytotoxins. The toxins found in cyanoHABs include cyclic peptides microcystins (MCs), and nodularins, and neurotoxins such as anatoxins (anatoxin-a, homoanatoxin-a, guanitoxin), ciguatoxins, saxitoxins, ß-methylamino-L-alanine (BMAA) and its isomers (2,4-diaminobutyric acid (2,4-DAB) and N-(2-aminoethyl)-glycine (AEG) [21]. More than 80 cyanobacteria species are known to be toxigenic, and assays for the detection and toxicity of cyanotoxins continue to develop.
One of the major sources of cyanotoxins for humans is drinking water. However, guidelines for water quality monitoring are limited to MCs [28]. Furthermore, exposure of grazing livestock with terrestrial cyanobacteria that may be present in fields used for livestock grazing (biofilms of Phormidium sp.) or can be fed to livestock directly [29,30,31,32], and of which the aggregates can bioaccumulate in cow’s milk [29,33] or bird eggs [34] and which increases the likelihood of human exposure [35]. There is an urgent need to detect other cyanobacterial toxins in drinking water and food and to understand how they are involved in the pathogenesis of chronic diseases such as neurodegenerative disorders.
Many cyanobacterial toxins still have to be discovered. For major toxins groups, new variants can be found and characterized. Thus, a structural variant of anatoxin-a, dihydro-anatoxin-a, has been recently identified in many samples of benthic cyanobacteria, even exceeding the abundance of anatoxin-a [36]. Of potential interest are compounds with neurotoxic effects, such as cyanostatins A and B, lipopeptides, isolated from cyanobacterial water blooms [37], anabaenopeptins [38], and microginins [39]. The identification of biologically active cyanotoxins, including many novel lipopeptides with neurotoxic effects such as jamaicamides and antillatoxins continues [40]. Vacuolar spongiform myelopathy has recently been linked to aetokthonotoxin (AETX) from epiphytic cyanobacterium Aetokhonos hydrillicola that is growing in man-made water bodies of the southeastern United States [41]. This finding warrants further research into the potential toxins produced by epiphytic and benthic cyanobacteria species.

2.1. Microcystins (MC) Family

A full structural chemical analysis of MCs was achieved in the 1980s through a combination of spectroscopy, nuclear magnetic resonance, mass spectrometry, and amino acid analysis and demonstrated that the chemical structure of MCs consists of a cyclic heptapeptide with two variable and five relatively conserved amino acids biosynthesized non-ribosomally via an MC synthetase gene cluster [42]. A universal nomenclature system was suggested based on the positions of amino acid residues 2 and 4 (denoted as X and Z in the original structure of MC, i.e., MC-XZ) [43]. By 2019 the identification of at least 279 different MC congeners was reported in the literature [42].
Among cyanobacterial toxins, MCs are the most diverse group and the best described, though MC-LR and MC-RR variants—are the only two widely researched. Other MC congeners demonstrate different toxicokinetic and toxicodynamic features [44]. Variations in toxicity between MC congeners can be attributed to the differences in their uptake by organic anion transporting polypeptides (OATP) transport as well as changes in their inhibition of serine/threonine protein phosphatases (PP) 1 and 2a [45]. The toxicity of MCs depends on variations in their chemical structure and ranges over six orders of magnitude [46].
MC toxicity affects not only the liver but also the brain [47] and other organs. Multiple neurotoxic effects of MC-LR were demonstrated using multiple biological models, including birds, fishes, and mammals [48,49,50]. For example, using murine brain cell line as a model, congener-dependent pronounced neurodegenerative effects were identified (MC-LF >> MC-LW > MC-LR) [51].

2.2. BMAA (β-N-methylamino-l-alanine) and Isomers

BMAA is a non-proteogenic amino acid produced by all known groups of free-living and symbiotic cyanobacteria [52]. The isomeric forms of BMAA, such as 2,4-DAB and AEG, can also be found in different species of cyanobacteria, including Anabaena, Leptolyngbya sp., Oscillatoria sp., Merismopedia sp. and Microcystis aeruginosa [53]. The isomers are detected in nature along with BMAA but are less studied. BMAA isomers are neurotoxic [54]. Recent research using larval zebrafish as a biological model identified 2,4-DAB as a more potent neurotoxin than AEG and BMAA [55].
Despite some contradictory research [56], an increasing body of experimental proof provides confirmation that BMAA plays a significant role in neurodegenerative diseases (ND) [57,58,59]. Some analytical methods related to liquid chromatography and fluorescence-based detection of BMAA, especially in older literature, may be prone to overestimation of BMAA concentration in studied samples and should be treated cautiously [60,61]. BMAA has been shown to contribute to protein misfolding, enzyme inhibition, and neuroinflammation [62]. BMAA toxic effects were found to be associated with the misincorporation of serine in multiple human proteins [63,64], which can lead to the formation of inclusion bodies in neurons [65]. Misincorporation in some serine sites has been reported to contribute to neuropathologies [66]. Even a low misincorporation rate (1 per 10,000 codons) can lead to neurodegeneration in a rodent model [67]. In their in vitro and in vivo studies, several groups that BMAA leads to the overexpression TDP-43 (TAR DNA-binding protein 43) encoded by the TARDBP gene [68,69,70].
Most animal models used for studying BMAA were based on investigating BMAA effects in the brain and other organs [59]. Thus, Xie and co-authors reported that in their experiments, less than 1% of total BMAA detected in adult mice plasma was taken in the brain [71], i.e., blood-brain barrier (BBB) was not easily permeable for BMAA. The mechanism of neurotoxicity may involve a direct action on the N-methyl-D-aspartate (NMDA) receptor, activation of glutamate receptor 5, and induction of oxidative stress.
Recently, Han and co-authors [72] found that BMAA can serve as a substrate for human alanyl-tRNA synthetase (AlaRS), avoiding the intrinsic editing activity of AlaRS, acting as a competitive inhibitor, and compromising the editing ability of AlaRS. Terminally differentiated cells, such as neurons, are particularly susceptible to mistranslation and accumulation and forming of misfolded and aggregated proteins [67,73].

2.3. Other Cyanobacterial Neurotoxins

Traditional neurotoxins from cyanobacteria with acute effects include alkaloid or organophosphorus compounds such as (a) anatoxin-a and homologs, which affect nicotinic acetylcholine alkaloid toxins and muscarinic acetylcholine receptors [74,75]; (b) saxitoxins can be produced by both dinoflagellates and by cyanobacteria from several genera including Aphanizomenon, Cylindrospermopsis, and Dolichospermum; (c) guanitoxins which are similar in structure to organophosphates and able to irreversibly inhibit acetylcholinesterase [76]; their presence was also found in desert assemblages [77,78]. Cylindrospermopsin is another frequent finding during fish kills resulting from cyanoHABs [79,80]. It is an alkaloid consisting of a tricyclic guanidine moiety combined with hydroxymethyluracil [81], which interferes with cellular metabolism and causes hepatotoxic and genotoxic effects, as well as neurotoxic effects [82].
Nodularins can be found and bioaccumulated in a wide range of organisms [83]. Acetylcholinesterase activity (AChE) is an indicator of neurotoxic effects, and exposure to a cyanobacterial pentopeptide nodularin affects the AChE activity in the Baltic clam Macoma balthica [84] and the mussel Mytilus edulis [85].
It is known that cyanobacteria are capable of producing different toxins which can be present during the same HABs [86]. However, it is not clear, how the environmental factors regulate the abundance of different MC congeners and isoforms of other toxins in a bloom [42]. During harmful blooms of cyanobacteria, different cyanobacteria species can co-occur, potentially producing various cyanotoxins, cyanopeptides, and other metabolites simultaneously. Thus, BMAA can co-occur with its isomers (2-DAB and AEG) and have synergistic neurotoxic effects, as demonstrated in in vitro cell line experiments [87]. Moreover, the joint presence of MC-LR and BMAA leads to their interaction in vivo and to the neurotoxic effect enhancement [88]. The development of methods allowing for the assessment of multiple toxins during algal blooms is needed [89].

2.4. Cyanopeptides

The majority of cyanobacterial secondary metabolites are peptides or include peptidic substructures. Cyanopeptides are non-ribosomal peptides rich with posttranslational modifications and non-proteinogenic amino acids and consist of linear, cyclic, or multicyclic molecules with basic, depsipeptidic, or lipopeptidic structures. More than 500 cyanopeptides ranging from app. 300 to 2000 Da had been structurally identified by 2019 [90]. Natural selection did not minimize the pool of peptides but favors the production of a wide array of different peptide structures. The biosynthetic non-ribosomal pathways of peptide synthesis are evolutionarily ancient and precede synthetic pathways of higher plants and animals. During HABs, cyanobacteria produce a tremendous amount of diverse cyanopeptides; however, their ecological significance is unclear. They can happen at surface waters in the same nanomolar concentration as MCs, exhibit toxicity towards grazers in the same micromolar range as MCs, and their production is synchronized with Microcystis sp. Although the abundance of MCs can be monitored successfully, more studies on cyanopeptides appearance and persistence during blooms [90,91] and their potential for chronic toxicity are needed. The bioactive compounds produced by cyanobacteria are not limited by peptides and also include alkaloids, cyclophanes, terpenes, lactones, etc. Cyanobacterial compounds have a broad bioactive spectrum, with many acting as serine protease inhibitors, trypsin and chymotrypsin inhibitors, and anti-cancer compounds capable of modulating infectious diseases [92].

2.5. Chronic Effects of Cyanobacterial Toxins

The epidemiological studies of human health impacts of chronic cyanobacterial toxins exposure are nascent. They have been associated with neurodegenerative diseases (ND), including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS) [93,94,95,96,97]. Clusters of ALS and ALS-like diseases have been reported in relation to cyanobacteria in Guam, France, Japan, New Hampshire, and Wisconsin (summarized in Table 1). The slow onset of ND (time distance between exposure and possible outcome) and problems with the assessment of environmental exposure interfere with our understanding of the role and significance of cyanotoxins in ND.
The link between BMAA and ND is yet to be further elucidated. Several studies have reported the presence of BMAA post-mortem in the brain tissues of patients who die from ALS/PD [102,121]. However, in ALS/AD disease [122], and other ND studies some researchers were not able to identify BMAA presence [123,124]. The ALS/PD neurodegenerative disorder, formerly hyperendemic in Guam-USA, Kii-Japan, and Papua-Indonesia associated with several cycad food toxins, including cyanotoxins [125]. BMAA and its isomers have now been identified in both aquatic and terrestrial eco-systems in North America [95,96], The Baltic Sea [126], France [113], Sweden [127], Peru [128], and Qatar [129]; and is produced by several different cyanobacteria [52], diatoms [127], and dinoflagellates [130].
Residential exposure to environmental pollutants may play an essential role in the etiology of ALS, which is supported by non-random distribution by addresses of ALS patients [131].

2.6. Stability of Cyanotoxins

Cyanotoxins possessing cyclic peptide structures, such as MCs and nodularins, are resistant to chemical degradation [132,133], highly stable, and may persist in aquatic ecosystems for weeks and months [2,133]. Thus, MCs can be retained in mussels (Mytilus californians) for up to eight weeks [134]. The high stability of MCs, cylindrospermopsin [135,136], and other cyanotoxins over a wide range of pH and temperature might have significant consequences for aquatic ecosystems and contribute to bioaccumulation of toxins to higher levels of food chains. These peptides are synthesized non-ribosomally and may contain non-proteinogenic amino acids [137,138,139].
Toxins undergoing attenuation via photodegradation may vary depending on the type of toxin, HABs timing, and environmental conditions [140]. Though cyanotoxins are resistant to chemical degradation, recent advances in molecular microbial communities research have found that toxic cyanoHABs favor the specific members of bacterioplankton with degrading abilities towards cyanotoxins [141,142]. More than 120 taxa of viruses, bacteria, microfungi, heterotrophic protists, and several eukaryotic microalgae negatively affect Microcystis growth [143]. The strains of the bacterial genera Sphingomonas (majority of MC-degrading bacteria), Rhodococcus, Brevibacterium, Burkholderia, Mycobacterium, Pseudomonas, Novosphyngobium and others can degrade MCs in time scale from hours to days [144,145,146,147,148,149]. The genomes of some MCs-degrading bacteria are sequenced [150,151], and mlr gene cluster have been implicated in playing a prominent role in the sequential hydrolysis of MCs peptide bonds [151,152].
Similar to MCs, nodularin and cylindrospermopsin can also be degraded by bacteria isolated from cyanobacterial blooms (Bacillus sp., Aeromonas sp.) [153,154,155]. Not only bacteria but fungi demonstrate algicidal activities such as Trichoderma citrinoviride degrading MCs [156]. Furthermore, Mohamed and co-authors [157] summarized data on six fungal species with biodegrading activities against MCs. Between zooplankton grazers, metazoans, such as Daphnia and copepods, are also susceptible to cyanotoxins [158,159]. Protozoa, on the other hand, are highly resistant to cyanotoxins and demonstrate great potential in controlling harmful cyanobacteria and improving phytoplankton composition in eutrophic waters [160,161].

2.7. Current Cyanotoxins Analytical Methods

Analytical techniques (fraction analysis, quality control) play a critical role in assessing the cyanotoxins’ effects and were summarized in many excellent reviews [162,163,164,165,166]. There are still significant data gaps in analytical methods, including (a) the absence of all the relevant standards [167]; (b) the need for validated methodologies for cyanotoxins outside the water samples: (c) the need for standardization of cyanotoxins in multicenter monitoring programs [168]; (d) the need for new technologies allowing simultaneous identification of as many toxins as possible; (e) and the need to improve robustness and a detection limit of detection methods. Another critical challenge is analyze cyanotoxins faster and feasibly in situ [169]. Unsuitable analytical methods may partly explain the lack of consensus over the widespread presence of some cyanotoxins (BMAA) in aquatic ecosystems [170].
Recently, high-resolution mass spectrometry (HRMS/MS) has become more available for researchers. Current methods rely on defined cyanotoxins and cyanopeptides targets and are generally inappropriate for detecting and identifying emerging novel compounds. The recent approach of the non-targeted analysis of pollutants and toxins in water focus on a comprehensive workflow for the acquisition and treatment of the data generated after liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS) analysis [171,172,173,174]. So-called suspect screening identifies novel compounds, including cyanotoxins and cyanopeptides based on the exact mass (m/z), presence of one or more charge states (z = 1, 2, etc.), expected isotope pattern and common adducts) as well as secondary fragmentation (MS2) even without reference chemicals [175,176,177,178].
Since the middle of the twentieth century, when the first MCs were identified and purified [179], many methods have been developed to analyze environmental samples for cyanotoxins [180]. Nowadays, the number of studied cyanotoxins and analytical methods for their qualitative and quantitative detections have increased. Detection approaches vary in terms of accuracy, sensitivity, and specificity. The most commonly used techniques for cyanotoxins detection are enzyme-linked immunosorbent assays (ELISA), protein phosphatase inhibition assay (PPIA), molecular assays - polymerase chain reaction (PCR), and quantitative real-time PCR (qPCR) for toxins producing genotypes for cyanobacteria identification, liquid chromatography (LC) and high-performance liquid chromatography (HPLC) combined with different detectors. Among these, liquid chromatography-mass spectrometry (LC-MS) takes a special place because it identifies the target cyanotoxins with high accuracy at a significantly low detection level (Table 2) [181,182]. Moreover, despite the structural diversity of cyanotoxins, LC-MS allows for determining groups of toxins simultaneously. That is one extra advantage of using this detection technique integrated with bioassays and molecular assays in complex environmental samples for complete water quality assessment [183,184].
As separation instruments, HPLC and UHPLC are usually used. UHPLC is faster due to the higher pressure applied, and the online SPE procedure provides reduced sample time processing [2,199]. HPLC-UV/PDA [191], and HPLC-DAD [204] have less LOD values (3–4 μg/L, 0.2–0.3 μg/L, respectively) for MCs than LC-MS/MS where minimum LOD values vary within 0.0003–0.1 µg/L [182,196,206]. Concerning other cyanotoxins, MS detection techniques also provide relatively low values of LOD and LOQ. The LC-MS method requires expensive instruments, lengthy operator training, and thorough sample preparation, making it time-consuming. That limits LC-MS techniques’ application as ubiquitous [2]. Nevertheless, this method remains preferable for precise quantitative analysis of cyanotoxins in water samples.

3. Toxin Exposure Pathways

Major cyanotoxins’ exposure routes include ingestion through drinking water or dermal contact with recreational waters [214,215], also through food, and inhalation since cyanotoxins were identified in aerosols generated by HABs [216]. Historically, cyanoHABs were considered a public health threat to freshwater lakes, rivers, and reservoirs. However, freshwater-sourced MCs can accumulate in marine mollusks in concentrations 100-fold greater than in surrounding water [10,134].

3.1. Transport of Cyanotoxins in Freshwater and Marine Systems

Recent studies demonstrated that cyanotoxins could persist during transport into estuarine and marine waters and directly affect marine ecosystems [10,134,217,218,219]. MCs and other toxins produced by freshwater cyanobacteria can enter the marine ecosystem via freshwater channels and outflows [10,218]. It changes the HABs’ management approach, requiring monitoring of multiple toxins across the freshwater-to-marine continuum and including cyanotoxins in marine and estuarine monitoring [220].

3.2. Toxin Exposure Pathways: Oral (Drinking Water)

When drinking water is impacted by cyanobacterial toxins resulting from HABs and not treated adequately to reduce the cyanotoxin levels, it can cause severe effects on the entire region [216].
The causes of cyanobacteria proliferation in urban environments are mainly due to the disposal of untreated domestic sewage in water reservoirs and surface runoff water from soils. In analyzing sewage disposal systems in the main cities of Kazakhstan-Almaty and Astana, the efficiency of biogenic compounds removal remain unsatisfactory, reaching only 30–40%. This eutrophication is due to the increase in nutrients, such as phosphorus and nitrogen, arising from human action, representing a serious risk to the health of living beings and drastically reducing water quality. To cope with this problem, the possibility of intensifying nitrogen and phosphorus removal using zeolite as a biofilm carrier in an activated sludge tank is examined [221].

3.3. Toxin Exposure Pathways: Oral (Food)

Food is an important source of cyanotoxin exposure [222,223] BMAA accumulation in shellfish and fish [130,223,224,225], and further transmission along the food chain (chicken tissues) [226] may lead to human exposure. Worryingly, exposure of crop plants to cyanotoxins through irrigation was already demonstrated [227,228,229]. For centuries some species of Nostoc—the symbiotic colonial cyanobacteria N. flagelliforme, N. commune, and N. sphaeroides—have been wild-harvested and consumed as a part of the traditional diet by indigenous people in different countries, including Peru, China, Ecuador, Mexico, Fiji, Philippines, Mongolia [230,231,232,233]. Cyanotoxins BMAA and its isomers were detected in dietary supplements [234,235,236].
Chronic dietary exposure to BMAA present in the traditional Chamorro diet was associated with the formation of both β-amyloid deposits and neurofibrils tangles (NFT) found in brain tissues of Chamorros people who died with ALS/Parkinson’s dementia complex (ALS/PD) [65]. BMAA occurs not only as a free amino acid at different levels of the trophic chain (cyanobacteria Nostoc sp., root symbioses, cycad seeds, flying foxes, and brain tissues of Chamorro people who passed away from ALS/PD) but can also be released by acid hydrolysis increasing in concentrations 10- to 240-fold [237]. Vervet monkeys fed for only six months with BMAA-dosed fruit developed β-amyloid deposits and NFT in the brain. Increasing the amount of L-serine in the vervets diet reduced the density of NFT and the risk of neurodegenerative pathological brain findings [65].

