Next Article in Journal
Polyoxyethylene Group-Dependent Surface Properties and Aggregation Behavior in Oleyl-Based Sulfosuccinate Systems
Previous Article in Journal
Chemical Fingerprints of Honey Fermented by Conventional and Non-Conventional Yeasts
Previous Article in Special Issue
Emerging Ferroptosis Involvement in Amyotrophic Lateral Sclerosis Pathogenesis: Neuroprotective Activity of Polyphenols
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Airborne Cyanobacterial Toxins and Their Links to Neurodegenerative Diseases

by
Zachary James Morris
1,*,
Elijah W. Stommel
2 and
James Spencer Metcalf
1,3
1
Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA
2
Department of Neurology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
3
Brain Chemistry Labs, Jackson, WY 83001, USA
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(11), 2320; https://doi.org/10.3390/molecules30112320
Submission received: 25 February 2025 / Revised: 13 May 2025 / Accepted: 16 May 2025 / Published: 26 May 2025

Abstract

:
Cyanobacteria can produce a wide range of toxins which have acute and chronic adverse health effects. Affecting a variety of mammalian systems, they are generally characterized according to their mode of action and the organs affected. Cyanobacterial neurotoxins are one cyanotoxin class that can negatively affect human health, and representatives of other cyanotoxins classes are increasingly showing neurotoxic effects. Of the various human exposure routes to cyanobacterial toxins, the significance of the airborne and inhalation route requires much greater clarity and understanding. People may be exposed to mixtures of cyanobacterial neurotoxins through the inhalation of sprays and dust, along with the potential to directly enter the central nervous system when crossing the blood-brain barrier. This review aims to summarize the current state of knowledge concerning airborne cyanobacterial neurotoxins, research gaps, health effects, and the need for management practices to protect human and animal health.

1. Introduction

Cyanobacteria are a diverse bacterial phylum which emerged ~3.5 billion years ago [1]. Thought to be responsible for oxygenating the early Earth atmosphere, cyanobacteria helped establish the conditions necessary for the evolution of the current biosphere. Cyanobacteria inhabit a wide range of aquatic and terrestrial habitats and can tolerate varied environmental conditions including hot and cold deserts, oceans, lakes, soil, and thermal features such as hot springs [2,3]. Many cyanobacteria exist as vegetative cells that are metabolically active and capable of reproduction, whereas some genera form differentiated cells such as heterocysts which specialize in nitrogen fixation [4].
Cyanobacteria are keystone organisms in most ecosystems as evidenced by their environmental ubiquity [3,5] and have adapted well to anthropogenic changes to the environment resulting from climate change and pollution [6,7,8,9]. Concerning climate change, increasing average global temperatures are likely to favor the growth of cyanobacteria over other photoautotrophic organisms [8,10]. Similarly, nutrient pollution has also increased [9,11,12], with nitrogen and phosphorous enrichment resulting in eutrophication, and permitting massive cyanobacterial blooms to form, sometimes reaching hundreds of square kilometers in area, such as in Lake Erie [13,14,15]. Often unsightly when environmental conditions deteriorate and blooms break down, cyanobacterial die-offs can lead to adverse water quality issues when bloom material is biologically degraded [16]. As cyanobacterial blooms grow and become more dense, toxic substances synthesized by cyanobacteria become more concentrated [17]. Through observations of wild and domestic animals (such as livestock), associations of cyanobacterial blooms with mass animal deaths have been derived [18]. These deaths are the result of small molecular weight secondary metabolites, known as cyanotoxins, which have adverse short and long-term health implications [19,20,21,22,23].
Many cyanobacteria are capable of synthesizing cyanotoxins which may vary in terms of chemical structure, mechanisms of toxicity, and subsequent adverse health conditions resulting from exposure. Of growing concern are cyanotoxins associated with neurodegenerative conditions such as Amyotrophic Lateral Sclerosis (ALS), Parkinson’s Disease (PD), and various forms of dementia such as Alzheimer’s Disease (AD). Cyanobacteria may synthesize toxins in response to variations in pH, temperature, light intensity, and available nitrogen and phosphorus [24,25,26,27,28,29], in addition to their potential roles as a defense mechanism against grazers or competitors. Exposure to neurotoxicologically relevant cyanotoxins may occur through various exposure routes including food [30,31,32,33,34,35], dietary supplements [36,37], drinking water [28,38], maternal transfer [39,40,41], and inhalation [42,43]. Of these exposure routes, exposure via inhalation is less studied, yet one of the most relevant, as cyanobacteria and cyanotoxins are likely to be easily transported through the atmosphere. As a result, humans and wildlife living near sites of harmful cyanobacterial blooms may be easily and frequently exposed. The purpose of this review is to outline the prominent cyanotoxins associated with neurological impairment and discuss atmospheric transfer and subsequent inhalation as an exposure route, in addition to mitigation strategies to prevent such exposures.

2. Cyanobacterial Neurotoxins

Cyanobacteria synthesize many secondary metabolites which are harmful to humans and wildlife. Several of these secondary metabolites have been studied extensively due to their potency and environmental relevance. This section considers cyanotoxins which may be the most harmful to human health from a neurological perspective, with deleterious effects observed in neuronal and non-neuronal systems (Table 1).

2.1. Anatoxin-a

The alkaloid cyanotoxin anatoxin-a (Figure 1) is synthesized by cyanobacterial genera including Anabaena, Lyngbya, Oscillatoria, Planktothrix, Raphidiopsis, and Woronichinia [91,92,93,94]. Previously called “very fast death factor”, anatoxin-a was characterized as a depolarizing agent in neurological tissues from multiple organisms [91,92,93,94] and bears some structural similarity to neurological stimulants such as cocaine [44,95]. Synthesis of anatoxin-a and variants may be positively correlated with environmental nitrogen and phosphorous concentrations [96,97,98], although some studies show that anatoxin-a synthesis may peak with moderate nitrogen stress [99]. Anatoxin-a mimics acetylcholine, a stimulatory neurotransmitter which interacts mainly with nicotinic acetylcholine receptors [44,45]. As acetylcholinesterase is unable to degrade anatoxin-a, neuronal stimulation cannot be attenuated, resulting in overstimulation [44]. In sufficient doses, exposure to anatoxin-a can result in paralysis, asphyxiation, abnormal muscular contraction, and death. Anatoxin-a is thought to be responsible for the deaths of animals such as domestic dogs and aquatic birds [100,101]. Apart from the known neurological effects, anatoxin-a is thought to induce cell death by apoptosis in rat thymocytes and fish lymphocytes, which may be caused by elevated concentrations of reactive oxygen species (ROS) upon exposure [46,47]. Fish lymphocytes exposed to 0.01 mg/L anatoxin-a also displayed significantly lower concentrations of enzymes which alleviate ROS stress, such as superoxide dismutase (SOD), catalase, glutathione reductase (GR) and glutathione peroxidase (GP) [47].

2.2. Guanitoxin

Guanitoxin (Figure 2) is an organophosphate synthesized by the cyanobacterial genera Dolichospermum (Anabaena), Sphaerospermopsis, Aphanizomenon, Cuspidothrix and possibly other genera such as Microcoleus [102,103,104]. Comparing the toxicity of guanitoxin with anatoxin-a revealed similar toxicological effects in a variety of tissues, in addition to profuse salivation in test organisms [44,92,102]. Formerly known as anatoxin-a (S) (“S” denoting salivation), the nomenclature was changed to “guanitoxin” due to significant differences from anatoxin-a in terms of structure, mechanism of toxicity, and lethal dose [48]. Unlike anatoxin-a, which mimics acetylcholine, guanitoxin prevents acetylcholine degradation by inhibiting acetylcholinesterase [102]. This inhibition can result in neurotoxicity by overstimulation, manifesting as paralysis, asphyxiation, and death. Although relatively little is known regarding the toxic effects of guanitoxin apart from inhibition of acetylcholinesterase, fish exposed to guanitoxin-producing cyanobacteria displayed changes in ROS-associated enzymes such as SOD, CAT, and glutathione-S-transferase (GST), the presence of micronuclei, and osmoregulatory disorders [49].

2.3. Saxitoxin

Saxitoxins (Figure 3) are synthesized by cyanobacterial genera such as Lyngbya, Aphanizomenon, Dolichospermum, Planktothrix, and Cylindrospermopsis [105,106]. The general structure consists of trialkyl tetrahydropurines with variable regions throughout the molecule [50,51]. Saxitoxins can be classified as C and G toxins, as well as the LW toxins produced by L. wollei [52]. Saxitoxins can block voltage-gated ion channels and prevent neural signaling [50,52]. The main target of saxitoxins are voltage-gated sodium channels, which take part in action potential generation. However, some variants of saxitoxin can block voltage-gated potassium and calcium channels, as well as nitric oxide synthases [50]. Toxic to many species, saxitoxins synthesized in marine environments are known to accumulate in shellfish and are the cause of paralytic shellfish poisoning [106,107,108]. Saxitoxins may also enact neurotoxicity through oxidative stress. Various organisms including rats, fish, Caenorhabditis elegans, and Daphnia magna show altered expression of enzymes such as catalase and synthesis of metabolites such as glutathione in neurological tissue when exposed to saxitoxins [53,54,55,109].

2.4. Neurotoxic Amino Acids

Neurotoxic amino acids (Figure 4) such as β-N-methylamino-L-alanine (BMAA), N-(2-aminoethyl)-glycine (AEG) and 2,4-diaminobutyric acid (DAB) are thought to be synthesized by most, if not all, cyanobacterial genera [30,110,111,112,113,114]. The synthesis of BMAA, AEG, and DAB may be linked to concentrations of available nitrogen, phosphorus, light, and pH [24,25,26,27,29]. BMAA can bioaccumulate through trophic levels in nature [30,31,32,33,115] and may accumulate in various regions of the brain in humans and other mammals [30,31,116]. In the eukaryotic cell, BMAA competes with the amino acid L-serine for misincorporation into proteins [56,57] and may cause protein misfolding and aggregation [116]. BMAA may also cause excitotoxicity by interacting with glutamate receptors [58,59,117,118] and is thought to selectively damage motor neurons [59,119,120]. Symptoms of ALS, PD, and dementia are associated with chronic exposure to BMAA in humans and non-human primates [30,56,121]. BMAA may also affect the proliferation of neural cancer cells, albeit with different effects observed depending on the cell line [122].
Some studies have shown that both DAB and AEG are more neurotoxic than BMAA, although toxicity can vary depending on the test organism and choice of bioassay [59,123,124,125]. AEG and DAB may be more neurotoxic than BMAA when tested in cortical cultures [59]. Although the exact mechanisms of AEG toxicity are not clear, DAB has been shown to be more excitotoxic than BMAA in the nervous systems of some invertebrates [125].
Exposure to neurotoxic amino acids may also induce toxicity by oxidative stress, epigenetic changes, impaired neurite development, apoptosis, neuronal inflammation, and morphological changes in neuronal mitochondria [59,60,61,62,63,64]. Like other toxins, BMAA is associated with elevated concentrations of enzymes associated with oxidative stress observed in some organisms, and elevated concentrations of ROS as well as DNA damage in neural stem cells [60,62]. BMAA and AEG can inhibit the transport of the glutathione precursor cystine into primary neuronal cultures [59]. Neuroinflammatory biomarkers such as elevated levels of COX2, nuclear NF-kB, TNF-alpha, and IL-1 beta expression are also associated with BMAA exposure. Finally, consistent with its role as a causative agent of ALS, exposure to BMAA is correlated with the presence of TDP-43 aggregates [63,64].

2.5. Microcystin

Microcystins are cyclic heptapeptides synthesized by cyanobacterial genera including Microcystis, Planktothrix, and Nostoc [126,127]. Over 240 microcystin congeners have been identified, due largely to amino acid substitutions, including at two positions in the peptide structure, “X” and “Z”, giving rise to the nomenclature of these toxins [128]. Microcystin synthesis is correlated with concentrations of available nitrogen and phosphorous [29,129]. Microcystins are hepatotoxins, inhibitors of phosphatases such as PP2A and tumor promoters [130,131,132]. Several mechanisms exist by which microcystin elicit toxicity. Microcystin-LR (MC-LR) (Figure 5) can cause ROS stress as evidenced by the dose dependent elevation of hydrogen peroxide in various cell types as well as altered expression of ROS-associated enzymes [65,66,67,68,69]. MC-LR can also induce neuroinflammation, as changes in expression of proinflammatory cytokines such as tumor necrosis factor alpha (TNF-alpha), interleukin-1 beta (IL-1 beta) and interleukin-6 (IL-6), along with enzymes such as nitric oxide synthase (NOS) and COX-2 have all been associated with MC-LR exposure [70,71,72,73,74,75]. The ability of microcystin to inhibit PP2A, whose function is necessary for proper neuronal function, could explain some neurological symptoms associated with MC exposure [133,134,135]. Chronic exposure to MC-LR can lead to disruption of the blood brain barrier (BBB) and neuroinflammation [71,72,136]. Microcystin effects on the BBB are likely brought on by disruption of tight junctions, increasing expression of matrix metalo-proteases (MMPs) and low levels of tight junction proteins such as Occludin and Claudin 5 [72]. Microcystins have also been shown to alter neuronal signaling in Aplysia and rats, albeit by different mechanisms. In Aplysia, MC-LR may prolong inward ion currents induced by serotonin and cyclic adenosine monophosphate [76], while in the rat hippocampal dentate gyrus, MC-LR inhibits long-term potentiation (LTP), leading to inhibition of spatial memory [77]. In rats, MC-LR may be responsible for the formation of Lewy bodies and cell-to-cell transfer of alpha-synuclein in the nervous system [71].

2.6. Cylindrospermopsin

Cylindrospermopsin (Figure 6) is an alkaloid cyanotoxin, consisting of a tricyclic guanidine moiety and a hydroxymethyluracil [137]. Synthesized by several cyanobacterial genera including Cylindrospermopsis, Umezakia, Aphanizomenon, Raphidiopsis, and Anabaena [78], cylindrospermopsin is of growing concern due to its potent cytotoxicity. One mechanism of cylindrospermopsin toxicity, translation inhibition, may lead to the buildup of reactive oxygen species (ROS), as free radical scavengers such as glutathione may not be synthesized, potentially leading to cell death by apoptosis and DNA damage [79,80,81,82,83,84,85]. It should be noted that the toxic effects of cylindrospermopsin may change depending on cell type [22,138,139,140,141,142]. While cylindrospermopsin has deleterious effects on multiple cell types including neurons [78], less is known regarding potential mechanisms of neuronal toxicity. Hinojosa et al. showed decreased viability in murine primary neuronal cultures exposed to cylindrospermopsin and proposed that cylindrospermopsin may disrupt synaptic integrity [86]. Cylindrospermopsin can also cross the BBB and disrupt acetylcholine activity in certain fish species [87,88,89]. Some evidence shows that cylindrospermopsin can cause neuroinflammation in BV-2 and N2a cells with elevated levels of TNF-alpha in each upon exposure [90].

