Next Article in Journal
Assessment of Suitable Habitats and Identification of Key Protection Areas for Polyplectron katsumatae in Jianfengling, Hainan Province, China
Previous Article in Journal
Injectables in the Therapy of Mid-Portion Achilles Tendinopathy, a Descriptive Review
Previous Article in Special Issue
Engineered Chlamydomonas reinhardtii Strains for Enhanced Astaxanthin Production
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Global Occurrence of Cyanotoxins in Drinking Water Systems: Recent Advances, Human Health Risks, Mitigation, and Future Directions

by
Jerikias Marumure
1,2,
Willis Gwenzi
3,4,5,
Zakio Makuvara
1,2,
Tinoziva T. Simbanegavi
6,
Richwell Alufasi
7,
Marvelous Goredema
7,
Claudious Gufe
8,
Rangarirayi Karidzagundi
9,
Piotr Rzymski
10 and
Dariusz Halabowski
11,*
1
Department of Physics, Geography and Environmental Science, School of Natural Sciences, Great Zimbabwe University, Masvingo, Zimbabwe
2
Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Pretoria 0002, South Africa
3
Formerly Alexander von Humboldt Fellow, Leibniz-Institut für Agrartechnik und Bioökonomie e.V. (ATB), Max-Eyth-Allee 100, D-14469 Potsdam, Germany
4
Formerly Alexander von Humboldt Fellow, Grassland Science and Renewable Plant Resources, Faculty of Organic Agricultural Sciences, Universität Kassel, Steinstraße 19, D-37213 Witzenhausen, Germany
5
Independent Researcher, Biosystems & Environmental Engineering Research Group, 380 New Adylin, Westgate, Harare, Zimbabwe
6
Department of Soil Science and Environment, Faculty of Agriculture, Environment, and Food Systems, University of Zimbabwe, P.O. Box MP 167, Mount Pleasant, Harare, Zimbabwe
7
Biological Sciences Department, Bindura University of Science Education, 741 Chimurenga Road, Off Trojan Road, Bindura, Zimbabwe
8
Department of Veterinary Technical Services, Central Veterinary Laboratories, P.O. Box CY55, 18A Borrowdale Road, Harare, Zimbabwe
9
Materials Development Unit, Zimbabwe Open University, P.O. Box MP1119, Mount Pleasant, Harare, Zimbabwe
10
Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland
11
University of Lodz, Faculty of Biology and Environmental Protection, Department of Ecology and Vertebrate Zoology, 90-237 Lodz, Poland
*
Author to whom correspondence should be addressed.
Life 2025, 15(5), 825; https://doi.org/10.3390/life15050825
Submission received: 5 April 2025 / Revised: 15 May 2025 / Accepted: 16 May 2025 / Published: 21 May 2025

Abstract

:
This paper applies a semi-quantitative approach to review the diversity, environmental controls, detection methods, human health risks, and mitigation of cyanotoxins in drinking water systems (DWSs). It discusses the environmental factors controlling the occurrence of cyanotoxins, presents the merits and limitations of emerging methods of their detection (qPCR, liquid chromatography–mass spectrometry, and electrochemical biosensors), and outlines the human exposure pathways and health outcomes with identification of high-risk groups and settings. High-risk groups include (1) communities relying on untreated drinking water from unsafe, polluted water sources and (2) low-income countries where cyanotoxins are not routinely monitored in DWSs. The fate and behavior processes are discussed, including removing cyanotoxins in DWSs based on conventional and advanced treatment processes. The available methods for cyanotoxin removal presented in this paper include (1) polymer-based adsorbents, (2) coagulation/flocculation, (3) advanced oxidation processes, (4) ultra- and nanofiltration, and (5) multi-soil layer systems. Future research should address (1) detection and fate in storage and conveyance facilities and at the point of consumption, (2) degradation pathways and toxicity of by-products or metabolites, (3) interactive health effects of cyanotoxins with legacy and emerging contaminants, (4) removal by low-cost treatment techniques (e.g., solar disinfection, boiling, bio-sand filtration, and chlorination), (5) quantitative health risk profiling of high-risk groups, and (6) epidemiological studies to link the prevalence of human health outcomes (e.g., cancer) to cyanotoxins in DWSs.

1. Introduction

Cyanobacterial Harmful Algal Blooms (CyanoHABs) occur in diverse surface waters, including those serving as drinking water sources [1]. These blooms are characterized by the rapid proliferation of Cyanobacteria, some of which produce toxic metabolites known as cyanotoxins [2]. They constitute a diverse group of chemical compounds that differ in mechanisms of toxic action.
The occurrence of cyanotoxins in different water systems has been extensively studied in high-income countries [3,4,5,6] and, to a lesser extent, in developing countries [7,8,9]. However, research remains limited in low-income regions like Africa, where studies have been conducted, e.g., in Zimbabwe [10,11], Ethiopia [12,13], Kenya [14,15], and Uganda [16,17,18] (Table S1). Some studies have also investigated and reported harmful algae and cyanotoxins in sources of drinking water [11,17], drinking water treatment facilities [19], containerized drinking water in households [20], and mineral water [21]. Cyanotoxins in drinking water pose risks to human health, primarily through ingestion.
Recent reviews have explored various aspects of cyanotoxins, including (1) their presence in aquatic ecosystems and organisms such as fish, with a limited focus on drinking water systems [9,22], (2) the diversity of Cyanobacteria and cyanotoxins [7,23], and (3) methods for removing cyanotoxins from drinking water [24,25,26]. However, a comprehensive review of the occurrence of cyanotoxins in drinking water systems (DWSs), their detection, exposure risks, health hazards, and mitigation strategies is currently lacking. DWSs include the entire continuum from source water (surface and groundwater) to water treatment, storage, distribution, and the point of use (e.g., tap water, containerized drinking water, and bottled mineral water).
The present work aims to (1) examine the nature, sources, and diversity of cyanotoxins and their global occurrence along the source water–drinking water continuum, with a particular focus on low- and middle-income regions, (2) review evidence on human health risks, including exposure routes and high-risk populations, (3) explore toxicokinetics using the adsorption–distribution–metabolism–excretion (ADME) framework and mechanisms of human toxicity, (4) analyze the fate, behavior, and mitigation of cyanotoxins in DWSs, (5) propose a mitigation framework and outline ten key knowledge gaps, including the potential roles of ethnomedicine and ethnotoxicology, and (6) identify future research directions and priorities. The focus of this paper is summarized in Figure 1.

2. Methods

This study applied a generic semi-quantitative approach adapted from earlier reviews by our group [27]. The approach comprised (1) Boolean search techniques and (2) subsequent qualitative data analysis.

Boolean Search and Retrieval Techniques

Boolean techniques were used to search and retrieve literature from English language scholarly databases (ScienceDirect®, Clarivate’s Web of Science®, Google Scholar®, Scopus®, and ResearchGate®). The focus on English language databases might have omitted some of these studies in other languages. However, given that English is the dominant language for scientific publication, it is believed that the literature and databases used accounted for the bulk of the articles. Particular attention was paid to recent articles covering a search period from 2000 to date, but earlier studies were included in cases where data were limited on particular aspects (e.g., Africa, human health risks).
The individual articles were then subjected to a preliminary qualitative screening and evaluation based on the study objectives. Nevertheless, there was limited evidence of drinking contamination by emerging contaminants before 2000. Therefore, subsequent searches were restricted to the period from 2000 to 2023.
Briefly, representative search strings for specific aspects were as follows:
Occurrence of cyanotoxins in drinking water systems
cyanotoxin(s) OR algal toxin(s) OR microcystin(s) or harmful alga(e)’ AND ‘drinking water systems OR drinking water OR potable water OR water sources OR groundwater OR surface water OR stored drinking water’ AND/AND NOT ‘Africa OR Europe OR North America OR Asia OR Australia OR Oceania OR south America OR Latin America OR Caribbea(n)’.
Cyanotoxin toxicokinetics
‘Cyanotoxins (and its variants)’ AND ‘Toxicokinetic(s) OR behavior OR ADME or adsorption–distribution–metabolism–excretion’ AND ‘human(s) OR human body OR mammal(s) OR in vivo OR in vitro OR preclinical OR rats OR mouse/mice OR pig(s)’.
Human exposure, toxicology, and health risks
‘Cyanotoxins (and its variant terms)’ AND ‘toxicity mechanisms (and variant terms (e.g., hepatotoxins, neurotoxins, dermatoxins, cytotoxins)’ OR ‘toxicology OR toxicological effects OR toxicity OR toxic effects OR health risks OR health hazards OR health effects OR health impacts OR health outcomes OR exposure pathway(s) OR exposure route(s)’ AND ‘human(s) OR rat(s) OR preclinical animals OR mammals OR mouse/mice OR pigs’.
Behavior, fate, and remediation
‘Cyanotoxins (and its variants)’ AND ‘behavior OR fate OR degradation OR removal OR remediation OR treatment OR treatment process/methods’ AND ‘drinking OR drinking water OR potable water OR drinking water systems’.
It is important to note that the presence of cyanotoxins in source waters (e.g., lakes, reservoirs, and rivers) was not assumed to directly imply contamination of formal drinking water systems (DWSs). Rather, our approach was to consider the broader source water–drinking water continuum, which includes both treated systems and regions where populations rely on untreated or minimally treated water sources. In many low- and middle-income regions, formal supply systems may be absent, and water is often collected directly from the environment for domestic use. We have therefore clarified in the manuscript that contamination in natural water bodies serves as a proxy for potential human exposure, with implications for both infrastructure-supported and informal water access scenarios.
The retrieved peer-reviewed articles were qualitatively screened for relevance to the study objectives, and relevant ones were retained for further analysis. Then, the relevant articles were reviewed in detail, while detailed tabulated results are in the Supplementary Materials (Table 1, Table 2, Table 3 , Table 4, Tables S1 and S2). Note that there is a large body of evidence on cyanotoxins in aquatic systems. Therefore, the data in Table 1, Table 2, Table 3 , Table 4, Tables S1 and S2 are not exhaustive. Instead, the data present an overview of the evidence drawn from several continents.

3. Cyanotoxins in DWSs

3.1. Diversity of Cyanotoxins

Cyanotoxins can be categorized into five general groups: (1) neurotoxins (e.g., alkaloids anatoxin-a and saxitoxins, and non-proteinogenic amino acid β-N-methylamino-L-alanine), (2) dermatotoxins (e.g., lyngbyatoxin-A, aplysiatoxin), (3) cytotoxins (e.g., polyketide alkaloid cylindrospermopsin, CYN), (4) hepatotoxins (e.g., cyclic peptides, microcystins (MCs), and nodularins), and (5) endotoxins (lipopolysaccharides) [3,97,98]. Toxin production by cyanobacterial species is geographically diversified, and certain species can produce different compounds based on their climate zone. However, particular strains from the same region can also vary in this respect [99]. For example, Raphidiopsis raciborskii is a producer of CYN in Australia, but its strains in South America are producing saxitoxins instead, while in Europe, no known cyanotoxin-producing strains of this species have ever been identified [100]. The factors controlling this behavior may warrant further research. Additionally, different cyanobacterial species produce different cyanotoxins, leading to a wide range of structural variants even within a single toxin class. Several studies have revealed the existence of more than 90 different MC structures [101,102,103], while other studies have reported over 240 variants [104] and even higher (e.g., 250 variants in [105] and 279 in [106]). Anatoxins also have been reported to have different structures (Figure 2) [107,108,109]. There are over 25 chemical variants of saxitoxin [110], while CYN has a few analogs that differ in functional groups [111].
One should note that only a fraction of the toxic cyanobacterial metabolites are likely known. The CyanoMetDB, a comprehensive public database of secondary metabolites from Cyanobacteria, comprises over 2000 chemical compounds, most of which have not been studied concerning their toxic properties and mechanisms of action [112,113]. Various in vitro and in vivo studies demonstrate that cyanobacterial strains incapable of producing any known cyanotoxin still reveal significant toxicities in experimental models [43,114,115]. Therefore, cyanobacterial species shall not be unequivocally considered non-toxic based only on the lack of detection of known cyanotoxins or genes responsible for their synthesis. It also underlines the need to continue research devoted to cyanobacterial toxicities, including identifying toxic species/strains, the molecular basis of toxin synthesis and the environmental conditions influencing it, and understanding mechanisms of action and risks arising from exposure to environmentally relevant concentrations.

3.2. Global Occurrence of Cyanotoxins in Drinking Water Systems

Globally, cyanotoxins have been detected in DWSs in Africa, Asia, Europe, Oceania/Australia, North America, Latin/South America, and even Antarctica [116,117,118,119]. Detailed data on the occurrence of cyanotoxins in Africa and other regions outside of Africa are presented in Tables S1 and S2 in the Supplementary Materials, respectively. However, several countries in Africa and elsewhere still lack data. Specifically, while extensive research and regulatory frameworks are in place in many industrialized countries, the occurrence and contamination of DWSs by Cyanobacteria and cyanotoxins in regions other than Africa are underreported and poorly understood [120]. This subsection aims to provide an overview of the global occurrence of cyanotoxins in DWSs.

3.2.1. Africa

Several studies have reported the presence of cyanotoxins and the associated cyanobacterial species in the water bodies that provide drinking water in the region (Tables S1 and S2 in the Supplementary Materials). The most frequent cyanotoxins detected are MCs. Of the 27 papers retrieved, all mentioned the presence of MCs in DWSs throughout Africa (Table S1). The presence of other cyanotoxins, such as anatoxins, CYN, and nodularin in DWSs has only been reported in a few studies conducted in only five countries (Botswana, Kenya, Tanzania, South Africa, and Zimbabwe) [121,122,123]. For instance, the neurotoxic anatoxin-a was found in Kenya’s Lakes Sonachi and Simbi at concentrations as high as 2.0 μg/g DW [121], and nodularin and CYN (0.004 to 0.01 µg/L) in Lake Victoria (Tanzania) [123].
According to studies [124,125,126], surface water in rivers, lakes, and reservoirs often contain higher concentrations of cyanotoxins, often beyond the drinking water recommendation guideline limit of 1 µg/L set by the World Health Organization (WHO), than other stages of the continuum. For example, six MC variants (MC-LA, MC-LR, MC-dmLR, MC-YR, MC-dmRR, and MC-RR) with concentrations ranging between 61.6 and 453.9 μg/L were detected in Ethiopia in the Legedadi Reservoir [12]. In a related study conducted in Zimbabwe, MCs with concentrations ranging from 18.0 to 22.5 µg/L were found in Lake Chivero [10].
However, many studies have shown a decrease in cyanotoxin levels from the source water to the point of use and consumption [11,125]. According to [127], most cyanotoxins are cell-bound and are easily eliminated by some water treatment techniques. For instance, in a Tanzanian study, no cyanotoxins were found in the piped water that had been treated. The water treatment techniques managed to eliminate the anatoxin-a, nodularin, and CYN found in the Lake Victoria source water [123]. Similarly, water treatment significantly decreased cyanobacterial cell counts and MC concentrations occurring at high levels in lakeshore water in Uganda [125]. However, there is little evidence that cyanotoxins can be eliminated or reduced to WHO-acceptable levels, especially in low-income settings in Africa. This is because this aspect has not been extensively investigated in low-income settings in Africa, where low-cost drinking water treatment processes such as chlorination, boiling, sand filtration, and solar disinfection are commonly used.
Several studies have detected cyanotoxins, in particular MCs, in various stages of the drinking water continuum at concentrations exceeding the WHO maximum guideline for human lifetime intake via drinking water [12,18,128]. According to [127], some water treatment methods, such as pre-chlorination and the use of algicides, cause the Cyanobacteria to lyse and release their toxins into the water. As a result, cyanotoxins may be present even in treated water. For instance, in a study carried out in Egypt, MCs (1–4 μg/L) were detected in the treated drinking water from the outflow tank [129]. Another study found MCs (4.3 μg/L) in South African treated tap water [130]. More importantly, it may be inferred that the conventional water treatment methods employed in some African countries are ineffective at removing or reducing the toxins to some acceptable levels, necessitating routine testing of treated water to give consumers access to safe drinking water.
Furthermore, it has also been observed that cyanotoxins can contaminate groundwater (Table S1). Some studies have revealed evidence that groundwater can become contaminated with MCs as a result of surface water and groundwater interaction [126,131,132]. This is a serious concern since many African communities rely on untreated groundwater for drinking, particularly from boreholes. Moreover, in many developing countries, residents often store treated drinking water in tanks or reservoirs, where cyanotoxins, particularly MCs, have been detected at concentrations exceeding the WHO guideline limit of 1 μg/L [62]. In South Africa, average values of 4.3 and 4.8 μg/L of MCs were found in communal tap water and treated water kept in tanks, respectively [130]. These results support regular toxicity monitoring of treated water stored in tanks, containers, and reservoirs.