3.4. Toxin Exposure Pathways: Air (Aerosolization)

Algae can be dispersed by air [238], and aerosol can be created from the algae during HABs [239]. The increase in the salinity of freshwater streams is likely to influence the abundance and diversity of aerosolized bacteria [240]. The cyanotoxins may be transported in aerosols from lakes with high concentrations of toxigenic cyanobacteria [238,241,242,243,244,245]. Airborne cyanobacteria persist in an urban environment and indoor living quarters [246,247,248,249]. The presence of airborne cyanobacteria in the nearby beach area is significant, and representatives of different taxa, including toxic microalgae, have been studied by many researchers worldwide [245,250]. The screening for toxins in extreme habitats demonstrated their presence in all general extremophile habitats [251].
Recent findings with rat models confirmed that BMAA exposure was insufficient in producing gross toxic effects; however, it still leaves the possibility of lifelong exposure via inhalation [252]. The health concerns associated with aerosolization remain understudied [253].
MC-LR exposure in the existing rodent models increases lung infiltration with granulocytes [254,255] and increases proinflammatory cytokine expression [256,257]. Recently, Breidenbach and co-authors [258] reported that the human airway epithelium response to MC-LR is represented by proinflammatory phenotype, including chemokines.
The aerosolization of cyanobacteria was proposed as a risk factor for ALS [112]. Aerial link of exposure was investigated with ALS/PD. BMAA and its isomers (DAB and AEG) were measured in air filters around lake Mascoma [96]. Moreover, Facciponte and co-authors [259] found that humans routinely inhale aerosolized cyanobacteria. Using PCR, authors identified cyanobacteria at high frequencies in the upper respiratory tract (93.20%) and central airway (79.31%). They concluded that cyanobacteria exposure might be a prevalent and chronic phenomenon and not necessarily restricted to water bodies.
Autoradiographic imaging in mice showed a distinct localization of radioactivity in olfactory mucosa and bulb following intranasal instillation of radiolabelled BMAA, confirming a direct transfer of BMAA via olfactory pathways to mice brain circumventing the BBB [260].

3.5. Natural Model of Toxin Exposure

The complexity of ND requires a deep understanding of the disease biology and makes it challenging to develop a model of cyanotoxin exposure close to neurodegenerative findings in humans due to the species-specific variations in the phosphorylation and cleavage of the tau protein [261]. Natural animal models should recapitulate two major features of human ND: Aβ deposition and NFT formation. Chronic low BMAA concentrations induce neurodegenerative changes in non-human primates [65,262]. BMAA can bioaccumulate in marine apex predators such as dolphins and sharks, and in humans [230,263,264]. It was detected in the brains of stranded dolphins with pathological hallmarks of AD at concentrations higher than those found post-mortem in individuals with ALS and AD [265]. Chronic low BMAA concentrations induce neurodegenerative changes in non-human primates [65,262]. There are increased numbers of β-amyloid+ and dystrophic neurites in the auditory cortex compared to the visual cortex and brainstem [265].

3.6. Cyanotoxins and Infections

BMAA can facilitate most of the mechanisms related to neurodegeneration [61]. Thus, Lobner and co-authors [266] demonstrated that BMAA at the 10–100 µmol potentiates neurotoxicity induced by β-amyloid and NMDA.
STX doubled the quantity of ZIKV-induced neural cell death in progenitor areas of human brain organoids, and the chronic ingestion of water contaminated with STX before and during gestation caused brain abnormalities in offspring of ZIKV-infected immunocompetent C57BL/6J mice. These results raised a public health concern regarding the consequences of arbovirus outbreaks in areas with droughts and/or frequent freshwater cyanobacterial blooms [267].
The outbreak of Zika syndrome coincided with a major drought in the region between 2012 and 2016. Characteristic of dry seasons, the concentration of nutrients from untreated effluents and lower volume of water, and an increase in atmospheric temperature allowed greater blooming of cyanobacteria. Consequently, the concentration of cyanotoxins, such as saxitoxins, increased. It led authors to formulate the hypothesis that cyanobacteria in the water supply would be a causal cofactor of zika-associated microcephaly.

4. Mechanisms of Brain Toxicity

The toxins produced by cyanobacteria are incredibly diverse. Well-studied neurotoxins of algal origin are alkaloids saxitoxins (STXs) that have been identified in dinoflagellates and several cyanobacterial genera, including Anabaena, Aphanizomenon, Planktothrix, Cylindrospermopsis, and Scytonema [268,269]. STXs are represented by more than 50 structural analogs commonly known as paralytic shellfish toxins (PSTs) [75,268,269]. They block the passage of sodium across a biological membrane and interfere with potassium and calcium-mediated ion channels [270].
Although the pathophysiology of some alkaloid and phosphororganic toxins (STXs, anatoxins, etc.) are relatively well studied, others, such as ciguatera-like toxins, are not clear [271,272]. Recently, the neurotoxic effects of cyanopeptides attracted more attention [90,273,274]. Some cyanopeptides have been found to have anti-proliferative effects on tubulin and microtubules [273,274,275,276,277], which are crucial components for neurons. For instance, the toxic cyclodepsipeptides known as cryptophycins [273], which are 100–1000 times more potent than paclitaxel and vinblastine, can impede the formation of a proper mitotic spindle by preventing the correct assembly of microtubules. This can lead to cell cycle arrest and, ultimately, cell death [275,276,277]. Another cyclic depsipeptide apratoxin isolated from Lyngbya sp. induces G1 cell cycle arrest [276,277]. Major mechanisms related to the neurotoxicity of cyanobacteria compounds include (1) blocking of essential proteins and channels; (2) inhibition of essential enzymes in mammalian cells such as protein phosphatases and phosphoprotein phosphatases [45,278,279]; (3) potentially molecular targets may include toll-like receptors (TLR) 4 and 8 [280], participants in neuronal conduction and neuroinflammation [281]. The hypothesis that L-BMAA can be misincorporated into proteins is discussed in detail by Dunlop and co-authors [63].
In the high-profile case of fatal human intoxication with MC in the hemodialysis unit in Brazil (1996), patients developed symptoms of acute neurointoxication (intermittent blindness, deafness. convulsions, tinnitus) and subsequential hepatotoxicity [14]. The mechanisms of MCs neurotoxicity were reviewed by different researchers [282,283]. Thus, MC-LR can induce apoptosis and atrophy of gonadotropin-releasing hormone (GnRH) neurons in rats’ hypothalamus [284,285]. Although the molecular mechanism of MC-LR- induced apoptosis remains elusive, growing evidence supports that MCs and other cyanobacterial toxins, such as cylindrospermopsin, act as endocrine disruptors [286,287]. Recently, Shi and co-authors [288] demonstrated that acutely administrated MC-LR induces pathological changes in rat’s hypothalamus and pituitary gland and alters the transcription of genes involved in hormone biosynthesis.
In recent decades an increasing number of studies showed MC-induced immune dysfunction and a disturbance in the production cytokines leading to inflammation in fishes [289,290] and rodents [291,292,293]. BMAA is proven to be neurotoxic for different biological models, including insects [294,295] and vertebrates (fish, rodents, dolphins, and birds) [40,296].

4.1. Neurodevelopmental Effects

The link between neurodegeneration and neonatal BMAA exposure, dose-dependent neuronal loss, beta-amyloid deposition, and behavioral deficits was recently demonstrated in a rat model [297]. Autoradiographic imaging confirmed transplacental uptake of radiolabelled BMAA and specific uptake in mouse fetuses [298]. Furthermore, in neonatal rats, the free BMAA concentration was higher in the neonatal brain than in peripheral tissues such as the thymus, pancreas, and spleen, except for the liver. The level of protein-associated BMAA was significantly higher in the hippocampus than in other brain regions [299]. The BMAA exposure to neural stem cells decreased neurite outgrowth and a number of neurites in neural stem cells (NSC) [300], and NSC were more sensitive to BMAA exposure than primary neurons [301]. The authors conclude that BMAA acts as a developmental toxin. BMAA can negatively impact NSC homeostasis, increasing susceptibility to neurodegenerative disease later in life [300]. Perinatal exposure in mice, even with low doses of BMAA, leads to neurobehavioral disturbances during the postnatal period and adulthood [302]. Moreover, BMAA modifies neuroblast organization increasing the number of neuroblasts clusters [303].
Recent studies have revealed that repeated rat treatment with MC-LR had a toxic effect on the development of a nervous system in the rat offspring [304].

4.2. Blood-Brain Barrier (BBB)

The BBB and the blood-cerebrospinal fluid (CSF) barrier separate the central nervous system (CNS) from blood and include the endothelial lining of the brain capillaries associated with astrocytes, pericytes, and neurons. The pericytes and astrocytes are closely associated with the endothelial cells and are required for capillary maturation (pericytes) and the maintenance of the permeability-barrier functions (astrocytes). The basement membrane (which contains laminin, proteoglycans, fibronectin, collagen IV, nidogen, and entactin) is essential for blood-barrier differentiation. BBB separates neurons from the circulating blood and maintains the internal chemical composition of the brain "milieu" responsible for the proper functioning of neuronal circuits, neurogenesis, angiogenesis, synaptic transmission, etc. BBB breakdown due to disruption of the tight junctions may result in synaptic and neuronal dysfunction and contribute to neurodegenerative disorders such as ALS, AD, Parkinson’s disease, and multiple sclerosis [305,306,307,308].
In adult rodent biological models, the rate of retention of BMAA is low [71,298,309,310]. Although Karlsson and co-authors [298,311] demonstrated a mild neuronal loss in the hippocampal regions of adult rats following BMAA exposure, no adult rodents models have successfully reproduced neuropathological changes typically seen in ND patients [312,313,314]. However, in rodent fetuses and neonates, early-life exposure to BMAA affects brain development with long-term consequences [33,298]. Moreover, neonatal exposure to BMAA during the critical period of neurogenesis caused β-amyloid deposition, neurofibrillary tangles of hyper-phosphorylated tau, Lewy bodies formation, microgliosis as well as neuronal loss in the hippocampal striatum, substantia nigra region and ventral horn of the spinal cord [315].
Berntzon and co-authors found BMAA in the CSF of a patient with ALS and some controls, though they did not confirm a prevalence of BMAA findings in the majority of ALS patients [121]. Significant amounts of BMAA were found in the brain tissues of ALS and AD patients, confirming the ability of BMAA to cross the BBB [122]. The other possible route of entry to the CNS is through the olfactory epithelium and the nasal passage or via the blood. The cyanobacterial neurotoxin BMAA can be directly transferred through olfactory pathways circumventing the BBB in mice and directly affecting olfactory neurons [260]. Recently, Garamszegi and co-authors [316] used triple quadrupole tandem mass spectrometry to demonstrate that BMAA and its isomers AEG and 2,4-DAB were detected in olfactory tissues of AD post-mortem brains. This finding contradicts early reports from Meneely and co-authors, who did not find BMAA in the brains of AD patients [124].
MC-LR has been confirmed to cause BBB disruption and enter the brain tissue, resulting in neurotoxicity and inducing structural and functional changes in neuronal cells [317,318,319]. Moreover, MC-LR had increased BBB permeability in mice inducing metalloproteinase-8 (MMP-8) expression and breaking through tight junctions [320]. MCs exposure inhibits serine- and threonine-specific protein phosphatases immune cortical neurons [321,322,323]. Recently, a study by Yan and co-authors [324] revealed that MC-LR disrupted the function of the neuronal ubiquitin-proteasome system in neurons, leading to the release of α-synuclein (α-syn) from neurons. The presence of α-syn in Lewy bodies has been associated with several ND [325,326]. In addition, α-syn was transported into glial cells through TLR4 receptors and activated the NLRP3 inflammasome [324].

4.3. Glia

Glial cells, including microglia, have long been suspected of playing a role in AD, but only because of their ability to react to neuronal dysfunctions (e.g., amyloid and Tau aggregates). Microglial activation and neuroinflammation are common to many NDs. This neurocentric view, which considered glial cells as secondary, has been challenged recently by the results of genetic association studies identifying genetic loci associated with the risk of AD that is associated with genes preferentially or exclusively expressed in glial cells [327].
The research on cyanopeptides effects on glia is limited. Chiu and co-authors [328,329] demonstrated a gliotoxicity of BMAA using the olfactory ensheathing cell as an in vitro model. A study conducted by Bubic and co-workers [330] showed that depsipeptide planktopeptin and anabaenopeptins impair the metabolic activities of normal human astrocytes via membrane perforation, oxidative stress, and changes in mitochondrial metabolism. Later, Mello and co-authors [331] showed cytotoxic effects of BMAA and MC-LR on primary astrocytes isolated from mixed adult brain cell cultures; and Soto, with co-workers [332], demonstrated damaging BMAA effects on Muller’s glial cells. Both glial cells and neurons can uptake and accumulate BMAA, as demonstrated using a specific polyclonal antibody against BMAA [333]. BMAA induces a proinflammatory response in astrocytes and microglial cells, causing a shift in the ratio of CD86/CD206 cells [303,334].
Takser and co-authors [335] demonstrated the effects of low doses of MC-LR, alone or as a part of a complex mixture with other cyanotoxins, such as cylindrospermopsin and anatoxin-A, on the viability of murine glial and neuroblastoma cell lines.
The role of dysfunctional astrocytes in the pathogenesis of ALS and other ND indicates that astrocytes may be targeted with strategies for their revival. These strategies may include direct intervention on astrocytes with modulatory medicines, exosomes, and miRNA-based therapies or their replacements.

5. Cyanotoxins, Cyanopeptides and Neurodegenerative Diseases

A central dogma of age-related ND is the claim that the accumulation and propagation of aggregated proteins cause neurodegeneration [336]. Recently, a mechanism that does not involve a specific neuropathogenic protein but is mediated by error-prone translation leading to stochastic near-cognate missense substitutions was suggested. Drummond and Wilke proposed in 2008 that tolerance to translation errors of certain proteins provides a new mechanism to explain their propensity to misfold pathologically [337]. Mistranslation destabilizes the proteome leading to misfolding and accumulation in the cells of potentially toxic protein aggregates [67,337]. The finding that translational error increases with age in some biological models (Drosophila) [338] may suggest the possibility that the rate of translation leading to aging-related proteostasis failure may be a key event in early ND diseases [339]. The mistranslating cells exhibit severely inhibited protein synthesis and formation of protein aggregates in the cellular ND model [340]. Aminoacyl-tRNA synthetases (AARSs) catalyze covalent binding tRNA with their cognate amino acids and are two-to-three orders of magnitude more selective than other amino acid-utilizing [341]. Altered translation fidelity impairs cellular homeostasis and has been implicated as a molecular mechanism underlying changes in synaptic function, selective loss of certain types of neurons, and the pathogenesis of numerous ND [342,343,344].
Hundreds of non-proteinogenic amino acids produced by cyanobacteria include BMAA and can, in principle, enter human protein synthesis through foods and drinking water. Many non-proteinogenic amino acids have structures similar to the standard amino acids and have the potential to be misincorporated into the proteins [345]. It has been speculated that microbial-produced non canonical amino acids could represent one of the environmental triggers for the onset of neurodegeneration [62,346].
The BMAA provides one of examples of multiple mimicry [345]. Earlier studies support the ability of BMAA to be incorporated into the proteins [57,347]. It has been hypothesized that BMAA is misincorporated at serine codons during protein synthesis [57]. The detection of non-canonical amino acids in protein sequence employed evaluation of misincorporation at the tRNA level using radiolabelled amino acids and tRNA microarrays to detect misacylation. This approach was used to prove that BMAA charges to both alanine and serine tRNAs and bypasses the proofreading ability of the alanine aaRs, suggesting misincorporation at alanine positions [72,188]. Han and co-authors [72] demonstrated that BMAA is a substrate for human alanyl-tRNA synthetase (AlaRS) and not a substrate for human seryl-tRNA synthetase (SerRS), and can form BMAA-tRNA-Ala by escaping from the intrinsic AlaRS proofreading activity (Figure 1).
Furthermore, BMAA inhibits the cognate amino acid activation, the editing functions of AlaRS, and the deacylation activity of HsAlaRS on Ser-tRNAAla [72]. The AlaRS possesses canonical and non-canonical cellular functions and is predominantly linked to loss in neuronal cells and neurodegenerative disorders in human and mouse models [67,343]. Moreover, using transcriptomic analysis, Wang and co-authors [348] confirmed that BMAA could alter the expression of major genes encoding components related to translation in prokaryotes (diazotrophic algae Anabaena).
We hypothesize that the ability of BMAA and other NCAA and cyanopeptides to affect protein homeostasis may have ancient evolutionary origins, initially serving to hinder the growth of neighboring microalgae in plankton communities. The inhibition of cell growth and progression in the cell cycle of eukaryotic cells was demonstrated in in vitro experiments [349]. The production of cyclic peptides, including non-proteinogenic amino acids, leads to the lysis of cyanobacteria and may be an effective control mechanism of cyanobacterial density during algal blooms [137]. A Trojan Horse entry of non-proteinogenic amino acids from cyanobacteria to eukaryotic proteins may be a more general event and not only limited by BMAA and may be reflected in associations with ND diseases (Figure 2).

6. Conclusions

The structural variety of cyanotoxins and cyanopeptides is produced during cyanobacterial blooms. Many structural aspects of key metabolites involved in the cyanotoxins pathways have yet to be elucidated. However, it is becoming clear that non-proteinogenic amino acids, free-existing or initially a part of cyanopeptides, may affect protein homeostasis and lead to mistranslation and misfolding of proteins in eukaryotic cells, building a link to ND development, including Parkinson’s and Alzheimer’s diseases. The gut symbiotic microorganisms become affected, and the development of chronic dysbiosis may increase gut and BBB permeability. Many aspects pertaining to the regulation, role, and function of cyanobacterial compounds also requires the development of innovative detection approaches. This knowledge may be harnessed to identify novel biomarkers for ND and new targets for interventions.

Author Contributions

G.N. and N.S.B. wrote an original draft and drew figures, E.D.P., E.F.-K. and S.A. edited the manuscript. S.A., E.F.-K. and N.S.B. contributed to the funding of the research. All authors have read and agreed to the published version of the manuscript.

Funding

This research received finding from Nazarbayev University (PURE ID: 16482715 and OPCRP2020018) and Kazakhstan MES grant AP14872088.

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.