3. Cyanotoxin Exposure Routes

3.1. Exposure to Cyanotoxins from Aquatic Environments

Globally, cyanotoxins have been implicated in intoxications of humans and wildlife. In 2014, roughly 500,000 Ohioans living in the Toledo area were without potable water due to the release of microcystins such as MC-LR from a large cyanobacterial bloom in the Western Basin of Lake Erie [28,38,143]. Animals which use cyanotoxin-contaminated water for drinking or as a habitat are also at risk of intoxication. For example, cyanotoxin exposure was thought to be responsible for the deaths of African elephants that drank from water sources known for cyanobacterial blooms [144]. Guanitoxin has been noted as the cause of waterfowl poisonings in Danish Lakes [52,145], and the deaths of livestock have been reported after consumption of water containing cylindrospermopsin [22]. Cyanotoxins may also be responsible for marine cetacean deaths [146].
Edible marine invertebrates such as shrimp and mollusks can accumulate BMAA in their tissues [33,147], and saxitoxins in shellfish may lead to shellfish poisoning [106,108]. Fish tissues can accumulate cyanotoxins in both marine and freshwater environments. BMAA was detected in the tissues of a variety of edible fish species in the Black Sea, as well as in carp from Lake Mascoma, New Hampshire amongst others [115,147,148,149]. More recently, BMAA has been detected in Lake Erie fish, which are consumed after being caught recreationally and commercially [150].
Cyanotoxins originating from aquatic environments may also impact terrestrial environments, albeit indirectly, as water contaminated with cyanotoxins may be used in agriculture. As cyanotoxins can accumulate in a variety of plants, exposure may occur through the consumption of contaminated plant matter after irrigation with cyanotoxin-containing water [30,151,152,153,154,155,156,157,158,159,160]. Mohammad et al. showed that BMAA contaminated water used to irrigate edible plants accumulated to detectable levels in both free and protein-associated states [158]. Microcystin can accumulate in edible plants such as lettuces, radishes, and carrots after irrigation with Microcystis and/or microcystin-containing water, which may hinder plant development depending on the concentration and duration of exposure [155]. The use of aquatic organisms for commercial animal feed can also lead to accumulation of BMAA in livestock [33].

3.2. Exposure to Cyanotoxins from Terrestrial Environments

Terrestrial cyanobacteria are capable of synthesizing many of the same toxins as aquatic species. On Guam, an unusually high number of individuals showed symptoms of Amyotrophic Lateral Sclerosis/Parkinsonism Dementia complex (ALS/PDC) which includes characteristic symptoms of ALS, AD, and PD [161,162]. Cox et al. examined Nostoc species inhabiting the coralloid roots of the cycad Cycas micronesica which were found to produce BMAA. BMAA was present at high concentrations in the seeds of the cycad which were consumed by flying foxes native to Guam. BMAA in the flying foxes and cycad seeds accumulated >1000-fold compared to that detected in the Nostoc associated with the cycad coralloid root. Cycad flour prepared by native Chamorro people also contained free and protein bound BMAA. Those who consumed flying foxes and cycad seed flour over extended periods of time accumulated BMAA in brain tissues [30,31].
Another possible exposure route to terrestrial cyanotoxins are biological soil crusts. Biological soil crusts are important in arid environments where they stabilize the uppermost soil surfaces, binding soil particles together and providing a physical barrier preventing erosion [163,164]. As cyanobacteria may constitute a large proportion of the biomass found in soil crusts [103,124,165,166,167,168], cyanotoxins have the potential to occur at varying concentrations and types depending on environmental conditions and the composition of genera present [167]. In the desert soils of Qatar, for example, prominent cyanobacterial genera included Microcoleus, Phormidium, and Chroococcus [166,168,169] and cyanotoxins such as microcystin, guanitoxin, BMAA, DAB, and AEG have been found in crust material [103]. In the Arctic, microcystin and anatoxin-a have been detected in biological soil crusts [170]. Biological soil crusts from loess deposits in Iran showed great diversity of cyanobacterial genera, but varying toxicity [167].
Other terrestrial sources of cyanotoxins include dry lake beds and cyanobacteria found in arctic regions. Dry and receding lake beds such as the Great Salt Lake in the State of Utah are thought to be sources of airborne BMAA, AEG, and DAB [171]. One final source of terrestrial cyanotoxins comes from cyanobacterial mats, such as those found in the Antarctic from which BMAA and microcystins were detected [172].

3.3. Exposure to Cyanotoxins from Food and Dietary Supplements

Dietary supplements containing or consisting entirely of cyanobacteria are popular due to their high nutrient content. Species of the genus Arthrospira are consumed in Central Africa [173] and are marketed in the United States along with Aphanizomenon flos-aquae as the dietary supplement “spirulina”. Despite being considered non-toxic, representatives of these cyanobacteria can synthesize a variety of toxins or may be contaminated with toxic cyanobacterial species before processing for consumption. In a screen for 37 cyanotoxins in supplements which included spirulina and Aphanizomenon flos-aquae, Fontaine et al. detected microcystins, DAB, anatoxin-a, and beta-amino-N-methylamine (BAMA) [35]. Roy-Lachapelle et al. also screened dietary supplements for cyanotoxins, with several detected above levels considered tolerable for daily intake [36]. However, as certain toxins such as BMAA have the potential to bioaccumulate in human tissues [30,31], repeated consumption of dietary supplements containing cyanobacteria may serve as a route of chronic exposure and potentially lead to neurological conditions. Metcalf et al. examined Arthrospira consumed by people in the African nation of Chad and provided evidence for the presence of microcystins and DAB [173]. Nostoc species such as Nostoc commune and Nostoc flagelliforme are consumed whole or as fa cai noodles in Peru and China respectively, and have both been shown to contain BMAA [34,174].

3.4. Exposure to Cyanotoxins in the Atmosphere

Although exposure to cyanotoxins from aquatic and terrestrial environments, as well as through food and dietary supplements, is well documented, exposure to cyanobacteria and cyanotoxins via the atmosphere is less well studied. Cyanobacteria may constitute a significant proportion of aerial microbial communities, as they can withstand a range of environmental conditions [169,175,176,177,178,179,180,181]. This presents unique health risks, as air currents can carry particles containing cyanobacteria and cyanotoxins over long distances [182], potentially being able to adversely affect individuals over broad spatial distributions. Cyanobacteria have also been detected in the atmosphere of indoor facilities, implying that air filtration methods may not be adequate at preventing airborne exposure [177,183,184,185,186]. Various mechanisms exist by which cyanobacteria and cyanotoxins can enter the atmosphere, and these can vary depending on whether their origin is terrestrial or aquatic.
Bubbles can be generated by wave action or the result of precipitation hitting the surface of water. After forming below the surface, the outer film of bubbles can accumulate bacteria [187]. Bacterial accumulation on bubbles is dependent on bubble size, bacterial concentrations in the water, and the distance the bubble travels before reaching the surface, with larger bubbles that travel greater distances accumulating more bacteria [188,189]. As the bubble bursts upon hitting the surface of the water, bacteria concentrated on the bubble are released by propulsion of jet and film drops. Jet drops are generated from the release of water from the inside of the bubble. Film drops are composed of drops from the surface of the water upon bubble bursting. Both types of drops are capable of ejecting bacteria and potentially toxic molecules into the atmosphere [187]. For example, microcystin congeners can enter the atmosphere with the bursting of bubbles, creating lake spray aerosols (LSAs) [190,191].
Terrestrial sources of atmospheric cyanobacteria and cyanotoxins are less well understood, but may include soil, biocrusts, plant matter, and lichen [30,103,165,166,170,182,192,193,194]. Physical disruption of soil surfaces and biocrusts can disperse cyanobacteria into the atmosphere [182]. In arid regions, dust particles containing cyanobacteria and associated toxins may enter the atmosphere after being swept up by air currents moving over soil surfaces. Furthermore, up to 109 bacterial cells may be present in a single gram of desert soil, suggesting that microbes will be more prevalent in the atmosphere after dust storms [169,195]. Some cyanotoxins have been detected on farmland and in groundwater [194,196], presenting the possibility of dispersal during agricultural practices. Some terrestrial Nostoc species form large colonies which are subject to desiccation, while others associate with fungi with forming lichen [192,193]. Desiccated or dead Nostoc and lichen as well as other abundant terrestrial cyanobacteria and cyanotoxins may also enter the atmosphere if they are physically disturbed.

4. Cyanobacteria and Cyanotoxins in the Atmosphere

Microalgae were observed as being present in the atmosphere as early as 1844 [197]. Since that time, the number of microorganisms identified in the atmosphere has greatly increased, including many cyanobacterial genera, with their presence dependent on both cellular and environmental characteristics [43,176,178,179,198]. Small cells may be more prevalent in the atmosphere according to some studies, as their small size may influence the ease with which they are swept up by air currents. Pico-cyanobacteria, which are 0.2–2 µm in diameter, are sometimes the predominant bacteria detected in aerosols [176,178]. Microcystis cells, which are between 3 and 4 µm in diameter, have been found during air sampling of the Baltic Sea and other habitats [199,200]. Bacterial cells of similar size have also been detected in mucosal samples (i.e., lung and nasal) from hospital patients [43], and Microcystis species including Microcystis aeruginosa were documented by Genitsaris et al. from inland samples [199]. Apart from cell size, members of the genus Microcystis may be more prone to aerosolization, as synthesis of gas vesicles allows them to float near the surface of aquatic environments, placing them near air currents moving across the surface of the water [21,201].
Although smaller cyanobacteria may predominate in the atmosphere, larger cyanobacteria may still be detected. Genitsaris et al. cataloged many cyanobacterial genera from atmospheric samples, which included Lyngbya, Phormidium, and Oscillatoria that form long filamentous trichomes [199]. Lyngbya species, which can reach up to 6 µm in width and commonly benthic [202], have been reported in tropical regions, but are more distinguishable in the atmosphere after harsh weather conditions such as monsoons [178,179]. Terrestrial Nostoc species have also been detected in aerosols [179,199]. 17 cyanobacterial species were detected in studies performed in Texas (USA) [175]. Some microalgae and cyanobacteria in this study were detected in samples taken from an airplane, implying that cell size may not be a significant factor for aerosolization and dispersal from terrestrial environments [175].
It may be assumed that greater wind speeds result in higher concentrations of cyanobacteria and cyanotoxins in the atmosphere. While bioaerosols may be positively correlated with wind speeds greater than 4 m/s, some studies suggest that wind speed may be negatively correlated with bioaerosols below 4 m/s [203,204]. Trout-Haney et al. observed a slightly negative correlation for wind speed and concentrations of aerosolized pico-cyanobacteria [176]. Wind speed may also be neutral under some circumstances, as Wood and Dietrich did not observe any correlation between wind speed and atmospheric concentrations of microcystin or nodularin [205]. Climatic factors can also influence cyanobacterial aerosolization. In India, for example, cyanobacterial genera detected in the atmosphere may change depending on the season, which is likely correlated with changes in temperature, precipitation, and humidity [179]. As mentioned above, certain genera were observed in tropical regions only after monsoons. Conditions characteristic of monsoons, such as high wind speed and precipitation, may provide the physical force necessary to aerosolize great numbers of bioparticles containing cyanobacteria [198].
Along with cyanobacteria, several cyanotoxins have been detected in air samples. Atmospheric cyanotoxins have been detected around water bodies known for harmful cyanobacterial blooms, as well as terrestrial environments. For example, microcystin and nodularin have been detected around several lakes worldwide during cyanobacterial blooms [205,206,207,208,209,210]. Anatoxin-a has been detected near a pond known for hypereutrophic conditions and harmful cyanobacterial blooms in New England [211]. BMAA has been detected in atmospheric samples around Lake Mascoma and surrounding water bodies in New England [115,198]. As various cyanotoxins have been detected in farm soil and dry lake beds such as the Great Salt Lake [171,194], inland soils may be a potential source of aerial cyanotoxins. Guanitoxin, to our knowledge, has not been detected in air samples. However, the presence of guanitoxin in desert biocrusts which may release cyanotoxins upon disruption may lend credence to guanitoxin possibly being present in the atmosphere derived from arid environments [103,166]. Limited data exists on the presence of aerosolized saxitoxin. A possible explanation is that some cyanobacteria that synthesize saxitoxin, such as L. wollei, are benthic, which may make entry into the atmosphere more difficult without extreme weather conditions (discussed above). Dabny did not detect saxitoxins in aerosol samples taken from a freshwater environment [212] however, other studies such as Yu et al. were able to detect saxitoxins in aerosol samples from a marine environment [213].

5. Health Implications of Atmospheric Cyanobacteria and Cyanotoxins

5.1. Possible Consequences of Exposure to Atmospheric Cyanobacteria and Associated Neurotoxins

Cyanobacteria that enter the atmosphere may present various health concerns. Cyanotoxins such as microcystin and BMAA, detected around some New Hampshire lakes, have been speculated to be causative agents of ALS cases in the surrounding area [115,214]. As mentioned above, cyanobacteria found in lung and nasal samples of hospital patients in New Hampshire were similar in size to Microcystis species [43]. In some cases, patients suffering from ALS were reported to live within 0.5 miles of lakes known for regular cyanobacterial blooms [214].
Cyanobacteria and cyanotoxins may also impact the health of individuals recreating on or near aquatic cyanobacterial blooms. Cheng et al. detected microcystin on air filters worn on the lapel of participants recreating near lakes with cyanobacterial blooms, implying that atmospheric microcystin was in close enough proximity to the face to be inhaled [208]. Backer et al. monitored the health of participants recreating on or near lakes with and without cyanobacterial blooms. While Backer et al. did not find a statistically significant difference in the number of participants who reported differences in health before and after recreation, many participants reported symptoms of dermal, respiratory, and digestive discomfort only after recreation, which were not experienced by participants recreating on control lakes with no visible bloom [207]. A separate study by Backer et al. showed similar results, with some participants only reporting health changes after recreation on bloom-containing lakes [206]. In both studies, microcystin was detected in atmospheric samples [206,207], supporting the idea that aerosolized microcystin may be a causative agent for the observed health changes.

5.2. Atmospheric Cyanobacteria and the Spread of Toxins

It should be noted that aerosolized cyanobacteria may spread hazardous substances other than cyanotoxins. Toxins such as polycyclic aromatic hydrocarbons (PAHs) and heavy metals may also be spread by phytoplankton [215,216,217], PAHs are flat planar molecules that can enter the environment through spilled fuel or biochar and are thought to be carcinogenic [218,219]. Heavy metals are present in many environments and may cause neurological complications such as ALS [220,221,222]. Studies performed on Microcystis aeruginosa support the hypothesis that both PAHs and heavy metals adhere to the cell surface, with some heavy metals possibly able to enhance PAH adsorption [217]. Tao et al. (2014) showed that metal salts such as copper nitrate and silver nitrate at concentrations of 500–5000 µmol/L enhanced the adsorption of PAHs such as phenanthrene onto M. aeruginosa cells [223]. As cyanobacteria have been detected in lung and nasal samples [43], it may be possible that exposed individuals may be inhaling multiple toxins.