3.2.2. Asia

Detecting toxic cyanotoxins and Cyanobacteria in drinking water sources is an emerging phenomenon in the region (Table S2 in the Supplementary Materials). These problems have been noted mostly in China but also in countries such as India, Japan, and Turkey [133]. China has had major problems with cyanobacterial blooms and cyanotoxin contamination in its drinking water sources. Cyanobacterial blooms and cyanotoxins have been found in water sources across India, including Dal Lake in Srinagar [134] and Ambazari Lake and Phutala Lake in Nagpur [135], and thus they are no longer used as sources of drinking water. The screening of water reservoirs in Pakistan also revealed the presence of several MC congeners, with MC-LR and MC-RR being most prevalent, as well as the presence of CYN [136]. The toxicity of bloom-forming Cyanobacteria (Raphidiopsis mediterranea) discovered in Japan (Biwa Lake) has been experimentally proven [137,138]. In Turkey, several cyanotoxins (MC-LF, MC-LW, MC-LA, MC-RR, and MC-LR) were found in Kovada Lake, the concentration of which in the water reached 98.9 µg/L [139]. Cyanobacteria Raphidiopsis raciborskii and Microcystis spp. were recorded in the Myanmar Dam in 2020, reportedly producing cyanotoxins such as CYN and MCs [140]. Other studies have detected cyanotoxins in DWSs in Singapore, Vietnam, Thailand, Bangladesh, and Korea [141,142,143,144,145], Mongolia [146,147], as well as Iran, Saudi Arabia, and Qatar in the Middle East [148,149].

3.2.3. Oceania/Australia

Cyanobacterial blooms and cyanotoxins in DWSs were a significant concern in Australia, especially from 1997 to 2003, when cyanotoxins were detected in many shallow lakes and reservoirs across the country. Concentrations of up to 18.9 µgL were recorded (Table S2). After this period, a serious incident was reported with the appearance of both saxitoxins and CYN in a large area (Murray River and its tributaries), which were used as sources of drinking water [150]. The serious threat to drinking water sources in Australia was the appearance of Raphidiopsis raciborskii in water bodies, releasing CYN into the water. In 1979, a major outbreak of gastroenteritis occurred in Palm Island, Australia, leading to the hospitalization of 10 adults and 140 children presenting symptoms of poisoning accompanied by elevated serum levels of liver enzymes, ketonuria, glycosuria, and hematuria. This case was linked to a massive Raphidiopsis raciborskii bloom in Solomon Dam, a drinking water reservoir, and the release of CYN toxin to the water column [151]. New Zealand has been affected by several incidents of cyanotoxins produced by Raphidiopsis raciborskii in the past [152,153,154,155], though the exact concentrations of this cyanotoxin are not known.

3.2.4. Europe

Studies have detected cyanotoxins and cyanobacterial blooms in water sources across Europe. Cyanotoxins, such as anatoxins, saxitoxins, nodularins, MCs, and CYN, have been detected in lakes, reservoirs, and rivers used as drinking water sources (Table S2). In southern Europe (Serbia and Italy), the highest cyanotoxin (MC) concentrations of 650 µg/L were recorded in the Ćelije Reservoir and 75.5 µg/L in Alto Flumendosa Lake, while in Spain, the highest MC concentration was 64.8 µg/L in the Cogotas Reservoir [156,157,158]. Central Europe (mainly Poland and the Czech Republic) was most affected by cyanobacterial blooms and cyanotoxins (MCs). Algal blooms appeared for many years in several reservoirs (Siemianówka, Jeziorsko, and Sulejów dams), and cyanotoxin concentrations up to 173.8 µg/L were recorded in DWSs [159,160,161,162]. The problem of the intensity of inland water cyanobacterial blooms in Central Europe and its threat to DWSs was well described in Poland [163], where out of 238 water bodies analyzed, 204 were dominated by Cyanobacteria, and 74 water bodies were contaminated with cyanotoxins. However, only four of these water bodies were a source of drinking water [163,164]. There is little evidence of cyanotoxins occurring in DWSs in northern Europe. In Immeln Lake in Sweden, the highest concentration of cyanotoxins (i.e., MC) detected in drinking water was 815.0 µg/L [165,166]. There are also reports on the occurrence of cyanotoxins in Russian reservoirs [167,168,169] as well as toxic Cyanobacteria in Ukraine, though in the latter case, the information on particular toxins is limited to microcystins [167,170,171].

3.2.5. South/Latin America

Besides Africa, South America appears to be the most affected continent in terms of the appearance of cyanotoxins in DWSs (Table S2). This is particularly evident in Brazil, where the greatest cyanotoxin (i.e., MC) concentrations in the water were as high as 303,053.9 µg/L in the Carpina Reservoir (Brazil) [172]. Recently, a report was published on the occurrence of MCs produced by Limnothrix planctonica in a source of drinking water in the Brazilian part of the Amazon River [173]. Although cyanotoxins appear infrequent on the continent, in Argentina, many drinking water sources have been reported to have been contaminated with MCs, nodularin, and anatoxin- [174,175]. In 1996, a fatal accident occurred in the Caruaru (Brazil) dialysis center. Lack of reverse osmosis in the filtration system resulted in the presence of MCs and CYN in the water supply, leading to the death of 76 exposed patients [38]. Cyanotoxins have also been detected in DWSs in several other countries in South America, including Colombia [176,177], Ecuador [178], Chile, Costa Rica, Dominican Republic, Mexico, El Salvador, Guatemala, Nicaragua, Panama, Uruguay, and Venezuela [179]. Some reviews also exist, focusing on the occurrence of cyanotoxins in aquatic systems in Latin America [180,181,182,183].

3.2.6. North America and Antarctica

Cyanobacterial blooms and cyanotoxins in water bodies and DWSs are also a significant concern in North America (Table S2). Studies have reported potentially toxic cyanotoxins and Cyanobacteria in both treated and untreated water supplies used for household consumption [184,185]. Several reports on cyanotoxins in DWSs exist in the U.S., including those in state agency reports. One of the better-documented examples of cyanotoxins affecting drinking water is Lake Erie in the Great Lakes basin (USA), which provides drinking water for about eleven million inhabitants [186]. Since 2002, there has been a severe problem with the occurrence of MCs in water in this lake, and in 2014 an intense cyanobacterial bloom manifested [187]. In Mexico, however, anatoxin-a and CYN were detected in addition to MCs, with concentrations in the Los Berros water treatment plant reaching 129 and 7 µg/L, respectively [188]. In the United States, NOD was recorded alongside MCs, whereas in Canada, only MCs were recorded [187]. Cyanotoxins have also been detected in cyanobacterial mats in meltwater ponds in Antarctica [116,117,118,119]. This indicates the widespread occurrence of cyanotoxins and the ability of Cyanobacteria to survive under extreme temperature conditions.

3.3. Factors Controlling Cyanotoxins in DWSs

3.3.1. Physical Factors Controlling Cyanotoxins in DWSs

Light and Temperature

Several studies have discovered that when light intensity is high, the Cyanobacteria produce more cyanotoxins, especially MCs, to protect themselves against reactive oxygen species [189,190,191]. It can be inferred that at low light intensity, the production of cyanotoxins is mainly supported by enhanced photosynthetic light reactions. Conversely, at high light intensity, algae are prompted to produce MCs as a response to photooxidative stress [192]. Moreover, various articles indicate that under increased temperature, the synthesis and release of cyanotoxins may be elevated [193,194,195]. However, there are also studies demonstrating that under decreased temperature, the MC quota per cell in M. aeruginosa can be increased [196]. In the temperate zone, the occurrence of Cyanobacteria associated with MC production was also noted during the winter season, indicating that winter blooms can be “cold and toxic” [197,198].

3.3.2. Chemical Factors Controlling Cyanotoxins in DWSs

Nutrients

In addition to being essential for cyanobacterial metabolism, nitrogen and phosphorus also directly influence the production of cyanotoxins, and depending on the toxin, both starvation and overloading can cause increased levels of these metabolites [199,200]. More importantly, the synthesis of cyanotoxins is impacted differently by various types of nitrogen [201]. N and P enrichment of aquatic systems leads to eutrophication, a global environmental concern [202,203]. In turn, eutrophication increases the amount of nutrients accessible to Cyanobacteria, which might increase cyanotoxin production. In one study by [203], cyanotoxins were found in several lakes, with the highest overall concentration detected in the most eutrophic lakes in central Canadian ecozones. Their concentrations increased as cyanobacterial biomass increased, as did total phosphorus, dissolved inorganic carbon, and water temperature. However, it should be noted that oligotrophic water bodies have recently also become exposed to the risk of toxic blooms, for example, such as Lake Baikal, Lake Superior, or Alpine lakes [204,205,206]. Identifying the environmental factors and taxa that increase the occurrence of cyanotoxins is critical for the effective treatment of cyanotoxins in DWSs.
Several trace elements are also essential for algal growth, especially iron, copper, zinc, and sulfate. Studies have demonstrated that these elements are linked to the generation of cyanotoxins, though only at a specific concentration [199,201]. Pharmaceuticals and personal care products, endocrine-disrupting substances, pesticides, polycyclic aromatic hydrocarbons, and antibiotics are some of the more recent pollutants thought to control cyanotoxins’ emergence [207,208]. In addition to being a necessary micronutrient for Cyanobacteria to grow and reproduce, nitrogen is one of the main ingredients in cyanotoxins, such as MCs. For example, algal growth and the synthesis of cyanotoxins are compromised by extremely low or high nitrogen content. High phosphorus concentrations might indirectly alter the generation of cyanotoxins by affecting the biomass of Cyanobacteria [199]. However, nitrogen and phosphorus limitations can lead to an increase in the generation of cyanotoxins [201].

3.3.3. Effects of Microbes on Cyanotoxins in DWSs

Cyanotoxins, such as MCs, are very stable compounds that defy physical breakdown at ambient pH and temperature [209,210]. However, certain microorganisms have been observed to break down MCs in water. These cyanotoxin-degrading bacteria are widely dispersed, and the degradation of cytotoxins by naturally existing bacteria is a viable option for toxin elimination [209,210]. As reported in several studies, the majority of cyanotoxin-biodegradation studies have been conducted on MCs, and several bacteria, such as probiotics and rumen bacteria, have been used to degrade MCs in DWSs at treatment plants [209,210,211]. These bacteria include Arthrobacter spp., Brevibacterium spp., Burkholderia spp., Methylobacillus spp., Morganella morganii, Paucibacter toxinivorans, Poterioochromonas spp., Pseudomonas aeruginosa, Ralstonia solanacearum, Rhodococcus spp., Sphingomonas spp., Sphingosinicella microcystinivoransSphingopyxis spp., and Stenotrophomonas spp. and have been reported to degrade MCs [209,210,211]. Despite claims of anatoxin-a biodegradation, only Pseudomonas has been demonstrated to digest anatoxin-a. In a study conducted by [211], it was discovered that Sphingopyxis, isolated from biological sand filters, played a crucial role in degrading MCs during water treatment processes. According to [212], 15 distinct fungal species exhibit significant lytic activity against Cyanobacteria, with 6 of these species capable of degrading MCs. Notably, selected fungal species, such as Aurobasidium pullulans and Trichoderma citrinoviride, have demonstrated the ability to specifically curtail the growth of Cyanobacteria while sparing beneficial algal species. Moreover, specific fungal strains like Trichaptum abietinum and Trichoderma citrinoviride have displayed a dual functionality, encompassing the lysis of Cyanobacteria and the breakdown of MCs produced from decaying cells [212]. Biological sand filters have been effectively utilized to purify drinking water by removing Cyanobacteria, cyanotoxins, and other microbial contaminants from the water supply.

3.3.4. Effect of Seasonality and Hydrodynamics on Cyanotoxins in DWSs

Cyanotoxin variation during different seasons and variances in their quantities might be linked to a range of environmental conditions and cyanobacterial biomass [213]. As a result, monitoring during peak seasons is critical for preventing human health hazards. Research conducted in tropical Africa by [214], spanning the hot–dry, cool–dry, and hot–wet seasons, showed that Cyanobacteria relative abundances were highest during the cool–dry season. This phenomenon was likely attributed to a reduction in river inflows and an increase in reservoir mixing that tends to occur during the cool–dry season. Furthermore, the cyanobacterial community’s structure was significantly influenced by various factors, including macrophyte cover, dissolved oxygen concentrations, water quality, reactive phosphorus, water depth, and chemical oxygen demand [214]. The study by [214] emphasizes that potentially dangerous and toxic algae species may proliferate under these climate change projections.
A recent study [215] revealed that the Mexican dam had the highest cyanobacterial biomass compared to Chlorophyta and Bacillariophyceae throughout the year. The Cyanobacteria production was linked to the prevalence of cyanotoxin-producing species as well as environmental factors such as rainfall, total dissolved solids, water temperature, and certain trace metals. Similarly, [216] revealed that fluctuations in dissolved MCs and MCs bound to algal cells follow seasonal patterns, exhibiting a distribution with a single peak during specific seasons. Notably, the peak in dissolved MCs in water was observed to occur approximately one month after the peak of algal cell-bound MCs. However, according to [217], Cyanobacteria and cyanotoxins have a bimodal seasonal distribution, with seasonal fluctuation patterns differing across European climatic zones. This suggests that Cyanobacteria and cyanotoxins can follow the same trend in temperate and tropical regions. However, more studies using the same harmonized sampling techniques, sampling times, and methods for analysis are needed to understand which regions are prone to cyanotoxins.
According to [218], thermal stratification and a significant fluctuation in dissolved oxygen levels between the top and bottom were observed in October 2017 in the Itupararanga Reservoir (Brazil), and a decrease in cyanobacterial biomass was seen at the temperature-stratified sample locations throughout the same period. Visser et al. [219] state that temperature stratification favors floating Cyanobacteria due to reduced mixing and increased light availability. Although cyanobacterial biomass decreased in areas where thermal stratification occurred, [220] discovered that some species with aerotopes were abundant and that changes were also linked to light availability due to decreased water transparency, which might have a major effect on controlling dominance.