Acknowledgments

We are thankful to Dmitry Malashenkov, Nazarbayev University for helpful discussion. The manuscript was improved by insightful comments of anonymous reviewers.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, S.A.; Mihali, T.K.; Neilan, B.A. Extraordinary conservation, gene loss, and positive selection in the evolution of an ancient neurotoxin. Mol. Biol. Evol. 2011, 28, 1173–1182. [Google Scholar] [CrossRef] [Green Version]
  2. Chorus, I.; Welker, M. Toxic Cyanobacteria in Water: A Guide to Their Public Health Consequences, Monitoring and Management, 2nd ed.; Taylor & Francis: Boca Raton, FL, USA, 2021. [Google Scholar] [CrossRef]
  3. Kozlowsky-Suzuki, B.; Wilson, A.E.; da Silva Ferrao-Filho, A. Biomagnification or biodilution of microcystins in aquatic foodwebs? Meta-analyses of laboratory and field studies. Harmful Algae 2012, 18, 47–55. [Google Scholar] [CrossRef]
  4. Pybus, M.; Hobson, D.; Onderka, D. Mass mortality of bats due to probable blue-green algal toxicity. J. Wildl. Dis. 1986, 22, 449–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mahmood, N.A.; Carmichael, W.W.; Pfahler, D. Anticholinesterase poisonings in dogs from a cyanobacterial (blue-green algae) bloom dominated by Anabaena flos-aquae. Am. J. Vet. Res. 1988, 49, 500–503. [Google Scholar]
  6. Henriksen, P.; Carmichael, W.W.; An, J.; Moestrup, O. Detection of an anatoxin-a (s)-like anticholinesterase in natural blooms and cultures of cyanobacteria/blue-green algae from Danish lakes and in the stomach contents of poisoned birds. Toxicon 1997, 35, 901–913. [Google Scholar] [CrossRef] [PubMed]
  7. Mez, K.; Beattie, K.A.; Codd, G.A.; Hanselmann, K.; Hauser, B.; Naegeli, H.; Preisig, H.R. Identification of a microcystin in benthic cyanobacteria linked to cattle deaths on alpine pastures in Switzerland. Eur. J. Phycol. 1997, 32, 111–117. [Google Scholar] [CrossRef]
  8. Saker, M.; Thomas, A.; Norton, J. Cattle mortality attributed to the toxic cyanobacterium Cylindrospermopsis raciborskii in an outback region of north Queensland. Environ. Toxicol. 1999, 14, 179–182. [Google Scholar] [CrossRef]
  9. Codd, G.A.; Lindsay, J.; Young, F.M.; Morrison, L.F.; Metcalf, J.S. Harmful cyanobacteria: From mass mortalities to management measures. In Harmful Cyanobacteria; Huisman, J., Matthijs, H.C.P., Visser, P.M., Eds.; Springer: Dordrecht, The Netherlands, 2005; pp. 1–23. [Google Scholar] [CrossRef]
  10. Miller, M.A.; Kudela, R.M.; Mekebri, A.; Crane, D.; Oates, S.C.; Tinker, M.T.; Staedler, M.; Miller, W.A.; Toy-Choutka, S.; Dominik, C.; et al. Evidence for a novel marine harmful algal bloom: Cyanotoxin (microcystin) transfer from land to sea otters. PLoS ONE 2010, 5, e12576. [Google Scholar] [CrossRef] [PubMed]
  11. Chatziefthimiou, A.D.; Richer, R.; Rowles, H.; Powell, J.T.; Metcalf, J.S. Cyanotoxins as a potential cause of dog poisonings in desert environments. Vet. Rec. 2014, 174, 484–485. [Google Scholar] [CrossRef]
  12. Danil, K.; Berman, M.; Frame, E.; Preti, A.; Fire, S.E.; Leighfield, T.; Carretta, J.; Carter, M.L.; Lefebvre, K. Marine algal toxins and their vectors in southern California cetaceans. Harmful Algae 2021, 103, 102000. [Google Scholar] [CrossRef] [PubMed]
  13. Rattner, B.A.; Wazniak, C.E.; Lankton, J.S.; McGowan, P.C.; Drovetski, S.V.; Egerton, T.A. Review of harmful algal bloom effects on birds with implications for avian wildlife in the Chesapeake Bay region. Harmful Algae 2022, 120, 102319. [Google Scholar] [CrossRef]
  14. Pouria, S.; de Andrade, A.; Barbosa, J.; Cavalcanti, R.; Barreto, V.; Ward, C.; Preiser, W.; Poon, G.K.; Neild, G.; Codd, G. Fatal microcystin intoxication in haemodialysis unit in Caruaru, Brazil. Lancet 1998, 352, 21–26. [Google Scholar] [CrossRef] [PubMed]
  15. Carmichael, W.W.; Azevedo, S.; An, J.S.; Molica, R.; Jochimsen, E.M.; Lau, S.; Rinehart, K.L.; Shaw, G.R.; Eaglesham, G.K. Human fatalities from cyanobacteria: Chemical and biological evidence for cyanotoxins. Environ. Health Perspect. 2001, 109, 663–668. [Google Scholar] [CrossRef]
  16. Briand, J.-F.; Jacquet, S.; Bernard, C.; Humbert, J.-F. Health hazards for terrestrial vertebrates from toxic cyanobacteria in surface water ecosystems. Vet. Res. 2003, 34, 361–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. McLellan, N.L.; Manderville, R.A. Toxic mechanisms of microcystins in mammals. Toxicol. Res. 2017, 6, 391–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. White, S.H.; Duivenvoorden, L.J.; Fabbro, L.D. A decision-making framework for ecological impacts associated with the accumulation of cyanotoxins (cylindrospermopsin and microcystin). Lakes Reserv. Res. Manag. 2005, 10, 25–37. [Google Scholar] [CrossRef]
  19. Paerl, H.W.; Huisman, J. Blooms like it hot. Science 2008, 320, 57–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Moe, S.J.; Haande, S.; Couture, R.-M. Climate change, cyanobacteria blooms and ecological status of lakes: A Bayesian network approach. Ecol. Model. 2016, 337, 330–347. [Google Scholar] [CrossRef] [Green Version]
  21. Huisman, J.; Codd, G.A.; Paerl, H.W.; Ibelings, B.W.; Verspagen, J.M.; Visser, P.M. Cyanobacterial blooms. Nat. Rev. Microbiol. 2018, 16, 471–483. [Google Scholar] [CrossRef]
  22. Taranu, Z.E.; Gregory-Eaves, I.; Leavitt, P.R.; Bunting, L.; Buchaca, T.; Catalan, J.; Domaizon, I.; Guilizzoni, P.; Lami, A.; McGowan, S. Acceleration of cyanobacterial dominance in north temperate-subarctic lakes during the Anthropocene. Ecol. Lett. 2015, 18, 375–384. [Google Scholar] [CrossRef] [PubMed]
  23. Trout-Haney, J.V.; Wood, Z.T.; Cottingham, K.L. Presence of the cyanotoxin microcystin in arctic lakes of Southwestern Greenland. Toxins 2016, 8, 256. [Google Scholar] [CrossRef] [Green Version]
  24. Ho, J.C.; Michalak, A.M.; Pahlevan, N. Widespread global increase in intense lake phytoplankton blooms since the 1980s. Nature 2019, 574, 667–670. [Google Scholar] [CrossRef] [PubMed]
  25. Gobler, C.J. Climate change and harmful algal blooms: Insights and perspective. Harmful Algae 2020, 91, 101731. [Google Scholar] [CrossRef]
  26. Bowling, L.; Baker, P. Major cyanobacterial bloom in the Barwon-Darling River, Australia, in 1991, and underlying limnological conditions. Mar. Freshw. Res. 1996, 47, 643–657. [Google Scholar] [CrossRef]
  27. Heisler, J.; Glibert, P.M.; Burkholder, J.M.; Anderson, D.M.; Cochlan, W.; Dennison, W.C.; Dortch, Q.; Gobler, C.J.; Heil, C.A.; Humphries, E.; et al. Eutrophication and harmful algal blooms: A scientific consensus. Harmful Algae 2008, 8, 3–13. [Google Scholar] [CrossRef] [Green Version]
  28. World Health Organization. Cyanobacterial Toxins: Microcystin-LR in Drinking-Water; No. WHO/SDE/WSH/03.04/57; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  29. Andersson, M.; Karlsson, O.; Banack, S.A.; Brandt, I. Transfer of developmental neurotoxin β-N-methylamino-L-alanine (BMAA) via milk to nursed offspring: Studies by mass spectrometry and image analysis. Toxicol. Lett. 2016, 258, 108–114. [Google Scholar] [CrossRef] [PubMed]
  30. Kulpys, J.; Paulauskas, E.; Pilipavicius, V.; Stankevicius, R. Influence of cyanobacteria Arthrospira (Spirulina) platensis biomass additives towards the body condition of lactation cows and biochemical milk indexes. Agric. Res. 2009, 7, 823–835. [Google Scholar]
  31. Christaki, E.; Florou-Paneri, P.; Bonos, E. Microalgae: A novel ingredient in nutrition. Int. J. Food Sci. 2011, 62, 794–799. [Google Scholar] [CrossRef]
  32. McGorum, B.C.; Pirie, R.S.; Glendinning, L.; McLachlan, G.; Metcalf, J.S.; Banack, S.A.; Cox, P.A.; Codd, G.A. Grazing livestock are exposed to terrestrial cyanobacteria. Vet. Res. 2015, 46, 16. [Google Scholar] [CrossRef] [Green Version]
  33. Andersson, M.; Karlsson, O.; Bergström, U.; Brittebo, E.B.; Brandt, I. Maternal transfer of the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) via milk to suckling offspring. PLoS ONE 2013, 8, e78133. [Google Scholar] [CrossRef] [Green Version]
  34. Nunn, P.B. 50 years of research on α-amino-β-methylaminopropionic acid (β-methylaminoalanine). Phytochemistry 2017, 144, 271–281. [Google Scholar] [CrossRef] [PubMed]
  35. Andersson, M.; Karlsson, O.; Brandt, I. The environmental neurotoxin β-N-methylamino-l-alanine (l-BMAA) is deposited into birds’ eggs. Ecotoxicol. Environ. Saf. 2018, 147, 720–724. [Google Scholar] [CrossRef] [PubMed]
  36. Puddick, J.; van Ginkel, R.; Page, C.D.; Murray, J.S.; Greenhough, H.E.; Bowater, J.; Selwood, A.I.; Wood, S.A.; Prinsep, M.R.; Truman, P. Acute toxicity of dihydroanatoxin-a from Microcoleus autumnalis in comparison to anatoxin-a. Chemosphere 2021, 263, 127937. [Google Scholar] [CrossRef]
  37. Sano, T.; Takagi, H.; Morrison, L.F.; Metcalf, J.S.; Codd, G.A.; Kaya, K. Leucine aminopeptidase M inhibitors, cyanostatin A and B, isolated from cyanobacterial water blooms in Scotland. Phytochemistry 2005, 66, 543–548. [Google Scholar] [CrossRef] [PubMed]
  38. Monteiro, P.R.; do Amaral, S.C.; Siqueira, A.S.; Xavier, L.P.; Santos, A.V. Anabaenopeptins: What we know so far. Toxins 2021, 13, 522. [Google Scholar] [CrossRef] [PubMed]
  39. Bownik, A.; Adamczuk, M.; Pawlik-Skowrońska, B. Behavioral disturbances induced by cyanobacterial oligopeptides microginin-FR1, anabaenopeptin-A and microcystin-LR are associated with neuromotoric and cytotoxic changes in Brachionus calyciflorus. J. Hazard. Mater. 2022, 438, 129472. [Google Scholar] [CrossRef]
  40. Metcalf, J.S.; Tischbein, M.; Cox, P.A.; Stommel, E.W. Cyanotoxins and the nervous system. Toxins 2021, 13, 660. [Google Scholar] [CrossRef]
  41. Breinlinger, S.; Phillips, T.J.; Haram, B.N.; Mareš, J.; Martínez Yerena, J.A.; Hrouzek, P.; Sobotka, R.; Henderson, W.M.; Schmieder, P.; Williams, S.M. Hunting the eagle killer: A cyanobacterial neurotoxin causes vacuolar myelinopathy. Science 2021, 371, eaax9050. [Google Scholar] [CrossRef]
  42. Bouaïcha, N.; Miles, C.O.; Beach, D.G.; Labidi, Z.; Djabri, A.; Benayache, N.Y.; Nguyen-Quang, T. Structural diversity, characterization and toxicology of microcystins. Toxins 2019, 11, 714. [Google Scholar] [CrossRef] [Green Version]
  43. Carmichael, W.W.; Beasley, V.; Bunner, D.L.; Eloff, J.N.; Falconer, I.; Gorham, P.; Harada, K.-I.; Krishnamurthy, T.; Min-Juan, Y.; Moore, R.E.; et al. Naming of cyclic heptapeptide toxins of cyanobacteria (blue-green algae). Toxicon 1988, 26, 971–973. [Google Scholar] [CrossRef] [PubMed]
  44. Díez-Quijada, L.; Prieto, A.I.; Guzmán-Guillén, R.; Jos, A.; Cameán, A.M. Occurrence and toxicity of microcystin congeners other than MC-LR and MC-RR: A review. Food Chem. Toxicol. 2019, 125, 106–132. [Google Scholar] [CrossRef] [PubMed]
  45. MacKintosh, C.; Beattie, K.A.; Klumpp, S.; Cohen, P.; Codd, G.A. Cyanobacterial microcystin-LR is a potent and specific inhibitor of protein phosphatases 1 and 2A from both mammals and higher plants. FEBS Lett. 1990, 264, 187–192. [Google Scholar] [CrossRef] [Green Version]
  46. Massey, I.Y.; Yang, F.; Ding, Z.; Yang, S.; Guo, J.; Al-Osman, M.; Kamegni, R.B.; Zeng, W. Exposure routes and health effects of microcystins on animals and humans: A mini-review. Toxicon 2018, 151, 156–162. [Google Scholar] [CrossRef]
  47. Hu, Y.; Chen, J.; Fan, H.; Xie, P.; He, J. A review of neurotoxicity of microcystins. Environ. Sci. Pollut. Res. 2016, 23, 7211–7219. [Google Scholar] [CrossRef] [PubMed]
  48. Tzima, E.; Serifi, I.; Tsikari, I.; Alzualde, A.; Leonardos, I.; Papamarcaki, T. Transcriptional and behavioral responses of zebrafish larvae to microcystin-LR exposure. Int. J. Mol. Sci. 2017, 18, 365. [Google Scholar] [CrossRef] [Green Version]
  49. Pašková, V.; Adamovský, O.; Pikula, J.; Skočovská, B.; Band’ouchová, H.; Horáková, J.; Babica, P.; Maršálek, B.; Hilscherová, K. Detoxification and oxidative stress responses along with microcystins accumulation in Japanese quail exposed to cyanobacterial biomass. Sci. Total Environ. 2008, 398, 34–47. [Google Scholar] [CrossRef]
  50. Zhao, S.; Li, G.; Chen, J. A proteomic analysis of prenatal transfer of microcystin-LR induced neurotoxicity in rat offspring. J. Proteom. 2015, 114, 197–213. [Google Scholar] [CrossRef] [PubMed]
  51. Feurstein, D.; Stemmer, K.; Kleinteich, J.; Speicher, T.; Dietrich, D.R. Microcystin congener–and concentration-dependent induction of murine neuron apoptosis and neurite degeneration. Toxicol. Sci. 2011, 124, 424–431. [Google Scholar] [CrossRef] [Green Version]
  52. Cox, P.A.; Banack, S.A.; Murch, S.J.; Rasmussen, U.; Tien, G.; Bidigare, R.R.; Metcalf, J.S.; Morrison, L.F.; Codd, G.A.; Bergman, B. Diverse taxa of cyanobacteria produce β-N-methylamino-L-alanine, a neurotoxic amino acid. Proc. Natl. Acad. Sci. USA 2005, 102, 5074–5078. [Google Scholar] [CrossRef] [Green Version]
  53. Violi, J.P.; Mitrovic, S.M.; Colville, A.; Main, B.J.; Rodgers, K.J. Prevalence of β-methylamino-L-alanine (BMAA) and its isomers in freshwater cyanobacteria isolated from eastern Australia. Ecotoxicol. Environ. Saf. 2019, 172, 72–81. [Google Scholar] [CrossRef] [PubMed]
  54. Schneider, T.; Simpson, C.; Desai, P.; Tucker, M.; Lobner, D. Neurotoxicity of isomers of the environmental toxin L-BMAA. Toxicon 2020, 184, 175–179. [Google Scholar] [CrossRef] [PubMed]
  55. Martin, R.M.; Bereman, M.S.; Marsden, K.C. The cyanotoxin 2, 4-DAB reduces viability and causes behavioral and molecular dysfunctions associated with neurodegeneration in larval zebrafish. Neurotox. Res. 2022, 40, 347–364. [Google Scholar] [CrossRef] [PubMed]
  56. Chernoff, N.; Hill, D.J.; Diggs, D.L.; Faison, B.D.; Francis, B.M.; Lang, J.R.; Larue, M.M.; Le, T.-T.; Loftin, K.A.; Lugo, J.N.; et al. A critical review of the postulated role of the non-essential amino acid, β-N-Methylamino-L-Alanine, in neurodegenerative disease in humans. J. Toxicol. Environ. Health Part B 2017, 20, 183–229. [Google Scholar] [CrossRef] [PubMed]
  57. Dunlop, R.A.; Banack, S.A.; Bishop, S.L.; Metcalf, J.S.; Murch, S.J.; Davis, D.A.; Stommel, E.W.; Karlsson, O.; Brittebo, E.B.; Chatziefthimiou, A.D.; et al. Is exposure to BMAA a risk factor for neurodegenerative diseases? A response to a critical review of the BMAA hypothesis. Neurotox. Res. 2021, 39, 81–106. [Google Scholar] [CrossRef]
  58. Sini, P.; Dang, T.B.C.; Fais, M.; Galioto, M.; Padedda, B.M.; Lugliè, A.; Iaccarino, C.; Crosio, C. Cyanobacteria, cyanotoxins, and neurodegenerative diseases: Dangerous liaisons. Inter. J. Mol. Sci. 2021, 22, 8726. [Google Scholar] [CrossRef] [PubMed]
  59. Courtier, A.; Potheret, D.; Giannoni, P. Environmental bacteria as triggers to brain disease: Possible mechanisms of toxicity and associated human risk. Life Sci. 2022, 304, 120689. [Google Scholar] [CrossRef]
  60. Faassen, E.J. Presence of the neurotoxin BMAA in aquatic ecosystems: What do we really know? Toxins 2014, 6, 1109–1138. [Google Scholar] [CrossRef] [Green Version]
  61. Lopicic, S.; Svirčev, Z.; Malešević, T.P.; Kopitović, A.; Ivanovska, A.; Meriluoto, J. Environmental neurotoxin β-N-Methylamino-L-alanine (BMAA) as a widely occurring putative pathogenic factor in neurodegenerative diseases. Microorganisms 2022, 10, 2418. [Google Scholar] [CrossRef]
  62. Nunes-Costa, D.; Magalhães, J.D.; G-Fernandes, M.; Cardoso, S.M.; Empadinhas, N. Microbial BMAA and the pathway for Parkinson’s disease neurodegeneration. Front. Aging Neurosci. 2020, 12, 26. [Google Scholar] [CrossRef] [Green Version]
  63. Dunlop, R.A.; Cox, P.A.; Banack, S.A.; Rodgers, K.J. The non-protein amino acid BMAA is misincorporated into human proteins in place of L-serine causing protein misfolding and aggregation. PLoS ONE 2013, 8, e75376. [Google Scholar] [CrossRef] [Green Version]
  64. Frøyset, A.K.; Khan, E.A.; Fladmark, K.E. Quantitative proteomics analysis of zebrafish exposed to sub-lethal dosages of β-methyl-amino-L-alanine (BMAA). Sci. Rep. 2016, 6, 29631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Cox, P.A.; Davis, D.A.; Mash, D.C.; Metcalf, J.S.; Banack, S.A. Dietary exposure to an environmental toxin triggers neurofibrillary tangles and amyloid deposits in the brain. Proc. Royal Soc. B Biol. Sci. 2016, 283, 20152397. [Google Scholar] [CrossRef] [Green Version]
  66. Haase, C.; Stieler, J.; Arendt, T.; Holzer, M. Pseudophosphorylation of tau protein alters its ability for self-aggregation. J. Neurochem. 2004, 88, 1509–1520. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, J.W.; Beebe, K.; Nangle, L.A.; Jang, J.; Longo-Guess, C.M.; Cook, S.A.; Davisson, M.T.; Sundberg, J.P.; Schimmel, P.; Ackerman, S.L. Editing-defective tRNA synthetase causes protein misfolding and neurodegeneration. Nature 2006, 443, 50–55. [Google Scholar] [CrossRef] [PubMed]