5.3. Occupational Exposure

Occupational exposure to cyanobacteria and cyanotoxins has been reported in some instances, but documentation of exposure specifically through inhalation is limited. Stewart et al. discussed several potential instances where cyanobacterial exposure may occur in laboratory settings and mass culture facilities, with those working with dehydrated or powdered cyanobacteria possibly being more at risk [224]. Other examples included soldiers performing canoeing exercises at a UK waterbody supporting a cyanobacteria bloom, resulting in atypical pneumonia in some recruits [225,226]. As these exercises involved full body immersion in cyanobacteria contaminated water, ingestion was the most likely source of exposure, although inhalation may have also occurred, which could explain the occurrence of pneumonia in some recruits [225,226].
In certain parts of the world, significant areas of land are covered with cyanobacterial biocrusts [227]. Exposure to these crusts and cyanotoxins may result in a higher risk of developing neurological conditions. For example, unusually high rates of ALS have been observed in deployed military personnel who served in the first Persian Gulf War (Operation Desert Shield) from 1990 to 1991 versus those who received the same training and were not deployed [228,229,230,231]. While some evidence suggests that instances of ALS in active service members may be the result of administered anti-chemical warfare agents [231], contributions from cyanotoxins such as BMAA, AEG, DAB, microcystins, and guanitoxin present in desert biocrusts becoming airborne and inhaled after physical disruption cannot be discounted [103,124,166].

5.4. Cyanobacteria and Cyanotoxins in Indoor Environments

While little is known about how atmospheric cyanobacteria and cyanotoxins may chronically or acutely adversely affect human health, more information is available demonstrating the proximity of potentially affected individuals to airborne contaminants. Particularly concerning is that cyanobacteria are amongst the most abundant photosynthetic microbes detected in the atmosphere of indoor environments [184,185,186,215]. As many as 40 algal taxa have been detected on indoor dust particles [184]. Chu et al. found that of 26 taxa detected indoors, cyanobacteria were the most abundant, with predominant genera including those possibly originating from terrestrial environments such as Phormidium [184]. Some genera potentially originating from aquatic environments, including Anabaena, have also been detected indoors [184,186].
As cyanobacteria may be transported to indoor environments on dust particles, it may be inferred that the same is true with cyanotoxins. Since soil is a heterogenous mixture of chemically diverse materials [232], cyanotoxins could enter indoor environments via adhesion to soil-originating dust. For example, some terrestrial cyanobacteria such as Nostoc are capable of synthesizing multiple toxins, including microcystin [127], and it may be inferred that cyanotoxins could enter the atmosphere on dust particles from areas with high concentrations of Nostoc. Further sources of indoor airborne cyanobacteria and cyanotoxins include areas that use dried cyanobacteria for the preparation of food [34,52,174].

5.5. Dose-Response for Inhaled Cyanotoxins

Although cyanobacteria and many of their associated toxins negatively impact the nervous system, the mode by which exposure occurs may influence the severity of symptoms. While data showing dose–response relationships for inhalation is not available for all classes of cyanotoxins discussed in this review, some cyanotoxins such as MC-LR, anatoxin-a, and BMAA have been derived. LD50 concentrations for MC-LR and anatoxin-a decrease upon exposure via nasal administration as opposed to other routes such as gastric intubation, reflecting the potency of direct inhalation [233,234]. Inhalation-based exposure to MC-LR resulted in increased liver size which is indicative of liver damage and increased proportionally to exposure concentrations [233]. Simultaneous exposure to MC-LR and anatoxin-a via inhalation can also result in synergistic toxicity which becomes more apparent with increasing toxin concentrations. However, it should be noted that mice exposed to aerosolized MC-LR did not show any adverse health defects, although this could be explained by technical limitations and the inability of mice to breathe large enough quantities of aerosolized toxins to exhibit symptoms of intoxication [233]. Although not administered via inhalation, Fawell et al. showed that mice exposed to anatoxin-a displayed dose-dependent changes in respiratory action at sublethal doses [235]. Benson et al. also showed a dose-dependent response in mice exposed to 200–265 µg/m3 of MC-LR, with exposed mice inhaling MC-LR for a period of seven days. Mice inhaling MC-LR for 0.5, 1, and 2 h displayed concurrent increases in the instances of nasal lesions and nasal epithelium necrosis, with both higher concentrations and longer periods of exposure [236].
BMAA has been shown to damage cells of the olfactory bulb and olfactory tract [42], but the degree to which BMAA may be toxic upon inhalation may be organism dependent. For example, Scott et al. showed that rats administered BMAA via inhalation did not exhibit any adverse health effects upon exposure to environmentally relevant concentrations. As concentrations of neurotoxic degradation products of BMAA such as 2,3-diaminoproprionic acid (DAP) increased proportionally to concentrations of inhaled BMAA, this implied that rats may show a degree of resistance to the toxic effects of inhaled BMAA [237]. However, Pierezan et al. showed that mice exposed intranasally to BMAA displayed selective damage to the olfactory bulb, as well as decreased viability and neurite growth in primary cultures of mice olfactory cells. It should be noted that Pierezan et al. did not observe adverse effects of BMAA at concentrations below 100µM in olfactory bulb neurons. Mixed cultures of olfactory neurons and glial cells showed decreased viability at 500µM concentrations, but not at 250µM, demonstrating the critical nature of BMAA dose on inhalation toxicity [238]. See Table 2 for a summary of the results discussed above.

5.6. Biomarkers for Inhalation of Cyanotoxins

Studies examining biomarkers for exposure to cyanotoxins, specifically through inhalation, are somewhat lacking, although it could be assumed that the biomarkers mentioned above for oxidative stress, neuroinflammation, and protein aggregation could be seen in cases of inhalation toxicity. One study reported nasal legions in mice exposed to MC-LR for seven days, in addition to increased expression of two unidentified proteins in mouse plasma which directly correlated with MC-LR exposure [236]. Although not conducted in the context of inhalation, changes in gene expression and the phosphorylation of enzymes involved in cell division have been documented in human-airway epithelial cells upon exposure to sublethal concentrations of cylindrospermopsin [239,240].

6. Mitigation of Cyanobacteria and Cyanotoxins in the Atmosphere

Several strategies exist which may lessen the impacts of harmful cyanobacterial blooms. However, few studies have been conducted with the intention of reducing cyanobacteria and cyanotoxins in the atmosphere. Since many cyanobacteria originate from aquatic environments, strategies aimed at reducing aquatic blooms may also reduce the amount of atmospheric cyanobacteria and cyanotoxins.
Current control methods utilize chemical agents which induce flocculation, the aggregation of dispersed bacterial cells in solution, forming flakes known as flocs [241]. Chemical agents such as aluminum or iron-based compounds and organic compounds like chitosan flocculate cyanobacteria by neutralizing the cell surface and precipitating them from solution [28,242,243,244]. A method known as Floc and Sink utilizes clay particles which act as ballast, causing the flocculated cyanobacteria to sink and removing them from the surface of waterbodies [28,245]. Although blooms may recover, temporarily removing cyanobacteria from surface waters may reduce aerosolized cyanobacteria, as fewer cells would be released into the atmosphere if only for a limited period. Flocculated cells may also not adhere to bubble films, possibly reducing the number of cells available to enter the atmosphere through mechanisms such as bubble bursting [187].
Reducing the amount of available nutrients in an aquatic environment may also reduce concentrations of cyanobacteria and thus cyanotoxins. To reduce bioavailable nutrients such as phosphates that enter aquatic environments as agricultural runoff, mitigation strategies include reducing fertilizer use on watershed farmland [245,246,247]. For example, cover crops such as tillage radishes grow extensive root systems capable of drawing nutrients upward from deeper soil layers to be utilized by cash crops, and could possibly reduce the total amount of phosphorus containing fertilizers needed for application to farmland [245,248,249,250]. Strategies that mitigate phosphates already present in waterbodies include the addition of compounds such as alum, clays, and lanthanum-modified bentonite, all of which precipitate phosphate anions from surface waters [28,251]. In either case, this limits bioavailable phosphorus and may reduce the growth of aquatic cyanobacteria, and the subsequent number of cells available to be liberated into the atmosphere.
Nie et al. investigated the presence of cyanobacteria on HVAC filters and found cyanobacteria of the genera Pseudanabaena, Nodularia, and Letpolyngbya [252]. These findings suggest that air-filtration methods may reduce concentrations of airborne cyanobacteria and lessen human exposure to cyanobacteria. Gaston et al. employed a filtration system in which a combination of high-efficiency particular air (HEPA) filters, air conditioning (AC) filter cassettes, and face mask pieces were used to filter airborne cyanobacteria. Roughly 80% of aerosolized cyanobacteria detected in this study were prevented from entering the atmosphere and were attached to one or more components of the filtration system [253], suggesting that in-house air filters along with personal protection face masks may help to minimize human exposure to airborne cyanobacteria.

7. Research Gaps

Although much is known about exposure to cyanobacterial toxins via drinking and recreational water as well as from contaminated crops and food, the contribution of the airborne cyanotoxin exposure route to toxicity is largely unknown. A further compounding factor is that, unlike exposure via water, avoiding toxic compounds within air is more difficult due to the necessity of inhalation and penetration of cyanobacteria into indoor environments.
If such exposure is unavoidable, current research gaps include accurate and simple means to detect and quantify cyanobacteria and cyanotoxins in air to provide an early warning system, like that provided by US national weather services for brevetoxins and wildfire smoke [254,255,256]. Such a system may be particularly important for large lakes, such as Lake Erie and the Great Salt Lake, or marine environments. Regarding Lake Erie, persistent cyanobacterial blooms, largely comprising the genus Microcystis, have the potential to cover large surface areas with microcystin contamination [12,257]. With smaller lakes, it may be sufficient to simply use the “precautionary principle” and post warning notices when toxic blooms are present or likely to be airborne. Although this may be sufficient for members of the public periodically recreating on lakes, individuals whose residences are near such waterbodies may be at increased risk of acute and chronic adverse health effects [214].
Other research gaps include the potential to filter air samples and remove cyanobacteria and their components as a preventative measure. Although some experiments have been performed in laboratory settings [253], publicly available technologies to specifically serve this purpose have, to our knowledge, not been made available. Non-specifically, such remedies may take the form of simple HEPA filters or bespoke filtration systems. It is likely that most airborne exposure occurs through the inhalation of water droplets, cellular material, and debris or dust particles, as shown by the presence of cyanobacteria in human lung samples and indoor environments [43,184,185,186]. Furthermore, the aerial exposure route may manifest in different toxicological outcomes if compounds are inhaled. This is largely because compounds may enter the body through the olfactory bulb and subsequently have the potential to bypass the BBB, resulting in neurotoxicity. BMAA, for example, was present in mouse olfactory bulbs following direct unilateral intranasal instillation, providing evidence that BMAA can bypass the BBB and enter the brain directly through the olfactory bulb [238]. Cyanotoxins such as BMAA, DAB, and AEG have also been detected in the olfactory bulbs of postmortem human subjects, which corresponded with the presence of proinflammatory proteins such as IL-6 and CASP1 in subjects with advanced Alzheimer’s Disease. Concurrently, the olfactory tracts of postmortem Alzheimer’s Disease patients showed pathologies such as neuropil vacuolation, gliosis, and the presence of reactive microglia and tauopathies [42].
Globally, the presence and persistence of cyanobacterial blooms is highly variable, with some lakes having large persistent blooms (e.g., Lake Erie), some serving as permanent cyanobacterial ecosystems (e.g., Rift Valley Lakes, Great Salt Lake), and the affected majority being small lakes with periodic/annual cyanobacterial blooms [38,258,259,260,261,262]. Therefore, a greater understanding of temporal and geographic ranges of blooms may aid in the identification of at-risk human populations. This includes the need to obtain accurate and rapid determinations of toxin concentrations to ascertain potential degrees of exposure. This can be easily achieved using low-cost, low-tech detection devices such as immunoassays, as evidenced by self-administered COVID-19 tests during the pandemic [263]. Furthermore, if properly calibrated, techniques such as qPCR may also be amenable to this task to increase the amount of available data for assessing disease patterns and trends in populations [264,265,266].
Although cases of acute toxicity from cyanobacterial toxin exposure are relatively easy to determine and investigate, understanding potential chronic effects requires greater effort. Under such scenarios, as a person may be exposed to cyanobacteria and their toxins at multiple times over multiple years, a better understanding of the amount and frequency of exposure would be invaluable [267,268,269]. Although this would require many analyses to be performed, the data may be amenable to the application of techniques such as machine learning to look for patterns in exposure without human bias or interference [267,268,269]. Another research gap with respect to chronic exposure concerns genetic differences within human populations which may affect susceptibility to cyanotoxin exposure. With some neurodegenerative diseases such as ALS and PD, a percentage of cases are thought to be caused by genetic predisposition with environmental triggers. By applying health assessment criteria such as those devised by Bradford-Hill, a better understanding of the ability of cyanobacteria and their toxins to cause adverse human health effects and disease can be achieved [270,271].
The potential list of diseases associated with exposure to cyanobacteria and their toxins continues to grow [52,267,268,269,271,272]. With the correct tools and surveillance networks in place, a better understanding of less-studied exposure routes can be achieved. The increasing analysis of cyanobacteria and their toxins in air indicates that this exposure route may be more important than previously considered. Having a better grasp on the occurrence and prevalence of cyanobacterial toxins in air may also lead to a more informed risk assessment. This may include a better understanding and partitioning of toxin allocation amounts for health protection and may also result in changes to recommended permissible cyanotoxin concentrations in water, food, and other compartments.
Perhaps the greatest research gap concerns the human exposome and the totality of exposure. Whether by water, air, or food, humans are exposed to a variety of compounds with potential acute and chronic deleterious effects. A better understanding of the variety of compounds that people are exposed to is required, along with data and research concerning potential synergistic toxicological effects [273]. To protect human health from the actions of cyanobacteria and cyanotoxins, the quantity of these compounds and how they interact in human bodies will provide the means to better protect human health. This is especially pertinent when combined with likely adverse effects of climate change on harmful cyanobacterial bloom occurrence and distribution.