3.3.5. Effect of Climate Change on Cyanotoxins in DWSs

Due to climate change, it is anticipated that Cyanobacteria will experience an increase in both their frequency and distribution, as this is likely to create favorable environments for their growth [221]. Furthermore, climate change’s impact might influence certain surface waters’ capacity to initiate phototransformation processes [221]. By fostering the perfect environment for Cyanobacteria to develop, climate change contributes to excessive cyanobacterial blooms and cyanotoxin production [222]. As reviewed by [223], human pollution and climate change cause Cyanobacteria to thrive, survive, and expand, potentially resulting in increased production of cyanotoxins. Warm waters are ideal for cyanobacterial growth; hence, as the planet’s temperatures rise, so do planetary water temperatures [222,223]. This is mainly because the topmost layer of the water stays considerably warmer than the rest of water column, which makes it more challenging for the water to mix, allowing Cyanobacteria to develop more efficiently [223]. Climate change-induced warming is increasing the thermal stratification of water columns. This confluence of variables (higher temperatures and stratification) supports cyanobacterial dominance, leading to cyanotoxin production [222,223]. This has the additional drawback of allowing this photosynthetic organism to readily take in sunlight and develop even more quickly, resulting in a thick layer on the water surface. Cyanobacteria can grow more readily when atmospheric carbon dioxide concentrations rise. The optimal environment for cyanobacterial growth, blooms, and cyanotoxin production is further created by increased water temperatures and carbon dioxide absorption [223]. Continental-wide analysis revealed that temperature is a major factor explaining the occurrence of cyanotoxins in Europe [224]. The frequency of cyanobacterial blooms during periods during which these events are usually not expected, e.g., in winter in the temperate zone, is also increasing [225].
Climate changes also impact precipitation rates and patterns. More storms may develop due to increased evaporation, although certain land regions will also be threatened by droughts [226]. Cyanobacterial blooms increase when more frequent nutrient contamination results from greater rainfall and storms [226]. Therefore, some actions that are accountable for the anthropogenic input of nutrients include fertilizing arable land, discharging sewage, using detergents extensively, and disposing of industrial effluents. Free et al. [227] discovered that the key characteristics determining cyanobacterial blooms in a shallow mesotrophic Mediterranean lake were seasonality, interannual fluctuation, lake level, water temperature, ocean oscillations, and concomitant rainfall. In addition, the same authors revealed that more extended periods of warmer temperatures can result in shorter-lasting blooms and subsequent production of cyanotoxins [227].
Vione and Rosario-Ortiz [221] conducted a study involving photochemical modeling in three distinct freshwater scenarios: (1) lakes undergoing summer stratification, (2) water exhibiting browning, and (3) water concentration due to evaporation. This investigation focused on two cyanotoxins, MC-LR and CYN, both characterized by distinct photoreaction kinetics and phototransformation pathways. Their results showed that the photodegradation of those cyanotoxins, which is significantly affected by reactivity with the triplet states of chromophoric dissolved organic matter, is quantitatively projected to be aided by anticipated changes in water-column conditions under changing environmental circumstances. This phenomenon, observed in the case of MC-LR, suggests that increased occurrence could be counteracted, to some extent, by accelerated photodegradation in specific environments [221]. However, the role of chromophoric dissolved organic matter in the degradation of other cyanotoxins remains poorly understood, indicating a knowledge gap in the intricate link between cyanotoxin photodegradation and changing climatic conditions [221].
According to a review conducted by [228], while climate change is predicted to enhance cyanobacterial blooms in some waterbodies, this is not the case in those with low concentrations of phosphorus or nitrogen. Such conditions meticulously restrict biomass, and even more so in shallower lakes and rivers than in stratified bodies of water. Limiting the release of nitrogen and phosphorus pollutants into water bodies is critical. In summary, instead of generalizations, management requires case-by-case evaluations focused on the implications of current and anticipated climatic changes on the specific waterbody, and these assessments should be carried out routinely and often as climate changes over time [228]. Moreover, it is crucial to understand and consider the fundamental factors and mechanisms driving the proliferation of Cyanobacteria and the production of cyanotoxins. This understanding is essential for formulating immediate and sustained strategies to mitigate global cyanotoxins’ increasing prevalence, severity, and health risks. As a result, it is prudent to develop an integrated nutrient management strategy that is consistent with different physical, biological, chemical, and integrated mitigation approaches, such as modifying water flow and flushing, dredging, applying chemicals, and integrating selective grazers.

3.4. Cyanotoxin Detection Methods

3.4.1. Conventional Methods

Cyanotoxins have been detected using a range of conventional methods (Table 1) and are based on biological, biochemical, and physicochemical characteristics of cyanotoxins [34,229,230]. However, the criteria for selecting the method rely on sensitivity and specificity [230]. Initially, cyanotoxins were detected using methods involving extracts, cell cultures, and cyanobacterial cells injected in animal models [34]. However, such methods have since been discredited due to (1) high cost, (2) ethics surrounding animal models, (3) time-consuming procedures, and (4) no specificity and low sensitivity [133]. Therefore, more direct methods, including immunological assays, whole-organism bioassays, and biochemical assays, were subsequently designed [35]. However, whole-organism bioassays lacked sensitivity and specificity and could not be applied in rapid, regular water monitoring [34]. A good example of this is the mouse bioassay, which was qualitative and only revealed overall toxicity and did not reveal the identity or variant of the toxin. Other direct methods, like the Brine shrimp assay, protein phosphatase inhibition colorimetric, fluorometric, and radiometric detection assays, had shortcomings with sample matrix interferences and were less effective at detecting cyanotoxins in water samples [132].
Analytical methods that later became popular are based on the physicochemical aspects of the toxins: functional groups, ultraviolet chromophores, and reactivities. Such methods include gas chromatography (GC), liquid chromatography (LC), mass spectrometry (MS), nuclear magnetic resonance (NMR), capillary electrophoresis, Matrix-Assisted Laser Desorption Time-of-Flight Mass Spectrometry (MALDI-TOF-MS), and high-performance liquid chromatography (HPLC) [32]. HPLC-based methods, such as HPLC-MS and HPLC with the photodiode array, have been recommended for being highly sensitive and reliable, but like most analytical methods, they suffer from a lack of available standards and long derivation times; thus, they are time-consuming [32,35]. GC and LC methods are inexpensive and sensitive; however, equipment setup renders them less popular [35,133]. The majority of analytical methods may be suitable for MC detection in DWSs but are difficult to work with due to the co-occurrence of various cyanobacterial species within algal blooms as well as the production of many cyanotoxin varieties, which result in complex compositions [29]. To cater to these shortfalls, methods have been coupled to carry out a single analysis to quantify and identify MCs, such as LC/MS/MS.
Gas chromatographic methods are similarly significantly sensitive and precise but suffer from interference by compounds such as metal salts and organic and inorganic compounds. These interactions with sample components result in downstream cleanup processes such as solid-phase extractions becoming laborious and time-consuming [132]. In addition, chromatographic methods also demand highly skilled analysts, cumbersome, time-consuming procedures, rare standards, and, most importantly, expensive equipment and operations [34]. High-performance capillary electrophoresis efficiently separates and quantifies variants of cyanotoxins based on charge and molecular mass. Variations of capillary electrophoresis (such as those coupled with pressurized ultraviolet) have successfully detected MCs (low detection limit of approximately 0.92 to 2.3 µg/L), particularly in aquatic environments [31,34,231]. Attempts at lowering detection levels to trace concentrations by capillary electrophoresis have succeeded by incorporating immunoaffinity chromatography, field-amplified sample stacking, and dynamic pH junction. Despite the efforts, there is still a need to improve the detection limit with this method so that it can be well robust for routine water monitoring of cyanotoxins [232,233].
Recently, routine analysis in water has been prevalently performed using the cheap, easy-to-perform, qualitative, and semi-quantitative Enzyme-Linked Immunosorbent Assay (ELISA) for the detection of MCs/NOD, CYN, ATX, and STXs with limits as low as 4 ng/L [232]. The enzymatic immunoassay uses commercial kits to screen ground and surface water, detecting toxins produced by active and inactive Cyanobacteria genotypes. However, the method is delimited by a lack of uniformity of cross-reactivity, which makes it semi-quantitative. This is because measured concentrations of cyanotoxins depend on various variables that may not be recognized during analysis. Assay kits targeting an MC moiety, 3S-amino-9S-methoxy-2S,6,8S-trimethyl-10-phenyldeca-4E,6E-dienoic acid (known as ADDA), have been reported to generate false positive results in detection [234]. Other assays based on antibodies and protein phosphatases are available but are less popular because they fail to distinguish MC variants [28,231]. In order to overcome the limitations of conventional methods, very sensitive, selective, quick (responding in a couple of seconds), and fouling-resistant systems have since been developed.

3.4.2. Advanced/Emerging Methods

Analytical methods that are robust and have now been designed to use micro-sensing devices, resulting in lower detection limits, improved target selectivity and specificity, and other benefits (Table 1). Biosensors are the latest technology that relies on the presence of (1) a bioreceptor in direct contact with a transducer and/or (2) a biological recognition element. These can be natural or artificial biological elements, such as antibodies, enzymes, nucleic acids, whole cells, and molecularly imprinted polymers [235]. Biosensors based on enzymes either rely on optical or electrochemical detection techniques. Those that employ antibodies are immunosensors, which can be detected via piezoelectric, NMR-based optical, and electrochemical sensors. Examples include immunoarray biosensors, luminescent immunosensors, Evanescent Wave fiber-optic immunosensors, surface plasmon resonance (SPR) immunosensors, and fluorescent immunosensors [36]. Those based on nucleic acids are electrochemical DNA and SPR-DNA biosensors.
Electrochemical biosensors achieve remarkably low detection limits due to the selective binding of a biological recognition element to the target analyte [32]. Two classes of electrochemical biosensors exist: (1) biocatalytic devices and (2) affinity sensors, which incorporate novel transducer and interface designs to yield remarkable results. These include metal and carbon-based composites that enhance electrode response [232]. Biocatalytic devices utilize enzyme target reactions to generate electroactive species, while affinity sensors, in contrast, oversee the interaction between the bioreceptor and the target to yield a quantifiable signal [232]. These sensors can employ labels for enhanced detection, categorized as ‘labelled’ or ‘label-free’ detection. Types of affinity sensors encompass aptasensors, DNA sensors, and immunosensors [29]. Biocatalytic devices utilize enzyme target reactions to generate electroactive species. In contrast, affinity sensors rely on the interaction between the bioreceptor and the target to yield a quantifiable signal [232]. These sensors can employ labels for enhanced detection, and thus can be categorized as ‘labelled’ or ‘label-free’ detection. Types of affinity sensors include aptasensors, DNA sensors, and immunosensors [29]. More recent technologies couple nanotechnology and biochemistry to increase signal transduction for detections as low as femtomolar concentrations.
Electrochemical biosensors are also categorized according to the mode of the electrical signal applied to the electrochemical cell. The categories include (1) potentiostatic (measures the current after controlled potential is applied to the electrochemical cell), (2) potentiometric (cell potential is measured under near-zero current), (3) impedance (potentially applied to the cell and the current response is measured to get complex resistance), and (4) galvanostatic (cell potential is measured following current application) [232]. In the past five years, impedance spectroscopy has gained popularity as it analyzes without the use of labels to create a detectable signal. The method is mostly preferred as it allows the study and interpretation of surface reactions and interface properties with simplicity during analysis due to its capacitive and resistive components. Examples of potentiostatic sensors are amperometry and voltammetry, and they have a dynamic range with low limits of detection. Voltammetric methods that have been used in detection are cyclic voltammetry, normal pulse voltammetry, differential pulse voltammetry, and square wave voltammetry [36].
One example of an electrochemical biosensor innovation with traceable efficiency is calf thymus DNA (ct-DNA), which was physically immobilized on a gold electrode following the discovery that MC-LR has a deleterious effect on ct-DNA. The method exhibited recoveries of around 107.6% while having detection levels as low as 1.4 ng/L in natural water samples [236]. Another novel method garnering popularity in automatic detection, particularly for onsite work, is the fiber-optical chemiluminescent biosensor system. The method utilizes a fiber optic bio-probe as both the biorecognition element, transducer, and Si-based photodiode detector, and its sensitivity achieves a low detection limit of 0.03 µg/L [237]. The method that uses a Surface-enhanced Raman Scattering (SERS) spectroscopic immunosensor that exhibits remarkable selectivity, sensitivity, and robustness is tailor-made for aquatic systems but operational with cultures of Microcystis aeruginosa. The SERS sensor attained a LOD of 0.014 µg/L [32,238].
The phosphorescent immunosensor utilizes antibodies and antigens as recognition units and Mn-ZnS RTP as sensing materials to bind specifically with MC-LR, rapidly detecting MC-LR at a low detection limit of 0.02 µg/L [32]. With this method, there is no significant obstruction from coexisting MC-LR pollutants in the water sample. In the detection of MCs in fresh crucian carp tissue and water, a novel method that uses Cu/Au/Pt trimetallic nanoparticles encapsulated in DNA hydrogel was prepared for colorimetric detection [32].
Aptasensors, an example of affinity biosensors that employ aptamers’ recognition element with high surface area and disposability, are now widespread. Nanoparticle technology has enabled the development of an aptasensor based on SERS. MC-LR aptamer and its corresponding complementary DNA fragments were conjugated to gold (signals) and magnetic (capture probes) nanoparticles. The method achieved a low detection limit of 0.002 ng/L. A low detection limit of 0.002 nM was achieved by employing a method that uses a dual signal amplification system with a ternary composite of AuNPs deposited on molybdenum disulfide (MoS2)-covered TiO2 nanobeads and horseradish peroxidase [133].
Another method utilizes a novel dual-mode aptasensor with MoS2-PtPd bimetallic nanoparticles and a zeolitic imidazolate framework (ZIF)-8-thionine (Thi)-Au (ZIF-8-Thi-Au) as signal material. A method based on non-radioactive energy transfer from an excited donor to an acceptor fluorophore, i.e., fluorescence resonance energy transfer (FRET), was utilized. The FRET-based method aptasensor positively correlated with the conventional ELISA method [133]. These methods are limited by temperature, pH, and ion concentration. A new sensor usable in real-world water systems was developed that uses a nanoprobe-based quantum-dot hapten sensing MC-LR at less than 0.03 µg/L. Interestingly, advanced fluorescent sensors have been developed that detect within 5 min and discriminate an array of cyanotoxins using smartphones. The technique integrates aptamers and single-stranded DNA dyes and has a low detection limit of less than 3 nM [232]. Aptasensor-based methods exhibit excellent recovery rates, indicating good stability against other components in water samples.
Quantitative real-time polymerase chain reaction (PCR) and DNA Microarray have improved sensitivity. They are based on the detection of the expression of toxin-encoding genes. Multiplex PCR is a recent alternative that amplifies and analyses many different genes in a single run [28]. This method is mostly suited for local authorities monitoring water bodies. Conventional PCR is only a qualitative (presence/absence) technique, while qPCR quantifies the reaction product in terms of gene copy numbers (with detection limits of 8.8 cells/mL of Microcystis spp.) [35]. The major limitation of qPCR is the reduction in amplification efficiency and the increasing length of PCR product. The presence of an amplified toxin-encoding gene does not necessarily indicate transcription, translation, or actual toxin expression. Therefore, it is essential to complement qPCR data with biochemical validation techniques. The microarray technique is a more recent high-throughput molecular method that screens for gene expression on a genomic scale. The technique is quick and efficient as it uses a chip loaded with DNA probes. Different adoptions of the technique enable gene expression studies using RNA and complementary DNA, and genetic variations using single-nucleotide polymorphisms or mutations. The major advantage of the microarray technique is the simultaneous analysis of the expression of many genes [34]. In one case, a microarray was used to simultaneously detect MC (mycE) and nodularin synthase genes (ndaF) expressed by hepatotoxin-producing Cyanobacteria. Similar studies were conducted using a microarray hybridized with a magnetic-capture hybridization technique that utilized bacterial magnetic particles. The technique is advantageous as it can be used as a monitoring strategy for cyanobacterial blooms in water systems; however, it is a costly method [35]. However, advanced analytical methods are often expensive, require high levels of skills to operate, and are often not readily available in low-income countries.
Potentially new toxins can be characterized while environmental monitoring can be carried out in DWSs by coupling existing emergent techniques with transcriptomic, proteomic, and metabolomic analyses. However, unlike indicator bacteria like E. coli and fecal coliforms, cyanotoxins are rarely included in routine drinking water quality monitoring, particularly in low-income nations. Furthermore, communities in distant and low-income areas frequently lack access to laboratory facilities for cyanotoxin testing. Therefore, there is a need to develop and validate low-cost analytical and detection tools to determine cyanotoxins in DWSs, including those based on (1) biosensors and (2) local knowledge systems (i.e., ‘ethno-toxicology, ethno-medicine’). In addition, further work is required to develop and apply maximum drinking water guidelines for total and individual cyanotoxins, especially in low-income countries where they are currently lacking.