  68. De Munck, E.; Munoz-Saez, E.; Miguel, B.G.; Solas, M.T.; Ojeda, I.; Martinez, A.; Gil, C.; Arahuetes, R.M. β-N-methylamino-l-alanine causes neurological and pathological phenotypes mimicking Amyotrophic Lateral Sclerosis (ALS): The first step towards an experimental model for sporadic ALS. Environ. Toxicol. Pharmacol. 2013, 36, 243–255. [Google Scholar] [CrossRef] [Green Version]
  69. Muñoz-Saez, E.; de Munck, E.; Arahuetes, R.M.; Solas, M.T.; Martínez, A.M.; Miguel, B.G. β-N-methylamino-L-alanine induces changes in both GSK3 and TDP-43 in human neuroblastoma. J. Toxicol. Sci. 2013, 38, 425–430. [Google Scholar] [CrossRef] [Green Version]
  70. Karlsson, O.; Jiang, L.; Ersson, L.; Malmström, T.; Ilag, L.L.; Brittebo, E.B. Environmental neurotoxin interaction with proteins: Dose-dependent increase of free and protein-associated BMAA (β-N-methylamino-L-alanine) in neonatal rat brain. Sci. Rep. 2015, 5, 15570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Xie, X.; Basile, M.; Mash, D.C. Cerebral uptake and protein incorporation of cyanobacterial toxin β-N-methylamino-L-alanine. Neuroreport 2013, 24, 779–784. [Google Scholar] [CrossRef]
  72. Han, N.-C.; Bullwinkle, T.J.; Loeb, K.F.; Faull, K.F.; Mohler, K.; Rinehart, J.; Ibba, M. The mechanism of β-N-methylamino-l-alanine inhibition of tRNA aminoacylation and its impact on misincorporation. J. Biol. Chem. 2020, 295, 1402–1410. [Google Scholar] [CrossRef]
  73. Rochet, J.-C. Errors in translation cause selective neurodegeneration. ACS Chem. Biol. 2006, 1, 562–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Carmichael, W.W.; Biggs, D.F.; Peterson, M.A. Pharmacology of anatoxin-a, produced by the freshwater cyanophyte Anabaena flos-aquae NRC-44-1. Toxicon 1979, 17, 229–236. [Google Scholar] [CrossRef]
  75. Christensen, V.G.; Khan, E. Freshwater neurotoxins and concerns for human, animal, and ecosystem health: A review of anatoxin-a and saxitoxin. Sci. Total Environ. 2020, 736, 139515. [Google Scholar] [CrossRef] [PubMed]
  76. Fiore, M.F.; de Lima, S.T.; Carmichael, W.W.; McKinnie, S.M.; Chekan, J.R.; Moore, B.S. Guanitoxin, re-naming a cyanobacterial organophosphate toxin. Harmful Algae 2020, 92, 101737. [Google Scholar] [CrossRef] [PubMed]
  77. Metcalf, J.; Richer, R.; Cox, P.; Codd, G. Cyanotoxins in desert environments may present a risk to human health. Sci. Total Environ. 2012, 421, 118–123. [Google Scholar] [CrossRef]
  78. Metcalf, J.; Banack, S.; Richer, R.; Cox, P. Neurotoxic amino acids and their isomers in desert environments. J. Arid. Environ. 2015, 112, 140–144. [Google Scholar] [CrossRef]
  79. Backer, L.C.; Manassaram-Baptiste, D.; LePrell, R.; Bolton, B. Cyanobacteria and algae blooms: Review of health and environmental data from the harmful algal bloom-related illness surveillance system (HABISS) 2007–2011. Toxins 2015, 7, 1048–1064. [Google Scholar] [CrossRef] [Green Version]
  80. Carmichael, W.W.; Boyer, G.L. Health impacts from cyanobacteria harmful algae blooms: Implications for the North American Great Lakes. Harmful Algae 2016, 54, 194–212. [Google Scholar] [CrossRef] [PubMed]
  81. Ohtani, I.; Moore, R.E.; Runnegar, M.T. Cylindrospermopsin: A potent hepatotoxin from the blue-green alga Cylindrospermopsis raciborskii. J. Amer. Chem. Soc. 1992, 114, 7941–7942. [Google Scholar] [CrossRef]
  82. Rzymski, P.; Poniedziałek, B. In search of environmental role of cylindrospermopsin: A review on global distribution and ecology of its producers. Wat. Res. 2014, 66, 320–337. [Google Scholar] [CrossRef]
  83. Chen, G.; Wang, L.; Wang, M.; Hu, T. Comprehensive insights into the occurrence and toxicological issues of nodularins. Mar. Pollut. Bull. 2021, 162, 111884. [Google Scholar] [CrossRef]
  84. Lehtonen, K.K.; Kankaanpää, H.; Leiniö, S.; Sipiä, V.O.; Pflugmacher, S.; Sandberg-Kilpi, E. Accumulation of nodularin-like compounds from the cyanobacterium Nodularia spumigena and changes in acetylcholinesterase activity in the clam Macoma balthica during short-term laboratory exposure. Aquat. Toxicol. 2003, 64, 461–476. [Google Scholar] [CrossRef]
  85. Kankaanpää, H.; Leiniö, S.; Olin, M.; Sjövall, O.; Meriluoto, J.; Lehtonen, K.K. Accumulation and depuration of cyanobacterial toxin nodularin and biomarker responses in the mussel Mytilus edulis. Chemosphere 2007, 68, 1210–1217. [Google Scholar] [CrossRef]
  86. Christensen, V.G.; Olds, H.T.; Norland, J.; Khan, E. Phytoplankton community interactions and cyanotoxin mixtures in three recurring surface blooms within one lake. J. Hazard. Mater. 2022, 427, 128142. [Google Scholar] [CrossRef]
  87. Martin, R.M.; Stallrich, J.; Bereman, M.S. Mixture designs to investigate adverse effects upon co-exposure to environmental cyanotoxins. Toxicology 2019, 421, 74–83. [Google Scholar] [CrossRef] [PubMed]
  88. Martin, R.M.; Bereman, M.S.; Marsden, K.C. BMAA and MCLR interact to modulate behavior and exacerbate molecular changes related to neurodegeneration in larval zebrafish. Toxicol. Sci. 2021, 179, 251–261. [Google Scholar] [CrossRef]
  89. Roy-Lachapelle, A.; Duy, S.V.; Munoz, G.; Dinh, 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, 5289–5300. [Google Scholar] [CrossRef] [Green Version]
  90. Janssen, E.M.-L. Cyanobacterial peptides beyond microcystins—A review on co-occurrence, toxicity, and challenges for risk assessment. Water Res. 2019, 151, 488–499. [Google Scholar] [CrossRef] [PubMed]
  91. Natumi, R.; Janssen, E.M.-L. Cyanopeptide co-production dynamics beyond mirocystins and effects of growth stages and nutrient availability. Environ. Sci. Technol. 2020, 54, 6063–6072. [Google Scholar] [CrossRef] [PubMed]
  92. Nagarajan, M.; Maruthanayagam, V.; Sundararaman, M. SAR analysis and bioactive potentials of freshwater and terrestrial cyanobacterial compounds: A review. J. Appl. Toxicol. 2013, 33, 313–349. [Google Scholar] [CrossRef] [PubMed]
  93. Caban-Holt, A.; Mattingly, M.; Cooper, G.; Schmitt, F.A. Neurodegenerative memory disorders: A potential role of environmental toxins. Neurol. Clin. 2005, 23, 485–521. [Google Scholar] [CrossRef]
  94. Shaw, C.A.; Höglinger, G.U. Neurodegenerative diseases: Neurotoxins as sufficient etiologic agents? Neuromolecular. Med. 2008, 10, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Caller, T.A.; Doolin, J.W.; Haney, J.F.; Murby, A.J.; West, K.G.; Farrar, H.E.; Ball, A.; Harris, B.T.; Stommel, E.W. A cluster of amyotrophic lateral sclerosis in New Hampshire: A possible role for toxic cyanobacteria blooms. Amyotrop. Lateral Scler. 2009, 10, 101–108. [Google Scholar] [CrossRef]
  96. Banack, S.A.; Caller, T.; Henegan, P.; Haney, J.; Murby, A.; Metcalf, J.S.; Powell, J.; Alan, P.; Stommel, E. Detection of cyanotoxins, β-N-methylamino-l-alanine and microcystins, from a lake surrounded by cases of amyotrophic lateral sclerosis. Toxins 2015, 7, 322–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Torbick, N.; Ziniti, B.; Stommel, E.; Linder, E.; Andrew, A.; Caller, T.; Haney, J.; Bradley, W.; Henegan, P.L.; Shi, X. Assessing cyanobacterial harmful algal blooms as risk factors for amyotrophic lateral sclerosis. Neurotox. Res. 2018, 33, 199–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Stanhope, J.M.; Brody, J.A.; Morris, C.E. Epidemiologic features of amyotrophic lateral sclerosis and parkinsonism-dementia in Guam, Mariana Islands. Int. J. Epidemiol. 1972, 1, 199–210. [Google Scholar] [CrossRef] [PubMed]
  99. Cox, P.A.; Sacks, O.W. Cycad neurotoxins, consumption of flying foxes, and ALS-PDC disease in Guam. Neurology 2002, 58, 956–959. [Google Scholar] [CrossRef] [PubMed]
  100. Yanagihara, R.T.; Garruto, R.M.; Gajdusek, D.C. Epidemiological surveillance of amyotrophic lateral sclerosis and parkinsonism-dementia in the commonwealth of the Northern Mariana Islands. Ann. Neurol. 1983, 13, 79–86. [Google Scholar] [CrossRef]
  101. Cox, P.A.; Banack, S.; Murch, S.J. Biomagnification of cyanobacterial neurotoxins and neurodegenerative disease among the Chamorro people of Guam. Proc. Natl. Acad. Sci. USA 2003, 100, 13380–13383. [Google Scholar] [CrossRef] [Green Version]
  102. Murch, S.J.; Cox, P.A.; Banack, S.A.; Steele, J.C.; Sacks, O.W. Occurrence of β-methylamino-l-alanine (BMAA) in. ALS/PDC patients from Guam. Acta Neurol. Scand. 2004, 110, 267–269. [Google Scholar] [CrossRef] [PubMed]
  103. Kimura, K.; Yase, Y.; Higashi, Y.; Yamamoto, K.; Sugiura, M.; Tsumoto, T.; Uno, S.; Yoshimura, S.; Namikawa, K.; Kumura, J.; et al. Endemiological and geomedical studies on amyotrophic lateral sclerosis and allied diseases in Kii Peninsula, Japan (preliminary report). Psychiatry Clin. Neurosci. 1961, 15, 175–181. [Google Scholar] [CrossRef] [PubMed]
  104. Yase, Y. The pathogenesis of amyotrophic lateral sclerosis. Lancet 1972, 300, 292–296. [Google Scholar] [CrossRef] [PubMed]
  105. Gunnarsson, L.-G.; Lygner, P.-E.; Veiga-Cabo, J.; de Pedro-Cuesta, J. An epidemic-like cluster of motor neuron disease in a Swedish county during the period 1973–1984. Neuroepidemiology 1996, 15, 142–152. [Google Scholar] [CrossRef]
  106. Sienko, D.G.; Davis, J.P.; Taylor, J.A.; Brooks, B.R. Amyotrophic lateral sclerosis: A case-control study following detection of a cluster in a small Wisconsin community. Arch. Neurol. 1990, 47, 38–41. [Google Scholar] [CrossRef] [PubMed]
  107. Corcia, P.; Jafari-Schluep, H.-F.; Lardillier, D.; Mazyad, H.; Giraud, P.; Clavelou, P.; Pouget, J.; Camu, W. A clustering of conjugal amyotrophic lateral sclerosis in southeastern France. Arch. Neurol. 2003, 60, 553–557. [Google Scholar] [CrossRef] [Green Version]
  108. Uccelli, R.; Binazzi, A.; Altavista, P.; Belli, S.; Comba, P.; Mastrantonio, M.; Vanacore, N. Geographic distribution of amyotrophic lateral sclerosis through motor neuron disease mortality data. Eur. J. Epidemiol. 2007, 22, 781–790. [Google Scholar] [CrossRef] [PubMed]
  109. Sabel, C.E.; Boyle, P.J.; Löytönen, M.; Gatrell, A.C.; Jokelainen, M.; Flowerdew, R.; Maasilta, P. Spatial clustering of amyotrophic lateral sclerosis in Finland at place of birth and place of death. Am. J. Epidemiol. 2003, 157, 898–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Horner, R.D.; Grambow, S.C.; Coffman, C.J.; Lindquist, J.H.; Oddone, E.Z.; Allen, K.D.; Kasarskis, E.J. Amyotrophic lateral sclerosis among 1991 Gulf War veterans: Evidence for a time-limited outbreak. Neuroepidemiology 2008, 31, 28–32. [Google Scholar] [CrossRef] [PubMed]
  111. Miranda, M.L.; Alicia Overstreet Galeano, M.; Tassone, E.; Allen, K.D.; Horner, R.D. Spatial analysis of the etiology of amyotrophic lateral sclerosis among 1991 Gulf War veterans. Neurotoxicology 2008, 29, 964–970. [Google Scholar] [CrossRef]
  112. Stommel, E.W.; Field, N.C.; Caller, T.A. Aerosolization of cyanobacteria as a risk factor for amyotrophic lateral sclerosis. Med. Hypotheses 2013, 80, 142–145. [Google Scholar] [CrossRef]
  113. Masseret, E.; Banack, S.; Boumédiène, F.; Abadie, E.; Brient, L.; Pernet, F.; Juntas-Morales, R.; Pageot, N.; Metcalf, J.; Cox, P. Dietary BMAA exposure in an amyotrophic lateral sclerosis cluster from southern France. PLoS ONE 2013, 8, e83406. [Google Scholar] [CrossRef] [Green Version]
  114. Caller, T.A.; Chipman, J.W.; Field, N.C.; Stommel, E.W. Spatial analysis of amyotrophic lateral sclerosis in Northern New England, USA, 1997–2009. Muscle Nerve 2013, 48, 235–241. [Google Scholar] [CrossRef] [PubMed]
  115. Torbick, N.; Hession, S.; Stommel, E.; Caller, T. Mapping amyotrophic lateral sclerosis lake risk factors across northern New England. Int. J. Health Geogr. 2014, 13, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Delzor, A.; Couratier, P.; Boumédiène, F.; Nicol, M.; Druet-Cabanac, M.; Paraf, F.; Méjean, A.; Ploux, O.; Leleu, J.-P.; Brient, L.; et al. Searching for a link between the L-BMAA neurotoxin and amyotrophic lateral sclerosis: A study protocol of the French BMAA-LS programme. BMJ Open 2014, 4, e005528. [Google Scholar] [CrossRef]
  117. Lee, S.; Choi, B.; Kim, S.J.; Kim, J.; Kang, D.; Lee, J. Relationship between freshwater harmful algal blooms and neurodegenerative disease incidence rates in South Korea. Environ. Health 2022, 21, 116. [Google Scholar] [CrossRef]
  118. Lannuzel, A.; Mecharles, S.; Tressières, B.; Demoly, A.; Alhendi, R.; Hédreville-Tablon, M.-A.; Alecu, C. Clinical varieties and epidemiological aspects of amyotrophic lateral sclerosis in the Caribbean island of Guadeloupe: A new focus of ALS associated with Parkinsonism. Amyotroph. Lateral Scler. Front. Degener. 2015, 16, 216–223. [Google Scholar] [CrossRef] [PubMed]
  119. Filippini, T.; Fiore, M.; Tesauro, M.; Malagoli, C.; Consonni, M.; Violi, F.; Arcolin, E.; Iacuzio, L.; Conti, G.O.; Cristaldi, A.; et al. Clinical and lifestyle factors and risk of amyotrophic lateral sclerosis: A population-based case-control study. Int. J. Environ. Res. Public Health 2020, 17, 857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Field, N.C.; Metcalf, J.S.; Caller, T.A.; Banack, S.A.; Cox, P.A.; Stommel, E.W. Linking β-methylamino-L-alanine exposure to sporadic amyotrophic lateral sclerosis in Annapolis, MD. Toxicon 2013, 70, 179–183. [Google Scholar] [CrossRef]
  121. Berntzon, L.; Ronnevi, L.; Bergman, B.; Eriksson, J. Detection of BMAA in the human central nervous system. Neuroscience 2015, 292, 137–147. [Google Scholar] [CrossRef] [Green Version]
  122. Pablo, J.; Banack, S.; Cox, P.; Johnson, T.; Papapetropoulos, S.; Bradley, W.; Buck, A.; Mash, D. Cyanobacterial neurotoxin BMAA in ALS and Alzheimer’s disease. Acta Psychiatr. Scand. 2009, 120, 216–225. [Google Scholar] [CrossRef] [PubMed]
  123. Montine, T.J.; Li, K.; Perl, D.P.; Galasko, D. Lack of β-methylamino-L-alanine in brain from controls, AD, or Chamorros with PDC. Neurology 2005, 65, 768–769. [Google Scholar] [CrossRef] [PubMed]
  124. Meneely, J.P.; Chevallier, O.P.; Graham, S.; Greer, B.; Green, B.D.; Elliott, C.T. β-methylamino-L-alanine (BMAA) is not found in the brains of patients with confirmed Alzheimer’s disease. Sci. Rep. 2016, 6, 36363. [Google Scholar] [CrossRef] [PubMed]
  125. Spencer, P.S.; Palmer, V.S.; Kisby, G.E. Western Pacific ALS-PDC: Evidence implicating cycad genotoxins. J. Neurol. Sci. 2020, 419, 117185. [Google Scholar] [CrossRef] [PubMed]
  126. Jonasson, S.; Eriksson, J.; Berntzon, L.; Spáčil, Z.; Ilag, L.L.; Ronnevi, L.-O.; Rasmussen, U.; Bergman, B. Transfer of a cyanobacterial neurotoxin within a temperate aquatic ecosystem suggests pathways for human exposure. Proc. Natl. Acad. Sci. USA 2010, 107, 9252–9257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Jiang, L.; Eriksson, J.; Lage, S.; Jonasson, S.; Shams, S.; Mehine, M.; Ilag, L.L.; Rasmussen, U. Diatoms: A novel source for the neurotoxin BMAA in aquatic environments. PLoS ONE 2014, 9, e84578. [Google Scholar] [CrossRef]
  128. Johnson, H.E.; King, S.R.; Banack, S.A.; Webster, C.; Callanaupa, W.J.; Cox, P.A. Cyanobacteria (Nostoc commune) used as a dietary item in the Peruvian highlands produce the neurotoxic amino acid BMAA. J. Ethnopharmacol. 2008, 118, 159–165. [Google Scholar] [CrossRef]
  129. Cox, P.A.; Richer, R.; Metcalf, J.S.; Banack, S.A.; Codd, G.A.; Bradley, W.G. Cyanobacteria and BMAA exposure from desert dust: A possible link to sporadic ALS among Gulf War veterans. Amyotroph. Lateral Scler. 2009, 10, 109–117. [Google Scholar] [CrossRef] [PubMed]
  130. Lage, S.; Costa, P.R.; Moita, T.; Eriksson, J.; Rasmussen, U.; Rydberg, S.J. BMAA in shellfish from two Portuguese transitional water bodies suggests the marine dinoflagellate Gymnodinium catenatum as a potential BMAA source. Aquat. Toxicol. 2014, 152, 131–138. [Google Scholar] [CrossRef] [PubMed]
  131. Bradley, W.G.; Miller, R.; Levine, T.; Stommel, E.; Cox, P. Studies of environmental risk factors in amyotrophic lateral sclerosis (ALS) and a phase I clinical trial of L-serine. Neurotox. Res. 2018, 33, 192–198. [Google Scholar] [CrossRef]
  132. Saitou, T.; Sugiura, N.; Itayama, T.; Inamori, Y.; Matsumura, M. Degradation characteristics of microcystins by isolated bacteria from Lake Kasumigaura. J. Water Supply Res. Technol. AQUA 2003, 52, 13–18. [Google Scholar] [CrossRef]
  133. Edwards, C.; Graham, D.; Fowler, N.; Lawton, L.A. Biodegradation of microcystins and nodularin in freshwaters. Chemosphere 2008, 73, 1315–1321. [Google Scholar] [CrossRef] [PubMed]