Author Contributions

Original draft preparation was carried out by Z.J.M. and J.S.M. with Z.J.M. as the primary author. Z.J.M., J.S.M. and E.W.S. contributed to review and editing. Investigation was carried out primarily by Z.J.M. with contributions and edits from J.S.M. and E.W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We would like to the thank the Center for Great Lakes and Watershed Studies for their support. We would also like to thank Chris Ward, Aspassia Chatziefthimiou, and Jari Willing for their insightful discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schopf, J.W.; Packer, B.M. Early Archean (3.3-billion to 3.5-billion-year-old) microfossils from Warrawoona Group, Australia. Science 1987, 237, 70. [Google Scholar] [CrossRef] [PubMed]
  2. Papke, R.T.; Ramsing, N.B.; Bateson, M.M.; Ward, D.M. Geographical isolation in hot spring cyanobacteria. Environ. Microbiol. 2003, 5, 650–659. [Google Scholar] [CrossRef] [PubMed]
  3. Kulasooriya, S.A. Cyanobacteria: Pioneers of planet earth. Ceylon J. Sci. (Biol. Sci.) 2012, 40, 71–88. [Google Scholar] [CrossRef]
  4. Álvarez, C.; Jiménez-Ríos, L.; Iniesta-Pallarés, M.; Jurado-Flores, A.; Molina-Heredia, F.P.; Ng, C.K.; Mariscal, V. Symbiosis between cyanobacteria and plants: From molecular studies to agronomic applications. J. Exp. Bot. 2023, 74, 6145–6157. [Google Scholar] [CrossRef]
  5. Gaysina, L.A.; Saraf, A.; Singh, P. Cyanobacteria in diverse habitats. In Cyanobacteria; Academic Press: Cambridge, MA, USA, 2019; pp. 1–28. [Google Scholar]
  6. Polyak, Y.; Zaytseva, T.; Medvedeva, N. Response of toxic cyanobacterium Microcystis aeruginosa to environmental pollution. Water Air Soil Pollut. 2013, 224, 1494. [Google Scholar] [CrossRef]
  7. Ramakrishnan, B.; Megharaj, M.; Venkateswarlu, K.; Naidu, R.; Sethunathan, N. The impacts of environmental pollutants on microalgae and cyanobacteria. Crit. Rev. Environ. Sci. Technol. 2010, 40, 699–821. [Google Scholar] [CrossRef]
  8. Fujimoto, N.; Sudo, R.; Sugiura, N.; Inamori, Y. Nutrient-limited growth of Microcystis aeruginosa and Phormidium tenue and competition under various N: P supply ratios and temperatures. Limnol. Oceanogr. 1997, 42, 250–256. [Google Scholar] [CrossRef]
  9. Carpenter, S.R.; Caraco, N.F.; Correll, D.L.; Howarth, R.W.; Sharpley, A.N.; Smith, V.H. Nonpoint pollution of surface waters with phosphorus and nitrogen. Ecol. Appl. 1998, 8, 559–568. [Google Scholar] [CrossRef]
  10. O’Neil, J.M.; Davis, T.W.; Burford, M.A.; Gobler, C.J. The rise of harmful cyanobacteria blooms: The potential roles of eutrophication and climate change. Harmful Algae 2012, 14, 313–334. [Google Scholar] [CrossRef]
  11. Grizzetti, B.; Bouraoui, F.; Aloe, A. Changes of nitrogen and phosphorus loads to European seas. Glob. Change Biol. 2012, 18, 769–782. [Google Scholar] [CrossRef]
  12. Grizzetti, B.; Billen, G.; Davidson, E.A.; Winiwarter, W.; de Vries, W.; Fowler, D.; Howard, C.M.; Bleeker, A.; Sutton, M.A.; Lassaletta, L.; et al. Global nitrogen and phosphorus pollution. In Just Enough Nitrogen: Perspectives on How to Get There for Regions with Too Much and Too Little Nitrogen; Springer: Berlin/Heidelberg, Germany, 2020; pp. 421–431. [Google Scholar]
  13. Michalak, A.M.; Anderson, E.J.; Beletsky, D.; Boland, S.; Bosch, N.S.; Bridgeman, T.B.; Chaffin, J.D.; Cho, K.; Confesor, R.; Daloğlu, I. Record-setting algal bloom in Lake Erie caused by agricultural and meteorological trends consistent with expected future conditions. Proc. Natl. Acad. Sci. USA 2013, 110, 6448–6452. [Google Scholar] [CrossRef] [PubMed]
  14. Watson, S.B.; Miller, C.; Arhonditsis, G.; Boyer, G.L.; Carmichael, W.; Charlton, M.N.; Confesor, R.; Depew, D.; Höök, T.O.; Ludsin, S.A. The re-eutrophication of Lake Erie: Harmful algal blooms and hypoxia. Harmful Algae 2016, 56, 44–66. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, H.; Boegman, L.; Scavia, D.; Culver, D.A. Spatial distributions of external and internal phosphorus loads in Lake Erie and their impacts on phytoplankton and water quality. J. Great Lakes Res. 2016, 42, 1212–1227. [Google Scholar] [CrossRef]
  16. Huisman, J.; Codd, G.A.; Paerl, H.W.; Ibelings, B.W.; Verspagen, J.M.H.; Visser, P.M. Cyanobacterial blooms. Nat. Rev. Microbiol. 2018, 16, 471–483. [Google Scholar] [CrossRef]
  17. Li, Z.; Yu, J.; Yang, M.; Zhang, J.; Burch, M.D.; Han, W. Cyanobacterial population and harmful metabolites dynamics during a bloom in Yanghe Reservoir, North China. Harmful Algae 2010, 9, 481–488. [Google Scholar] [CrossRef]
  18. Lomeo, D.; Tebbs, E.J.; Babayani, N.D.; Chadwick, M.A.; Gondwe, M.J.; Jungblut, A.D.; McCulloch, G.P.; Morgan, E.R.; Schillereff, D.N.; Simis, S.G.H.; et al. Remote sensing and spatial analysis reveal unprecedented cyanobacteria bloom dynamics associated with elephant mass mortality. Sci. Total Environ. 2024, 957, 177525. [Google Scholar] [CrossRef] [PubMed]
  19. Backer, L.C.; Landsberg, J.H.; Miller, M.; Keel, K.; Taylor, T.K. Canine cyanotoxin poisonings in the United States (1920s–2012): Review of suspected and confirmed cases from three data sources. Toxins 2013, 5, 1597–1628. [Google Scholar] [CrossRef]
  20. Falconer, I.R.; Humpage, A.R. Health risk assessment of cyanobacterial (blue-green algal) toxins in drinking water. Int. J. Environ. Res. Public Health 2005, 2, 43–50. [Google Scholar] [CrossRef]
  21. Negri, A.P.; Jones, G.J.; Hindmarsh, M. Sheep mortality associated with paralytic shellfish poisons from the cyanobacterium Anabaena circinalis. Toxicon 1995, 33, 1321–1329. [Google Scholar] [CrossRef]
  22. Thomas, A.; Saker, M.; Norton, J.; Olsen, R. Cyanobacterium Cylindrospermopsis raciborskii as a probable cause of death in cattle in northern Queensland. Aust. Vet. J. 1998, 76, 592–594. [Google Scholar] [CrossRef]
  23. Wood, S.A.; Heath, M.W.; Holland, P.T.; Munday, R.; McGregor, G.B.; Ryan, K.G. Identification of a benthic microcystin-producing filamentous cyanobacterium (Oscillatoriales) associated with a dog poisoning in New Zealand. Toxicon 2010, 55, 897–903. [Google Scholar] [CrossRef] [PubMed]
  24. Chatziefthimiou, A.D.; Metcalf, J.S.; Glover, W.B.; Powell, J.T.; Banack, S.A.; Cox, P.A.; Ladjimi, M.; Sultan, A.A.; Chemaitelly, H.; Richer, R.A. Cyanotoxin accumulation and growth patterns of biocrust communities under variable environmental conditions. Toxicon X 2024, 23, 100199. [Google Scholar] [CrossRef] [PubMed]
  25. Koksharova, O.A.; Butenko, I.O.; Pobeguts, O.V.; Safronova, N.A.; Govorun, V.M. The first proteomic study of Nostoc sp. PCC 7120 exposed to cyanotoxin BMAA under nitrogen starvation. Toxins 2020, 12, 310. [Google Scholar] [CrossRef] [PubMed]
  26. Koksharova, O.A.; Butenko, I.O.; Pobeguts, O.V.; Safronova, N.A.; Govorun, V.M. Proteomic insights into starvation of nitrogen-replete cells of Nostoc sp. PCC 7120 under β-N-methylamino-L-alanine (BMAA) treatment. Toxins 2020, 12, 372. [Google Scholar] [CrossRef]
  27. Downing, S.; Banack, S.A.; Metcalf, J.S.; Cox, P.A.; Downing, T.G. Nitrogen starvation of cyanobacteria results in the production of β-N-methylamino-L-alanine. Toxicon 2011, 58, 187–194. [Google Scholar] [CrossRef]
  28. Song, L.; Jia, Y.; Qin, B.; Li, R.; Carmichael, W.W.; Gan, N.; Xu, H.; Shan, K.; Sukenik, A. Harmful cyanobacterial blooms: Biological traits, mechanisms, risks, and control strategies. Annu. Rev. Environ. Resour. 2023, 48, 123–147. [Google Scholar] [CrossRef]
  29. Scott, L.L.; Downing, S.; Phelan, R.R.; Downing, T.G. Environmental modulation of microcystin and β-N-methylamino-L-alanine as a function of nitrogen availability. Toxicon 2014, 87, 1–5. [Google Scholar] [CrossRef]
  30. Cox, P.A.; Banack, S.A.; 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]
  31. Murch, S.J.; Cox, P.A.; Banack, S.A. A mechanism for slow release of biomagnified cyanobacterial neurotoxins and neurodegenerative disease in Guam. Proc. Natl. Acad. Sci. USA 2004, 101, 12228–12231. [Google Scholar] [CrossRef]
  32. Jonasson, S.; Eriksson, J.; Berntzon, L.; Spácil, 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]
  33. 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]
  34. 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 (Suppl. S2), 44–49. [Google Scholar] [CrossRef] [PubMed]
  35. Chatziefthimiou, A.D.; Deitch, E.J.; Glover, W.B.; Powell, J.T.; Banack, S.A.; Richer, R.A.; Cox, P.A.; Metcalf, J.S. Analysis of neurotoxic amino acids from marine waters, microbial mats, and seafood destined for human consumption in the Arabian Gulf. Neurotox. Res. 2018, 33, 143–152. [Google Scholar] [CrossRef] [PubMed]
  36. Fontaine, J.; Duy, S.V.; Troncy, C.; Dinh, Q.T.; Simon, D.F.; Munoz, G.; Sauvé, S. Screening of multi-class cyanotoxins in algal dietary supplements marketed in North America. Algal Res. 2023, 73, 103162. [Google Scholar] [CrossRef]
  37. Roy-Lachapelle, A.; Solliec, M.; Bouchard, M.F.; Sauvé, S. Detection of cyanotoxins in algae dietary supplements. Toxins 2017, 9, 76. [Google Scholar] [CrossRef]
  38. Wynne, T.T.; Stumpf, R.P. Spatial and temporal patterns in the seasonal distribution of toxic cyanobacteria in Western Lake Erie from 2002–2014. Toxins 2015, 7, 1649–1663. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. 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]
  40. 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]
  41. Andersson, M.; Ersson, L.; Brandt, I.; Bergström, U. Potential transfer of neurotoxic amino acid β-N-methylamino-alanine (BMAA) from mother to infant during breast-feeding: Predictions from human cell lines. Toxicol. Appl. Pharmacol. 2017, 320, 40–50. [Google Scholar] [CrossRef]
  42. 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]
  43. 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] [PubMed]
  44. Barnard, M.A.; Chaffin, J.D.; Plaas, H.E.; Boyer, G.L.; Wei, B.; Wilhelm, S.W.; Rossignol, K.L.; Braddy, J.S.; Bullerjahn, G.S.; Bridgeman, T.B.; et al. Roles of nutrient limitation on Western Lake Erie cyanoHAB toxin production. Toxins 2021, 13, 47. [Google Scholar] [CrossRef] [PubMed]
  45. Soliakov, L.; Gallagher, T.; Wonnacott, S. Anatoxin-a-evoked [3H] dopamine release from rat striatal synaptosomes. Neuropharmacology 1995, 34, 1535–1541. [Google Scholar] [CrossRef] [PubMed]
  46. Lakshmana Rao, P.; Bhattacharya, R.; Gupta, N.; Parida, M.; Bhaskar, A.; Dubey, R. Involvement of caspase and reactive oxygen species in cyanobacterial toxin anatoxin-a-induced cytotoxicity and apoptosis in rat thymocytes and Vero cells. Arch. Toxicol. 2002, 76, 227–235. [Google Scholar] [CrossRef]
  47. Zhong, Y.; Shen, L.; Ye, X.; Zhou, D.; He, Y.; Li, Y.; Ding, Y.; Zhu, W.; Ding, J.; Zhang, H. Neurotoxic anatoxin-a can also exert immunotoxicity by the induction of apoptosis on Carassius auratus lymphocytes in vitro when exposed to environmentally relevant concentrations. Front. Physiol. 2020, 11, 316. [Google Scholar] [CrossRef]
  48. Fiore, M.F.; de Lima, S.T.; Carmichael, W.W.; McKinnie, S.M.K.; Chekan, J.R.; Moore, B.S. Guanitoxin, re-naming a cyanobacterial organophosphate toxin. Harmful Algae 2020, 92, 101737. [Google Scholar] [CrossRef]
  49. Passos, L.S.; Gomes, L.C.; Pereira, T.M.; Sadauskas-Henrique, H.; Dal Pont, G.; Ostrensky, A.; Pinto, E. Response of Oreochromis niloticus (Teleostei: Cichlidae) exposed to a guanitoxin-producing cyanobacterial strain using multiple biomarkers. Sci. Total Environ. 2022, 835, 155471. [Google Scholar] [CrossRef]
  50. 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]
  51. Llewellyn, L.E. Saxitoxin, a toxic marine natural product that targets a multitude of receptors. Nat. Prod. Rep. 2006, 23, 200–222. [Google Scholar] [CrossRef]
  52. Metcalf, J.S.; Tischbein, M.; Cox, P.A.; Stommel, E.W. Cyanotoxins and the nervous system. Toxins 2021, 13, 660. [Google Scholar] [CrossRef]
  53. Ramos, P.B.; Diehl, F.; dos Santos, J.M.; Monserrat, J.M.; Yunes, J.S. Oxidative stress in rats induced by consumption of saxitoxin contaminated drink water. Harmful Algae 2014, 37, 68–74. [Google Scholar] [CrossRef]
  54. Wu, H.; Prithiviraj, B.; Tan, Z. Physiological effects of oxidative stress caused by saxitoxin in the nematode Caenorhabditis elegans. Mar. Drugs 2023, 21, 544. [Google Scholar] [CrossRef] [PubMed]