4. Human Health Risks

4.1. Exposure Pathways

Human exposure to cyanotoxins via DWSs is a cause for concern. Accordingly, several studies [98,239,240] have revealed human exposure to cyanotoxins through various pathways. Notably, ingestion (i.e., contaminated drinking water or food that has been prepared with it) and dermal exposure during routine activities such as showering, bathing, or swimming are all examples of exposure pathways (Figure 3) [241]. The global occurrence of cyanotoxins in various DWSs (Tables S1 and S2) makes human exposure to cyanotoxins difficult to avoid. For example, groundwater is typically extracted straight from the ground via a borehole to a standing tap and used for various activities without prior treatment [242]. The currently recognized exposure routes are skin contact (dermal route), inhalation, hemodialysis, and ingestion with water or food.

4.1.1. Ingestion

Chronic and accidental ingestion of cyanotoxin-contaminated drinking water is the dominant route of human exposure to cyanotoxins. Direct ingestion is a frequent route of cyanotoxin intake, as evidenced by their occurrence even in treated drinking water. Several epidemiological studies conducted in various countries linked some diseases such as liver cancer (Serbia) [243], diarrhea, headache, vomiting, fever, muscular and abdominal pain (Sweden) [244], gastroenteritis (Serbia, Sweden, and Brazil) [38,244,245], and gastroenteritis-like ailments (Australia) [151,246] to consumption of contaminated water. Furthermore, exposure to cyanotoxins is also via ingestion of food prepared or cooked by contaminated water [247].

4.1.2. Skin Contact (Dermal Route)

The skin has been identified as another route for cyanotoxin infection in humans, involving DWSs during bathing and recreational activities such as swimming. Recently, the ability of cyanotoxins, particularly those with small molecular sizes, to penetrate the skin has been demonstrated [248]. For instance, anatoxin-a, which has a log p value of 0.8 and a low molecular weight, may enter the body through passive diffusion through healthy skin. Some studies have also used human and animal models to assess the effects of cyanobacterial extracts on the skin [54,57,248]. Notably, 5–15% of people had significant skin reactions, as did the tested animals. In addition, irritant reactions on abdominal skin and ear tissue were shown, confirming the risk of dermal exposure. However, no quantitative dose–response relationship was reported. Studies in Australia indicated the occurrence of Lyngbya majuscula (a producer of lyngbyatoxin) bloom caused skin itching, redness, burning, blistering, and swelling in exposed marine recreational users [249].

4.1.3. Intravenous (Hemodialysis)

The unique intravenous pathway via hemodialysis has placed humans at significant risk of exposure to cyanotoxins (Figure 3). The most fatal documented case of human cyanotoxin exposure occurred in Brazil, where out of 130 hemodialysis patients, 76 of 100 patients who developed acute liver failure lost their lives due to the use of water contaminated with MCs during their treatment [38]. The water employed for dialysis revealed a high concentration of MCs (19.5 µg/L), surpassing the World Health Organization’s safety threshold of 1.0 µg/L for drinking water supplies by 19.5 times [38,250,251]. A comparison of victims’ symptoms and pathology with data from animal studies led to the conclusion that intravenous exposure to MCs, specifically MC-YR, MC-LR, and MC-AR, was the major contributing factor to the mortality of dialysis patients [38]. However, the number of fatalities reported for the Brazilian incident seems to vary among studies, i.e., 52 in [251] versus 60 in [252] versus 76 in [38]. The source of this discrepancy is unclear, but could be attributed to the fact that some studies reported the total fatalities of all the people exposed to MCs via dialysis fluid, while others reported those who died due to specific human health conditions confirmed following an autopsy.

4.1.4. Inhalation

According to [253], humans regularly breathe in aerosolized Cyanobacteria and cyanotoxins into their lungs. This is supported by recent studies that revealed that DWSs, particularly natural surface waters and artificial reservoirs, can produce bioaerosols that might contain various cyanobacterial water bloom components, which could potentially expose individuals through inhalation [254,255,256]. For example, cyanotoxin MCs (<35–415 fg/m3), endotoxins (0.13–0.64 EU/m3), cyanobacterial DNA (101–103 cells/m3) [256], and various Cyanobacteria-producing MC-LR species (maximum abundance of 1685 cells/m3) [255] were quantified in bioaerosols collected above surface waters. Similarly, Cyanobacteria and cyanotoxins have been detected in laboratory-aerosolized lake water [254,257]. As a result, the literature evidence such as the induction of respiratory irritation and inflammatory injuries [256], as well as the detection of Cyanobacteria in epithelial cells of the central airway and the upper respiratory tract [253,256], all point to bioaerosol exposure via inhalation. However, future studies are necessary to determine the extent of exposure in various DWSs along the continuum. More importantly, active water sports have also been identified as a poisoning pathway. Backer et al. [258] demonstrated that cyanotoxins can be transmitted from water to air by bursting bubbles on the water surface. The concentration of cyanotoxin in human breath samples was 2.9 ng/m3 compared to up to 5 ng/L detected in nasal swabs after participating in recreational activities on lakes containing 10–500 μg/L of MCs [42]. Koreivienė et al. [42] detected low MC concentrations in post-exposure nose swabs but not in blood samples, thereby confirming the inhalation route. In some cases, cyanotoxin exposure has also been associated with lung inflammation [259,260].

4.2. Potential High-Risk Settings and Groups

Identifying high-risk settings and groups for cyanotoxin is critical for decision-making, including devising, prioritizing, and implementing preventative and control measures. However, systematic studies investigating and characterizing settings and groups with a high risk of exposure to cyanotoxins and their health hazards at global, regional, and national scales are scarce. Potential high-risk settings and groups in low- and middle-income regions include (1) those relying on untreated drinking water from unsafe, polluted water sources, (2) countries where cyanotoxins are not routinely monitored in DWSs, (3) regions with high concentrations and diversity of cyanotoxins in drinking water at the point-of-consumption, and (4) healthcare settings where dialysis fluids with poor quality control and quality assurance are commonly used in hemodialysis patients. These high-risk settings and groups warrant attention in future research focusing on risk profiling and understanding the human exposure and health hazards of cyanotoxins.

4.3. Evidence of Human Health Risks

Human exposure to cyanotoxins is reportedly associated with a variety of illnesses, such as skin irritations, allergic reactions, gastrointestinal illnesses, liver failure, neurotoxic effects, and, in extreme cases, death (Figure 3, Table 2 and Table 3). Note that evidence on health risks of cyanotoxins is largely based on (1) clinical animal models such as mice, rats, and pigs, (2) indirect data on humans obtained from forensic and epidemiological studies, and (3) cases due to chronic exposure or teratogenic impacts on populations at risk [261]. The common health risk is gastroenteritis, resulting from consuming contaminated water [242]. Some of the earliest cases linking gastroenteritis to MC exposure were reported in 1931 in towns located along the Ohio River, USA [262], and from 1960 to 1965 in Harare, Zimbabwe [11,263]. Apart from directly consuming contaminated water, exposure to Cyanobacteria and their metabolites through recreational activities also causes gastroenteritis [132,264,265]. In addition, an increase in exposure time and the density of Cyanobacteria increases the incidence of gastroenteritis [264]. Contrary to the above findings, no significant hazards from exposure to cyanobacterial blooms in recreational waters were observed in some studies conducted in the United Kingdom and Australia [266]. The differences in results could be attributed to the species in the studied water bodies, which could have responded differently in the different environmental conditions, resulting in different toxicity levels.
Besides gastroenteritis, exposure to cyanotoxins may potentially be associated with liver cancers or chronic liver damage [47,245,267]. The International Agency for Research on Cancer classified MCs as Group 2B (possibly carcinogenic to humans), mostly due to their ability to disrupt cellular architecture and adverse effects on cell division and repair, as supported by the tumor promotion data [268]. Particularly, MC-LR exposure has been linked to the origin and progression of hepatocellular carcinoma and intrahepatic cholangiocarcinoma [269]. Other cyanobacterial metabolites were associated with allergic reactions, neurotoxic effects, and skin irritations [270]. However, the effects of long-term exposure to cyanotoxins are not fully understood. Scattered experimental and epidemiological studies correlated contact with cyanotoxin-contaminated water with various conditions affecting the liver [37,267]. Long-term consumption of cyanobacterial metabolites (e.g., in ng/L or µg/L levels), exclusively or in combination with other factors, causes chronic adverse effects such as liver cancers or chronic liver damage and diseases [37,267]. Laboratory-based experiments on children living in the Three Georges Reservoir Region in China assessed the relationship between chronic MC exposure and liver damage [38]. The results showed that chronic exposure to MCs can cause liver cell damage and ultimately significantly increase serum liver enzyme levels.
Neurotoxins are fast-acting compounds, and reports indicate that they interfere with the neuro-muscular system by inhibiting the transfer of excitatory signals, causing paralysis of the respiratory muscles, and death by respiratory failure within min to a few hours post-exposure [42,271]. For example, saxitoxins cause sensation loss by blocking sodium channels in nerve axons [271]. In addition, rat experiments have shown that MCs can cross brain barriers, accumulate in the brain, and cause changes in nerves and the brain [272].
Besides causing symptoms of varying severity, there are also reports of human fatalities related to cyanotoxin exposures [37,272,273]. The most tragic documented event incident in this regard occurred in 1996 in Caruaru, Brazil, when 76 patients died because of dialysate contamination by MCs [37]. The patients showed a host of conditions, including disruption of liver plates, mixed leukocyte infiltration, necrosis, liver cell deformity, cytoplasmic vacuolization, apoptosis, cholestasis, and multinucleated hepatocytes as well as intracellular edema, mitochondrial changes, rough and smooth endoplasmic reticulum injuries, lipid vacuoles, and residual bodies [262]. Although MCs were detected in blood serum and liver, further analyses implicated CYN in the poisoning, suggesting a possibility of multiple cyanotoxin exposures [274]. Consumption of cyanotoxin-contaminated portable water resulted in 2000 cases of gastroenteritis and 88 deaths in Bahia, Brazil, in 1988 [262,271]. Reports indicate that Dolichospermum and Microcystis genera Cyanobacteria had contaminated the water in the Itapirca Dam reservoir, but the studies did not identify the specific metabolites that caused acute gastroenteritis.

4.4. Behaviour and Fate of Cyanotoxins in the Human Body

4.4.1. Cyanotoxin Toxicokinetics: The ADME Concept

The toxicokinetics of various cyanotoxins have attracted significant research attention [273,275,276]. Toxicokinetics describes the time-dependent behavior and fate of a toxicant (in this case, cyanotoxins) from the intake into the bloodstream, movement, or distribution to final excretion as a function of exposure dose or concentration (Figure 3). The ADME concept is widely used to assess toxicokinetics. Thus, four processes are central to the ADME toxicokinetic concept [276]: (1) absorption of the cyanotoxins from the exposure source into the human body or bloodstream, (2) the movement and distribution or partitioning of cyanotoxins from the blood into the tissues, including target organs and receptors, (3) the metabolism or (bio)chemical transformation of cyanotoxin into metabolites, and (4) the excretion rates or removal mechanisms of cyanotoxins from the human body. Some studies have investigated aspects of the toxicokinetics of cyanotoxins in humans and other organisms, including mammals such as rats, mice, and pigs [273,275]. However, for brevity, a comprehensive review of the toxicokinetics of all the individual cyanotoxins is beyond the scope of the present study. Here, we focus on the toxicokinetics of MCs as one example of the most toxic cyanotoxins often detected in drinking water systems. Here, the ADME concept is applied to discuss the evidence on four aspects of the toxicokinetics of cyanotoxins, with a focus on MCs as an example.

Absorption

Depending on the use of DWSs, cyanotoxins enter the human body mainly via the oral route [277]. However, at the point of use, DWSs serve multiple functions, including bathing, cooking, and medical applications (e.g., as a source of dialysis water). Thus, the exposure mechanisms could be complex and involve multiple sources and exposure pathways. In the literature, potential human exposure via inhalation and dermal intake has also been discussed [278]. Intravenous exposure via poorly treated dialysis water obtained from contaminated DWSs has been reported in healthcare facilities, including cases of fatalities (e.g., the Caruaru case in Brazil) [251]. Thus, absorption into the bloodstream may occur via the digestive and respiratory systems and intravenously. However, comprehensive information on the absorption of cyanotoxins in the human body is still lacking. The few available studies show low intestinal absorption of MCs. For example, one study using Caco-2 cells to understand the transport of MCs across the apical–basolateral pathway observed rapid uptake of 24 to 40% in the apical part, but only a small fraction (0.3–1.3%) reached the basolateral part [279]. This low efflux of MCs into the basolateral part is the key mechanism accounting for low intestinal permeability reported in other studies [280].
Other studies based on preclinical in vivo mammal models provide further insights into the absorption of cyanotoxins, particularly MCs. One study used a radio-labeled form of dihydro-MC-LR (MC-LR) administered orally to mice [281]. In the same study, the highest proportion of the MCs was detected in the liver (~0.68%), while the large and small intestines had nearly half the amount (i.e., ~0.25% each). About 38% of the radio-labeled dihydro-MC-LR was detected in the gastrointestinal tract contents, while the bulk of it (67%) was unaccounted for. These results point to the low oral bioavailability of MCs. Similar results were also observed in pigs exposed to MCs via gavage, where no MCs were observed in the serum following exposure to 2 µg/kg [275]. In the same study, about 1.1% of the total dose of the MC administered was detected in the liver, where it was covalently bonded to the protein phosphatases, while part of the unbound one was detected in the kidneys and large intestines. These absorption patterns control the distribution of cyanotoxins in the various organs and tissues. Besides studies using animal models, data on cyanotoxin adsorption in the human body are still limited. Moreover, the few available studies using preclinical animal models have focused on MCs, particularly MC-LR, possibly due to their frequent detection compared to other cyanotoxins. However, other MC variants (e.g., MC-LA, MC-RR, or MC-YR) often co-occur with MC-LR and have been detected in some studies [282,283,284,285,286]. In addition, although neurotoxic blooms are less common than hepatotoxic blooms, anatoxins and saxitoxins have been detected in Australia, North America, and Europe [287,288], and have been associated with deaths of wild and domestic animals [289,290]. Thus, besides hepatotoxins, neurotoxins could also be relevant to DWSs [289].