  134. Gibble, C.M.; Peacock, M.B.; Kudela, R.M. Evidence of freshwater algal toxins in marine shellfish: Implications for human and aquatic health. Harmful Algae 2016, 59, 59–66. [Google Scholar] [CrossRef] [Green Version]
  135. Chiswell, R.K.; Shaw, G.R.; Eaglesham, G.; Smith, M.J.; Norris, R.L.; Seawright, A.A.; Moore, M.R. Stability of cylindrospermopsin, the toxin from the cyanobacterium, Cylindrospermopsis raciborskii: Effect of pH, temperature, and sunlight on decomposition. Environ. Toxicol. 1999, 14, 155–161. [Google Scholar] [CrossRef]
  136. Adamski, M.; Żmudzki, P.; Chrapusta, E.; Bober, B.; Kaminski, A.; Zabaglo, K.; Latkowska, E.; Bialczyk, J. Effect of pH and temperature on the stability of cylindrospermopsin. Characterization of decomposition products. Algal Res. 2016, 15, 129–134. [Google Scholar] [CrossRef]
  137. Sedmak, B.; Carmeli, S.; Eleršek, T. "Non-toxic" cyclic peptides induce lysis of cyanobacteria—An effective cell population density control mechanism in cyanobacterial blooms. Microb. Ecol. 2008, 56, 201–209. [Google Scholar] [CrossRef]
  138. Sedmak, B.; Sukenik, A.; Eleršek, T.; Kosi, G. The biological role of cyclic hepatotoxic and non-hepatotoxic cyanopeptides and its ecological consequences. In Ecotoxicology Research Developments; Santos, E.B., Ed.; Nova Science Pub Inc.: London, UK, 2009; pp. 169–300. [Google Scholar]
  139. Bionda, N. Cyclic Lipodepsipeptides as Lead Structures for the Discovery of New Antibiotics. Ph.D. Thesis, Florida Atlantic University, Boca Raton, FL, USA, 2013. [Google Scholar]
  140. Kurtz, T.; Zeng, T.; Rosario-Ortiz, F.L. Photodegradation of cyanotoxins in surface waters. Water Res. 2021, 192, 116804. [Google Scholar] [CrossRef] [PubMed]
  141. Li, H.; Xing, P.; Wu, Q.L. The high resilience of the bacterioplankton community in the face of a catastrophic disturbance by a heavy Microcystis bloom. FEMS Microbiol. Ecol. 2012, 82, 192–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Kormas, K.A.; Lymperopoulou, D.S. Cyanobacterial toxin degrading bacteria: Who are they? BioMed Res. Int. 2013, 2013, 463894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Van Wichelen, J.; Vanormelingen, P.; Codd, G.A.; Vyverman, W. The common bloom-forming cyanobacterium Microcystis is prone to a wide array of microbial antagonists. Harmful Algae 2016, 55, 97–111. [Google Scholar] [CrossRef]
  144. Mazur-Marzec, H.; Plinski, M. Do toxic cyanobacteria blooms pose a threat to the Baltic ecosystem? Oceanologia 2009, 51, 293–319. [Google Scholar] [CrossRef] [Green Version]
  145. Stolz, A. Molecular characteristics of xenobiotic-degrading sphingomonads. Appl. Microbiol. Biotechnol. 2009, 81, 793–811. [Google Scholar] [CrossRef] [PubMed]
  146. Kato, H.; Imanishi, S.Y.; Tsuji, K.; Harada, K.-I. Microbial degradation of cyanobacterial cyclic peptides. Water Res. 2007, 41, 1754–1762. [Google Scholar] [CrossRef]
  147. Yang, F.; Huang, F.; Feng, H.; Wei, J.; Massey, I.Y.; Liang, G.; Zhang, F.; Yin, L.; Kacew, S.; Zhang, X.; et al. A complete route for biodegradation of potentially carcinogenic cyanotoxin microcystin-LR in a novel indigenous bacterium. Water Res. 2020, 174, 115638. [Google Scholar] [CrossRef] [PubMed]
  148. Massey, I.Y.; Yang, F. A mini review on microcystins and bacterial degradation. Toxins 2020, 12, 268. [Google Scholar] [CrossRef] [Green Version]
  149. Hu, C.; Zuo, Y.; Peng, L.; Gan, N.; Song, L. Widespread distribution and adaptive degradation of microcystin degrader (mlr-genotype) in Lake Taihu, China. Toxins 2021, 13, 864. [Google Scholar] [CrossRef] [PubMed]
  150. Okano, K.; Shimizu, K.; Maseda, H.; Kawauchi, Y.; Utsumi, M.; Itayama, T.; Zhang, Z.; Sugiura, N. Whole-genome sequence of the microcystin-degrading bacterium Sphingopyxis sp. strain C-1. Genome Announc. 2015, 3, e0083815. [Google Scholar] [CrossRef] [Green Version]
  151. Qin, L.; Zhang, X.; Chen, X.; Wang, K.; Shen, Y.; Li, D. Isolation of a novel microcystin-degrading bacterium and the evolutionary origin of mlr gene cluster. Toxins 2019, 11, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Maghsoudi, E.; Fortin, N.; Greer, C.; Maynard, C.; Page, A.; Duy, S.V.; Sauve, S.; Prevost, M.; Dorner, S. Cyanotoxin degradation activity and mlr gene expression profiles of a Sphygopyxis sp. isolated from Lake Champlain, Canada. Environ. Sci. Process. Impacts 2016, 18, 1417–1426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Imanishi, S.; Kato, H.; Mizuno, M.; Tsuji, K.; Harada, K.-I. Bacterial degradation of microcystins and nodularin. Chem. Res. Toxicol. 2005, 18, 591–598. [Google Scholar] [CrossRef] [PubMed]
  154. Mohamed, Z.A.; Alamri, S.A. Biodegradation of cylindrospermopsin toxin by microcystin-degrading bacteria isolated from cyanobacterial blooms. Toxicon 2012, 60, 1390–1395. [Google Scholar] [CrossRef] [PubMed]
  155. Dziga, D.; Kokocinski, M.; Maksylewicz, A.; Czaja-Prokop, U.; Barylski, J. Cylindrospermopsin biodegradation abilities of Aeromonas sp. isolated from Rusałka Lake. Toxins 2016, 8, 55. [Google Scholar] [CrossRef] [Green Version]
  156. Mohamed, Z.A.; Hashem, M.; Alamri, S.A. Growth inhibition of the cyanobacterium Microcystis aeruginosa and degradation of its microcystin toxins by the fungus Trichoderma citrinoviride. Toxicon 2014, 86, 51–58. [Google Scholar] [CrossRef] [PubMed]
  157. Mohamed, Z.A.; Hashem, M.; Alamri, S.; Campos, A.; Vasconcelos, V. Fungal biodegradation and removal of cyanobacteria and microcystins: Potential applications and research needs. Environ. Sci. Pollut. Res. 2021, 28, 37041–37050. [Google Scholar] [CrossRef] [PubMed]
  158. Kurmayer, R.; Jüttner, F. Strategies for the co-existence of zooplankton with the toxic cyanobacterium Planktothrix rubescens in Lake Zurich. J. Plankton Res. 1999, 21, 659–683. [Google Scholar] [CrossRef]
  159. Akbar, S.; Huang, J.; Zhou, Q.; Gu, L.; Sun, Y.; Zhang, L.; Lyu, K.; Yang, Z. Elevated temperature and toxic Microcystis reduce Daphnia fitness and modulate gut microbiota. Environ. Pollut. 2021, 271, 116409. [Google Scholar] [CrossRef] [PubMed]
  160. Combes, A.; Dellinger, M.; Cadel-six, S.; Amand, S.; Comte, K. Ciliate Nassula sp. grazing on a microcystin-producing cyanobacterium (Planktothrix agardhii): Impact on cell growth and in the microcystin fractions. Aquatic Toxicol. 2013, 126, 435–441. [Google Scholar] [CrossRef]
  161. Zhang, L.; Wang, Z.; Wang, N.; Gu, L.; Sun, Y.; Huang, Y.; Chen, Y.; Yang, Z. Mixotrophic Ochromonas addition improves the harmful Microcystis-dominated phytoplankton community in in situ microcosms. Environ. Sci. Technol. 2020, 54, 4609–4620. [Google Scholar] [CrossRef] [PubMed]
  162. Kaushik, R.; Balasubramanian, R. Methods and approaches used for detection of cyanotoxins in environmental samples: A review. Crit. Rev. Environ. Sci. Technol. 2013, 43, 1349–1383. [Google Scholar] [CrossRef]
  163. Duncan, M.W. Good mass spectrometry and its place in good science. J. Mass Spectrom. 2012, 47, 795–809. [Google Scholar] [CrossRef] [PubMed]
  164. Faassen, E.J.; Antoniou, M.G.; Beekman-Lukassen, W.; Blahova, L.; Chernova, E.; Christophoridis, C.; Combes, A.; Edwards, C.; Fastner, J.; Harmsen, J. A collaborative evaluation of LC-MS/MS based methods for BMAA analysis: Soluble bound BMAA found to be an important fraction. Mar. Drugs 2016, 14, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Sanseverino, I.; António, D.C.; Loos, R.; Lettieri, T. Cyanotoxins: Methods and Approaches for Their Analysis and Detection; Centre, J.R., Ed.; Publications Office of the European Union: Luxembourg, 2017. [Google Scholar] [CrossRef]
  166. 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]
  167. Van Apeldoorn, M.E.; Van Egmond, H.P.; Speijers, G.J.; Bakker, G.J. Toxins of cyanobacteria. Mol. Nutr. Food Res. 2007, 51, 7–60. [Google Scholar] [CrossRef] [PubMed]
  168. Testai, E.; Buratti, F.M.; Funari, E.; Manganelli, M.; Vichi, S.; Arnich, N.; Biré, R.; Fessard, V.; Sialehaamoa, A. Review and analysis of occurrence, exposure and toxicity of cyanobacteria toxins in food. EFSA Support. Publ. 2016, 13, 998E. [Google Scholar] [CrossRef]
  169. Merel, S.; Walker, D.; Chicana, R.; Snyder, S.; Baurès, E.; Thomas, O. State of knowledge and concerns on cyanobacterial blooms and cyanotoxins. Environ. Int. 2013, 59, 303–327. [Google Scholar] [CrossRef] [PubMed]
  170. Abbes, S.; Vo Duy, S.; Munoz, G.; Dinh, Q.T.; Simon, D.F.; Husk, B.; Baulch, H.M.; Vinçon-Leite, B.; Fortin, N.; Greer, C.W.; et al. Occurrence of BMAA isomers in bloom-impacted lakes and reservoirs of Brazil, Canada, France, Mexico, and the United Kingdom. Toxins 2022, 14, 251. [Google Scholar] [CrossRef]
  171. Altenburger, R.; Ait-Aissa, S.; Antczak, P.; Backhaus, T.; Barceló, D.; Seiler, T.-B.; Brion, F.; Busch, W.; Chipman, K.; de Alda, M.L.; et al. Future water quality monitoring-adapting tools to deal with mixtures of pollutants in water resource management. Sci. Total Environ. 2015, 512, 540–551. [Google Scholar] [CrossRef] [Green Version]
  172. Hollender, J.; Schymanski, E.L.; Singer, H.P.; Ferguson, P.L. Nontarget screening with high resolution mass spectrometry in the environment: Ready to go? Environ. Sci. Technol. 2017, 51, 11505–11512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Escher, B.I.; Stapleton, H.M.; Schymanski, E.L. Tracking complex mixtures of chemicals in our changing environment. Science 2020, 367, 388–392. [Google Scholar] [CrossRef]
  174. Dom, I.; Biré, R.; Hort, V.; Lavison-Bompard, G.; Nicolas, M.; Guérin, T. Extended targeted and non-targeted strategies for the analysis of marine toxins in mussels and oysters by (LC-HRMS). Toxins 2018, 10, 375. [Google Scholar] [CrossRef] [Green Version]
  175. Krauss, M.; Singer, H.; Hollender, J. LC–high resolution MS in environmental analysis: From target screening to the identification of unknowns. Anal. Bioanal. Chem. 2010, 397, 943–951. [Google Scholar] [CrossRef] [Green Version]
  176. Bogialli, S.; Bortolini, C.; Di Gangi, I.M.; Di Gregorio, F.N.; Lucentini, L.; Favaro, G.; Pastore, P. Liquid chromatography-high resolution mass spectrometric methods for the surveillance monitoring of cyanotoxins in freshwaters. Talanta 2017, 170, 322–330. [Google Scholar] [CrossRef]
  177. Gonzalez-Gaya, B.; Lopez-Herguedas, N.; Bilbao, D.; Mijangos, L.; Iker, A.; Etxebarria, N.; Irazola, M.; Prieto, A.; Olivares, M.; Zuloaga, O. Suspect and non-target screening: The last frontier in environmental analysis. Anal. Methods 2021, 13, 1876–1904. [Google Scholar] [CrossRef] [PubMed]
  178. Picardo, M.; Sanchís, J.; Núñez, O.; Farré, M. Suspect screening of natural toxins in surface and drinking water by high performance liquid chromatography and high-resolution mass spectrometry. Chemosphere 2020, 261, 127888. [Google Scholar] [CrossRef] [PubMed]
  179. Bishop, C.T.; Anet, E.F.L.J.; Gorham, P.R. Isolation and identification of the fast-death factor in Microcystis aeruginosa NRC-1. Can. J. Biochem. Physiol. 1959, 37, 453–471. [Google Scholar] [CrossRef] [PubMed]
  180. He, Q.; Wang, W.; Xu, Q.; Liu, Z.; Teng, J.; Yan, H.; Liu, X. Microcystins in water: Detection, microbial degradation strategies, and mechanisms. Int. J. Environ. Res. Public Health 2022, 19, 13175. [Google Scholar] [CrossRef] [PubMed]
  181. 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]
  182. Van Hassel, W.H.R.; Huybrechts, B.; Masquelier, J.; Wilmotte, A.; Andjelkovic, M. Development, validation and application of a targeted LC-MS method for quantification of microcystins and nodularin: Towards a better characterization of drinking water. Water 2022, 14, 1195. [Google Scholar] [CrossRef]
  183. Panda, D.; Dash, B.P.; Manickam, S.; Boczkaj, G. Recent advancements in LC-MS based analysis of biotoxins: Present and future challenges. Mass Spectrom. Rev. 2022, 41, 766–803. [Google Scholar] [CrossRef]
  184. Triantis, T.; Tsimeli, K.; Kaloudis, T.; Thanassoulias, N.; Lytras, E.; Hiskia, A. Development of an integrated laboratory system for the monitoring of cyanotoxins in surface and drinking waters. Toxicon 2010, 55, 979–989. [Google Scholar] [CrossRef]
  185. Zhang, L.; Liu, J.; Zhang, D.; Luo, L.; Liao, Q.; Yuan, L.; Wu, N. Seasonal and spatial variations of microcystins in Poyang Lake, the largest freshwater lake in China. Environ. Sci. Pollut. Res. Int. 2018, 25, 6300–6307. [Google Scholar] [CrossRef]
  186. Turner, A.D.; Dhanji-Rapkova, M.; O’Neill, A.; Coates, L.; Lewis, A.; Lewis, K. Analysis of microcystins in cyanobacterial blooms from freshwater bodies in England. Toxins 2018, 10, 39. [Google Scholar] [CrossRef] [Green Version]
  187. Mantzouki, E.; Lurling, M.; Fastner, J.; de Senerpont Domis, L.; Wilk-Wozniak, E.; Koreiviene, J.; Seelen, L.; Teurlincx, S.; Verstijnen, Y.; Krzton, W.; et al. Temperature effects explain continental scale distribution of cyanobacterial toxins. Toxins 2018, 10, 156. [Google Scholar] [CrossRef] [Green Version]
  188. Main, B.J.; Bowling, L.C.; Padula, M.P.; Bishop, D.P.; Mitrovic, S.M.; Guillemin, G.J.; Rodgers, K.J. Detection of the suspected neurotoxin beta-methylamino-l-alanine (BMAA) in cyanobacterial blooms from multiple water bodies in Eastern Australia. Harmful Algae 2018, 74, 10–18. [Google Scholar] [CrossRef] [PubMed]
  189. Pitois, F.; Fastner, J.; Pagotto, C.; Dechesne, M. Multi-toxin occurrences in ten French water resource reservoirs. Toxins 2018, 10, 283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Turner, A.D.; Waack, J.; Lewis, A.; Edwards, C.; Lawton, L. Development and single-laboratory validation of a UHPLC-MS/MS method for quantitation of microcystins and nodularin in natural water, cyanobacteria, shellfish and algal supplement tablet powders. J. Chromatogr. B 2018, 1074–1075, 111–123. [Google Scholar] [CrossRef]
  191. Thuret-Benoist, H.; Pallier, V.; Feuillade-Cathalifaud, G. Quantification of microcystins in natural waters by HPLC-UV after a pre-concentration step: Validation of the analytical performances and study of the interferences. Environ. Toxicol. Pharmacol. 2019, 72, 103223. [Google Scholar] [CrossRef]
  192. Botha, C.J.; Laver, P.; Singo, A.; Venter, E.; Ferreira, G.C.H.; Rösemann, M.; Myburgh, J.G. Evaluation of a Norwegian-developed ELISA to determine microcystin concentrations in fresh water. Water Supply 2019, 19, 743–752. [Google Scholar] [CrossRef]
  193. 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]
  194. Leon, C.; Penuela, G.A. Detected cyanotoxins by UHPLC MS/MS technique in tropical reservoirs of northeastern Colombia. Toxicon 2019, 167, 38–48. [Google Scholar] [CrossRef]
  195. Vo Duy, S.; Munoz, G.; Dinh, Q.T.; Tien Do, D.; Simon, D.F.; Sauve, S. Analysis of the neurotoxin beta-N-methylamino-L-alanine (BMAA) and isomers in surface water by FMOC derivatization liquid chromatography high resolution mass spectrometry. PLoS ONE 2019, 14, e0220698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. McKindles, K.M.; Zimba, P.V.; Chiu, A.S.; Watson, S.B.; Gutierrez, D.B.; Westrick, J.; Kling, H.; Davis, T.W. A multiplex analysis of potentially toxic cyanobacteria in Lake Winnipeg during the 2013 bloom season. Toxins 2019, 11, 587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Yan, B.; Liu, Z.; Huang, R.; Xu, Y.; Liu, D.; Wang, W.; Zhao, Z.; Cui, F.; Shi, W. Impact factors on the production of beta-methylamino-L-alanine (BMAA) by cyanobacteria. Chemosphere 2020, 243, 125355. [Google Scholar] [CrossRef] [PubMed]
  198. Abbas, F.; Porojan, C.; Mowe, M.A.D.; Lehane, M.; Mitrovic, S.M.; Lim, R.P.; Yeo, D.C.J.; Furey, A. Sample extraction and liquid chromatography–tandem mass spectrometry (LC-MS/MS) method development and validation for the quantitative detection of cyanobacterial hepatotoxins and neurotoxins in Singapore’s reservoirs. Mar. Freshw. Res. 2020, 71, 673–688. [Google Scholar] [CrossRef]
  199. Stoyneva-Gärtner, M.; Stefanova, K.; Descy, J.-P.; Uzunov, B.; Radkova, M.; Pavlova, V.; Mitreva, M.; Gärtner, G. Microcystis aeruginosa and M. wesenbergii were the primary planktonic microcystin producers in several Bulgarian waterbodies (August 2019). Appl. Sci. 2020, 11, 357. [Google Scholar] [CrossRef]
  200. Zervou, S.K.; Moschandreou, K.; Paraskevopoulou, A.; Christophoridis, C.; Grigoriadou, E.; Kaloudis, T.; Triantis, T.M.; Tsiaoussi, V.; Hiskia, A. Cyanobacterial toxins and peptides in Lake Vegoritis, Greece. Toxins 2021, 13, 394. [Google Scholar] [CrossRef] [PubMed]