  55. da Silva, C.A.; de Morais, E.C.P.; Costa, M.D.M.; Ribas, J.L.C.; Guiloski, I.C.; Ramsdorf, W.A.; Zanata, S.M.; Cestari, M.M.; Ribeiro, C.A.O.; Magalhães, V.F. Saxitoxins induce cytotoxicity, genotoxicity and oxidative stress in teleost neurons in vitro. Toxicon 2014, 86, 8–15. [Google Scholar] [CrossRef] [PubMed]
  56. 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. R. Soc. B Biol. Sci. 2016, 283, 20152397. [Google Scholar] [CrossRef]
  57. 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]
  58. Metcalf, J.S.; Lobner, D.; Banack, S.A.; Cox, G.A.; Nunn, P.B.; Wyatt, P.B.; Cox, P.A. Analysis of BMAA enantiomers in cycads, cyanobacteria, and mammals: In vivo formation and toxicity of d-BMAA. Amino Acids 2017, 49, 1427–1439. [Google Scholar] [CrossRef]
  59. 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]
  60. Esterhuizen-Londt, M.; Wiegand, C.; Downing, T.G. β-N-methylamino-L-alanine (BMAA) uptake by the animal model, Daphnia magna and subsequent oxidative stress. Toxicon 2015, 100, 20–26. [Google Scholar] [CrossRef]
  61. Esterhuizen-Londt, M.; Pflugmacher, S.; Downing, T.G. The effect of β-N-methylamino-L-alanine (BMAA) on oxidative stress response enzymes of the macrophyte Ceratophyllum demersum. Toxicon 2011, 57, 803–810. [Google Scholar] [CrossRef]
  62. 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-methylamino-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]
  63. Tian, K.W.; Jiang, H.; Wang, B.B.; Zhang, F.; Han, S. Intravenous injection of L-BMAA induces a rat model with comprehensive characteristics of amyotrophic lateral sclerosis/Parkinson–dementia complex. Toxicol. Res. 2016, 5, 79–96. [Google Scholar] [CrossRef] [PubMed]
  64. Silva, D.F.; Candeias, E.; Esteves, A.R.; Magalhães, J.D.; Ferreira, 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. Neuroinflamm. 2020, 17, 332. [Google Scholar] [CrossRef] [PubMed]
  65. Botha, N.; Gehringer, M.M.; Downing, T.G.; van de Venter, M.; Shephard, E.G. The role of microcystin-LR in the induction of apoptosis and oxidative stress in CaCo2 cells. Toxicon 2004, 43, 85–92. [Google Scholar] [CrossRef]
  66. Pflugmacher, S. Promotion of oxidative stress in the aquatic macrophyte Ceratophyllum demersum during biotransformation of the cyanobacterial toxin microcystin-LR. Aquat. Toxicol. 2004, 70, 169–178. [Google Scholar] [CrossRef]
  67. Jayaraj, R.; Anand, T.; Rao, P.L. Activity and gene expression profile of certain antioxidant enzymes to microcystin-LR induced oxidative stress in mice. Toxicology 2006, 220, 136–146. [Google Scholar] [CrossRef] [PubMed]
  68. Jiang, J.; Gu, X.; Song, R.; Zhang, Q.; Geng, J.; Wang, X.; Yang, L. Time-dependent oxidative stress and histopathological changes in Cyprinus carpio L. exposed to microcystin-LR. Ecotoxicology 2011, 20, 1000–1009. [Google Scholar] [CrossRef]
  69. Pinho, G.L.L.; Da Rosa, C.M.; Maciel, F.E.; Bianchini, A.; Yunes, J.S.; Proença, L.A.D.O.; Monserrat, J.M. Antioxidant responses and oxidative stress after microcystin exposure in the hepatopancreas of an estuarine crab species. Ecotoxicol. Environ. Saf. 2005, 61, 353–360. [Google Scholar] [CrossRef]
  70. Mondal, A.; Saha, P.; Bose, D.; Chatterjee, S.; Seth, R.K.; Xiao, S.; Porter, D.E.; Brooks, B.W.; Scott, G.I.; Nagarkatti, M. 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]
  71. 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. 2023, 60, 884–900. [Google Scholar] [CrossRef]
  72. 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]
  73. Mondal, A.; Bose, D.; Saha, P.; Seth, R.; Scott, G.; Xiao, S.; Porter, D.; Brooks, B.; Chatterjee, S. Microcystin-LR exacerbates neuroinflammation and neurodegeneration in nonalcoholic fatty liver disease. FASEB J. 2021, 35. [Google Scholar] [CrossRef]
  74. Li, X.B.; Zhang, X.; Ju, J.; Li, Y.; Yin, L.; Pu, Y. Alterations in neurobehaviors and inflammation in hippocampus of rats induced by oral administration of microcystin-LR. Environ. Sci. Pollut. Res. 2014, 21, 12419–12425. [Google Scholar] [CrossRef] [PubMed]
  75. 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. Hazard. Mater. 2019, 373, 504–518. [Google Scholar] [CrossRef] [PubMed]
  76. Ichinose, M.; Endo, S.; Critz, S.D.; Shenolikar, S.; Byrne, J.H. Microcystin-LR, a potent protein phosphatase inhibitor, prolongs the serotonin-and cAMP-induced currents in sensory neurons of Aplysia californica. Brain Res. 1990, 533, 137–140. [Google Scholar] [CrossRef]
  77. Wang, J.; Lin, F.; Cai, F.; Yan, W.; Zhou, Q.; Xie, L. Microcystin-LR inhibited hippocampal long-term potential via regulation of the glycogen synthase kinase-3β pathway. Chemosphere 2013, 93, 223–229. [Google Scholar] [CrossRef]
  78. Pichardo, S.; Devesa, V.; Puerto, M.; Vélez, D.; Cameán, A.M. Intestinal transport of cylindrospermopsin using the Caco-2 cell line. Toxicol. Vitro 2017, 38, 142–149. [Google Scholar] [CrossRef]
  79. Guzmán-Guillén, R.; Prieto, A.I.; Vasconcelos, V.M.; Cameán, A.M. Cyanobacterium producing cylindrospermopsin cause oxidative stress at environmentally relevant concentrations in sub-chronically exposed tilapia (Oreochromis niloticus). Chemosphere 2013, 90, 1184–1194. [Google Scholar] [CrossRef]
  80. Žegura, B.; Gajski, G.; Štraser, A.; Garaj-Vrhovac, V. Cylindrospermopsin induced DNA damage and alteration in the expression of genes involved in the response to DNA damage, apoptosis and oxidative stress. Toxicon 2011, 58, 471–479. [Google Scholar] [CrossRef]
  81. López-Alonso, H.; Rubiolo, J.A.; Vega, F.; Vieytes, M.R.; Botana, L.M. Protein synthesis inhibition and oxidative stress induced by cylindrospermopsin elicit apoptosis in primary rat hepatocytes. Chem. Res. Toxicol. 2013, 26, 203–212. [Google Scholar] [CrossRef]
  82. Hinojosa, M.G.; Cascajosa-Lira, A.; Prieto, A.I.; Gutiérrez-Praena, D.; Vasconcelos, V.; Jos, A.; Cameán, A.M. Cytotoxic effects and oxidative stress produced by a cyanobacterial cylindrospermopsin producer extract versus a cylindrospermopsin non-producing extract on the neuroblastoma SH-SY5Y cell line. Toxins 2023, 15, 320. [Google Scholar] [CrossRef]
  83. Zhang, X.; Hu, J.; Jian, J.; Liu, H.; Zhou, X.; Zhang, Y.; Zou, G.; Zhou, L.; Wang, J. Exploring the neurotoxic effects of cylindrospermopsin in early development of zebrafish: An integrated impact of oxidative stress, inflammatory response, and apoptosis. Ecotoxicol. Environ. Saf. 2025, 293, 118021. [Google Scholar] [CrossRef] [PubMed]
  84. Puerto, M.; Jos, A.; Pichardo, S.; Moyano, R.; Blanco, A.; Cameán, A.M. Acute exposure to pure cylindrospermopsin results in oxidative stress and pathological alterations in tilapia (Oreochromis niloticus). Environ. Toxicol. 2014, 29, 371–385. [Google Scholar] [CrossRef]
  85. Sieroslawska, A.; Rymuszka, A. Cylindrospermopsin induces oxidative stress and genotoxic effects in the fish CLC cell line. J. Appl. Toxicol. 2015, 35, 426–433. [Google Scholar] [CrossRef] [PubMed]
  86. Hinojosa, M.G.; Prieto, A.I.; Muñoz-Castro, C.; Sánchez-Mico, M.V.; Vitorica, J.; Cameán, A.M.; Jos, Á. Cytotoxicity and effects on the synapsis induced by pure cylindrospermopsin in an E17 embryonic murine primary neuronal culture in a concentration- and time-dependent manner. Toxins 2022, 14, 175. [Google Scholar] [CrossRef] [PubMed]
  87. Silva, R.d.C.d.; Grötzner, S.R.; Costa, D.D.M.; Garcia, J.R.E.; Muelbert, J.; de Magalhães, V.F.; Neto, F.F.; Ribeiro, C.A.d.O. Comparative bioaccumulation and effects of purified and cellular extract of cylindrospermopsin to freshwater fish Hoplias malabaricus. J. Toxicol. Environ. Health Part A 2018, 81, 620–632. [Google Scholar] [CrossRef]
  88. Guzmán-Guillén, R.; Manzano, I.L.; Moreno, I.M.; Ortega, A.I.P.; Moyano, R.; Blanco, A.; Cameán, A.M. Cylindrospermopsin induces neurotoxicity in tilapia fish (Oreochromis niloticus) exposed to Aphanizomenon ovalisporum. Aquat. Toxicol. 2015, 161, 17–24. [Google Scholar] [CrossRef]
  89. Rabelo, J.C.S.; Hanusch, A.L.; de Jesus, L.W.O.; Mesquita, L.A.; Franco, F.C.; Silva, R.A.; Sabóia-Morais, S.M.T. DNA damage induced by cylindrospermopsin on different tissues of the biomonitor fish Poecilia reticulata. Environ. Toxicol. 2021, 36, 1125–1134. [Google Scholar] [CrossRef]
  90. 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]
  91. Carmichael, W.W.; Biggs, D.F.; Gorham, P.R. Toxicology and pharmacological action of Anabaena flos-aquae toxin. Science 1975, 187, 542–544. [Google Scholar] [CrossRef]
  92. Carmichael, W.W.; Gorham, P.R. Anatoxins from clones of Anabaena flos-aquae isolated from lakes of western Canada. Int. Ver. Für Theor. Angew. Limnol. Mitteilungen 1978, 21, 285–295. [Google Scholar]
  93. Devlin, J.P.; Edwards, O.E.; Gorham, P.R.; Hunter, N.R.; Pike, R.K.; Stavric, B. Anatoxin-a, a toxic alkaloid from Anabaena flos-aquae NRC-44h. Can. J. Chem. 1977, 55, 1367–1371. [Google Scholar] [CrossRef]
  94. Paerl, H.W.; Otten, T.G. Harmful cyanobacterial blooms: Causes, consequences, and controls. Microb Ecol. 2013, 65, 995–1010. [Google Scholar] [CrossRef] [PubMed]
  95. Wonnacott, S.; Gallagher, T. The chemistry and pharmacology of anatoxin-a and related homotropanes with respect to nicotinic acetylcholine receptors. Mar. Drugs 2006, 4, 228–254. [Google Scholar] [CrossRef]
  96. 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]
  97. Heath, M.; Wood, S.A.; Young, R.G.; Ryan, K.G. The role of nitrogen and phosphorus in regulating Phormidium sp. (cyanobacteria) growth and anatoxin production. FEMS Microbiol. Ecol. 2016, 92, fiw021. [Google Scholar] [CrossRef]
  98. Kramer, B.J.; Jankowiak, J.G.; Nanjappa, D.; Harke, M.J.; Gobler, C.J. Nitrogen and phosphorus significantly alter growth, nitrogen fixation, anatoxin-a content, and the transcriptome of the bloom-forming cyanobacterium, Dolichospermum. Front. Microbiol. 2022, 13, 955032. [Google Scholar] [CrossRef]
  99. Gagnon, A.; Pick, F.R. Effect of nitrogen on cellular production and release of the neurotoxin anatoxin-a in a nitrogen-fixing cyanobacterium. Front. Microbiol. 2012, 3, 211. [Google Scholar] [CrossRef]
  100. Edwards, C.; Beattie, K.A.; Scrimgeour, C.M.; Codd, G.A. Identification of anatoxin-a in benthic cyanobacteria (blue-green algae) and in associated dog poisonings at Loch Insh, Scotland. Toxicon 1992, 30, 1165–1175. [Google Scholar] [CrossRef]
  101. Metcalf, J.S.; Morrison, L.F.; Krienitz, L.; Ballot, A.; Krause, E.; Kotut, K.; Pütz, S.; Wiegand, C.; Pflugmacher, S.; Codd, G.A. Analysis of the cyanotoxins anatoxin-a and microcystins in Lesser Flamingo feathers. Toxicol. Environ. Chem. 2006, 88, 159–167. [Google Scholar] [CrossRef]
  102. Mahmood, N.A.; Carmichael, W.W. The pharmacology of anatoxin-a(s), a neurotoxin produced by the freshwater cyanobacterium Anabaena flos-aquae NRC 525-17. Toxicon 1986, 24, 425–434. [Google Scholar] [CrossRef]
  103. Metcalf, J.S.; Richer, R.; Cox, P.A.; Codd, G.A. Cyanotoxins in desert environments may present a risk to human health. Sci. Total Environ. 2012, 421–422, 118–123. [Google Scholar] [CrossRef] [PubMed]
  104. Fernandes, K.A.; Fadul, J.C.; Fiore, M.F.; Pinto, E. A systematic review on guanitoxin: General characteristics and ecological risks. Chemosphere 2024, 352, 141277. [Google Scholar] [CrossRef] [PubMed]
  105. Carmichael, W.W.; Evans, W.R.; Yin, Q.Q.; Bell, P.; Moczydlowski, E. Evidence for paralytic shellfish poisons in the freshwater cyanobacterium Lyngbya wollei (Farlow ex Gomont) comb. nov. Appl. Environ. Microbiol. 1997, 63, 3104–3110. [Google Scholar] [CrossRef] [PubMed]
  106. 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]
  107. Lipkind, G.M.; Fozzard, H.A. A structural model of the tetrodotoxin and saxitoxin binding site of the Na+ channel. Biophys. J. 1994, 66, 1–13. [Google Scholar] [CrossRef]
  108. O’Neill, K.; Musgrave, I.F.; Humpage, A. Low dose extended exposure to saxitoxin and its potential neurodevelopmental effects: A review. Environ. Toxicol. Pharmacol. 2016, 48, 7–16. [Google Scholar] [CrossRef]
  109. Pinto, A.; Macário, I.P.; Marques, S.M.; Lourenço, J.; Domingues, I.; Botelho, M.J.; Asselman, J.; Pereira, P.; Pereira, J.L. A short-term exposure to saxitoxin triggers a multitude of deleterious effects in Daphnia magna at levels deemed safe for human health. Sci. Total Environ. 2024, 951, 175431. [Google Scholar] [CrossRef]