Distribution

Evidence on the distribution of cyanotoxins is predominantly based on studies conducted on preclinical animal species such as rats and mice [273,291]. These studies largely focused on MCs administered via intravenous or intraperitoneal routes. In earlier investigations conducted in the 1990s, the results showed that MCs bioaccumulate in the liver within about an hour after administration [281,292]. For example, about 71.5% of radio-labeled dihydro-MC-LR administered to mice via the intraperitoneal route accumulated in the liver within one hour [281]. A comparable accumulation of 67% within 1 h was also reported in mice following intravenous administration of MC-LR [292]. A few exceptions exist, where lower values were detected in the liver in other preclinical animal species. For example, one study showed that MC-LR distribution in Wistar rats, two hours following intravenous administration, decreased in the following order: muscle had the highest concentration (17.0%), then liver (6.1%), kidneys (3.7%), intestines (0.5%), lungs (0.3%), and finally gonads (0.1%) [291]. Another study based on intravenous administration of radio-labeled MCs showed that in albino rats, the distribution was highest in the liver (19.2%), followed by the gut contents (9.4%), and then the kidneys (5.3%) [293].
Studies have also investigated the half-life of MCs in preclinical animal species. One study showed that the percentage of MCs in the liver remained at 66% for six days after exposure, indicating a high retention time [292]. In other studies, half-lives of MCs of 0.8 min and 2.1 min for alpha particles and 6.9, 42, and 49 min for beta particles have been reported [293,294]. Further evidence based on LC-MS/MS analysis shows that the MC-LR concentrations in the liver remained unchanged two to seven days after exposure [295]. The long residence time indicates that MCs are bioaccumulated and are retained in the liver with minimal excretion. The bioaccumulation and long retention time in the liver may be due to (1) selective hepatic organic anion transporting polypeptide (OATP) transporters that promote transfer into the liver and (2) the absence of robust efflux pumps or transporters to excrete MCs from the liver [273,296,297]. Indeed, studies in both rodents and humans have demonstrated that hepatic OATP1B2 in rodents and hepatic OATP1B1 and OATP1B3 in humans facilitate the transport of MCs [296,297]. In mice, the Oatp1b2-null transporters were observed to be resistant to the hepatotoxicity of MCs, thus ensuring their continued functionality even in the presence of MCs [296,297]. On the contrary, one study showed that only one efflux transporter (MRP2) transported MC-LR out of a total of seven key efflux transporters investigated [298]. Therefore, the predominance of influx transporter systems for MCs relative to efflux transporters accounts for the high uptake and accumulation of MCs in the liver relative to their efflux.
A point to note is the difference in MC distribution among closely related rodent species (rats versus mice), but the reasons for this observation are unclear. However, differences among species have been reported in other studies (e.g., fish versus humans) and have been explained by several hypotheses related to differences in the capacity of MCs to bind to plasma proteins [299]. For example, one study observed that the LD50 of MC-LR in mammals was about 100 times lower than that of fish because the MC-plasma protein binding in the former was 16 to 28% compared to only 4% in the latter [299]. Further evidence shows that human albumin has the highest binding capacity for MC variants (MC-LR, MC-RR) compared to that of other species (e.g., bovine, porcine, and carp) [299]. Humans also have higher amounts of albumin than other species (e.g., fish). In addition, the facilitated uptake and dissociation of MCs bound to albumin, and possibly mediated by OATP transporters, may further explain the species differences [273]. The role of OATPs in the facilitated transport of MCs, even across the brain barrier, has been reported in some studies [300]. These differences among species raise questions about the transferability of toxicokinetic data based on non-human species often used in preclinical studies to humans. Further research is required to understand the role of albumin and OATPs in the transport, distribution, and partitioning of cyanotoxins in human organs. Such studies should move beyond MCs to include other cyanotoxins detected in DWSs.

Metabolism/(Bio)Chemical Transformation

The metabolism of cyanotoxins, particularly MCs, has been reported in the literature [273]. Metabolic pathways for MCs include the conjugation of glutathione (GSH) by glutathione-S-transferases (GSTs) to produce soluble and polar metabolites that are easily excreted [301]. In vivo studies show that the GSH conjugate undergoes further metabolism to a cysteinyl conjugate as the predominant metabolite [301,302,303]. Two human GSTs (GSTA1 and GSTT1) have excellent catalytic efficiency for MCs of 0.0012 and 0.0022 µM/min, respectively [304]. However, some in vivo studies suggest that the process is reversible, as evidenced by the detection of the parent form of MCs in tissues, urine, feces, and plasma following intraperitoneal injection of the GSH form of MC-RR [84].
MCs’ metabolic rate and overall metabolism appear low because the parent form of MCs is the dominant one detected in the liver following administration [295,305]. For example, a study conducted in rats showed the predominance of MC-LR in the liver in the first 24 h after administration, which dropped to about 50% of the total dose in three to seven days [295]. Another study based on LC-MS/MS also showed that MC-LR was still the dominant form detected in the liver two days following oral administration of MC-LR in rats [305]. A few exceptions exist—one study observed that about 83% of the radio-labeled MC-LR detected in the liver was covalently bonded to cytosolic components within 24 h. However, 83% likely included MC-LR metabolites and MC-LR conjugated to protein phosphatases [292]. In another study, higher MC-LR concentrations in the liver in rats relative to other organs were attributed to the inhibition of GSH synthesis, although this did not affect the concentrations of conjugate and GSH [306].
Evidence suggests that the toxicity of cysteinyl and GSH conjugates is less than that of parent MCs. In one study, the cysteinyl and GSH conjugates of MC-LR were 3 to 10 times less toxic concerning the inhibition of protein phosphatase than the parent MCs [307]. In another study, the LD50 values for cysteinyl and GSH conjugates of two MC variants (MC-YR and MC-LR) were 2.4 to 16.5 times higher than that of the parent MC following intravenous administration in mice [84]. Thus, the metabolic breakdown of MCs via the conjugation of GSH contributes to the detoxification of MCs. Further research is required to understand the human body’s metabolism and the metabolites’ toxicity based on typical concentrations detected in human tissue and organs.

Excretion

The excretion of MCs and parent compounds or metabolites occurs via feces and urine (Figure 3) [273]. The parent compound dominates the excreted MCs, followed by cysteinyl conjugate [84,308]. This indicates that the GSH conjugate of MCs is an unstable intermediate state that readily transforms to the cysteinyl conjugate or even back to the parent MC. One study showed that a total of 24%, comprising about 9% and 15% of radio-labeled MC-LR, was excreted via urine and feces, respectively, while about 83% bioaccumulated in the liver [292]. In the same study, 74% of the MC-LR excreted via urine occurred within 12 h, and approximately 63% and 30% of that was in the form of the parent compound and metabolites, respectively. The corresponding fecal excretion after 6 h and 12 h was 0.9% and 0.5%, respectively, but thereafter, the excretion dropped to only 1% per day for the next six days [292]. Comparable low urinary excretion of 1.9% of the total dose in 120 min was reported following intravenous administration of MCs in rats [293].
The fecal excretion of MCs following intraperitoneal or intravenous administration points to the biliary excretion of MCs [273]. This is further supported by data showing that, during 1 h liver perfusion, approximately 1.7% of radio-labeled MC-LR was excreted into the bile [309]. Still, the bulk of the MCs in the perfusate and bile excretion were in the form of the parent toxins, while about 85% of the toxins detected in the liver were in the polar form. Excretion of MCs in the bile has been reported in other studies using rats and mice [294,310]. The total amount of MCs excreted via feces and urine is relatively low, while the bulk of the MCs bioaccumulate in body organs, especially the liver. In light of this, options to minimize systemic exposure and human toxicity following exposure could be based on: (1) enhancing the metabolism or biochemical transformation to fewer toxic metabolites and/or (2) devising methods to enhance the excretion of both parent toxins and the metabolites.

4.4.2. Toxicity Effects and Mechanisms

Individual Effects of Cyanotoxins

Several studies, including reviews, have investigated the toxicity mechanisms of cyanotoxins, particularly MCs (Table 3) [63,273,311]. Other studies have used omics techniques to investigate the toxicology of cyanotoxins [63]. Based on the toxicological or receptor target, cyanotoxins are classified into four main categories: (1) dermatoxins causing skin irritation on contact, (2) hepatotoxins acting on the liver, (3) neurotoxins causing injury or toxicity to the nervous system, and (4) cytotoxins causing both neurotoxic and hepatotoxic effects [312,313,314]. MC variants or congeners (MC-LR, MC-RR, and MC-YR) and nodularins are considered hepatotoxins [315]. MCs also damage the kidneys and reproductive system [233,238]. CYNs are cytotoxins that mainly target and damage the liver and nervous systems, as well as the kidneys, although they reveal a broad spectrum of adverse effects on various cells through the induction of oxidative stress [111,316,317]. Anatoxins (homoanatoxin-a, anatoxin-a(s), and anatoxin-a) are neurotoxins that target and bind the neuronal nicotinic acetylcholine receptors [318,319]. Aplysiatoxins and lyngbyatoxins are dermatoxins, while lipopolysaccharides are regarded as inflammatory substances [320]. Lyngbyatoxins are vesicatory and highly inflammatory substances, thus causing dermatitis and dermal lesions, and are also strong tumor promoters or carcinogens that activate the protein kinase C [321].
From a public health perspective, the broad categorization of cyanotoxins based on toxicological targets could be misleading. This is because most cyanotoxins have multiple toxicological receptors or targets rather than a single one. Specifically, several cyanotoxins have also been linked to other adverse health effects on histopathologic, cardiac, reproductive, gastrointestinal, and endocrine or hormonal systems (Table 3) [240,273,310,322,323]. Experimental studies and reviews on molecular toxicology have also reported several toxicity mechanisms for MCs alone, including (1) inhibition of protein phosphatases, (2) oxidative stress induced by reactive oxygen species, (3) cell death or apoptosis, and (4) cytoskeleton disruption [238,324,325]. MC variants are also well-known potent inhibitors for the protein phosphatase (PPP) enzyme system with the median lethal dose (LD50) of 0.1–1 nM for the PP1 and PP2A enzymes [49,326,327]. This inhibition of the PPP enzyme systems occurs via MCs covalently binding or bonding to the catalytic subunit of serine/threonine protein [326,327]. However, the extent of inhibition varies among MC variants as a function of the degree of methylation or demethylation status of the amino acids [328,329].
MC-induced oxidative stress is characterized by (1) excessive generation of reactive oxygen species, (2) depletion of GSH, (3) lipid peroxidation or malondialdehyde production, and (4) lactate dehydrogenase (LDH) leakage [330,331]. Thus, one study showed that administering an antioxidant to mice exposed to MC-LR mitigated hepatoxicity by reducing oxidative stress and downregulating the associated processes [332]. One study investigated the reproductive toxicity of MC-LR in mice and concluded that MC-LR significantly impaired spermatogenesis [333]. The possible mechanisms include (1) indirect or direct inhibition of the biosynthesis of gonadotropin-releasing hormones at the hypothalamic level and (2) a reduction in the luteinizing hormone concentration in serum. Collectively, the two processes suppress testosterone production in the testis. MCs also induce dose-dependent cell death, including apoptosis, necrosis, and/or autophagy [310,330,334]. For example, in HepG2 and Vero-E6 cells, MC-induced autophagy was detected at 25 µM and 50 µM, which are less than the critical threshold concentrations for inducing autophagy in HepG2 and Vero-EG cells, respectively [335]. In the same study, equal proportions of necrosis and apoptosis were detected at MC concentrations exceeding the corresponding critical threshold values. MC-induced cell death has been attributed to several mechanisms, including (1) oxidative stress, (2) inhibition of the protein phosphatases, and (3) disruption of the calcium homeostasis and cytoskeleton [264,336,337,338].
The cytoskeletal disruption caused by MCs involves the reorganization of microfilaments, microtubules, and intermediate filaments, which, in turn, lead to the deterioration in intercellular interactions and cell morphology [339,340]. Several mechanisms account for MC-induced cytoskeletal disruption: (1) oxidative stress or ROS generation and degradation of cell organelles and biomolecules, (2) the hyper-phosphorylation of cytoskeletal proteins, and (3) altered expression of cytoskeletal genes [322,341]. Several studies, including reviews on data in non-humans, particularly aquatic animals such as zebrafish, have also shown that MCs are endocrine-disrupting compounds (EDCs) [323,342]. A recent review of 61 studies showed that MCs meet eight out of the ten characteristics of EDCs in terms of interfering with six hormonal processes in the body with respect to (1) biosynthesis, (2) release, (3) circulation or distribution, (4) metabolism or biochemical degradation, (5) binding, and (6) action of natural hormones. The other two EDC characteristics associated with MCs were (1) epigenetic modification and (2) altering or disrupting the fate and behavior of the producing (source) and/or the responding (receptor) cells [323].

Interactive Effects of Cyanotoxins and Other Toxicants

In aquatic environments, including DWSs, cyanotoxins often occur as a mixture of diverse forms and chemical structures. These cyanotoxins also often co-occur with legacy and emerging contaminants, including organics, inorganics, and microplastics. Evidence shows that microplastics adsorb and act as vectors or carriers of MCs [343,344]. The interactions among cyanotoxins and between cyanotoxins and health stressors, including legacy and emerging contaminants, may give rise to complex synergistic and interactive toxicities or health effects. Limited studies have investigated the interactive human health risks of cyanotoxins and other human health stressors. One in vitro study investigated the genotoxicity and mutagenicity of a mixture of two cyanotoxins (MC-LR, CYN) and concluded that the toxicity of mixtures cannot be extrapolated from data based on individual cyanotoxins [51].
A study investigating the toxicity of a combination of chlorpyrifos (pesticide) and CYN on the differentiated SH-SY5Y human neuroblastoma cell showed dose-dependent individual and interactive effects of the two toxicants on viability and induction of necrosis and apoptosis [55]. Another study was conducted using mice to understand the interactions of MC-LR and cadmium exposures on the risk of chronic kidney disease as part of a case-control study in central China [345]. The results showed that exposure to MC-LR induced dose–response kidney injury in mice via the signaling pathway of phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR). Comparison of the highest Cd dose quartile to the lowest value also showed dose–response effects of Cd. Moreover, positive additive or synergistic interactive effects were observed between Cd and MC-LR, indicating relative additional risk due to the combined effects of the two toxicants. The authors concluded that exposure to MC-LR was an independent risk factor for chronic kidney disease and had synergistic interactive effects with Cd, and both exhibited dose–response relationships. In one pre-print paper based on metabolomics and using the Asian clam as a bioassay species, a mixture of microplastics and MCs caused alterations in several metabolites in hepatopancreas compared to the control [323]. The authors concluded that these changes reflected perturbations in several molecular pathways, such as oxidative stress, amino and energy metabolism, and protein degradation/tissue damage. These early results highlight the need for specific toxicity evaluation of mixtures of cyanotoxins and even cyanotoxins and other contaminants.
The multiple toxicity mechanisms and receptor targets, coupled with the diversity of cyanotoxins in terms of origins and chemical structure, make the human health effects even more complex. The potential interactive health risks of cyanotoxins with other contaminants exacerbate this complexity. Due to their chemical diversity, data on toxicity and human health risks of cyanotoxins are limited to only a subset of the cyanotoxins dominated by MCs, which are the most common ones. Thus, information on the toxicity mechanisms of several cyanotoxins detected in DWSs is still limited. Further in vitro and in vivo research is required to better understand the interactive health risks of cyanotoxins with other human health stressors, risk factors, and underlying health conditions. Such information is critical for profiling and mitigation of public health risks.

4.4.3. A Critique of the Evidence on Human Health Risks

Overall, available evidence on human health risks, including cyanotoxins’ toxicokinetics and toxicity mechanisms, has several drawbacks. For example, most of the data on toxicokinetics and toxicity mechanisms are based on extracted individual ‘pure’ cyanotoxins. Thus, such evidence fails to account for other co-occurring cyanotoxins and their interactions. Most toxicological studies tend to be short-term; hence, the results do not account for long-term and inter-generational health effects. Moreover, while human exposure to cyanotoxins is complex and involves multiple pathways, data on human health risks are often inferred from studies based on a single or a few exposure routes. For example, there is a lack of toxicological evidence on the ADME framework based on experiments simulating human exposure via dialysis fluids. Related to inter-generational health effects, the concept of Developmental Origins of Health and Disease (DOHaD) postulates that periconceptual, fetal, and early infant stages of life have an adverse impact on long-term health outcomes [346]. However, limited data exist validating the DOHaD concept for cyanotoxins. In real drinking water systems, cyanotoxins may exist as a complex mixture of various types and variants. However, few studies have partitioned the relative contribution of individual cyanotoxins in mixtures to human health outcomes. Moreover, given that some cyanotoxins have similar toxicity mechanisms while others have multiple health effects, this could create ‘equifinality’ and ‘multifinality’ problems. Multifinality implies that one contaminant may induce more than one human health condition or outcome, while equifinality implies that several emerging organic contaminants and even conventional pollutants may induce similar human health effects or outcomes [347]. The concepts of multifinality and equifinality complicate the interpretation of toxicological data of cyanotoxins but are rarely addressed in existing evidence. Note that these limitations are not limited to cyanotoxins but have been discussed in one of our earlier papers focusing on emerging contaminants in DWSs [347]. This calls for developing new conceptual tools and frameworks to address the complexity of understanding the human health risks of cyanotoxins in DWSs.