  201. Zhang, Y.; Husk, B.R.; Duy, S.V.; Dinh, Q.T.; Sanchez, J.S.; Sauve, S.; Whalen, J.K. Quantitative screening for cyanotoxins in soil and groundwater of agricultural watersheds in Quebec, Canada. Chemosphere 2021, 274, 129781. [Google Scholar] [CrossRef]
  202. Choi, J.-W.; Jang, J.-H.; Kim, A.-K.; Lee, S.-H. Determination of β-N-Methylamino-L-Alanine (BMAA) levels in surface water and treated water by liquid chromatography-tandem mass spectrometry. J. Environ. Anal. Health Toxicol. 2021, 24, 171–175. [Google Scholar] [CrossRef]
  203. Dinh, Q.T.; Munoz, G.; Simon, D.F.; Vo Duy, S.; Husk, B.; Sauve, S. Stability issues of microcystins, anabaenopeptins, anatoxins, and cylindrospermopsin during short-term and long-term storage of surface water and drinking water samples. Harmful Algae 2021, 101, 101955. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, T.; Xie, H.; Cao, Y.; Xu, Q.; Gan, N. Magnetic solid phase extraction coupled with high-performance liquid chromatography-diode array detection based on assembled magnetic covalent organic frameworks for selective extraction and detection of microcystins in aquatic foods. J. Chromatogr. A 2022, 1685, 463614. [Google Scholar] [CrossRef] [PubMed]
  205. Pan, L.; Huang, J.J.; Chen, J.; He, X.; Wang, Y.; Wang, J.; Wang, B. Trace determination of multiple hydrophilic cyanotoxins in freshwater by off- and on-line solid phase extraction coupled to liquid chromatography-tandem mass spectrometry. Sci. Total Environ. 2022, 853, 158545. [Google Scholar] [CrossRef]
  206. Van Hassel, W.H.R.; Andjelkovic, M.; Durieu, B.; Marroquin, V.A.; Masquelier, J.; Huybrechts, B.; Wilmotte, A. A summer of cyanobacterial blooms in Belgian waterbodies: Microcystin quantification and molecular characterizations. Toxins 2022, 14, 61. [Google Scholar] [CrossRef]
  207. Choi, J.-W.; Jang, J.-H.; Lee, S.-H.; Yoon, M.-A. Determination of cylindrospermopsin in surface and treated water using liquid chromatography-tandem mass spectrometry. Environ. Anal. Health Toxicol. 2022, 25, 71–76. [Google Scholar] [CrossRef]
  208. Duy, S.V.; Munoz, G.; Dinh, Q.T.; Zhang, Y.; Simon, D.F.; Sauve, S. Fast screening of saxitoxin, neosaxitoxin, and decarbamoyl analogues in fresh and brackish surface waters by on-line enrichment coupled to HILIC-HRMS. Talanta 2022, 241, 123267. [Google Scholar] [CrossRef] [PubMed]
  209. Aparicio-Muriana, M.M.; Carmona-Molero, R.; Lara, F.J.; Garcia-Campana, A.M.; Del 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]
  210. Zhao, P.; Qiu, J.; Li, A.; Yan, G.; Li, M.; Ji, Y. Matrix effect of diverse biological samples extracted with different extraction ratios on the detection of beta-N-methylamino-L-alanine by two common LC-MS/MS analysis methods. Toxins 2022, 14, 387. [Google Scholar] [CrossRef] [PubMed]
  211. Emmons, R.V.; Karaj, E.; Cudjoe, E.; Bell, D.S.; Tillekeratne, L.M.V.; Gionfriddo, E. Leveraging multi-mode microextraction and liquid chromatography stationary phases for quantitative analysis of neurotoxin beta-N-methylamino-L-alanine and other non-proteinogenic amino acids. J. Chromatogr. A 2022, 1685, 463636. [Google Scholar] [CrossRef] [PubMed]
  212. Zhang, H.; Li, Y.; Abdallah, M.F.; Tan, H.; Li, J.; Liu, S.; Zhang, R.; Sun, F.; Li, Y.; Yang, S. Novel one-point calibration strategy for high-throughput quantitation of microcystins in freshwater using LC-MS/MS. Sci. Total Environ. 2023, 858, 159345. [Google Scholar] [CrossRef]
  213. Kulabhusan, P.K.; Campbell, K. Recent trends in the detection of freshwater cyanotoxins with a critical note on their occurrence in Asia. Trends Environ. Anal. Chem. 2021, 32, e00150. [Google Scholar] [CrossRef]
  214. Lee, J.; Lee, S.; Jiang, X. Cyanobacterial toxins in freshwater and food: Important sources of exposure to humans. Annu. Rev. Food Sci. Technol. 2017, 8, 281–304. [Google Scholar] [CrossRef] [PubMed]
  215. Nielsen, M.C.; Jiang, S.C. Can cyanotoxins penetrate human skin during water recreation to cause negative health effects? Harmful Algae 2020, 98, 101872. [Google Scholar] [CrossRef]
  216. Tatters, A.O.; Howard, M.D.; Nagoda, C.; Busse, L.; Gellene, A.G.; Caron, D.A. Multiple stressors at the land-sea interface: Cyanotoxins at the land-sea interface in the Southern California Bight. Toxins 2017, 9, 95. [Google Scholar] [CrossRef] [Green Version]
  217. Tatters, A.O.; Howard, M.D.; Nagoda, C.; Fetscher, A.E.; Kudela, R.M.; Caron, D.A. Heterogeneity of toxin-producing cyanobacteria and cyanotoxins in coastal watersheds of southern California. Estuaries Coasts 2019, 42, 958–975. [Google Scholar] [CrossRef]
  218. Tatters, A.O.; Smith, J.; Kudela, R.M.; Hayashi, K.; Howard, M.D.; Donovan, A.R.; Loftin, K.A.; Caron, D.A. The tide turns: Episodic and localized cross-contamination of a California coastline with cyanotoxins. Harmful Algae 2021, 103, 102003. [Google Scholar] [CrossRef]
  219. Howard, M.D.; Smith, J.; Caron, D.A.; Kudela, R.M.; Loftin, K.; Hayashi, K.; Fadness, R.; Fricke, S.; Kann, J.; Roethler, M.; et al. Integrative monitoring strategy for marine and freshwater harmful algal blooms and toxins across the freshwater-to-marine continuum. Integr. Environ. Assess. Manag. 2022, 1, 1–19. [Google Scholar] [CrossRef] [PubMed]
  220. Andraka, D.; Ospanov, K.; Myrzakhmetov, M. Current state of communal sewage treatment in the republic of Kazakhstan. J. Ecol. Eng. 2015, 16, 101–109. [Google Scholar] [CrossRef] [Green Version]
  221. Gutiérrez-Praena, D.; Jos, Á.; Pichardo, S.; Moreno, I.M.; Cameán, A.M. Presence and bioaccumulation of microcystins and cylindrospermopsin in food and the effectiveness of some cooking techniques at decreasing their concentrations: A review. Food Chem. Toxicol. 2013, 53, 139–152. [Google Scholar] [CrossRef]
  222. Abdallah, M.F.; Van Hassel, W.H.; Andjelkovic, M.; Wilmotte, A.; Rajkovic, A. Cyanotoxins and food contamination in developing countries: Review of their types, toxicity, analysis, occurrence and mitigation strategies. Toxins 2021, 13, 786. [Google Scholar] [CrossRef] [PubMed]
  223. Downing, S.; Contardo-Jara, V.; Pflugmacher, S.; Downing, T.G. The fate of the cyanobacterial toxin β-N-methylamino-L-alanine in freshwater mussels. Ecotoxicol. Environ. Safety 2014, 101, 51–58. [Google Scholar] [CrossRef] [PubMed]
  224. Baptista, M.S.; Vasconcelos, R.G.; Ferreira, P.C.; Almeida, C.M.R.; Vasconcelos, V.M. Assessment of the non-protein amino acid BMAA in Mediterranean mussel Mytilus galloprovincialis after feeding with estuarine cyanobacteria. Environ. Sci. Pollution Res. 2015, 22, 12501–12510. [Google Scholar] [CrossRef] [PubMed]
  225. Lance, E.; Arnich, N.; Maignien, T.; Biré, R. Occurrence of β-N-methylamino-l-alanine (BMAA) and isomers in aquatic environments and aquatic food sources for humans. Toxins 2018, 10, 83. [Google Scholar] [CrossRef] [Green Version]
  226. Kim, S.Y.; Rydberg, S. Transfer of the neurotoxin β-N-methylamino-l-alanine (BMAA) in the agro–aqua cycle. Mar. Drugs 2020, 18, 244. [Google Scholar] [CrossRef] [PubMed]
  227. Crush, J.; Briggs, L.; Sprosen, J.; Nichols, S. Effect of irrigation with lake water containing microcystins on microcystin content and growth of ryegrass, clover, rape, and lettuce. Environ. Toxicol. 2008, 23, 246–252. [Google Scholar] [CrossRef]
  228. Mohamed, Z.A.; Al Shehri, A.M. Microcystins in groundwater wells and their accumulation in vegetable plants irrigated with contaminated waters in Saudi Arabia. J. Hazard. Mater. 2009, 172, 310–315. [Google Scholar] [CrossRef]
  229. Kittler, K.; Schreiner, M.; Krumbein, A.; Manzei, S.; Koch, M.; Rohn, S.; Maul, R. Uptake of the cyanobacterial toxin cylindrospermopsin in Brassica vegetables. Food Chem. 2012, 133, 875–879. [Google Scholar] [CrossRef]
  230. Roney, B.R.; Renhui, L.; Banack, S.A.; Murch, S.; Honegger, R.; Cox, P.A. Consumption of fa cai Nostoc soup: A potential for BMAA exposure from Nostoc cyanobacteria in China? Amyotroph. Lateral Scler. 2009, 10, 44–49. [Google Scholar] [CrossRef]
  231. Han, D.; Deng, Z.; Lu, F.; Hu, Z. Biology and biotechnology of edible Nostoc. In Handbook of Microalgal Culture: Applied Phycology and Biotechnology, 2nd ed.; Richmond, A., Hu, Q., Eds.; John Wiley & Sons: Hoboken, NJ, USA, 2013; pp. 433–444. [Google Scholar] [CrossRef]
  232. Fidor, A.; Konkel, R.; Mazur-Marzec, H. Bioactive peptides produced by cyanobacteria of the genus Nostoc: A review. Mar. Drugs 2019, 17, 561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Pérez-Lloréns, J.L. Microalgae: From staple foodstuff to avant-garde cuisine. Inter. J. Gastr. Food Sci. 2020, 21, 100221. [Google Scholar] [CrossRef]
  234. McCarron, P.; Logan, A.C.; Giddings, S.D.; Quilliam, M.A. Analysis of β-N-methylamino-L-alanine (BMAA) in spirulina-containing supplements by liquid chromatography-tandem mass spectrometry. Aquat. Biosyst. 2014, 10, 5. [Google Scholar] [CrossRef] [Green Version]
  235. Roy-Lachapelle, A.; Solliec, M.; Bouchard, M.F.; Sauvé, S. Detection of cyanotoxins in algae dietary supplements. Toxins 2017, 9, 76. [Google Scholar] [CrossRef] [Green Version]
  236. 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]
  237. Murch, S.J.; Cox, P.A.; Banack, S.A. A mechanism for slow release of biomagnified cyanobacterial neurotoxins and neuro-degenerative disease in Guam. Proc. Natl. Acad. Sci. USA 2004, 101, 12228–12231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Brown, R.M., Jr.; Larson, D.A.; Bold, H.C. Airborne algae: Their abundance and heterogeneity. Science 1964, 143, 583–585. [Google Scholar] [CrossRef] [PubMed]
  239. Olson, N.E.; Cooke, M.E.; Shi, J.H.; Birbeck, J.A.; Westrick, J.A.; Ault, A.P. Harmful algal bloom toxins in aerosol generated from inland lake water. Environ. Sci. Technol. 2020, 54, 4769–4780. [Google Scholar] [CrossRef] [PubMed]
  240. Harb, C.; Pan, J.; DeVilbiss, S.; Badgley, B.; Marr, L.C.; Schmale, D.G., III; Foroutan, H. Increasing freshwater salinity impacts aerosolized bacteria. Environ. Sci. Technol. 2021, 55, 5731–5741. [Google Scholar] [CrossRef] [PubMed]
  241. Backer, L.C.; McNeel, S.V.; Barber, T.; Kirkpatrick, B.; Williams, C.; Irvin, M.; Zhou, Y.; Johnson, T.B.; Nierenberg, K.; Aubel, M.; et al. Recreational exposure to microcystins during algal blooms in two California lakes. Toxicon 2010, 55, 909–921. [Google Scholar] [CrossRef]
  242. Brodie, E.L.; DeSantis, T.Z.; Parker, J.P.M.; Zubietta, I.X.; Piceno, Y.M.; Andersen, G.L. Urban aerosols harbor diverse and dynamic bacterial populations. Proc. Natl. Acad. Sci. USA 2007, 104, 299–304. [Google Scholar] [CrossRef] [Green Version]
  243. Wood, S.A.; Dietrich, D.R. Quantitative assessment of aerosolized cyanobacterial toxins at two New Zealand lakes. J. Environ. Monit. 2011, 13, 1617–1624. [Google Scholar] [CrossRef] [Green Version]
  244. Genitsaris, S.; Kormas, K.A.; Moustaka-Gouni, M. Airborne algae and cyanobacteria: Occurrence and related health effects. Front. Biosci.-Elite 2011, 3, 772–787. [Google Scholar]
  245. Wisniewska, K.A.; Śliwińska-Wilczewska, S.; Lewandowska, A.U. Airborne microalgal and cyanobacterial diversity and composition during rain events in the southern Baltic Sea region. Sci. Rep. 2022, 12, 2029. [Google Scholar] [CrossRef]
  246. Plaas, H.E.; Paerl, R.W.; Baumann, K.; Karl, C.; Popendorf, K.J.; Barnard, M.A.; Chang, N.Y.; Curtis, N.P.; Huang, H.; Mathieson, O.L.; et al. Harmful cyanobacterial aerosolization dynamics in the airshed of a eutrophic estuary. Sci. Total Environ. 2022, 852, 158383. [Google Scholar] [CrossRef]
  247. Berstein, I.I.; Safferman, R.S. Viable algae in house dust. Nature 1970, 227, 851–852. [Google Scholar] [CrossRef]
  248. Després, V.; Huffman, J.A.; Burrows, S.M.; Hoose, C.; Safatov, A.; Buryak, G.; Fröhlich-Nowoisky, J.; Elbert, W.; Andreae, M.; Pöschl, U.; et al. Primary biological aerosol particles in the atmosphere: A review. Tellus B Chem. Phys. Meteorol. 2012, 64, 15598. [Google Scholar] [CrossRef] [Green Version]
  249. Wisniewska, K.; Lewandowska, A.U.; Śliwińska-Wilczewska, S. The importance of Cyanobacteria and microalgae present in aerosols to human health and the environment—Review study. Environ. Int. 2019, 131, 104964. [Google Scholar] [CrossRef]
  250. Wiśniewska, K.A.; Śliwińska-Wilczewska, S.; Lewandowska, A.U. The first characterization of airborne cyanobacteria and microalgae in the Adriatic Sea region. PLoS ONE 2020, 15, e0238808. [Google Scholar] [CrossRef] [PubMed]
  251. Gärtner, G.; Stoyneva-Gärtner, M.; Uzunov, B. Algal toxic compounds and their aeroterrestrial, airborne and other extremophilic producers with attention to soil and plant contamination: A review. Toxins 2021, 13, 322. [Google Scholar] [CrossRef]
  252. Scott, L.L.; Downing, S.; Downing, T.G. The evaluation of BMAA inhalation as a potential exposure route using a rat model. Neurotox. Res. 2018, 33, 6–14. [Google Scholar] [CrossRef]
  253. Plaas, H.E.; Paerl, H.W. Toxic cyanobacteria: A growing threat to water and air quality. Environ. Sci. Technol. 2020, 55, 44–64. [Google Scholar] [CrossRef] [PubMed]
  254. Oliveira, V.R.; Mancin, V.G.; Pinto, E.F.; Soares, R.M.; Azevedo, S.M.; Macchione, M.; Carvalho, A.R.; Zin, W.A. Repeated intranasal exposure to microcystin-LR affects lungs but not nasal epithelium in mice. Toxicon 2015, 104, 14–18. [Google Scholar] [CrossRef]
  255. Li, X.; Xu, L.; Zhou, W.; Zhao, Q.; Wang, Y. Chronic exposure to microcystin-LR affected mitochondrial DNA maintenance and caused pathological changes of lung tissue in mice. Environ. Pollut. 2016, 210, 48–56. [Google Scholar] [CrossRef] [PubMed]
  256. Carvalho, G.M.; Oliveira, V.R.; Soares, R.M.; Azevedo, S.M.; Lima, L.M.; Barreiro, E.J.; Valença, S.S.; Saldiva, P.H.; Faffe, D.S.; Zin, W.A. Can LASSBio 596 and dexamethasone treat acute lung and liver inflammation induced by microcystin-LR? Toxicon 2010, 56, 604–612. [Google Scholar] [CrossRef] [PubMed]
  257. Zhao, S.; Sun, H.; Yan, W.; Xu, D.; Shen, T. A proteomic study of the pulmonary injury induced by microcystin-LR in mice. Toxicon 2018, 150, 304–314. [Google Scholar] [CrossRef]
  258. Breidenbach, J.D.; French, B.W.; Gordon, T.T.; Kleinhenz, A.L.; Khalaf, F.K.; Willey, J.C.; Hammersley, J.R.; Wooten, R.M.; Crawford, E.L.; Modyanov, N.N. Microcystin-LR aerosol induces inflammatory responses in healthy human primary airway epithelium. Environ. Int. 2022, 169, 107531. [Google Scholar] [CrossRef]
  259. Facciponte, D.N.; Bough, M.W.; Seidler, D.; Carroll, J.L.; Ashare, A.; Andrew, A.S.; Tsongalis, G.J.; Vaickus, L.J.; Henegan, P.L.; Butt, T.H. Identifying aerosolized cyanobacteria in the human respiratory tract: A proposed mechanism for cyanotoxin-associated diseases. Sci. Total Environ. 2018, 645, 1003–1013. [Google Scholar] [CrossRef]
  260. Pierozan, P.; Piras, E.; Brittebo, E.; Karlsson, O. The cyanobacterial neurotoxin β-N-methylamino-l-alanine (BMAA) targets the olfactory bulb region. Arch. Toxicol. 2020, 94, 2799–2808. [Google Scholar] [CrossRef]
  261. Sarasa, M.; Pesini, P. Natural non-transgenic animal models for research in Alzheimer’s disease. Curr. Alzheimer Res. 2009, 6, 171–178. [Google Scholar] [CrossRef] [Green Version]
  262. Davis, D.A.; Cox, P.A.; Banack, S.A.; Lecusay, P.D.; Garamszegi, S.P.; Hagan, M.J.; Powell, J.T.; Metcalf, J.S.; Palmour, R.M.; Beierschmitt, A.; et al. L-serine reduces spinal cord pathology in a vervet model of preclinical ALS/MND. J. Neuropathol. Exp. Neurol. 2020, 79, 393–406. [Google Scholar] [CrossRef] [PubMed]
  263. Mondo, K.; Hammerschlag, N.; Basile, M.; Pablo, J.; Banack, S.A.; Mash, D.C. Cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) in shark fins. Mar. Drugs 2012, 10, 509–520. [Google Scholar] [CrossRef] [Green Version]
  264. Davis, D.A.; Garamszegi, S.P.; Banack, S.A.; Dooley, P.D.; Coyne, T.M.; McLean, D.W.; Rotstein, D.S.; Mash, D.C.; Cox, P.A. BMAA, Methylmercury, and mechanisms of neurodegeneration in dolphins: A natural model of toxin exposure. Toxins 2021, 13, 697. [Google Scholar] [CrossRef] [PubMed]
  265. Davis, D.A.; Mondo, K.; Stern, E.; Annor, A.K.; Murch, S.J.; Coyne, T.M.; Brand, L.E.; Niemeyer, M.E.; Sharp, S.; Bradley, W.G.; et al. Cyanobacterial neurotoxin BMAA and brain pathology in stranded dolphins. PLoS ONE 2019, 14, e0213346. [Google Scholar] [CrossRef]
  266. Lobner, D.; Piana, P.M.T.; Salous, A.K.; Peoples, R.W. β-N-methylamino-L-alanine enhances neurotoxicity through multiple mechanisms. Neurobiol. Dis. 2007, 25, 360–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Pedrosa, C.D.S.G.; Souza, L.R.Q.; Gomes, T.A.; de Lima, C.V.F.; Ledur, P.F.; Karmirian, K.; Barbeito-Andres, J.; Costa, M.D.N.; Higa, L.M.; Rossi, Á.D.; et al. The cyanobacterial saxitoxin exacerbates neural cell death and brain malformations induced by Zika virus. PLoS Negl. Trop. Dis. 2020, 14, e0008060. [Google Scholar] [CrossRef] [Green Version]