  110. Banack, S.A.; Metcalf, J.S.; Jiang, L.; Craighead, D.; Ilag, L.L.; Cox, P.A. Cyanobacteria produce N-(2-aminoethyl) glycine, a backbone for peptide nucleic acids which may have been the first genetic molecules for life on earth. PLoS ONE 2012, 7, e49043. [Google Scholar] [CrossRef]
  111. 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]
  112. Esterhuizen, M.; Downing, T.G. β-N-methylamino-L-alanine (BMAA) in novel South African cyanobacterial isolates. Ecotoxicol. Environ. Saf. 2008, 71, 309–313. [Google Scholar] [CrossRef]
  113. Metcalf, J.S.; Banack, S.A.; Lindsay, J.; Morrison, L.F.; Cox, P.A.; Codd, G.A. Co-occurrence of β-N-methylamino-L-alanine, a neurotoxic amino acid with other cyanobacterial toxins in British waterbodies, 1990–2004. Environ. Microbiol. 2008, 10, 702–708. [Google Scholar] [CrossRef]
  114. 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]
  115. Banack, S.A.; Caller, T.; Henegan, P.; Haney, J.; Murby, A.; Metcalf, J.S.; Powell, J.; Cox, P.A.; 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]
  116. 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. Off. J. Soc. Toxicol. 2012, 130, 391–404. [Google Scholar] [CrossRef] [PubMed]
  117. Weiss, J.H.; Choi, D.W. Beta-N-methylamino-L-alanine neurotoxicity: Requirement for bicarbonate as a cofactor. Science 1988, 241, 973–975. [Google Scholar] [CrossRef]
  118. Weiss, J.H.; Koh, J.Y.; Choi, D.W. Neurotoxicity of β-N-methylamino-L-alanine (BMAA) and β-N-oxalylamino-L-alanine (BOAA) on cultured cortical neurons. Brain Res. 1989, 497, 64–71. [Google Scholar] [CrossRef]
  119. 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]
  120. Rao, S.D.; Banack, S.A.; Cox, P.A.; Weiss, J.H. BMAA selectively injures motor neurons via AMPA/kainate receptor activation. Exp. Neurol. 2006, 201, 244–252. [Google Scholar] [CrossRef]
  121. 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]
  122. Burton, B.; Collins, K.; Brooks, J.; Marx, K.; Renner, A.; Wilcox, K.; Moore, E.; Osowski, K.; Riley, J.; Rowe, J.; et al. The biotoxin BMAA promotes dysfunction via distinct mechanisms in neuroblastoma and glioblastoma cells. PLoS ONE 2023, 18, e0278793. [Google Scholar] [CrossRef]
  123. Main, B.J.; Rodgers, K.J. Assessing the combined toxicity of BMAA and its isomers 2,4-DAB and AEG in vitro using human neuroblastoma cells. Neurotox. Res. 2018, 33, 33–42. [Google Scholar] [CrossRef] [PubMed]
  124. Metcalf, J.S.; Banack, S.A.; Richer, R.; Cox, P.A. Neurotoxic amino acids and their isomers in desert environments. J. Arid Environ. 2015, 112, 140–144. [Google Scholar] [CrossRef]
  125. Spasic, S.; Stanojevic, M.; Nesovic Ostojic, J.; Kovacevic, S.; Prostran, M.; Lopicic, S. Extensive depolarization and lack of recovery of leech Retzius neurons caused by 2,4 diaminobutyric acid. Aquat. Toxicol. 2018, 199, 269–275. [Google Scholar] [CrossRef] [PubMed]
  126. Harke, M.J.; Steffen, M.M.; Gobler, C.J.; Otten, T.G.; Wilhelm, S.W.; Wood, S.A.; Paerl, H.W. A review of the global ecology, genomics, and biogeography of the toxic cyanobacterium, Microcystis spp. Harmful Algae 2016, 54, 4–20. [Google Scholar] [CrossRef]
  127. Kurmayer, R. The toxic cyanobacterium Nostoc sp. strain 152 produces highest amounts of microcystin and nostophycin under stress conditions 1. J. Phycol. 2011, 47, 200–207. [Google Scholar] [CrossRef]
  128. 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]
  129. Zhou, B.; Wang, Z. Effects of nitrogen and phosphorus on Microcystis aeruginosa growth and microcystin production. Green Process. Synth. 2022, 11, 64–70. [Google Scholar] [CrossRef]
  130. Lone, Y.; Koiri, R.K.; Bhide, M. An overview of the toxic effect of potential human carcinogen microcystin-LR on testis. Toxicol. Rep. 2015, 2, 289–296. [Google Scholar] [CrossRef]
  131. Xu, Y.; Xu, W.; Hu, X.; Su, H.; Wen, G.; Yang, K.; Cao, Y. Toxicity of the microcystin-producing cyanobacteria Microcystis aeruginosa to shrimp Litopenaeus vannamei. Ecotoxicology 2022, 31, 1403–1412. [Google Scholar] [CrossRef]
  132. Zhou, M.; Tu, W.; Xu, J. Mechanisms of microcystin-LR-induced cytoskeletal disruption in animal cells. Toxicon 2015, 101, 92–100. [Google Scholar] [CrossRef]
  133. Javadpour, P.; Dargahi, L.; Ahmadiani, A.; Ghasemi, R. To be or not to be: PP2A as a dual player in CNS functions, its role in neurodegeneration, and its interaction with brain insulin signaling. Cell. Mol. Life Sci. 2019, 76, 2277–2297. [Google Scholar] [CrossRef] [PubMed]
  134. Nematullah, M.; Hoda, M.N.; Khan, F. Protein phosphatase 2A: A double-faced phosphatase of cellular system and its role in neurodegenerative disorders. Mol. Neurobiol. 2018, 55, 1750–1761. [Google Scholar] [CrossRef] [PubMed]
  135. Verbinnen, I.; Vaneynde, P.; Reynhout, S.; Lenaerts, L.; Derua, R.; Houge, G.; Janssens, V. Protein phosphatase 2A (PP2A) mutations in brain function, development, and neurologic disease. Biochem. Soc. Trans. 2021, 49, 1567–1588. [Google Scholar] [CrossRef]
  136. Wang, W.; Zhang, H.; Wei, L.; Ma, Y.; Jiang, H.; Yuen, C.N.T.; Zhang, J.; Wu, H.; Shu, Y. Microcystin-leucine arginine causes brain injury and functional disorder in Lithobates catesbeianus tadpoles by oxidative stress and inflammation. Aquat. Toxicol. 2023, 258, 106509. [Google Scholar] [CrossRef] [PubMed]
  137. Ohtani, I.; Moore, R.E.; Runnegar, M.T.C. Cylindrospermopsin: A potent hepatotoxin from the blue-green alga Cylindrospermopsis raciborskii. J. Am. Chem. Soc. 1992, 114, 7941–7942. [Google Scholar] [CrossRef]
  138. Gutiérrez-Praena, D.; Pichardo, S.; Jos, Á.; Cameán, A.M. Toxicity and glutathione implication in the effects observed by exposure of the liver fish cell line PLHC-1 to pure cylindrospermopsin. Ecotoxicol. Environ. Saf. 2011, 74, 1567–1572. [Google Scholar] [CrossRef]
  139. Gutiérrez-Praena, D.; Pichardo, S.; Jos, Á.; Moreno, F.J.; Cameán, A.M. Biochemical and pathological toxic effects induced by the cyanotoxin cylindrospermopsin on the human cell line Caco-2. Water Res. 2012, 46, 1566–1575. [Google Scholar] [CrossRef]
  140. 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]
  141. Puerto, M.; Prieto, A.I.; Maisanaba, S.; Gutiérrez-Praena, D.; Mellado-García, P.; Jos, Á.; Cameán, A.M. Mutagenic and genotoxic potential of pure cylindrospermopsin by a battery of in vitro tests. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2018, 121, 413–422. [Google Scholar] [CrossRef]
  142. Roos, W.P.; Kaina, B. DNA damage-induced cell death by apoptosis. Trends Mol. Med. 2006, 12, 440–450. [Google Scholar] [CrossRef]
  143. Brittain, S.M.; Wang, J.; Babcock-Jackson, L.; Carmichael, W.W.; Rinehart, K.L.; Culver, D.A. Isolation and characterization of microcystins, cyclic heptapeptide hepatotoxins from a Lake Erie Strain of Microcystis aeruginosa. J. Great Lakes Res. 2000, 26, 241–249. [Google Scholar] [CrossRef]
  144. Veerman, J.; Kumar, A.; Mishra, D.R. Exceptional landscape-wide cyanobacteria bloom in Okavango Delta, Botswana in 2020 coincided with a mass elephant die-off event. Harmful Algae 2022, 111, 102145. [Google Scholar] [CrossRef] [PubMed]
  145. Henrikson, T.; Ekberg, E.C.; Nilner, M. Symptoms and signs of temporomandibular disorders in girls with normal occlusion and Class II malocclusion. Acta Odontol. Scand. 1997, 55, 229–235. [Google Scholar] [CrossRef] [PubMed]
  146. 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]
  147. Jiang, L.; Kiselova, N.; Rosén, J.; Ilag, L.L. Quantification of neurotoxin BMAA (β-N-methylamino-L-alanine) in seafood from Swedish markets. Sci. Rep. 2014, 4, 6931. [Google Scholar] [CrossRef]
  148. Brand, L.E.; Pablo, J.; Compton, A.; Hammerschlag, N.; Mash, D.C. Cyanobacterial blooms and the occurrence of the neurotoxin, beta-N-methylamino-L-alanine (BMAA), in South Florida aquatic food webs. Harmful Algae 2010, 9, 620–635. [Google Scholar] [CrossRef]
  149. Sandhu, P.K.; Solonenka, J.T.; Murch, S.J. Neurotoxic non-protein amino acids in commercially harvested lobsters (Homarus americanus H. Milne-Edwards). Sci. Rep. 2024, 14, 8017. [Google Scholar] [CrossRef]
  150. Reside, A. Characterizing the Neurotoxic Potential of the algal toxin beta-methylamino-L-alanine (BMAA) in fish. Doctoral Dissertation, University of Guelph, Guelph, ON, Canada, 2022. [Google Scholar]
  151. Ai, Y.; Lee, S.; Lee, J. Drinking water treatment residuals from cyanobacteria bloom-affected areas: Investigation of potential impact on agricultural land application. Sci. Total Environ. 2020, 706, 135756. [Google Scholar] [CrossRef]
  152. Codd, G.; Bell, S.; Kaya, K.; Ward, C.; Beattie, K.; Metcalf, J. Cyanobacterial toxins, exposure routes and human health. Eur. J. Phycol. 1999, 34, 405–415. [Google Scholar] [CrossRef]
  153. Contardo-Jara, V.; Schwanemann, T.; Pflugmacher, S. Uptake of a cyanotoxin, β-N-methylamino-L-alanine, by wheat (Triticum aestivum). Ecotoxicol. Environ. Saf. 2014, 104, 127–131. [Google Scholar] [CrossRef]
  154. Contardo-Jara, V.; Schwanemann, T.; Esterhuizen-Londt, M.; Pflugmacher, S. Protein association of β-N-methylamino-L-alanine in Triticum aestivum via irrigation. Food Addit. Contam. Part A 2018, 35, 732–740. [Google Scholar] [CrossRef] [PubMed]
  155. Levizou, E.; Papadimitriou, T.; Papavasileiou, E.; Papadimitriou, N.; Kormas, K.A. Root vegetables bioaccumulate microcystins-LR in a developmental stage-dependent manner under realistic exposure scenario: The case of carrot and radish. Agric. Water Manag. 2020, 240, 106274. [Google Scholar] [CrossRef]
  156. Esterhuizen-Londt, M.; Pflugmacher, S. Vegetables cultivated with exposure to pure and naturally occurring β-N-methylamino-L-alanine (BMAA) via irrigation. Environ. Res. 2019, 169, 357–361. [Google Scholar] [CrossRef] [PubMed]
  157. Li, B.; Yu, S.; Li, G.; Chen, X.; Huang, M.; Liao, X.; Li, H.; Hu, F.; Wu, J. Transfer of a cyanobacterial neurotoxin, β-methylamino-L-alanine from soil to crop and its bioaccumulation in Chinese cabbage. Chemosphere 2019, 219, 997–1001. [Google Scholar] [CrossRef]
  158. Mohamed, Z.A.; Elnour, R.O.; Alamri, S.; Hashem, M.; Alshehri, A.M.; Campos, A.; Vasconcelos, V.; Badawye, H. Occurrence of β-N-methylamino-L-alanine (BMAA) toxin in irrigation water and field vegetable plants and assessing its potential risk to human health. Water Air Soil Pollut. 2024, 235, 72. [Google Scholar] [CrossRef]
  159. Samardzic, K.; Steele, J.R.; Violi, J.P.; Colville, A.; Mitrovic, S.M.; Rodgers, K.J. Toxicity and bioaccumulation of two non-protein amino acids synthesised by cyanobacteria, β-N-Methylamino-L-alanine (BMAA) and 2,4-diaminobutyric acid (DAB), on a crop plant. Ecotoxicol. Environ. Saf. 2021, 208, 111515. [Google Scholar] [CrossRef]
  160. Weralupitiya, C.; Wanigatunge, R.P.; Gunawardana, D.; Vithanage, M.; Magana-Arachchi, D. Cyanotoxins uptake and accumulation in crops: Phytotoxicity and implications on human health. Toxicon 2022, 211, 21–35. [Google Scholar] [CrossRef]
  161. Reed, D.M.; Brody, J.A. Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia on Guam, 1945–1972: I. Descriptive Epidemiology. Am. J. Epidemiol. 1975, 101, 287–301. [Google Scholar] [CrossRef]
  162. Spencer, P.S. Parkinsonism and motor neuron disorders: Lessons from Western Pacific ALS/PDC. J. Neurol. Sci. 2022, 433, 120021. [Google Scholar] [CrossRef]
  163. Belnap, J. Biological soil crusts and wind erosion. In Biological Soil Crusts: Structure, Function, and Management; Springer: Berlin/Heidelberg, Germany, 2003; pp. 339–347. [Google Scholar]
  164. Weber, B.; Belnap, J.; Büdel, B.; Antoninka, A.J.; Barger, N.N.; Chaudhary, V.B.; Darrouzet-Nardi, A.; Eldridge, D.J.; Faist, A.M.; Ferrenberg, S.; et al. What is a biocrust? A refined, contemporary definition for a broadening research community. Biol. Rev. 2022, 97, 1768–1785. [Google Scholar] [CrossRef]
  165. Chatziefthimiou, A.D.; Banack, S.A.; Cox, P.A. Biocrust-produced cyanotoxins are found vertically in the desert soil profile. Neurotox. Res. 2021, 39, 42–48. [Google Scholar] [CrossRef] [PubMed]
  166. 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 (Suppl. S2), 109–117. [Google Scholar] [CrossRef] [PubMed]
  167. Dulić, T.; Meriluoto, J.; Malešević, T.P.; Gajić, V.; Važić, T.; Tokodi, N.; Obreht, I.; Kostić, B.; Kosijer, P.; Khormali, F.; et al. Cyanobacterial diversity and toxicity of biocrusts from the Caspian Lowland loess deposits, North Iran. Quat. Int. 2017, 429, 74–85. [Google Scholar] [CrossRef]