5. Risk Mitigation Strategy

A three-step framework based on hazard identification, risk assessment, and mitigation is proposed to mitigate cyanotoxins’ human exposure and health risks. A detailed discussion of related risk assessment and mitigation frameworks was presented in studies focusing on legacy and emerging contaminants [348,349,350]. Thus, for brevity, only an overview of the three key steps in the context of cyanotoxins is summarized here.

5.1. Hazard Identification

This entails identifying and characterizing the nature and hotspots of cyanotoxins in the drinking water value chain, including drinking water sources, water treatment systems, storage, and conveyance facilities, and at the point of use (e.g., households).

5.2. Risk Assessment

Risk assessment seeks to delineate high-risk settings, communities, and age groups (infants, elderly, and those with underlying health conditions) characterized by (1) prevalence of cyanotoxins (hazard) and (2) high likelihood (probability) of human exposure via DWSs. The risk assessment of cyanotoxins can be estimated using qualitative and quantitative risk assessment tools commonly used for other contaminants. Such risk assessment can be coupled to spatial GIS analysis, and the outputs can be visualized as spatial maps that can be used as decision-support tools.

5.3. Preventive and Control Methods

Several potential mitigation measures may be recommended depending on the socio-economic settings, including (1) adopting the precautionary principle in low-income settings lacking current drinking water treatment and data on the occurrence of cyanotoxins due to a lack of analytical laboratories. In such settings, the precautionary principle requires that one must assume the worst-case scenario and consider the drinking water as contaminated. Hence, mandatory drinking water treatment using the best available technology may be recommended. In cases where data are available, tools such as the Hazard Analysis and Critical Control Points (HACCPs), widely used in the food industry, and DWSs can be used for systematic risk assessment and mitigation/management [351,352]. Applying HACCPs could be part of a broader long-term strategy to develop drinking water safety plans at the regional or national levels. In light of the knowledge gaps highlighted, there is also a need for further research to better understand the health risks of cyanotoxins and their toxicity mechanisms. Such an understanding is critical for improved hazard identification and risk characterization. In cases where analytical laboratory facilities exist, cyanotoxins need to be included among routine parameters tested in drinking water, especially in regions with a known prevalence of harmful algal blooms. In such cases, drinking water should conform to current maximum guideline limits for cyanotoxins, and water violating such limits should be treated to achieve acceptable quality for drinking.

5.4. Removal of Cyanotoxins

Cyanotoxins have been removed from DWSs using a variety of techniques (Table 4). The cyanotoxin removal methods are classified into (1) conventional, (2) membrane-based methods/adsorption (carbon-based methods), (3) advanced oxidation technologies, (4) biodegradation, and (5) plant-based coagulants. Drinking water treatment with these methods can remove intracellular cyanotoxins (undamaged cyanobacteria cells) and extracellular dissolved toxins of several Cyanobacteria [69,83,94,129,353]. Although conventional methods have been shown to remove both extracellular and intracellular cyanotoxins, it was reported that methods such as flocculation/coagulation promote the release of MCs into the water [62,129]. Additionally, conventional MCs, which are typically applied post-flocculation/coagulation, such as chlorination, were reported to partially remove MCs and, therefore, result in concentrations of cyanotoxins (1.1 to 3.6 μg/L) above the permissible limits (1 μg/L) recommended by the WHO [69,129]. However, combining conventional methods and advanced methods has significantly reduced the cyanotoxins in drinking water. For instance, extracellular MCs are rarely removed from the water by coagulation, and it was reported that between 2.5 and 7.9% of MCs are removed by coagulation alone. When ozonation and coagulation were applied, between 6.8 and 11.7% of MCs were removed [67]. Moreover, chlorine and ultraviolet exposure led to lysis of 26.2% and 6.6% of M. aeruginosa cells, respectively. However, when ultraviolet/chlorine (with 0.2 mg/L of chlorine) was applied, the damage increased by 51.9% [69].
Various chemicals, such as oxidants, algaecides, chlorine, and permanganate, are commonly used in water treatment facilities to manage Cyanobacteria and cyanotoxins [354,355,356,357]. A crucial aspect of controlling cyanotoxins in DWSs involves reducing the availability of chemicals that Cyanobacteria require for growth. However, introducing an oxidant at the water intake presents challenges regarding cyanotoxin removal. One primary concern is avoiding the rupture of cyanobacterial cells. The consensus suggests that it is most effective to eliminate the maximum quantity of cyanotoxins while the cells are still intact and in the form of particles before the toxins are released [357]. Chlorine has demonstrated the ability to remove cyanobacterial cells, achieving a 90% removal rate within 10 min of oxidation [328,354]. However, limited data exist on the release and fate of the cyanotoxins following cell damage. Hence, the release and fate of cyanotoxins following cell damage warrant further research. On the other hand, permanganate does not react with CYN [354,356], and its reaction with anatoxin-a is rapid, although the rate is influenced by pH, with the rate doubling between pH 8 and 10 [358]. According to a study by [359], the effectiveness of permanganate oxidation in eliminating MCs depends on temperature, characterized by an activation energy of 16 ± 5 kJ/mol rather than pH (within the range of 6 to 10) or alkalinity.
Ozonolysis is generally quite a fast, effective process applied to eliminate cyanotoxins in DWSs. It uses the two oxidation mechanisms of ozone and hydroxyl radicals [360]. However, the extent of oxidation depends on ozone dose, temperature, and pH [360]. In addition, chlorine dioxide has been used to hydrolyze cyanotoxins in DWSs, but it was found to be ineffective at a concentration recommended for drinking water treatment. Most types of activated carbon have generally been effective in removing the most common and toxic cyanotoxins (MCs, anatoxin-a, and CYN) from DWSs. Even though the adsorption of cyanotoxins onto activated carbon varies by carbon type and source water chemistry, each application is unique; activated carbons must be tested to determine effectiveness [361,362]. Aluminum salts like aluminum sulfate are employed to control phosphorus levels in DWSs, limiting their availability as a nutrient for Cyanobacteria. Alum floc binds with sedimentary phosphorus, creating an aluminum–phosphorus compound that immobilizes phosphorus, reducing algae and enhancing water clarity as the floc settles. The effective aluminum dosage depends on water alkalinity, maintaining a pH between 5.5 and 9.0 [361,362].
Using algaecides to remove Cyanobacteria might not be optimal for reducing cyanotoxins, as they rupture cells, releasing higher cyanotoxin concentrations. This could decrease the efficiency of conventional filtration for cyanotoxin removal, requiring alternatives like activated carbon or oxidation [363,364,365,366,367]. Copper sulfate is a common algaecide due to its cost-effectiveness and ease of application, but its use raises concerns about sediment accumulation and harm to aquatic organisms. Chelated copper algaecides address this but pose similar environmental risks [363,364,365,366,367]. Peroxide-based algaecides may offer a relatively eco-friendly alternative to copper, while pollutants like metal ions, pesticides, and antibiotics also impact algal growth and cyanotoxin production [368,369]. However, peroxide-based algaecides may have potential adverse ecological health effects on non-target organisms. Moreover, the efficacy of peroxides depends on the physicochemical properties of the contaminated water [370]. For example, one study shows that Microcystis is highly sensitive to H2O2 at higher than lower light and under nutrient-enriched conditions [370]. Further research is needed to unravel the complex interactions among these factors affecting cyanobacterial growth and toxin production.

6. Future Perspectives

6.1. Harnessing Ethnomedicine and Ethnotoxicology of Cyanotoxins

There is a notable lack of studies documenting indigenous knowledge systems related to cyanotoxins, particularly in the fields of ‘ethnotoxicology’ and ‘ethnomedicine’ for mitigation strategies. Ethnomedicine refers to traditional medical knowledge and practices that interpret human health, disease, and healing within cultural contexts [371,372]. Ethnotoxicology, therefore, encompasses traditional knowledge on the health impacts of toxins, including cyanotoxins, on humans.
To date, research on ethnomedicinal and ethnotoxicological knowledge of cyanotoxins remains limited, even in low- and middle-income regions such as Africa, Southeast Asia, and Latin America, where traditional medicine and herbal remedies are widely used [373]. It is plausible that indigenous communities in these regions possess valuable knowledge on the health effects and potential mitigation of cyanotoxins. Therefore, documenting, validating, and integrating this knowledge into public health and water management strategies could enhance cyanotoxin risk mitigation, particularly in resource-limited settings.

6.2. Research Needs

Future research should focus on the following:
  • Understanding cyanotoxin occurrence, fate, and transport along the entire DWS chain—from the source to the point of use—including storage and conveyance infrastructure.
  • Investigating potential interactive human health effects of cyanotoxins with other pollutants, particularly emerging contaminants.
  • Further exploration of degradation pathways and their by-products, as well as the conditions of formation of these by-products and their toxicity compared to the parent compound.
  • Developing and evaluating novel, low-cost cyanotoxin removal methods, such as biochar and metallic iron, which have demonstrated efficiency in removing various water contaminants.
  • Exploring the potential methods to modify the cyanotoxins to regulate their toxic effects and make high-value products [374].
  • Process modeling of cyanotoxin occurrence, dissemination, fate, and behavior in DWSs to identify human exposure hotspots, especially in low-income regions.
  • Applying quantitative microbial risk assessment tools and disability-adjusted life years to profile cyanotoxin-related health risks in high-risk populations.
  • Developing and validating simple, cost-effective cyanotoxin indicators based on easily measurable water quality parameters, including those informed by indigenous knowledge, for implementation in low-income settings.

7. Conclusions

Cyanotoxins are emerging contaminants in DWSs, posing significant public health risks. Despite their widespread occurrence, research gaps remain in understanding their fate, human exposure, and mitigation strategies. This review provided a semi-quantitative analysis of cyanotoxin diversity, detection methods, health risks, and removal processes along the source water–drinking water continuum.
Evidence shows that cyanotoxins such as MCs, CYN, anatoxins, and saxitoxins are frequently detected in DWSs, with high-risk populations including communities relying on untreated water and those without routine monitoring systems. Advancements in detection technologies, including qPCR, LC-MS, and biosensors, have improved monitoring, but their accessibility remains a challenge in low-income settings. Conventional and advanced treatment methods show promise for cyanotoxin removal, yet cost-effective and scalable solutions are still needed, particularly in resource-limited regions.
Looking forward, key research priorities include (1) better characterization of cyanotoxin occurrence in storage, distribution systems, and at the point of use; (2) assessment of human health risks, particularly the impact of chronic exposure and interactions with other contaminants; (3) development of affordable and efficient removal strategies, leveraging both conventional and indigenous knowledge systems; and (4) exploration of ethnomedicine and ethnotoxicology as alternative approaches to understanding and mitigating cyanotoxin risks. Addressing these gaps will enhance global efforts to ensure safe drinking water and mitigate the health hazards posed by cyanotoxins. Future research should adopt a multidisciplinary approach, integrating environmental science, toxicology, public health, and traditional knowledge to develop holistic and sustainable solutions.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/life15050825/s1. Table S1: Occurrence of cyanotoxins in drinking water systems in Africa; Table S2: Summary data on the occurrence of cyanotoxins in drinking water systems in other regions outside of Africa.