  268. Wiese, M.; D’agostino, P.M.; Mihali, T.K.; Moffitt, M.C.; Neilan, B.A. Neurotoxic alkaloids: Saxitoxin and its analogs. Mar. Drugs 2010, 8, 2185–2211. [Google Scholar] [CrossRef] [Green Version]
  269. Smith, F.M.; Wood, S.A.; van Ginkel, R.; Broady, P.A.; Gaw, S. First report of saxitoxin production by a species of the freshwater benthic cyanobacterium, Scytonema Agardh. Toxicon 2011, 57, 566–573. [Google Scholar] [CrossRef]
  270. Cusick, K.D.; Sayler, G.S. An overview on the marine neurotoxin, saxitoxin: Genetics, molecular targets, methods of detection and ecological functions. Mar. Drugs 2013, 11, 991–1018. [Google Scholar] [CrossRef] [Green Version]
  271. Roué, M.; Gugger, M.; Golubic, S.; Amzil, Z.; Aráoz, R.; Turquet, J.; Chinain, M.; Laurent, D. Marine cyanotoxins potentially harmful to human health. In Outstanding Marine Molecules; Chapter 1; Wiley-VCH Verlag GmbH & Co. KGaA: Weinheim, Germany, 2014; pp. 1–22. [Google Scholar] [CrossRef]
  272. Bonnard, I.; Bornancin, L.; Dalle, K.; Chinain, M.; Zubia, M.; Banaigs, B.; Roué, M. Assessment of the chemical diversity and potential toxicity of benthic cyanobacterial blooms in the lagoon of Moorea Island (French Polynesia). J. Mar. Sci. Eng. 2020, 8, 406. [Google Scholar] [CrossRef]
  273. Weiss, C.; Figueras, E.; Borbely, A.N.; Sewald, N. Cryptophycins: Cytotoxic cyclodepsipeptides with potential for tumor targeting. J. Pept. Sci. 2017, 23, 514–531. [Google Scholar] [CrossRef] [Green Version]
  274. Kust, A.; Řeháková, K.; Vrba, J.; Maicher, V.; Mareš, J.; Hrouzek, P.; Chiriac, M.C.; Benedová, Z.; Tesařová, B.; Saurav, K. Insight into unprecedented diversity of cyanopeptides in eutrophic ponds using an ms/ms networking approach. Toxins 2020, 12, 561. [Google Scholar] [CrossRef]
  275. Panda, D.; Himes, R.H.; Moore, R.E.; Wilson, L.; Jordan, M.A. Mechanism of action of the unusually potent microtubule inhibitor cryptophycin 1. Biochemistry 1997, 36, 12948–12953. [Google Scholar] [CrossRef]
  276. Costa, M.; Costa-Rodrigues, J.; Fernandes, M.H.; Barros, P.; Vasconcelos, V.; Martins, R. Marine cyanobacteria compounds with anticancer properties: A review on the implication of apoptosis. Mar. Drugs 2012, 10, 2181–2207. [Google Scholar] [CrossRef] [Green Version]
  277. Luesch, H.; Chanda, S.K.; Raya, R.M.; DeJesus, P.D.; Orth, A.P.; Walker, J.R.; Izpisúa Belmonte, J.C.; Schultz, P.G. A functional genomics approach to the mode of action of apratoxin A. Nat. Chem. Biol. 2006, 2, 158–167. [Google Scholar] [CrossRef]
  278. Hastie, C.J.; Borthwick, E.B.; Morrison, L.F.; Codd, G.A.; Cohen, P.T.W. Inhibition of several protein phosphatases by a non-covalently interacting microcystin and a novel cyanobacterial peptide, nostocyclin. Biochim. Biophys. Acta 2005, 1726, 187–193. [Google Scholar] [CrossRef] [PubMed]
  279. Maynes, J.T.; Luu, H.A.; Cherney, M.M.; Andersen, R.J.; Williams, D.; Holmes, C.F.; James, M.N. Crystal structures of protein phosphatase-1 bound to motuporin and dihydromicrocystin-LA: Elucidation of the mechanism of enzyme inhibition by cyanobacterial toxins. J. Mol. Biol. 2006, 356, 111–120. [Google Scholar] [CrossRef]
  280. Tapia-Larios, C.; Mendoza, E.G.; Rubio-Armendáriz, M.C.; Olivero-Verbel, J. Identification of novel molecular targets of four microcystin variants by high-throughput virtual screening. Appl. Sci. 2022, 12, 2612. [Google Scholar] [CrossRef]
  281. Liu, L.; Xu, T.C.; Zhao, Z.A.; Zhang, N.N.; Li, J.; Chen, H.S. Toll-like receptor 4 signaling in neurons mediates cerebral ischemia/reperfusion injury. Mol. Neurobiol. 2022, 60, 864–874. [Google Scholar] [CrossRef]
  282. Mello, F.D.; Braidy, N.; Marçal, H.; Guillemin, G.; Nabavi, S.M.; Neilan, B.A. Mechanisms and effects posed by neurotoxic products of cyanobacteria/microbial eukaryotes/dinoflagellates in algae blooms: A review. Neurotox. Res. 2018, 33, 153–167. [Google Scholar] [CrossRef] [PubMed]
  283. Hinojosa, M.G.; Gutiérrez-Praena, D.; Prieto, A.I.; Guzmán-Guillén, R.; Jos, A.; Cameán, A.M. Neurotoxicity induced by microcystins and cylindrospermopsin: A review. Sci. Total Environ. 2019, 668, 547–565. [Google Scholar] [CrossRef] [PubMed]
  284. Chen, L.; Shi, T.; Wang, Y.T.; He, J.; Zhao, X.; Wang, Y.K.; Giesy, J.P.; Chen, F.; Chen, Y.; Tuo, X.; et al. Effects of acute exposure to microcystins on hypothalamic-pituitary-adrenal (HPA), -gonad (HPG) and -thyroid (HPT) axes of female rats. Sci. Total Environ. 2021, 778, 145196. [Google Scholar] [CrossRef]
  285. Jin, H.; How, J.; Meng, X.; Ma, T.; Wang, B.; Liu, Z.; Sha, X.; Ding, J.; Han, X. Microcystin-leucine arginine induced the apoptosis of GnRH neurons by activating the endoplasmic reticulum stress resulting in a decrease of serum testosterone level in mice. Ecotoxicol. Environ. Saf. 2021, 208, 111748. [Google Scholar] [CrossRef]
  286. Zhang, S.; Liu, H.; Du, X.; Chen, X.; Petlulu, P.; Tian, Z.; Shi, L.; Zhang, B.; Yuan, S.; Guo, X.; et al. A new identity of microcystins: Environmental endocrine disruptors? An evidence-based review. Sci. Total Environ. 2022, 851, 158262. [Google Scholar] [CrossRef] [PubMed]
  287. Casas-Rodriguez, A.; Cameán, A.M.; Jos, A. Potential endocrine disruption of cyanobacterial toxins, microcystins and cylindrospermopsin: A review. Toxins 2022, 14, 882. [Google Scholar] [CrossRef]
  288. Shi, T.; Xu, L.L.; Chen, L.; He, J.; Wang, Y.K.; Chen, F.; Chen, Y.; Giesy, J.P.; Wang, Y.T.; Wu, Q.H.; et al. Acute exposure to microcystins affects hypothalamic-pituitary axes of male rats. Environ. Pollut. 2023, 318, 120843. [Google Scholar] [CrossRef]
  289. Wei, L.; Sun, B.; Chang, M.; Liu, Y.; Nie, P. Effects of cyanobacterial toxin microcystin-LR on the transcription levels of immune-related genes in grass carp Ctenopharyngodon idella. Environ. Biol. Fish. 2009, 85, 231–238. [Google Scholar] [CrossRef]
  290. Rymuszka, A.; Adaszek, Ł. Cytotoxic effects and changes in cytokine gene expression induced by microcystin-containing extract in fish immune cells–An in vitro and in vivo study. Fish Shellfish Immun. 2013, 34, 1524–1532. [Google Scholar] [CrossRef]
  291. Saha, P.; Bose, D.; Stebliankin, V.; Cickovski, T.; Seth, R.K.; Porter, D.E.; Brooks, B.W.; Mathee, K.; Narasimhan, G.; Colwell, R.; et al. Prior exposure to microcystin alters host gut resistome and is associated with dysregulated immune homeostasis in translatable mouse models. Sci. Rep. 2022, 12, 11516. [Google Scholar] [CrossRef]
  292. Diez-Quijada, L.; Benítez-González, M.D.M.; Puerto, M.; Jos, A.; Cameán, A.M. Immunotoxic effects induced by microcystins and cylindrospermopsin: A review. Toxins 2021, 13, 711. [Google Scholar] [CrossRef]
  293. Yan, M.; Jin, H.; Pan, C.; Hang, H.; Li, D.; Han, X. Movement disorder and neurotoxicity induced by chronic exposure to microcystin-LR in mice. Mol. Neurobiol. 2022, 59, 5516–5531. [Google Scholar] [CrossRef] [PubMed]
  294. Goto, J.J.; Koenig, J.H.; Ikeda, K. The physiological effect of ingested β-N-methylamino-L-alanine on a glutamatergic synapse in an in vivo preparation. Comp. Biochem. Physiol. C Toxicol. Pharm. 2012, 156, 171–177. [Google Scholar] [CrossRef] [PubMed]
  295. Okle, O.; Rath, L.; Galizia, C.G.; Dietrich, D.R. The cyanobacterial neurotoxin beta-N-methylamino-l-alanine (BMAA) induces neuronal and behavioral changes in honeybees. Toxicol. Appl. Pharm. 2013, 270, 9–15. [Google Scholar] [CrossRef] [PubMed]
  296. Wang, S.; Qiu, J.; Zhao, M.; Li, F.; Yu, R.; Li, A. Accumulation and distribution of neurotoxin BMAA in aquatic animals and effect on the behavior of zebrafish in a T-maze test. Toxicon 2020, 173, 39–47. [Google Scholar] [CrossRef] [PubMed]
  297. Scott, L.; Downing, T. Dose-dependent adult neurodegeneration in a rat model after neonatal exposure to β-n-methylamino-l-alanine. Neurotox. Res. 2019, 35, 711–723. [Google Scholar] [CrossRef] [PubMed]
  298. Karlsson, O.; Lindquist, N.G.; Brittebo, E.B.; Roman, E. Selective brain uptake and behavioral effects of the cyanobacterial toxin BMAA (β-N-methylamino-L-alanine) following neonatal administration to rodents. Toxicol. Sci. 2009, 109, 286–295. [Google Scholar] [CrossRef] [Green Version]
  299. Karlsson, O.; Berg, A.-L.; Hanrieder, J.; Arnerup, G.; Lindström, A.-K.; Brittebo, E.B. Intracellular fibril formation, calcification, and enrichment of chaperones, cytoskeletal, and intermediate filament proteins in the adult hippocampus CA1 following neonatal exposure to the nonprotein amino acid BMAA. Arch. Toxicol. 2015, 89, 423–436. [Google Scholar] [CrossRef] [Green Version]
  300. Pierozan, P.; Karlsson, O. Mitotically heritable effects of BMAA on striatal neural stem cell proliferation and differentiation. Cell Death Dis. 2019, 10, 478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Pierozan, P.; Cattani, D.; Karlsson, O. Hippocampal neural stem cells are more susceptible to the neurotoxin BMAA than primary neurons: Effects on apoptosis, cellular differentiation, neurite outgrowth, and DNA methylation. Cell Death Dis. 2020, 11, 910. [Google Scholar] [CrossRef] [PubMed]
  302. Laugeray, A.; Oummadi, A.; Jourdain, C.; Feat, J.; Meyer-Dilhet, G.; Menuet, A.; Plé, K.; Gay, M.; Routier, S.; Mortaud, S.; et al. Perinatal exposure to the cyanotoxin β-N-méthylamino-l-alanine (BMAA) results in long-lasting behavioral changes in offspring—Potential involvement of DNA damage and oxidative stress. Neurotox. Res. 2018, 33, 87–112. [Google Scholar] [CrossRef]
  303. Méresse, S.; Larrigaldie, V.; Oummadi, A.; de Concini, V.; Morisset-Lopez, S.; Reverchon, F.; Menuet, A.; Montécot-Dubourg, C.; Mortaud, S. β-N-Methyl-Amino-L-Alanine cyanotoxin promotes modification of undifferentiated cells population and disrupts the inflammatory status in primary cultures of neural stem cells. Toxicology 2022, 482, 153358. [Google Scholar] [CrossRef]
  304. Li, X.; Zhang, X.; Ju, J.; Li, Y.; Yin, L.; Pu, Y. Maternal repeated oral exposure to microcystin-LR affects neurobehaviors in developing rats. Environ. Toxicol. Chem. 2015, 34, 64–69. [Google Scholar] [CrossRef]
  305. Montagne, A.; Zhao, Z.; Zlokovic, B.V. Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J. Exp. Med. 2017, 214, 3151–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Al-Bachari, S.; Naish, J.H.; Parker, G.J.; Emsley, H.C.; Parkes, L.M. Blood–brain barrier leakage is increased in Parkinson’s disease. Front. Physiol. 2020, 11, 593026. [Google Scholar] [CrossRef]
  307. Pan, Y.; Nicolazzo, J.A. Altered blood–brain barrier and blood–spinal cord barrier dynamics in amyotrophic lateral sclerosis: Impact on medication efficacy and safety. Br. J. Pharmacol. 2022, 179, 2577–2588. [Google Scholar] [CrossRef] [PubMed]
  308. Barisano, G.; Montagne, A.; Kisler, K.; Schneider, J.A.; Wardlaw, J.M.; Zlokovic, B.V. Blood–brain barrier link to human cognitive impairment and Alzheimer’s disease. Nat. Cardiovasc. Res. 2022, 1, 108–115. [Google Scholar] [CrossRef] [PubMed]
  309. Smith, Q.R.; Nagura, H.; Takada, Y.; Duncan, M.W. Facilitated transport of the neurotoxin, β-N-methylamino-L-alanine, across the blood-brain barrier. J. Neurochem. 1992, 58, 1330–1337. [Google Scholar] [CrossRef]
  310. Myhre, O.; Eide, D.M.; Kleiven, S.; Utkilen, H.C.; Hofer, T. Repeated five-day administration of L-BMAA, microcystin-LR, or as mixture, in adult C57BL/6 mice-lack of adverse cognitive effects. Sci. Rep. 2018, 8, 2308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  311. Karlsson, O.; Berg, A.L.; Lindström, A.K.; Hanrieder, J.; Arnerup, G.; Roman, E.; Bergquist, J.; Lindquist, N.G.; Brittebo, E.B.; Andersson, M. Neonatal exposure to the cyanobacterial toxin BMAA induces changes in protein expression and neurodegeneration in adult hippocampus. Toxicol. Sci. 2012, 130, 391–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Perry, T.L.; Bergeron, C.; Biro, A.J.; Hansen, S. Chronic oral administration of β-N-methylamino-l-alanine is not neurotoxic to mice. J. Neurol. Sci. 1989, 94, 173–180. [Google Scholar] [CrossRef]
  313. Duncan, M.W.; Villacreses, N.E.; Pearson, P.G.; Wyatt, L.; Rapoport, S.I.; Kopin, I.J.; Markey, S.P.; Smith, Q.R. 2-Amino-3-(methylamino)-propanoic acid (BMAA) pharmacokinetics and blood-brain barrier permeability in the rat. J. Pharmacol. Exp. Ther. 1991, 258, 27–35. [Google Scholar]
  314. Cruz-Aguado, R.; Winkler, D.; Shaw, C.A. Lack of behavioural and neuropathological effects of dietary β-methylamino-l-alanine (BMAA) in mice. Pharmacol. Biochem. Behav. 2006, 84, 294–299. [Google Scholar] [CrossRef]
  315. Scott, L.L.; Downing, T.G. A single neonatal exposure to BMAA in a rat model produces neuropathology consistent with neurodegenerative diseases. Toxins 2017, 10, 22. [Google Scholar] [CrossRef] [Green Version]
  316. Garamszegi, S.P.; Banack, S.A.; Duque, L.L.; Metcalf, J.S.; Stommel, E.W.; Cox, P.A.; Davis, D.A. Detection of β-N-methylamino-L-alanine in postmortem olfactory bulbs of Alzheimer’s disease patients using UHPLC-MS/MS: An autopsy case-series study. Toxicol. Rep. 2023, 10, 87–96. [Google Scholar] [CrossRef]
  317. Fischer, W.J.; Altheimer, S.; Cattori, V.; Meier, P.J.; Dietrich, D.R.; Hagenbuch, B. Organic anion transporting polypeptides expressed in liver and brain mediate uptake of microcystin. Toxicol. Appl. Pharmacol. 2005, 203, 257–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Mondal, A.; Saha, P.; Bose, D.; Chatterjee, S.; Seth, R.K.; Xiao, S.; Porter, D.E.; Brooks, B.W.; Scott, G.I.; Nagarkatti, M.; et al. Environmental microcystin exposure in underlying NAFLD-induced exacerbation of neuroinflammation, blood-brain barrier dysfunction, and neurodegeneration are NLRP3 and S100B dependent. Toxicology 2021, 461, 152901. [Google Scholar] [CrossRef]
  319. Wang, J.; Chen, Y.; Zhang, C.; Xiang, Z.; Ding, J.; Han, X. Learning and memory deficits and Alzheimer’s disease-like changes in mice after chronic exposure to microcystin-LR. J. Hazardous Mater. 2019, 373, 504–518. [Google Scholar] [CrossRef] [PubMed]
  320. Wang, J.; Zhang, C.; Zhu, J.; Ding, J.; Chen, Y.; Han, X. Blood-brain barrier disruption and inflammation reaction in mice after chronic exposure to microcystin-LR. Sci. Total Environ. 2019, 689, 662–678. [Google Scholar] [CrossRef] [PubMed]
  321. Feurstein, D.; Holst, K.; Fischer, A.; Dietrich, D.R. Oatp-associated uptake and toxicity of microcystins in primary murine whole brain cells. Toxicol. Appl. Pharmacol. 2009, 234, 247–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. Feurstein, D.; Kleinteich, J.; Heussner, A.H.; Stemmer, K.; Dietrich, D.R. Investigation of microcystin congener–dependent uptake into primary murine neurons. Environ. Health Perspect. 2010, 118, 1370–1375. [Google Scholar] [CrossRef]
  323. Rudrabhatla, P.; Albers, W.; Pant, H.C. Peptidyl-prolyl isomerase 1 regulates protein phosphatase 2A-mediated topographic phosphorylation of neurofilament proteins. J. Neurosci. 2009, 29, 14869–14880. [Google Scholar] [CrossRef] [Green Version]
  324. Yan, M.; Jin, H.; Pan, C.; Han, X. Chronic microcystin-LR-induced α-synuclein promotes neuroinflammation through activation of the NLRP3 inflammasome in microglia. Mol. Neurobiol. 2022, 60, 884–900. [Google Scholar] [CrossRef]
  325. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501. [Google Scholar] [CrossRef]
  326. Bougea, A. Synuclein in neurodegeneration. Adv. Clin. Chem. 2001, 103, 97–134. [Google Scholar] [CrossRef]
  327. Verheijen, J.; Sleegers, K. Understanding Alzheimer disease at the interface between genetics and transcriptomics. Trends Genet. 2018, 34, 434–447. [Google Scholar] [CrossRef] [Green Version]
  328. Chiu, A.S.; Gehringer, M.M.; Braidy, N.; Guillemin, G.J.; Welch, J.H.; Neilan, B.A. Gliotoxicity of the cyanotoxin, β-methyl-amino-L-alanine (BMAA). Sci. Rep. 2013, 3, 1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Chiu, A.S.; Braidy, N.; Marçal, H.; Welch, J.H.; Gehringer, M.M.; Guillemin, G.J.; Neilan, B.A. Global cellular responses to β-methyl-amino-l-alanine (BMAA) by olfactory ensheathing glial cells (OEC). Toxicon 2015, 99, 136–145. [Google Scholar] [CrossRef] [PubMed]
  330. Bubik, A.; Sedmak, B.; Novinec, M.; Lenarčič, B.; Lah, T.T. Cytotoxic and peptidase inhibitory activities of selected non-hepatotoxic cyclic peptides from cyanobacteria. Biol. Chem. 2008, 389, 1339–1346. [Google Scholar] [CrossRef] [PubMed]