  168. Richer, R.; Banack, S.A.; Metcalf, J.S.; Cox, P.A. The persistence of cyanobacterial toxins in desert soils. J. Arid Environ. 2015, 112, 134–139. [Google Scholar] [CrossRef]
  169. Powell, J.T.; Chatziefthimiou, A.D.; Banack, S.A.; Cox, P.A.; Metcalf, J.S. Desert crust microorganisms, their environment, and human health. J. Arid Environ. 2015, 112, 127–133. [Google Scholar] [CrossRef]
  170. Chrapusta, E.; Węgrzyn, M.; Zabaglo, K.; Kaminski, A.; Adamski, M.; Wietrzyk, P.; Bialczyk, J. Microcystins and anatoxin-a in Arctic biocrust cyanobacterial communities. Toxicon 2015, 101, 35–40. [Google Scholar] [CrossRef]
  171. Metcalf, J.S.; Banack, S.A.; Cox, P.A. Cyanotoxin analysis of air samples from the Great Salt Lake. Toxins 2023, 15, 659. [Google Scholar] [CrossRef]
  172. Jungblut, A.D.; Wilbraham, J.; Banack, S.A.; Metcalf, J.S.; Codd, G.A. Microcystins, BMAA and BMAA isomers in 100-year-old Antarctic cyanobacterial mats collected during Captain RF Scott’s Discovery Expedition. Eur. J. Phycol. 2018, 53, 115–121. [Google Scholar] [CrossRef]
  173. Metcalf, J.S.; Dunlop, R.A.; Banack, S.A.; Souza, N.R.; Cox, P.A. Cyanotoxin analysis and amino acid profiles of cyanobacterial food items from Chad. Neurotox. Res. 2021, 39, 72–80. [Google Scholar] [CrossRef]
  174. 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]
  175. 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]
  176. 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]
  177. 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]
  178. Sahu, N.; Tangutur, A.D. Airborne algae: Overview of the current status and its implications on the environment. Aerobiologia 2015, 31, 89–97. [Google Scholar] [CrossRef]
  179. Sharma, N.K.; Singh, S.; Rai, A.K. Diversity and seasonal variation of viable algal particles in the atmosphere of a subtropical city in India. Environ. Res. 2006, 102, 252–259. [Google Scholar] [CrossRef]
  180. Sharma, N.K.; Rai, A.K.; Singh, S.; Brown, R.M., Jr. Airborne algae: Their present status and relevance 1. J. Phycol. 2007, 43, 615–627. [Google Scholar] [CrossRef]
  181. Sharma, N.K.; Singh, S. Differential aerosolization of algal and cyanobacterial particles in the atmosphere. Indian J. Microbiol. 2010, 50, 468–473. [Google Scholar] [CrossRef]
  182. Metcalf, J.S.; Chatziefthimiou, A.D.; Souza, N.R.; Cox, P.A. Desert Dust as a Vector for Cyanobacterial Toxins. The Arabian Seas: Biodiversity, Environmental Challenges and Conservation Measures; Springer: Berlin/Heidelberg, Germany, 2021; pp. 161–178. [Google Scholar]
  183. Bernstein, I.L.; Safferman, R.S. Viable algae in house dust. Nature 1970, 227, 851–852. [Google Scholar] [CrossRef]
  184. Chu, W.L.; Tneh, S.Y.; Ambu, S. A survey of airborne algae and cyanobacteria within the indoor environment of an office building in Kuala Lumpur, Malaysia. Grana 2013, 52, 207–220. [Google Scholar] [CrossRef]
  185. Holland, R.D.; Walne, P.L.; Richardson, C.B.; Hornsby, R.P. Viable Algae from Housedust-Possible Causal Agents in Human Allergenicity. J. Phycol. 1973, 9, 11–12. [Google Scholar]
  186. Tiberg, E.; Bergman, B.; Wictorin, B.; Willen, T. Occurrence of microalgae in indoor and outdoor environments in Sweden. Nord. Aerobiol. 1983, 24–29. [Google Scholar]
  187. Blanchard, D.C.; Syzdek, L.D. Electrostatic collection of jet and film drops 1. Limnol. Oceanogr. 1975, 20, 762–774. [Google Scholar] [CrossRef]
  188. Blanchard, D.C.; Syzdek, L.D. Water-to-air transfer and enrichment of bacteria in drops from bursting bubbles. Appl. Environ. Microbiol. 1982, 43, 1001–1005. [Google Scholar] [CrossRef] [PubMed]
  189. Blanchard, D.C. The size and height to which jet drops are ejected from bursting bubbles in seawater. J. Geophys. Res. Ocean. 1989, 94, 10999–11002. [Google Scholar] [CrossRef]
  190. 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]
  191. Shi, J.H.; Olson, N.E.; Birbeck, J.A.; Pan, J.; Peraino, N.J.; Holen, A.L.; Ledsky, I.R.; Jacquemin, S.J.; Marr, L.C.; Schmale, D.G. Aerosolized Cyanobacterial Harmful Algal Bloom Toxins: Microcystin Congeners Quantified in the Atmosphere. Environ. Sci. Technol. 2023, 57, 21801–21814. [Google Scholar] [CrossRef]
  192. Oksanen, I.; Jokela, J.; Fewer, D.P.; Wahlsten, M.; Rikkinen, J.; Sivonen, K. Discovery of rare and highly toxic microcystins from lichen-associated cyanobacterium Nostoc sp. strain IO-102-I. Appl. Environ. Microbiol. 2004, 70, 5756–5763. [Google Scholar] [CrossRef]
  193. Kaasalainen, U.; Fewer, D.P.; Jokela, J.; Wahlsten, M.; Sivonen, K.; Rikkinen, J. Cyanobacteria produce a high variety of hepatotoxic peptides in lichen symbiosis. Proc. Natl. Acad. Sci. USA 2012, 109, 5886–5891. [Google Scholar] [CrossRef]
  194. Zhang, Y.; Husk, B.R.; Duy, S.V.; Dinh, Q.T.; Sanchez, J.S.; Sauvé, 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]
  195. Whitman, W.B.; Coleman, D.C.; Wiebe, W.J. Prokaryotes: The unseen majority. Proc. Natl. Acad. Sci. USA 1998, 95, 6578–6583. [Google Scholar] [CrossRef]
  196. Chatziefthimiou, A.D.; Metcalf, J.S.; Glover, W.B.; Banack, S.A.; Dargham, S.R.; Richer, R.A. Cyanobacteria and cyanotoxins are present in drinking water impoundments and groundwater wells in desert environments. Toxicon 2016, 114, 75–84. [Google Scholar] [CrossRef]
  197. Ehrenberg, C.G. Bericht über die zur Bekanntmachung geeigneten Verhandlungen der Königlich-Preussischen. Akad. Wiss. Berl. 1844, 9, 182–207. [Google Scholar]
  198. Murby, A.L.; Haney, J.F. Field and laboratory methods to monitor lake aerosols for cyanobacteria and microcystins. Aerobiologia 2016, 32, 395–403. [Google Scholar] [CrossRef]
  199. Genitsaris, S.; Kormas, K.A.; Moustaka-Gouni, M. Airborne algae and cyanobacteria: Occurrence and related health effects. Front. Biosci (Elite Ed.) 2011, 3, 772–787. [Google Scholar] [CrossRef] [PubMed]
  200. Lewandowska, A.U.; Śliwińska-Wilczewska, S.; Woźniczka, D. Identification of cyanobacteria and microalgae in aerosols of various sizes in the air over the Southern Baltic Sea. Mar. Pollut. Bull. 2017, 125, 30–38. [Google Scholar] [CrossRef]
  201. Brookes, J.D.; Ganf, G.G. Variations in the buoyancy response of Microcystis aeruginosa to nitrogen, phosphorus and light. J. Plankton Res. 2001, 23, 1399–1411. [Google Scholar] [CrossRef]
  202. Ahern, K.S.; Ahern, C.R.; Savige, G.M.; Udy, J.W. Mapping the distribution, biomass and tissue nutrient levels of a marine benthic cyanobacteria bloom (Lyngbya majuscula). Mar. Freshw. Res. 2007, 58, 883–904. [Google Scholar] [CrossRef]
  203. Vignati, E.; de Leeuw, G.; Schulz, M.; Plate, E. Characterization of aerosols at a coastal site near Vindeby (Denmark). J. Geophys. Res. Ocean. 1999, 104, 3277–3287. [Google Scholar] [CrossRef]
  204. Dueker, M.E.; O’Mullan, G.D.; Martínez, J.M.; Juhl, A.R.; Weathers, K.C. Onshore wind speed modulates microbial aerosols along an urban waterfront. Atmosphere 2017, 8, 215. [Google Scholar] [CrossRef]
  205. 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]
  206. Backer, L.C.; Carmichael, W.; Kirkpatrick, B.; Williams, C.; Irvin, M.; Zhou, Y.; Johnson, T.B.; Nierenberg, K.; Hill, V.R.; Kieszak, S.M. Recreational exposure to low concentrations of microcystins during an algal bloom in a small lake. Mar. Drugs 2008, 6, 389–406. [Google Scholar] [CrossRef] [PubMed]
  207. Backer, L.C.; McNeel, S.V.; Barber, T.; Kirkpatrick, B.; Williams, C.; Irvin, M.; Zhou, Y.; Johnson, T.B.; Nierenberg, K.; Aubel, M. Recreational exposure to microcystins during algal blooms in two California lakes. Toxicon 2010, 55, 909–921. [Google Scholar] [CrossRef] [PubMed]
  208. Cheng, Y.S.; Zhou, Y.; Irvin, C.M.; Kirkpatrick, B.; Backer, L.C. Characterization of aerosols containing microcystin. Mar. Drugs 2007, 5, 136–150. [Google Scholar] [CrossRef] [PubMed]
  209. Gambaro, A.; Barbaro, E.; Zangrando, R.; Barbante, C. Simultaneous quantification of microcystins and nodularin in aerosol samples using high-performance liquid chromatography/negative electrospray ionization tandem mass spectrometry. Rapid Commun. Mass Spectrom. 2012, 26, 1497–1506. [Google Scholar] [CrossRef]
  210. Hu, Y.; Chen, J.; Fan, H.; Xie, P.; He, J. A review of the neurotoxicity of microcystins. Environ. Sci. Pollut. Res. 2016, 23, 7211–7219. [Google Scholar] [CrossRef]
  211. Sutherland, J.W.; Turcotte, R.J.; Molden, E.; Moriarty, V.; Kelly, M.; Aubel, M.; Foss, A. The detection of airborne anatoxin-a (ATX) on glass fiber filters during a harmful algal bloom. Lake Reserv. Manag. 2021, 37, 113–119. [Google Scholar]
  212. Dabney, A.D. Detection of Microcystin Aerosol Particles. Master’s Thesis, University of South Carolina, Columbia, SC, USA, 2022. [Google Scholar]
  213. Yu, S.; Zhou, X.; Hu, P.; Chen, H.; Shen, F.; Yu, C.; Meng, H.; Zhang, Y.; Wu, Y. Inhalable particle-bound marine biotoxins in a coastal atmosphere: Concentration levels, influencing factors and health risks. J. Hazard. Mater. 2022, 434, 128925. [Google Scholar] [CrossRef]
  214. 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. Amyotroph. Lateral Scler. 2009, 10 (Suppl. S2), 101–108. [Google Scholar] [CrossRef]
  215. Wiśniewska, 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]
  216. Kirso, U.; Paalme, L.; Voll, M.; Urbas, E.; Irha, N. Accumulation of carcinogenic hydrocarbons at the sediment-water interface. Mar. Chem. 1990, 30, 337–341. [Google Scholar] [CrossRef]
  217. Kowalewska, G.; Konat, J. Distribution of polynuclear aromatic hydrocarbons (PAHs) in sediments of the southern Baltic Sea. Oceanogr. Lit. Rev. 1997, 12, 1559. [Google Scholar]
  218. Baird, W.M.; Hooven, L.A.; Mahadevan, B. Carcinogenic polycyclic aromatic hydrocarbon-DNA adducts and mechanism of action. Env. Mol Mutagen. 2005, 45, 106–114. [Google Scholar] [CrossRef] [PubMed]
  219. ATSDR Agency for Toxic Substances and Disease Registry. Public Health Statement for Polycyclic Aromatic Hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons (PAHs)|Public Health Statement|ATSDR. Available online: https://www.scirp.org/reference/referencespapers?referenceid=202673 (accessed on 28 August 2014).
  220. EPA United States Environmental Protection Agency. Metals. Metals|US EPA. Available online: https://www.researchgate.net/figure/United-State-Environmental-Protection-Agency-USEPA-maximum-contamination-levels-for_tbl1_349899836 (accessed on 7 February 2025).
  221. Andrew, A.S.; O’Brien, K.M.; Jackson, B.P.; Sandler, D.P.; Kaye, W.E.; Wagner, L.; Stommel, E.W.; Horton, D.K.; Mehta, P.; Weinberg, C.R. Keratinous biomarker of mercury exposure associated with amyotrophic lateral sclerosis risk in a nationwide U.S. study. Amyotroph Lateral Scler Front. Degener. 2020, 21, 420–427. [Google Scholar] [CrossRef] [PubMed]
  222. Hoffman, H.I.; Bradley, W.G.; Chen, C.Y.; Pioro, E.P.; Stommel, E.W.; Andrew, A.S. Amyotrophic lateral sclerosis risk, family income, and fish consumption estimates of mercury and omega-3 PUFAs in the United States. Int. J. Environ. Res. Public Health 2021, 18, 4528. [Google Scholar] [CrossRef]
  223. Tao, Y.; Xue, B.; Yang, Z.; Yao, S.; Li, S. Effects of heavy metals on the sorption of polycyclic aromatic hydrocarbons by. J. Environ. Qual. 2014, 43, 1953–1962. [Google Scholar] [CrossRef]
  224. Stewart, I.; Carmichael, W.W.; Sadler, R.; McGregor, G.B.; Reardon, K.; Eaglesham, G.K.; A Wickramasinghe, W.; A Seawright, A.; Shaw, G.R. Occupational and environmental hazard assessments for the isolation, purification and toxicity testing of cyanobacterial toxins. Environ. Health 2009, 8, 52. [Google Scholar] [CrossRef]
  225. Stewart, I.; Webb, P.M.; Schluter, P.J.; Shaw, G.R. Recreational and occupational field exposure to freshwater cyanobacteria–a review of anecdotal and case reports, epidemiological studies and the challenges for epidemiologic assessment. Environ. Health 2006, 5, 6. [Google Scholar] [CrossRef]
  226. Turner, P.C.; Gammie, A.J.; Hollinrake, K.; Codd, G.A. Pneumonia associated with contact with cyanobacteria. Br. Med. J. 1990, 300, 1440. [Google Scholar] [CrossRef]
  227. Richer, R.; Anchassi, D.; El-Assaad, I.; El-Matbouly, M.; Ali, F.; Makki, I.; Metcalf, J.S. Variation in the coverage of biological soil crusts in the State of Qatar. J. Arid Environ. 2012, 78, 187–190. [Google Scholar] [CrossRef]