Author Contributions

Conceptualization, W.G.; methodology, W.G.; investigation and writing—original draft preparation, all authors; writing—review and editing, W.G., D.H., and P.R.; visualization, W.G.; project administration, J.M.; resources, P.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Berdalet, E.; Kudela, R.; Urban, E.; Enevoldsen, H.; Banas, N.S. GlobalHAB: A New Program to Promote International Research, Observations, and Modeling of Harmful Algal Blooms in Aquatic Systems. Oceanography 2017, 30, 70–81. [Google Scholar] [CrossRef]
  2. Antoniou, M.G.; Cruz, A.A.d.l.; Dionysiou, D.D. Cyanotoxins: New Generation of Water Contaminants. J. Environ. Eng. 2005, 131, 1239–1243. [Google Scholar] [CrossRef]
  3. Loftin, K.A.; Graham, J.L.; Hilborn, E.D.; Lehmann, S.C.; Meyer, M.T.; Dietze, J.E.; Griffith, C.B. Cyanotoxins in inland lakes of the United States: Occurrence and potential recreational health risks in the EPA National Lakes Assessment 2007. Harmful Algae 2016, 56, 77–90. [Google Scholar] [CrossRef] [PubMed]
  4. Bormans, M.; Amzil, Z.; Mineaud, E.; Brient, L.; Savar, V.; Robert, E.; Lance, E. Demonstrated transfer of cyanobacteria and cyanotoxins along a freshwater-marine continuum in France. Harmful Algae 2019, 87, 101639. [Google Scholar] [CrossRef]
  5. 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]
  6. Fastner, J.; Teikari, J.; Hoffmann, A.; Köhler, A.; Hoppe, S.; Dittmann, E.; Welker, M. Cyanotoxins associated with macrophytes in Berlin (Germany) water bodies—Occurrence and risk assessment. Sci. Total Environ. 2023, 858, 159433. [Google Scholar] [CrossRef] [PubMed]
  7. Huo, D.; Gan, N.; Geng, R.; Cao, Q.; Song, L.; Yu, G.; Li, R. Cyanobacterial blooms in China: Diversity, distribution, and cyanotoxins. Harmful Algae 2021, 109, 102106. [Google Scholar] [CrossRef]
  8. Wang, J.; Zhang, S.; Mu, X.; Hu, X.; Ma, Y. Research Characteristics on Cyanotoxins in Inland Water: Insights from Bibliometrics. Water 2022, 14, 667. [Google Scholar] [CrossRef]
  9. Falfushynska, H.; Kasianchuk, N.; Siemens, E.; Henao, E.; Rzymski, P. A Review of Common Cyanotoxins and Their Effects on Fish. Toxics 2023, 11, 118. [Google Scholar] [CrossRef]
  10. Ndebele, M.R.; Magadza, C.H.D. The occurrence of microcystin-LR in Lake Chivero, Zimbabwe. Lakes Reserv. Res. Manag. 2006, 11, 57–62. [Google Scholar] [CrossRef]
  11. Mhlanga, L.; Day, J.; Cronberg, G.; Chimbari, M.; Siziba, N.; Annadotter, H. Cyanobacteria and cyanotoxins in the source water from Lake Chivero, Harare, Zimbabwe, and the presence of cyanotoxins in drinking water. Afr. J. Aquat. Sci. 2006, 31, 165–173. [Google Scholar] [CrossRef]
  12. Habtemariam, H.; Kifle, D.; Leta, S.; Beekman, W.; Lürling, M. Cyanotoxins in drinking water supply reservoir (Legedadi, Central Ethiopia): Implications for public health safety. SN Appl. Sci. 2021, 3, 328. [Google Scholar] [CrossRef]
  13. Willén, E.; Ahlgren, G.; Tilahun, G.; Spoof, L.; Neffling, M.-R.; Meriluoto, J. Cyanotoxin production in seven Ethiopian Rift Valley lakes. Inland Waters 2011, 1, 81–91. [Google Scholar] [CrossRef]
  14. Mwaura, F.; Koyo, A.O.; Zech, B. Cyanobacterial blooms and the presence of cyanotoxins in small high altitude tropical headwater reservoirs in Kenya. J. Water Health 2004, 2, 49–57. [Google Scholar] [CrossRef]
  15. Raffoul, M.H.; Enanga, E.M.; Senar, O.E.; Creed, I.F.; Trick, C.G. Assessing the potential health risk of cyanobacteria and cyanotoxins in Lake Naivasha, Kenya. Hydrobiologia 2020, 847, 1041–1056. [Google Scholar] [CrossRef]
  16. Muyodi, F.J.; Hecky, R.E.; Kitamirike, J.M.; Odong, R. Trends in health risks from water-related diseases and cyanotoxins in Ugandan portion of Lake Victoria basin. Lakes Reserv. Res. Manag. 2009, 14, 247–257. [Google Scholar] [CrossRef]
  17. Poste, A. Microcystin in Ugandan Lakes: Production Dynamics, Accumulation in Fish, and Risk Evaluation. Ph.D. Thesis, University of Waterloo: Waterloo, ON, Canada, 2010. [Google Scholar]
  18. Zhao, X.; Liu, Y.; Guo, Y.-M.; Xu, C.; Chen, L.; Codd, G.A.; Chen, J.; Wang, Y.; Wang, P.-Z.; Yang, L.-W.; et al. Meta-analysis reveals cyanotoxins risk across African inland waters. J. Hazard. Mater. 2023, 451, 131160. [Google Scholar] [CrossRef] [PubMed]
  19. Pantelić, D.; Svirčev, Z.; Simeunović, J.; Vidović, M.; Trajković, I. Cyanotoxins: Characteristics, production and degradation routes in drinking water treatment with reference to the situation in Serbia. Chemosphere 2013, 91, 421–441. [Google Scholar] [CrossRef] [PubMed]
  20. Fosso-Kankeu, E.; Jagals, P.; Preez, H.D. Exposure of rural households to toxic cyanobacteria in container-stored water. Water SA 2008, 34, 631–636. [Google Scholar] [CrossRef]
  21. Flores, C.; Caixach, J.; Barca, S.; Vieira-Lanero, R.; Cobo, F. Occurrence of Cyanotoxins in Mineral Water Sources and Hot Springs from NW Iberian Peninsula. Biol. Life Sci. Forum 2022, 14, 26. [Google Scholar] [CrossRef]
  22. Lovin, L.M.; Brooks, B.W. Global scanning of anatoxins in aquatic systems: Environment and health hazards, and research needs. Mar. Freshw. Res. 2019, 71, 689–700. [Google Scholar] [CrossRef]
  23. Sundaravadivelu, D.; Sanan, T.T.; Venkatapathy, R.; Mash, H.; Tettenhorst, D.; DAnglada, L.; Frey, S.; Tatters, A.O.; Lazorchak, J. Determination of Cyanotoxins and Prymnesins in Water, Fish Tissue, and Other Matrices: A Review. Toxins 2022, 14, 213. [Google Scholar] [CrossRef] [PubMed]
  24. Abbas, T.; Kajjumba, G.W.; Ejjada, M.; Masrura, S.U.; Marti, E.J.; Khan, E.; Jones-Lepp, T.L.; Abbas, T.; Kajjumba, G.W.; Ejjada, M.; et al. Recent Advancements in the Removal of Cyanotoxins from Water Using Conventional and Modified Adsorbents—A Contemporary Review. Water 2020, 12, 2756. [Google Scholar] [CrossRef]
  25. Wang, S.; Jiao, Y.; Rao, Z. Selective removal of common cyanotoxins: A review. Environ. Sci. Pollut. Res. 2021, 28, 28865–28875. [Google Scholar] [CrossRef]
  26. Agathokleous, E.; Peñuelas, J. Monitoring, Regulation, and Mitigation of Cyanotoxins in the Environment to Protect Human Health and Wildlife. Environ. Sci. Technol. 2022, 56, 14225–14227. [Google Scholar] [CrossRef]
  27. Gwenzi, W.; Marumure, J.; Makuvara, Z.; Simbanegavi, T.T.; Njomou-Ngounou, E.L.; Nya, E.L.; Kaetzl, K.; Noubactep, C.; Rzymski, P. The pit latrine paradox in low-income settings: A sanitation technology of choice or a pollution hotspot? Sci. Total Environ. 2023, 879, 163179. [Google Scholar] [CrossRef]
  28. Chia, M.A.; Ameh, I.; George, K.C.; Balogun, E.O.; Akinyemi, S.A.; Lorenzi, A.S. Genetic Diversity of Microcystin Producers (Cyanobacteria) and Microcystin Congeners in Aquatic Resources across Africa: A Review Paper. Toxics 2022, 10, 772. [Google Scholar] [CrossRef]
  29. Pestana, C.J.; Moura, D.S.; Capelo-Neto, J.; Edwards, C.; Dreisbach, D.; Spengler, B.; Lawton, L.A. Potentially Poisonous Plastic Particles: Microplastics as a Vector for Cyanobacterial Toxins Microcystin-LR and Microcystin-LF. Environ. Sci. Technol. 2021, 55, 15940. [Google Scholar] [CrossRef]
  30. Zaffiro, A.; Rosenblum, L.; Wendelken, S. Method 546: Determination of Total Microcystins and Nodularins in Drinking Water and Ambient Water by Adda Enzyme-Linked Immunosorbent Assay; United States Environmental Protection Agency: Columbus, OH, USA, 2016. [Google Scholar]
  31. Ballot, A.; Sandvik, M.; Rundberget, T.; Botha, C.J.; Miles, C.O. Diversity of cyanobacteria and cyanotoxins in Hartbeespoort Dam, South Africa. Mar. Freshw. Res. 2013, 65, 175–189. [Google Scholar] [CrossRef]
  32. Massey, I.Y.; Wu, P.; Wei, J.; Luo, J.; Ding, P.; Wei, H.; Yang, F. A Mini-Review on Detection Methods of Microcystins. Toxins 2020, 12, 641. [Google Scholar] [CrossRef]
  33. Wendelken, S. Method 545: Determination of Cylindrospermopsin and Anatoxin- in Drinking Water by Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometry (LC/ESI-MS/MS); United States Envirnonmental Protection Agency: Columbus, OH, USA, 2015. [Google Scholar]
  34. Kaushik, R.; Balasubramanian, R. Methods and Approaches Used for Detection of Cyanotoxins in Environmental Samples: A Review. Crit. Rev. Environ. Sci. Technol. 2013, 43, 1349–1383. [Google Scholar] [CrossRef]
  35. Sanseverino, I.; António, D.C.; Loos, R.; Lettieri, T. Cyanotoxins: Methods and Approaches for Their Analysis and Detection; JRC Technology Report; European Commission: Brussels, Belgium, 2017; pp. 1–70. [Google Scholar] [CrossRef]
  36. Mthembu, Z.; Govender, H.; Hendricks, N.; Omotola, E.O.; Kowtharapu, L.P.; Katari, N.K.; Gumbi, B. Detection of Microcystins in South African surface waters by high performance liquid chromatography in the light of Quality by Design statical tool. Res. Sq. 2023. [Google Scholar] [CrossRef]
  37. Li, Y.; Chen, J.-a.; Zhao, Q.; Pu, C.; Qiu, Z.; Zhang, R.; Shu, W. A Cross-Sectional Investigation of Chronic Exposure to Microcystin in Relationship to Childhood Liver Damage in the Three Gorges Reservoir Region, China. Environ. Health Perspect. 2011, 119, 1483–1488. [Google Scholar] [CrossRef] [PubMed]
  38. Carmichael, W.W.; Azevedo, S.M.; An, J.S.; Molica, R.J.; Jochimsen, E.M.; Lau, S.; Rinehart, K.L.; Shaw, G.R.; Eaglesham, G.K. Human fatalities from cyanobacteria: Chemical and biological evidence for cyanotoxins. Environ. Health Perspect. 2001, 109, 663–668. [Google Scholar] [CrossRef] [PubMed]
  39. Turner, P.C.; Gammie, A.J.; Hollinrake, K.; Codd, G.A. Pneumonia associated with contact with cyanobacteria. Br. Med. J. 1990, 300, 1440–1441. [Google Scholar] [CrossRef] [PubMed]
  40. Mrdjen, I.; Morse, M.A.; Ruch, R.J.; Knobloch, T.J.; Choudhary, S.; Weghorst, C.M.; Lee, J. Impact of Microcystin-LR on Liver Function Varies by Dose and Sex in Mice. Toxins 2018, 10, 435. [Google Scholar] [CrossRef]
  41. Labine, M.; Gong, Y.; Minuk, G.Y. Long-Term, Low-Dose Exposure to Microcystin-LR Does not Cause or Increase the Severity of Liver Disease in Rodents. Ann. Hepatol. 2017, 16, 959–965. [Google Scholar] [CrossRef]
  42. Koreivienė, J.; Anne, O.; Kasperovičienė, J.; Burškytė, V. Cyanotoxin management and human health risk mitigation in recreational waters. Environ. Monit. Assess. 2014, 186, 4443–4459. [Google Scholar] [CrossRef]
  43. Rzymski, P.; Poniedziałek, B.; Mankiewicz-Boczek, J.; Faassen, E.J.; Jurczak, T.; Gągała-Borowska, I.; Ballot, A.; Lürling, M.; Kokociński, M. Polyphasic toxicological screening of Cylindrospermopsis raciborskii and Aphanizomenon gracile isolated in Poland. Algal Res. 2017, 24, 72–80. [Google Scholar] [CrossRef]
  44. Fastner, J.; Heinze, R.; Humpage, A.R.; Mischke, U.; Eaglesham, G.K.; Chorus, I. Cylindrospermopsin occurrence in two German lakes and preliminary assessment of toxicity and toxin production of Cylindrospermopsis raciborskii (Cyanobacteria) isolates. Toxicon 2003, 42, 313–321. [Google Scholar] [CrossRef]
  45. Zhao, Y.; Yan, Y.; Xie, L.; Wang, L.; He, Y.; Wan, X.; Xue, Q. Long-term environmental exposure to microcystins increases the risk of nonalcoholic fatty liver disease in humans: A combined fisher-based investigation and murine model study. Environ. Int. 2020, 138, 105648. [Google Scholar] [CrossRef] [PubMed]
  46. Zhao, Y.; Xue, Q.; Su, X.; Xie, L.; Yan, Y.; Wang, L.; Steinman, A.D. First Identification of the Toxicity of Microcystins on Pancreatic Islet Function in Humans and the Involved Potential Biomarkers. Environ. Sci. Technol. 2016, 50, 3137–3144. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, X.; Xie, P.; Zhang, X.; Zhou, W.; Zhao, S.; Zhao, Y.; Cai, Y. Toxic effects of microcystin-LR on the HepG2 cell line under hypoxic and normoxic conditions. J. Appl. Toxicol. 2013, 33, 1180–1186. [Google Scholar] [CrossRef] [PubMed]
  48. Sedan, D.; Laguens, M.; Copparoni, G.; Aranda, J.O.; Giannuzzi, L.; Marra, C.A.; Andrinolo, D. Hepatic and intestine alterations in mice after prolonged exposure to low oral doses of Microcystin-LR. Toxicon 2015, 104, 26–33. [Google Scholar] [CrossRef] [PubMed]
  49. Hastie, C.J.; Borthwick, E.B.; Morrison, L.F.; Codd, G.A.; Cohen, P.T.W. Inhibition of several protein phosphatases by a non-covalently interacting microcystin and a novel cyanobacterial peptide, nostocyclin. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2005, 1726, 187–193. [Google Scholar] [CrossRef] [PubMed]
  50. Xu, Y.; Cui, J.; Yu, H.; Zong, W.; Xu, Y.; Cui, J.; Yu, H.; Zong, W. Insight into the Molecular Mechanism for the Discrepant Inhibition of Microcystins (MCLR, LA, LF, LW, LY) on Protein Phosphatase 2A. Toxins 2022, 14, 390. [Google Scholar] [CrossRef]
  51. Díez-Quijada, L.; Prieto, A.I.; Puerto, M.; Jos, Á.; Cameán, A.M. In Vitro Mutagenic and Genotoxic Assessment of a Mixture of the Cyanotoxins Microcystin-LR and Cylindrospermopsin. Toxins 2019, 11, 318. [Google Scholar] [CrossRef]
  52. Yang, F.; Wen, C.; Zheng, S.; Yang, S.; Chen, J.; Feng, X. Involvement of MAPK/ERK1/2 pathway in microcystin-induced microfilament reorganization in HL7702 hepatocytes. J. Toxicol. Environ. Health Part A 2018, 81, 1135–1141. [Google Scholar] [CrossRef]
  53. Torokne, A.; Palovics, A.; Bankine, M. Allergenic (sensitization, skin and eye irritation) effects of freshwater cyanobacteria—Experimental evidence. Environ. Toxicol. 2001, 16, 512–516. [Google Scholar] [CrossRef]
  54. Stewart, I.; Seawright, A.A.; Schluter, P.J.; Shaw, G.R. Primary irritant and delayed-contact hypersensitivity reactions to the freshwater cyanobacterium Cylindrospermopsis raciborskii and its associated toxin cylindrospermopsin. BMC Dermatol. 2006, 6, 5. [Google Scholar] [CrossRef]
  55. Hinojosa, M.G.; Gutiérrez-Praena, D.; López, S.; Prieto, A.I.; Moreno, F.J.; Jos, Á.; Cameán, A.M. Toxic effects of the cylindrospermopsin and chlorpyrifos combination on the differentiated SH-SY5Y human neuroblastoma cell line. Toxicon 2023, 227, 107091. [Google Scholar] [CrossRef] [PubMed]
  56. Mankiewicz-Boczek, J.; Palus, J.; Gągała, I.; Izydorczyk, K.; Jurczak, T.; Dziubałtowska, E.; Stępnik, M.; Arkusz, J.; Komorowska, M.; Skowron, A.; et al. Effects of microcystins-containing cyanobacteria from a temperate ecosystem on human lymphocytes culture and their potential for adverse human health effects. Harmful Algae 2011, 10, 356–365. [Google Scholar] [CrossRef]