  331. D‘Mello, F.; Braidy, N.; Marçal, H.; Guillemin, G.; Rossi, F.; Chinian, M.; Laurent, D.; Teo, C.; Neilan, B.A. Cytotoxic effects of environmental toxins on human glial cells. Neurotox. Res. 2017, 31, 245–258. [Google Scholar] [CrossRef] [PubMed]
  332. Soto, T.; Buzzi, E.D.; Rotstein, N.P.; German, O.L.; Politi, L.E. Damaging effects of BMAA on retina neurons and Müller glial cells. Exp. Eye Res. 2021, 202, 108342. [Google Scholar] [CrossRef] [PubMed]
  333. Tan, V.X.; Mazzocco, C.; Varney, B.; Bodet, D.; Guillemin, T.A.; Bessede, A.; Guillemin, G.J. Detection of the cyanotoxins L-BMAA uptake and accumulation in primary neurons and astrocytes. Neurotox. Res. 2018, 33, 55–61. [Google Scholar] [CrossRef]
  334. Dias, F.R.P.; de Souza Almeida, R.R.; Sovrani, V.; Thomaz, N.K.; Gonçalves, C.A.; Quincozes-Santos, A.; Bobermin, L.D. Glioprotective effects of resveratrol against BMAA-induced astroglial dysfunctions. Neurotox. Res. 2022, 40, 530–541. [Google Scholar] [CrossRef]
  335. Takser, L.; Benachour, N.; Husk, B.; Cabana, H.; Gris, D. Cyanotoxins at low doses induce apoptosis and inflammatory effects in murine brain cells: Potential implications for neurodegenerative diseases. Toxicol. Rep. 2016, 3, 180–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Forum on Neuroscience and Nervous System Disorders; Board on Health Sciences Policy; Institute of Medicine. Neurodegeneration: Exploring Commonalities across Diseases: Workshop Summary; National Academies Press: Washington, DC, USA, 2013. [Google Scholar] [CrossRef]
  337. Drummond, D.A.; Wilke, C.O. Mistranslation-induced protein misfolding as a dominant constraint on coding-sequence evolution. Cell 2008, 134, 341–352. [Google Scholar] [CrossRef] [Green Version]
  338. Martinez-Miguel, V.E.; Lujan, C.; Espie, T.; Martinez-Martinez, D.; Moore, S.; Backes, C.; Gonzalez, S.; Galimov, E.R.; Brown, A.E.; Halic, M. Increased fidelity of protein synthesis extends lifespan. Cell Metab. 2021, 33, 2288–2300. [Google Scholar] [CrossRef]
  339. Brilkova, M.; Nigri, M.; Kumar, H.S.; Moore, J.; Mantovani, M.; Keller, C.; Grimm, A.; Eckert, A.; Shcherbakov, D.; Akbergenov, R.; et al. Error-prone protein synthesis recapitulates early symptoms of Alzheimer disease in aging mice. Cell Rep. 2022, 40, 111433. [Google Scholar] [CrossRef]
  340. Lant, J.T.; Kiri, R.; Duennwald, M.L.; O’Donoghue, P. Formation and persistence of polyglutamine aggregates in mistranslating cells. Nucleic Acids Res. 2021, 49, 11883–11899. [Google Scholar] [CrossRef] [PubMed]
  341. Tawfik, D.S.; Gruic-Sovulj, I. How evolution shapes enzyme selectivity–lessons from aminoacyl-tRNA synthetases and other amino acid utilizing enzymes. FEBS J. 2020, 287, 1284–1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  342. Kapur, M.; Ackerman, S.L. mRNA translation gone awry: Translation fidelity and neurological disease. Trends Genet. 2018, 34, 218–231. [Google Scholar] [CrossRef] [PubMed]
  343. Zhang, H.; Yang, X.L.; Sun, L. The uniqueness of AlaRS and its human disease connections. RNA Biol. 2021, 18, 1501–1511. [Google Scholar] [CrossRef]
  344. Terrey, M.; Adamson, S.I.; Chuang, J.H.; Ackerman, S.L. Defects in translation-dependent quality control pathways lead to convergent molecular and neurodevelopmental pathology. Elife 2021, 10, e66904. [Google Scholar] [CrossRef] [PubMed]
  345. Song, Y.; Zhou, H.; Vo, M.N.; Shi, Y.; Nawaz, M.H.; Vargas-Rodriguez, O.; Diedrich, J.K.; Yates, J.R.; Kishi, S.; Musier-Forsyth, K.; et al. Double mimicry evades tRNA synthetase editing by toxic vegetable-sourced non-proteinogenic amino acid. Nat. Commun. 2017, 8, 2281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  346. Rodgers, K.J. Non-protein amino acids and neurodegeneration: The enemy within. Exp. Neurol. 2014, 253, 192–196. [Google Scholar] [CrossRef] [PubMed]
  347. Glover, W.; Mash, D.C.; Murch, S.J. The natural non-protein amino acid N-β-methylamino-L-alanine (BMAA) is incorporated into protein during synthesis. Amino Acids 2014, 46, 2553–2559. [Google Scholar] [CrossRef]
  348. Wang, Z.-Q.; Zhang, C.-C. A tRNA t6A modification system contributes to the sensitivity towards the toxin β-N-methylamino-L-alanine (BMAA) in the cyanobacterium Anabaena sp. PCC 7120. Aquat. Toxicol. 2022, 245, 106121. [Google Scholar] [CrossRef]
  349. Okamoto, S.; Esumi, S.; Hamaguchi-Hamada, K.; Hamada, S. β-N-methylamino-L-alanine (BMAA) suppresses cell cycle progression of non-neuronal cells. Sci. Rep. 2018, 8, 17995. [Google Scholar] [CrossRef] [Green Version]
  350. Meyer, J.L.; Gunasekera, S.P.; Brown, A.L.; Ding, Y.; Miller, S.; Teplitski, M.; Paul, V.J. Cryptic diversity of Black Band Disease cyanobacteria in Siderastrea siderea corals revealed by chemical ecology and comparative genome-resolved metagenomics. Mar. Drugs 2023, 21, 76. [Google Scholar] [CrossRef]
  351. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  352. Morais, L.H.; Schreiber, H.L., IV; Mazmanian, S.K. The gut microbiota-brain axis in behavior and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
  353. Silva, D.F.; Candeias, E.; Esteves, A.R.; Magalhaes, J.D.; Fereira, I.L.; Nunes-Costa, D.; Rego, A.C.; Empadinhas, N.; Cardoso, S.M. Microbial BMAA elicits mitochondrial dysfunction, innate immunity activation, and Alzheimer’s disease features in cortical neurons. J. Neuroinflammation 2020, 17, 332. [Google Scholar] [CrossRef] [PubMed]
  354. Esteves, A.R.; Munoz-Pinto, M.F.; Nunes-Costa, D.; Candeias, E.; Silva, D.F.; Magalhães, J.D.; Pereira-Santos, A.R.; Ferreira, I.L.; Alarico, S.; Tiago, I.; et al. Footprints of a microbial toxin from the gut microbiome to mesencephalic mitochondria. Gut 2023, 72, 73–89. [Google Scholar] [CrossRef] [PubMed]
  355. Fu, Y.; Li, A.; Qiu, J.; Yan, W.; Yan, C.; Zhang, L.; Li, M. Effects of the neurotoxin β-N-methylamino-L-alanine (BMAA) on the early embryonic development of marine shellfish and fish. Frontiers Mar. Sci. 2022, 9, 1033851. [Google Scholar] [CrossRef]
Figure 1. Fate of non-canonical amino acids (NCAA) produced by cyanobacteria in the neuronal cell. Misfolded proteins can be refolded via chaperones or degraded; forming of aggregates can lead to neuronal cell death.
Figure 1. Fate of non-canonical amino acids (NCAA) produced by cyanobacteria in the neuronal cell. Misfolded proteins can be refolded via chaperones or degraded; forming of aggregates can lead to neuronal cell death.
Toxins 15 00233 g001
Figure 2. “Cyanopeptides Hypothesis”. NCAA and cyanopeptides production by cyanobacteria that leads to mistranslation, proteostasis, and cellular growth delay may be an ancient mechanism developed initially to regulate the growth of other microorganisms and outcompete them. Outcompeting of symbiotic microorganisms may lead to disease development (examples: (1) Black Band disease of corals associated with cyclic peptides production by Roseophilum sp. cyanobacteria [350]; (2) association of the gut dysbiosis and ND such as Parkinson’s and Alzheimer’s diseases [351,352]). The mitochondria (likely evolved from prokaryotic endosymbionts) and cells that are high-energy demanding, such as neurons and cardiomyocytes, are likely to be affected by NCAA (example: BMAA induce mitochondrial dysfunction in neurons with cardiolipin exposure [353,354] and in embryonic cardiomyocytes leading to cardiac developmental defects [355].
Figure 2. “Cyanopeptides Hypothesis”. NCAA and cyanopeptides production by cyanobacteria that leads to mistranslation, proteostasis, and cellular growth delay may be an ancient mechanism developed initially to regulate the growth of other microorganisms and outcompete them. Outcompeting of symbiotic microorganisms may lead to disease development (examples: (1) Black Band disease of corals associated with cyclic peptides production by Roseophilum sp. cyanobacteria [350]; (2) association of the gut dysbiosis and ND such as Parkinson’s and Alzheimer’s diseases [351,352]). The mitochondria (likely evolved from prokaryotic endosymbionts) and cells that are high-energy demanding, such as neurons and cardiomyocytes, are likely to be affected by NCAA (example: BMAA induce mitochondrial dysfunction in neurons with cardiolipin exposure [353,354] and in embryonic cardiomyocytes leading to cardiac developmental defects [355].
Toxins 15 00233 g002
Table 1. ALS/PD clusters related to environmental factors and cyanobacteria.
Table 1. ALS/PD clusters related to environmental factors and cyanobacteria.
LocationPeriodALS/PD CasesWater QualityToxic Food or Dietary
Components
Key FindingsReference
Guam1945–1969492Mn↑Cycad flourCycad toxic effect;
Biochemical and neuropathologic abnormalities in ALS/PD diagnosed locals
[98]
Guam1940s–1960s--Cycad flour, flying foxes; food containing phytotoxinsAccumulation of cycad neurotoxins (BMAA, cycasin) in flying foxes;
Flying foxes consumption → ALS-PDC
[99]
Guam and other Mariana Islands1956–198039--Similar genotypic composition of Chamorros on all the Mariana Islands but different
mortality rates of ALS/PD on Saipan than on Guam;
Environmental factors of ALS > genetic
[100]
Guam, Canada-23HABs: BMAACycad flour, flying foxes (for Guam)BMAA in tissues from frontal cortex;
BMAA-containing food relates to ALS/PDC;
HABs → cyanobacterial contamination water supplies →BMAA biomagnification
[101,102]
Kii Peninsula,
Japan
1961>4Ca, Mg, Na, KHCO3, Cl ↓-Low mineral content in water supplies possibly leads to ND; > ALS—Mitogawa area[103]
KII Peninsula, Japan197240Mn↑-Possible association of Mn to ALS. [104]
Skaraborg, Sweden1973–198470 males--Cluster of MND in Skaraborg;
Agricultural occupation → MND risk.
[105]
Two Rivers, Small Wisconsin, USA1975–19836-fishPolychlorinated biphenyl
Contaminated fish consumption → ALS risk.
[106]
France1975–199918--ALS cluster in south-eastern France;
Infections or environmental factors of ALS > genetic.
[107]
Italy1980–2001634--16 ALS clusters;
Low efficiency in detoxification systems;
Environmental factors of ALS (toxins)
[108]
Finland1985–1995576Pb, Cd, Zn↑-Two ALS clusters;
Environmental factors of ALS.
[109]
Enfield, NH, northeastern USA1990–2007278HABs: BMAA, MCFish, shellfishHigh ALS incidence near Lake Mascoma;
Chronic exposure to cyanotoxins → ALS;
Combined impact of multiple cyanotoxins.
[95]
Iraq, Saudi Arabia1991–200148BMAA-48 ALS cases in Persian Gulf war veterans linked to
desert’s crust contains BMAA;
Aerosolization of cyanobacteria → inhalation of dust → BMAA exposure
[110,111,112]
Southern France, Hérault district1994–2009381HABs: BMAAshellfishALS cluster in Thau lagoon; Association with
high concentrations of BMAA in mussels and oysters
[113]
Northern New England, USA1997–2009688HABs, [CH3Hg]+-11 clusters of ALS grouped in 4 regions;
Location of ALS cases are close to water bodies where HABs occurs;
Environmental factors → ALS risk
[114]
Northern New England, USA1997–2009>800HABs-HABs → water-quality → ALS risk[115]
Northern New England, USA1999–2009-HABs: BMAA-Mapping cyanobacterial HABs for northern New England lakes; Cyanotoxins increase ALS risk.[97]
Western NH, USA--HABs: BMAAfishHigh concentrations of BMAA and DAB were found in the Lake Mascoma fish;
BMAA, DAB, AEG in the air filters;
ALS linked to BMAA.
[96]
France2003–201172HABs: BMAA Nine ALS clusters;
ALS linked to BMAA.
[116]
South Korea2005–2017-HABs: BMAA mycrocystin, and other cyanotoxins-HABs severity → ND occurrence;
HABs→ long-term impacts on human health
[117]
Guadeloupe1996–201163--The highest incidence of ALS - Marie-Galante island;
Environmental factor(s) → ALS risk
[118]
Northern and Southern Italy2002–201295-dietary supplements Private wells using → ALS risk↑;
Amino acid supplements → ALS risk
[119]
Annapolis, Maryland, USA20133HABs: BMAAblue crabHigh concentrations of BMAA in the crabs originated Chesapeake Bay
HABs exposure → ALS occurrence
[120]
Table 2. Chemical detection methods for cyanotoxins in water samples.
Table 2. Chemical detection methods for cyanotoxins in water samples.
CyanotoxinsDetection
Techniques
SensivityReference
LODLOQ
1.MC-LR and 2 congenersUHPLC-MS/MS0.02–0.04 ng/mL-[185]
2.MC-LR and 11 congenersUHPLC-MS/MS-0.2 µg/L[186]
3.MC-LR and 4 congenersLC-MS/MS0.005–0.0817 µg/L0.005–0.0817 µg/L[187]
Nodularin0.0048 µg/L0.0048 µg/L
Anatoxin-a0.0001 µg/L0.0004 µg/L
Cylindrospermopsin0.0001 µg/L0.0004 µg/L
4.BMAAUHPLC-MS/MS0.02 pg/µL0.05 pg/µL[188]
2,4-DAB0.04 pg/µL0.13 pg/µL
5.MC-LR and 7 congenersLC-MS/MS-0.04–0.5 µg/L[189]
Anatoxin-a-0.02 µg/L
Cylindrospermopsin (and deoxyCYN)-0.01–0.02 µg/L
Saxitoxins (4 congeners), GTX (5 congeners), decarbamoylgonyautoxin, N-sulfogonyautoxins-1 and -2-0.1–2 µg/L
6.MC-LR and 11 congenersHPLC-MS/MS0.01 ± 0.01–0.19 ± 0.2 μg/L0.04 ± 0.04–0.64 ± 0.65 μg/L[190]
Nodularin0.04 ± 0.02 μg/L0.13 ± 0.06 μg/L
7.MC-LR and 2 congenersHPLC-UV/PDA3–4 μg/L9–13 μg/L[191]
8.MC-LR and 2 congenersHPLC-HRMS0.002 μg/L-[192]
9.Anatoxin-a,HILIC-MS/MS0.004 ng/mL0.01 ng/mL[193]
Cylindrospermopsin0.07 ng/mL0.23 ng/mL
Saxitoxin0.01 ng/mL0.04 ng/mL
MC-LR and 4 congenersRPLC- MS/MS0.02–0.08 ng/mL0.07–0.28 ng/mL
Nodularin0.05 ng/mL0.18 ng/mL
10.MC-LR and 5 congenersUHPLC-MS/MS-0.046–0.052 µg/L[194]
Nodularin-0.049 µg/L
Cylindrospermopsin-0.052 µg/L
11.BMAAUHPLC-HRMS5 ng/L10 ng/L[195]
DAB3 ng/L5 ng/L
AEG2 ng/L5 ng/L
BAMA5 ng/L10 ng/L
12.MC-LR and 5 congenersHPLC-MS/MS0.0003–0.0009 µg/L-[196]
Cylindrospermopsin0.0005 µg/L-
Saxitoxin, dcSTX0.0009–0.0013 µg/L-
13.BMAALC-MS/MS10 ng/L-[197]
14.MC-LR and 5 congenersLC-MS/MS0.04–0.8 μg/L0.1–2.3 μg/L[198]
Nodularin0.3 μg/L0.9 μg/L
Anatoxin-a0.27 μg/L0.81 μg/L
Cylindrospermopsin0.14 μg/L0.4 μg/L
15.MC-LR and 2 congenersHPLC-DAD0.08–0.15 µg/L-[199]
16.MC-LR and 11 congenersLC-MS/MS0.001–0.007 μg/L0.003–0.020 μg/L[200]
Nodularin0.002 μg/L0.006 μg/L
Anatoxin-a0.001 μg/L0.003 μg/L
Cylindrospermopsin0.001 μg/L0.003 μg/L
17.MCsUPLC-MS/MS0.005 µg/L-[201]
Anatoxin-a0.02 µg/L-
Cylindrospermopsin0.02 µg/L-
Saxitoxin0.8 µg/L-
BMAA0.03 µg/L-
18.BMAALC-MS/MS0.030 μg/L0.096 μg/L[202]
19.MC-LR and 11 congenerson-line SPE – UHPLC-HRMS5–37 ng/L15–130 ng/L[203]
Anatoxin-a15–18 ng/L50–60 ng/L
Homoanatoxin-a11–12 ng/L30–40 ng/L
Cylindrospermopsin41–53 ng/L130–170 ng/L
20.MC-LR and 1 congenerHPLC-DAD0.2-0.3 μg/L-[204]
21.Anatoxin-aUHPLC-MS/MS1.1 ng/L2.5 ng/L[205]
Cylindrospermopsin10.9 ng/L21.7 ng/L
Saxitoxins (4 congeners)3.5-9 ng/L7.1–26.9 ng/L
GTX (7 congeners)18.5–54.5 ng/L42.2–227.6 ng/L
22.MC-LR and 7 congenersUHPLC-MS/MS0.1 µg/L0.5 µg/L[181]
Nodularin0.1 µg/L0.5 µg/L
23.MC-LR and 2 congenersUHPLC-MS/MS0.1 µg/L24 µg/L[206]
24.CylindrospermopsinUHPLC-MS/MS0.029 μg/L0.091 μg/L[207]
25.Saxitoxins (4 congeners)on-line SPE–HILIC-HRMS0.72–3.9 ng/L2.4–13 ng/L[208]
26.MC-LR and 1 congenertandem-SPE-HILIC-MS/MS0.0012–0.0021 μg/L0.004–0.007 μg/L[209]
Nodularin0.0021 μg/L0.007 μg/L
Anatoxin-a0.03 μg/L0.1 μg/L
Cylindrospermopsin0.0012 μg/L0.004 μg/L
BMAA0.015 μg/L0.05 μg/L
DAB0.009 μg/L0.03 μg/L
AEG0.006 μg/L0.02 μg/L
27.BMAALC-MS/MS2.8 ng/mL-[210]
DAB1.7 ng/mL-
28.BMAAon-line SPE-UHPLC-HRMS10 ng/L-[211]
BAMA10 ng/L-
DAB10 ng/L-
AEG5 ng/L-
29.BMAAUHPLC-MS/MS-2.5 µg/L[212]
AEG-2.5 µg/L
DABA-5 µg/L
30.MC-LR and 7 congenersUHPLC-MS/MS (ESI)0.02–0.2 µg/L0.05–0.5 µg/L[213]
LOD—limit of detection; LOQ—limit of quantification; UHPLC—ultra high-performance liquid chromatography; HILIC-MS/MS—hydrophilic interaction liquid chromatography-tandem mass spectrometry; RPLC-MS/MS—reverse phase chromatography tandem mass spectrometry; UV/PDA—ultraviolet/photodiode array detection; DAD—diode array detector; ESI—electrospray ionization; SPE—solid phase extraction; BAMA—β-amino-N-methylalanine; GTX—gonyautoxins; dcSTX—decarbamoylsaxitoxin.
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

Nugumanova, G.; Ponomarev, E.D.; Askarova, S.; Fasler-Kan, E.; Barteneva, N.S. Freshwater Cyanobacterial Toxins, Cyanopeptides and Neurodegenerative Diseases. Toxins 2023, 15, 233. https://doi.org/10.3390/toxins15030233

AMA Style

Nugumanova G, Ponomarev ED, Askarova S, Fasler-Kan E, Barteneva NS. Freshwater Cyanobacterial Toxins, Cyanopeptides and Neurodegenerative Diseases. Toxins. 2023; 15(3):233. https://doi.org/10.3390/toxins15030233

Chicago/Turabian Style

Nugumanova, Galina, Eugene D. Ponomarev, Sholpan Askarova, Elizaveta Fasler-Kan, and Natasha S. Barteneva. 2023. "Freshwater Cyanobacterial Toxins, Cyanopeptides and Neurodegenerative Diseases" Toxins 15, no. 3: 233. https://doi.org/10.3390/toxins15030233

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