  228. Horner, R.; Kamins, K.; Feussner, J.; Grambow, S.; Hoff-Lindquist, J.; Harati, Y.; Mitsumoto, H.; Pascuzzi, R.; Spencer, P.; Tim, R.; et al. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology 2003, 61, 742–749. [Google Scholar] [CrossRef]
  229. 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]
  230. Coffman, C.J.; Horner, R.D.; Grambow, S.C.; Lindquist, J. Estimating the occurrence of amyotrophic lateral sclerosis among Gulf War (1990–1991) veterans using capture-recapture methods. Neuroepidemiology 2005, 24, 141–150. [Google Scholar] [CrossRef] [PubMed]
  231. Miranda, M.L.; Galeano, M.A.O.; 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] [PubMed]
  232. Vernham, G.; Bailey, J.J.; Chase, J.M.; Hjort, J.; Field, R.; Schrodt, F. Understanding trait diversity: The role of geodiversity. Trends Ecol. Evol. 2023, 38, 736–748. [Google Scholar] [CrossRef]
  233. Fitzgeorge, R.B.; Clark, S.A.; Keevil, C.W. Routes of Intoxication. In Detection Methods for Cyanobacterial Toxins; The Royal Society of Chemistry: London, UK, 1994; pp. 69–74. [Google Scholar]
  234. Creasia, D.A. Acute inhalation toxicity of microcystin-LR with mice. Toxicon 1990, 28, 605. [Google Scholar]
  235. Fawell, J.K.; Mitchell, R.E.; Hill, R.E.; Everett, D.J. The toxicity of cyanobacterial toxins in the mouse: II anatoxin-a. Hum. Exp. Toxicol. 1999, 18, 168–173. [Google Scholar] [CrossRef]
  236. Benson, J.M.; Hutt, J.A.; Rein, K.; Boggs, S.E.; Barr, E.B.; Fleming, L.E. The toxicity of microcystin-LR in mice following 7 days of inhalation exposure. Toxicon 2005, 45, 691–698. [Google Scholar] [CrossRef]
  237. 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]
  238. 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]
  239. Ziesemer, S.; Meyer, S.; Edelmann, J.; Vennmann, J.; Gudra, C.; Arndt, D.; Effenberg, M.; Hayas, O.; Hayas, A.; Thomassen, J.S. Target mechanisms of the cyanotoxin cylindrospermopsin in immortalized human airway epithelial cells. Toxins 2022, 14, 785. [Google Scholar] [CrossRef]
  240. Kubickova, B.; Laboha, P.; Hildebrandt, J.P.; Hilscherová, K.; Babica, P. Effects of cylindrospermopsin on cultured immortalized human airway epithelial cells. Chemosphere 2019, 220, 620–628. [Google Scholar] [CrossRef] [PubMed]
  241. Ojima, Y.; Nguyen, M.H.; Yajima, R.; Taya, M. Flocculation of Escherichia coli cells in association with enhanced production of outer membrane vesicles. Appl. Environ. Microbiol. 2015, 81, 5900–5906. [Google Scholar] [CrossRef] [PubMed]
  242. Lürling, M.; Kang, L.; Mucci, M.; van Oosterhout, F.; Noyma, N.P.; Miranda, M.; Huszar, V.L.; Waajen, G.; Marinho, M.M. Coagulation and precipitation of cyanobacterial blooms. Ecol. Eng. 2020, 158, 106032. [Google Scholar] [CrossRef]
  243. Sukenik, A.; Kaplan, A. Cyanobacterial harmful algal blooms in aquatic ecosystems: A comprehensive outlook on current and emerging mitigation and control approaches. Microorganisms 2021, 9, 1472. [Google Scholar] [CrossRef]
  244. Zang, X.; Zhang, H.; Liu, Q.; Li, L.; Li, L.; Zhang, X. Harvesting of Microcystis flos-aquae using chitosan coagulation: Influence of proton-active functional groups originating from extracellular and intracellular organic matter. Water Res. 2020, 185, 116272. [Google Scholar] [CrossRef] [PubMed]
  245. Liu, J.; Khalaf, R.; Ulén, B.; Bergkvist, G. Potential phosphorus release from catch crop shoots and roots after freezing-thawing. Plant Soil 2013, 371, 543–557. [Google Scholar] [CrossRef]
  246. EPA United States Environmental Protection Agency. Lake Erie Binational Nutrient Management Strategy. Lake Erie Binational Nutrient Management Strategy|US EPA. Available online: https://www.epa.gov/greatlakes/lake-erie-binational-nutrient-management-strategy (accessed on 16 September 2024).
  247. Smith, D.R.; King, K.W.; Williams, M.R. What is causing the harmful algal blooms in Lake Erie? J. Soil Water Conserv. 2015, 70, 27A–29A. [Google Scholar] [CrossRef]
  248. White, C.M.; Weil, R.R. Forage radish cover crops increase soil test phosphorus surrounding radish taproot holes. Soil Sci. Soc. Am. J. 2011, 75, 121–130. [Google Scholar] [CrossRef]
  249. Carver, R.E.; Nelson, N.O.; Roozeboom, K.L.; Kluitenberg, G.J.; Tomlinson, P.J.; Kang, Q.; Abel, D.S. Cover crop and phosphorus fertilizer management impacts on surface water quality from a no-till corn-soybean rotation. J. Environ. Manag. 2022, 301, 113818. [Google Scholar] [CrossRef]
  250. Soltangheisi, A.; Teles, A.P.B.; Sartor, L.R.; Pavinato, P.S. Cover cropping may alter legacy phosphorus dynamics under long-term fertilizer addition. Front. Environ. Sci. 2020, 8, 13. [Google Scholar] [CrossRef]
  251. Wang, C.; Yang, Y.; Hou, J.; Wang, P.; Miao, L.; Wang, X.; Guo, L. Optimization of cyanobacterial harvesting and extracellular organic matter removal utilizing magnetic nanoparticles and response surface methodology: A comparative study. Algal Res. 2020, 45, 101756. [Google Scholar] [CrossRef]
  252. Nie, C.; Geng, X.; Zhang, R.; Wang, L.; Li, L.; Chen, J. Abundant cyanobacteria in autumn adhering to the heating, ventilation, and air-conditioning (HVAC) in Shanghai. Microorganisms 2023, 11, 1835. [Google Scholar] [CrossRef] [PubMed]
  253. Gaston, C.J.; Royer, H.M.; Leibensperger, R.J., III; Maizel, D.; Lanpher, K.B.; Solo-Gabriele, H.; Brand, L.E.; Zhai, R.G.; Caban-Martinez, A.J.; Popendorf, K.J. Filtration efficiency of air conditioner filters and face masks to limit exposure to aerosolized algal toxins. Aerosol Air Qual. Res. 2021, 21, 210016. [Google Scholar] [CrossRef]
  254. Hardison, D.R.; Holland, W.C.; Currier, R.D.; Kirkpatrick, B.; Stumpf, R.; Fanara, T.; Burris, D.; Reich, A.; Kirkpatrick, G.J.; Litaker, R.W. HABscope: A tool for use by citizen scientists to facilitate early warning of respiratory irritation caused by toxic blooms of Karenia brevis. PLoS ONE 2019, 14, e0218489. [Google Scholar] [CrossRef]
  255. Campbell, L.; Henrichs, D.W.; Olson, R.J.; Sosik, H.M. Continuous automated imaging-in-flow cytometry for detection and early warning of Karenia brevis blooms in the Gulf of Mexico. Environ. Sci. Pollut. Res. 2013, 20, 6896–6902. [Google Scholar] [CrossRef]
  256. Ko, B.; Park, J.; Nam, J.Y. Spatiotemporal bag-of-features for early wildfire smoke detection. Image Vis. Comput. 2013, 31, 786–795. [Google Scholar] [CrossRef]
  257. Bridgeman, T.B.; Chaffin, J.D.; Filbrun, J.E. A novel method for tracking western Lake Erie Microcystis blooms, 2002–2011. J. Great Lakes Res. 2013, 39, 83–89. [Google Scholar] [CrossRef]
  258. Coffer, M.M.; Schaeffer, B.A.; Darling, J.A.; Urquhart, E.A.; Salls, W.B. Quantifying national and regional cyanobacterial occurrence in US lakes using satellite remote sensing. Ecol. Indic. 2020, 111, 105976. [Google Scholar] [CrossRef]
  259. Ganf, G.G. Phytoplankton biomass and distribution in a shallow eutrophic lake (Lake George, Uganda). Oecologia 1974, 16, 9–29. [Google Scholar] [CrossRef]
  260. Vareschi, E. The ecology of Lake Nakuru (Kenya) I. Abundance and feeding of the Lesser Flamingo. Oecologia 1978, 32, 11–35. [Google Scholar] [CrossRef]
  261. Vareschi, E. The ecology of Lake Nakuru (Kenya) III. Abiotic factors and primary production. Oecologia 1982, 55, 81–101. [Google Scholar] [CrossRef] [PubMed]
  262. Vareschi, E.; Jacobs, J. The ecology of Lake Nakuru (Kenya) V. Production and consumption of consumer organisms. Oecologia 1984, 61, 83–98. [Google Scholar] [CrossRef] [PubMed]
  263. Vaeth, M.J.E.; Cheema, M.; Omer, S.; Gupta, I.; Sun, K.J.; Mitchell, A.; Elhabashy, M.; Foyez, M.; Cheema, A.; Javed, B. Self-administered versus clinician-performed BinaxNOW COVID rapid test: A comparison of accuracy. Microbiol. Spectr. 2024, 12, e02525-23. [Google Scholar] [CrossRef] [PubMed]
  264. Hodinka, R.L.; Kaiser, L. Point-Counterpoint: Is the era of viral culture over in the clinical microbiology laboratory? J. Clin. Microbiol. 2013, 51, 2–8. [Google Scholar] [CrossRef]
  265. Pacheco, A.B.F.; Guedes, I.A.; Azevedo, S.M. Is qPCR a reliable indicator of cyanotoxin risk in freshwater? Toxins 2016, 8, 172. [Google Scholar] [CrossRef]
  266. Rudko, S.P.; Reimink, R.L.; Peter, B.; White, J.; Hanington, P.C. Democratizing water monitoring: Implementation of a community-based qPCR monitoring program for recreational water hazards. PLoS ONE 2020, 15, e0229701. [Google Scholar] [CrossRef]
  267. Clements, E.; Thompson, K.A.; Hannoun, D.; Dickenson, E.R. Classification machine learning to detect de facto reuse and cyanobacteria at a drinking water intake. Sci. Total Environ. 2024, 948, 174690. [Google Scholar] [CrossRef]
  268. Fournier, C.; Quesada, A.; Cirés, S.; Saberioon, M. Discriminating bloom-forming cyanobacteria using lab-based hyperspectral imagery and machine learning: Validation with toxic species under environmental ranges. Sci. Total Environ. 2024, 932, 172741. [Google Scholar] [CrossRef]
  269. Mellios, N.; Moe, S.J.; Laspidou, C. Machine learning approaches for predicting health risk of cyanobacterial blooms in northern european lakes. Water 2020, 12, 1191. [Google Scholar] [CrossRef]
  270. Hill, A.B. The environment and disease: Association or causation? Proc. R. Soc. Med. 1965, 58, 295–300. [Google Scholar] [CrossRef]
  271. Newell, M.E.; Adhikari, S.; Halden, R.U. Systematic and state-of the science review of the role of environmental factors in Amyotrophic Lateral Sclerosis (ALS) or Lou Gehrig’s Disease. Sci. Total Environ. 2022, 817, 152504. [Google Scholar] [CrossRef] [PubMed]
  272. 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. Int. J. Mol. Sci. 2021, 22, 8726. [Google Scholar] [CrossRef] [PubMed]
  273. Metcalf, J.S.; Codd, G.A. Co-occurrence of cyanobacteria and cyanotoxins with other environmental health hazards: Impacts and implications. Toxins 2020, 12, 629. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure of anatoxin-a.
Figure 1. Structure of anatoxin-a.
Molecules 30 02320 g001
Figure 2. Structure of guanitoxin.
Figure 2. Structure of guanitoxin.
Molecules 30 02320 g002
Figure 3. General structure of saxitoxin.
Figure 3. General structure of saxitoxin.
Molecules 30 02320 g003
Figure 4. Structure of BMAA.
Figure 4. Structure of BMAA.
Molecules 30 02320 g004
Figure 5. Structure of Microcystin-LR (MC-LR).
Figure 5. Structure of Microcystin-LR (MC-LR).
Molecules 30 02320 g005
Figure 6. Structure of Cylindrospermopsin.
Figure 6. Structure of Cylindrospermopsin.
Molecules 30 02320 g006
Table 1. Cyanotoxins and neurological conditions.
Table 1. Cyanotoxins and neurological conditions.
CyanotoxinOxidative StressNeuro-InflammationProtein MisfoldingNeuro-StimulationNeuro-InhibitionReferences
Anatoxin-a+--+-[44,45,46,47]
Guanitoxin+--+-[48,49]
Saxitoxin++--+[50,51,52,53,54,55]
Amino Acids *++++-[56,57,58,59,60,61,62,63,64]
Microcystin++-++[65,66,67,68,69,70,71,72,73,74,75,76,77]
Cylindrospermopsin++--+[78,79,80,81,82,83,84,85,86,87,88,89,90]
* Includes BMAA and isomers.
Table 2. Toxicological analysis of inhaled anatoxin-a, MC-LR, and BMAA in rodent models.
Table 2. Toxicological analysis of inhaled anatoxin-a, MC-LR, and BMAA in rodent models.
CyanotoxinRouteLD50Test OrganismReference
Anatoxin-aInhalation2000 µg/kgMouse[233]
Microcystin-LRInhalation250 µg/kgMouse[233]
Microcystin-LRAerosolNA *Mouse[233]
Microcystin-LRInhalation43 µg/kgMouse[234]
BMAAInhalationNARat[236]
* This study was limited to aerosol exposure at 0.0005 µg/kg. No deaths were observed at 0.0005 µg/kg.
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

Morris, Z.J.; Stommel, E.W.; Metcalf, J.S. Airborne Cyanobacterial Toxins and Their Links to Neurodegenerative Diseases. Molecules 2025, 30, 2320. https://doi.org/10.3390/molecules30112320

AMA Style

Morris ZJ, Stommel EW, Metcalf JS. Airborne Cyanobacterial Toxins and Their Links to Neurodegenerative Diseases. Molecules. 2025; 30(11):2320. https://doi.org/10.3390/molecules30112320

Chicago/Turabian Style

Morris, Zachary James, Elijah W. Stommel, and James Spencer Metcalf. 2025. "Airborne Cyanobacterial Toxins and Their Links to Neurodegenerative Diseases" Molecules 30, no. 11: 2320. https://doi.org/10.3390/molecules30112320

APA Style

Morris, Z. J., Stommel, E. W., & Metcalf, J. S. (2025). Airborne Cyanobacterial Toxins and Their Links to Neurodegenerative Diseases. Molecules, 30(11), 2320. https://doi.org/10.3390/molecules30112320

Article Metrics

Back to TopTop