  57. Pilotto, L.; Hobson, P.; Burch, M.D.; Ranmuthugala, G.; Attewell, R.; Weightman, W. Acute skin irritant effects of cyanobacteria (blue-green algae) in healthy volunteers. Aust. N. Z. J. Public Health 2004, 28, 220–224. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, W.; Liu, J.; Xiao, Y.; Zhang, Y.; Yu, Y.; Zheng, Z.; Liu, Y.; Li, Q. The Impact of Cyanobacteria Blooms on the Aquatic Environment and Human Health. Toxins 2022, 14, 658. [Google Scholar] [CrossRef]
  59. Vela, L.; Sevilla, E.; Gonzalez, C.; Bes, M.T.; Fillat, M.F.; Peleato, M.L. Exploring the interaction of microcystin-LR with proteins and DNA. Toxicol. Vitr. 2008, 22, 1714–1718. [Google Scholar] [CrossRef] [PubMed]
  60. Gao, H.; Zhu, N.; Deng, S.; Du, C.; Tang, Y.; Tang, P.; Xu, S.; Liu, W.; Shen, M.; Xiao, X.; et al. Combination Effect of Microcystins and Arsenic Exposures on CKD: A Case-Control Study in China. Toxins 2023, 15, 144. [Google Scholar] [CrossRef] [PubMed]
  61. Rapala, J.; Lahti, K.; Räsänen, L.A.; Esala, A.-L.; Niemelä, S.I.; Sivonen, K. Endotoxins associated with cyanobacteria and their removal during drinking water treatment. Water Res. 2002, 36, 2627–2635. [Google Scholar] [CrossRef] [PubMed]
  62. Mohamed, Z.A.; Deyab, M.A.; Abou-Dobara, M.I.; El-Raghi, W.M. Occurrence of toxic cyanobacteria and microcystin toxin in domestic water storage reservoirs, Egypt. J. Water Supply Res. Technol.-Aqua 2016, 65, 431–440. [Google Scholar] [CrossRef]
  63. Ma, Y.; Liu, H.; Du, X.; Shi, Z.; Liu, X.; Wang, R.; Zhang, S.; Tian, Z.; Shi, L.; Guo, H.; et al. Advances in the toxicology research of microcystins based on Omics approaches. Environ. Int. 2021, 154, 106661. [Google Scholar] [CrossRef]
  64. Zamyadi, A.; Dorner, S.; Sauvé, S.; Ellis, D.; Bolduc, A.; Bastien, C.; Prévost, M. Species-dependence of cyanobacteria removal efficiency by different drinking water treatment processes. Water Res. 2013, 47, 2689–2700. [Google Scholar] [CrossRef]
  65. Xagoraraki, I. Fate of pharmaceuticals during water chlorination. In Proceedings of the Water Quality Technology Conference, Charlotte, NC, USA, 4–8 November 2007. [Google Scholar]
  66. Mohamed, Z.A. Breakthrough of Oscillatoria limnetica and microcystin toxins into drinking water treatment plants—Examples from the Nile River, Egypt. Water SA 2016, 42, 161–165. [Google Scholar] [CrossRef]
  67. Teixeira, M.R.; Rosa, M.J. Comparing dissolved air flotation and conventional sedimentation to remove cyanobacterial cells of Microcystis aeruginosa: Part II. The effect of water background organics. Sep. Purif. Technol. 2007, 53, 126–134. [Google Scholar] [CrossRef]
  68. Yuan, B.-L.; Qu, J.-H.; Fu, M.-L. Removal of cyanobacterial microcystin-LR by ferrate oxidation–coagulation. Toxicon 2002, 40, 1129–1134. [Google Scholar] [CrossRef] [PubMed]
  69. Sun, J.; Bu, L.; Deng, L.; Shi, Z.; Zhou, S. Removal of Microcystis aeruginosa by UV/chlorine process: Inactivation mechanism and microcystins degradation. Chem. Eng. J. 2018, 349, 408–415. [Google Scholar] [CrossRef]
  70. Lambert, T.W.; Holmes, C.F.B.; Hrudey, S.E. Adsorption of microcystin-LR by activated carbon and removal in full scale water treatment. Water Res. 1996, 30, 1411–1422. [Google Scholar] [CrossRef]
  71. Kumar, P.; Cledon, M.; Brar, S.K. A low-cost graphitized sand filter to deliver MC-LR-free potable water: Water treatment plants and household perspective. Sci. Total Environ. 2020, 747, 141135. [Google Scholar] [CrossRef]
  72. Mouchet, P.; Bonnelye, V. Solving algae problems: French expertise and worldwide applications. J. Water Supply Res. Technol. 1998, 47, 125–141. [Google Scholar] [CrossRef]
  73. Ho, L.; Meyn, T.; Keegan, A.; Hoefel, D.; Brookes, J.; Saint, C.P.; Newcombe, G. Bacterial degradation of microcystin toxins within a biologically active sand filter. Water Res. 2006, 40, 768–774. [Google Scholar] [CrossRef]
  74. Gijsbertsen-Abrahamse, A.J.; Schmidt, W.; Chorus, I.; Heijman, S.G.J. Removal of cyanotoxins by ultrafiltration and nanofiltration. J. Membr. Sci. 2006, 276, 252–259. [Google Scholar] [CrossRef]
  75. Drogui, P.; Daghrir, R.; Simard, M.-C.; Sauvageau, C.; Blais, J.F. Removal of microcystin-LR from spiked water using either activated carbon or anthracite as filter material. Environ. Technol. 2012, 33, 381–391. [Google Scholar] [CrossRef]
  76. Mashile, P.P.; Mpupa, A.; Nomngongo, P.N. Adsorptive removal of microcystin-LR from surface and wastewater using tyre-based powdered activated carbon: Kinetics and isotherms. Toxicon 2018, 145, 25–31. [Google Scholar] [CrossRef] [PubMed]
  77. Kohler, E.; Villiger, J.; Posch, T.; Derlon, N.; Shabarova, T.; Morgenroth, E.; Pernthaler, J.; Blom, J.F. Biodegradation of Microcystins during Gravity-Driven Membrane (GDM) Ultrafiltration. PLoS ONE 2014, 9, e111794. [Google Scholar] [CrossRef] [PubMed]
  78. Chow, C.W.K.; Panglisch, S.; House, J.; Drikas, M.; Burch, M.D.; Gimbel, R. A study of membrane filtration for the removal of cyanobacterial cells. Aqua 1997, 46, 324–334. [Google Scholar]
  79. Xiong, Q.; Liu, Z.H.; Zhang, Y.H.; Liang, H.F.; Yang, S.; Li, J.L. Algaecidal effect of a nanomaterial sheet. Huanjing Kexue 2006, 27, 715–719. [Google Scholar] [PubMed]
  80. Wang, H.; Ho, L.; Lewis, D.M.; Brookes, J.D.; Newcombe, G. Discriminating and assessing adsorption and biodegradation removal mechanisms during granular activated carbon filtration of microcystin toxins. Water Res. 2007, 41, 4262–4270. [Google Scholar] [CrossRef] [PubMed]
  81. Lopes, W.S.; Buriti, J.S.; Cebalos, B.S.O.; Sousa, J.T.; Leite, V.D.; Vieira, F.F. Removal of Microcystin-LR from Drinking Water Using a System Involving Oxidation and Adsorption. Water Air Soil Pollut. 2017, 228, 1–14. [Google Scholar] [CrossRef]
  82. Jian, Z.; Bai, Y.; Chang, Y.; Liang, J.; Qu, J. Removal of micropollutants and cyanobacteria from drinking water using KMnO4 pre-oxidation coupled with bioaugmentation. Chemosphere 2019, 215, 1–7. [Google Scholar] [CrossRef]
  83. Bai, M.; Zheng, Q.; Zheng, W.; Li, H.; Lin, S.; Huang, L.; Zhang, Z. •OH Inactivation of Cyanobacterial Blooms and Degradation of Toxins in Drinking Water Treatment System. Water Res. 2019, 154, 144–152. [Google Scholar] [CrossRef] [PubMed]
  84. Li, H.; Bai, M.; Yang, X.; Zhong, Z.; Gao, M.; Tian, Y. OH pre-treatment of algae blooms and degradation of microcystin-LR in a drinking water system of 480 m3/day: Comparison with ClO2. Chem. Eng. J. 2019, 367, 189–197. [Google Scholar] [CrossRef]
  85. Hoeger, S.J.; Shaw, G.; Hitzfeld, B.C.; Dietrich, D.R. Occurrence and elimination of cyanobacterial toxins in two Australian drinking water treatment plants. Toxicon 2004, 43, 639–649. [Google Scholar] [CrossRef]
  86. Momani, F.A.A.; Jarrah, N. Treatment and kinetic study of cyanobacterial toxin by ozone. J. Environ. Sci. Health Part A 2010, 45, 719–731. [Google Scholar] [CrossRef] [PubMed]
  87. Lawton, L.A.; Robertson, P.K.J.; Cornish, B.J.P.A.; Marr, I.L.; Jaspars, M. Processes influencing surface interaction and photocatalytic destruction of microcystins on titanium dioxide photocatalysts. J. Catal. 2003, 213, 109–113. [Google Scholar] [CrossRef]
  88. Yuan, B.; Li, Y.; Huang, X.; Liu, H.; Qu, J. Fe(VI)-assisted photocatalytic degradating of microcystin-LR using titanium dioxide. J. Photochem. Photobiol. A Chem. 2006, 178, 106–111. [Google Scholar] [CrossRef]
  89. Mohamed, Z.A.; Alamri, S.A. Biodegradation of cylindrospermopsin toxin by microcystin-degrading bacteria isolated from cyanobacterial blooms. Toxicon 2012, 60, 1390–1395. [Google Scholar] [CrossRef] [PubMed]
  90. Dziga, D.; Kokocinski, M.; Maksylewicz, A.; Czaja-Prokop, U.; Barylski, J. Cylindrospermopsin Biodegradation Abilities of Aeromonas sp. Isolated from Rusałka Lake. Toxins 2016, 8, 55. [Google Scholar] [CrossRef]
  91. Somdee, T.; Thunders, M.; Ruck, J.; Lys, I.; Allison, M.; Page, R. Degradation of [Dha7]MC-LR by a Microcystin Degrading Bacterium Isolated from Lake Rotoiti, New Zealand. Int. Sch. Res. Not. 2013, 2013, 596429. [Google Scholar] [CrossRef]
  92. Nhut, H.T.; Hung, N.T.Q.; Lap, B.Q.; Han, L.T.N.; Tri, T.Q.; Bang, N.H.K.; Hiep, N.T.; Ky, N.M. Use of Moringa oleifera seeds powder as bio-coagulants for the surface water treatment. Int. J. Environ. Sci. Technol. 2020, 18, 2173–2180. [Google Scholar] [CrossRef]
  93. Wan, J.; Chakraborty, T.; Xu, C.C.; Ray, M.B. Treatment train for tailings pond water using Opuntia ficus-indica as coagulant. Sep. Purif. Technol. 2019, 211, 448–455. [Google Scholar] [CrossRef]
  94. Bouaidi, W.E.; Essalhi, S.; Douma, M.; Tazart, Z.; Ounas, A.; Enaime, G.; Yaacoubi, A.; Loudiki, M. Evaluation of the potentiality of Vicia faba and Opuntia ficus indica as eco-friendly coagulants to mitigate Microcystis aeruginosa blooms. Desalination Water Treat. 2020, 196, 198–213. [Google Scholar] [CrossRef]
  95. Teixeira, M.R.; Camacho, F.P.; Sousa, V.S.; Bergamasco, R. Green technologies for cyanobacteria and natural organic matter water treatment using natural based products. J. Clean. Prod. 2017, 162, 484–490. [Google Scholar] [CrossRef]
  96. Camacho, F.; Bongiovani, M.; Amorim, M.P.; Silva, M.; Coldebella, P.; Bergamasco, R. Coagulation/Flocculation/Flotation/Nanofiltration Processes Using Moringa Oleifera as Coagulant of Eutrophized River. Chem. Eng. Trans. 2015, 43, 1123–1128. [Google Scholar] [CrossRef]
  97. Corbel, S.; Mougin, C.; Bouaïcha, N. Cyanobacterial toxins: Modes of actions, fate in aquatic and soil ecosystems, phytotoxicity and bioaccumulation in agricultural crops. Chemosphere 2014, 96, 1–15. [Google Scholar] [CrossRef] [PubMed]
  98. Codd, G.A.; Testai, E.; Funari, E.; Svirčev, Z. Cyanobacteria, Cyanotoxins, and Human Health. In Water Treatment for Purification from Cyanobacteria and Cyanotoxins, Hiskia, A.E., Triantis, T.M., Antoniou, M.G., Kaloudis, T., Dionysiou, D.D., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2020; pp. 37–68. [Google Scholar]
  99. Daly, R.I.; Ho, L.; Brookes, J.D. Effect of Chlorination on Microcystis aeruginosa Cell Integrity and Subsequent Microcystin Release and Degradation. Key Eng. Mater. 2007, 277–279, 606–611. [Google Scholar] [CrossRef] [PubMed]
  100. Antunes, J.T.; Leão, P.N.; Vasconcelos, V.M. Cylindrospermopsis raciborskii: Review of the distribution, phylogeography, and ecophysiology of a global invasive species. Front. Microbiol. 2015, 6, 473. [Google Scholar] [CrossRef]
  101. Jiang, Y.; Ji, B.; Wong, R.N.S.; Wong, M.H. Statistical study on the effects of environmental factors on the growth and microcystins production of bloom-forming cyanobacterium—Microcystis aeruginosa. Harmful Algae 2008, 7, 127–136. [Google Scholar] [CrossRef]
  102. Fotiou, T.; Triantis, T.M.; Kaloudis, T.; O’Shea, K.E.; Dionysiou, D.D.; Hiskia, A. Assessment of the roles of reactive oxygen species in the UV and visible light photocatalytic degradation of cyanotoxins and water taste and odor compounds using C–TiO2. Water Res. 2016, 90, 52–61. [Google Scholar] [CrossRef]
  103. Yusof, T.N.; Rafatullah, M.; Mohamad, R.; Ismail, N.; Zainuddin, Z.; Lalung, J. Cyanobacteria Characteristics and Methods for Isolation and Accurate Identification of Cyanotoxins: A Review Article. Avicenna J. Environ. Health Eng. 2017, 4, 10051. [Google Scholar] [CrossRef]
  104. Spoof, L.; Catherine, A. Appendix 3 Tables of MCs and Nodularins. In Handbook of Cyanobacterial Monitoring and Cyanotoxin Analysis, 1st ed.; Meriluoto, J., Spoof, L., Codd, G.A., Eds.; John Wiley & Sons, Ltd.: Chichester, UK, 2017. [Google Scholar]
  105. Chorus, I.; Welker, M. Toxic Cyanobacteria in Water. A Guide to Their Public Health Consequences, Monitoring and Management, 2nd ed.; CRC Press: London, UK, 2021. [Google Scholar]
  106. 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]
  107. Wonnacott, S.; Jackman, S.; Swanson, K.L.; Rapoport, H.; Albuquerque, E.X. Nicotinic pharmacology of anatoxin analogs. II. Side chain structure-activity relationships at neuronal nicotinic ligand binding sites. J. Pharmacol. Exp. Ther. 1991, 259, 387–391. [Google Scholar] [CrossRef]
  108. Méjean, A.; Paci, G.; Gautier, V.; Ploux, O. Biosynthesis of anatoxin-a and analogues (anatoxins) in cyanobacteria. Toxicon 2014, 91, 15–22. [Google Scholar] [CrossRef]
  109. Bruno, M.; Ploux, O.; Metcalf, J.S.; Mejean, A.P.; Awlik-Skowronska, B.; Furey, A. Anatoxin-a, Homoanatoxin-a, and Natural Analogues. In Handbook of Cyanobacterial Monitoring and Cyanotoxin Analysis; Wiley Online Library: Hoboken, NJ, USA, 2016; pp. 138–147. [Google Scholar] [CrossRef]
  110. Robillot, C.; Llewellyn, L.E. Water analysis, algal and microbial toxins. In Encyclopedia of Analytical Science, 2nd ed.; Worsfold, P., Townshend, A.P., Eds.; Elsevier Ltd.: Amsterdam, The Netherlands, 2012; pp. 343–349. [Google Scholar]
  111. Poniedziałek, B.; Rzymski, P.; Kokociński, M. Cylindrospermopsin: Water-linked potential threat to human health in Europe. Environ. Toxicol. Pharmacol. 2012, 34, 651–660. [Google Scholar] [CrossRef] [PubMed]
  112. Jones, M.R.; Pinto, E.; Torres, M.A.; Dörr, F.; Mazur-Marzec, H.; Szubert, K.; Tartaglione, L.; Dell’Aversano, C.; Miles, C.O.; Beach, D.G.; et al. CyanoMetDB, a comprehensive public database of secondary metabolites from cyanobacteria. Water Res. 2021, 196, 117017. [Google Scholar] [CrossRef]
  113. Jones, R.J.; Pinto, E.; Torres, F.D.; Mazur-Marzec, H.; Szybert, K.; Tartaglione, L.; Cel’Aversano, C.; Miles, C.O.; Beach, D.G.; McCarron, P.; et al. Comprehensive database of secondary metabolites from cyanobacteria. bioRiv 2020. [Google Scholar] [CrossRef]
  114. Hrouzek, P.; Kapuścik, A.; Vacek, J.; Voráčová, K.; Paichlová, J.; Kosina, P.; Voloshko, L.; Ventura, S.; Kopecký, J. Cytotoxicity evaluation of large cyanobacterial strain set using selected human and murine in vitro cell models. Ecotoxicol. Environ. Saf. 2016, 124, 177–185. [Google Scholar] [CrossRef]
  115. Falfushynska, H.; Horyn, O.; Osypenko, I.; Rzymski, P.; Wejnerowski, Ł.; Dziuba, M.K.; Sokolova, I.M. Multibiomarker-based assessment of toxicity of central European strains of filamentous cyanobacteria Aphanizomenon gracile and Raphidiopsis raciborskii to zebrafish Danio rerio. Water Res. 2021, 194, 116923. [Google Scholar] [CrossRef] [PubMed]
  116. Hitzfeld, B.C.; Lampert, C.S.; Spaeth, N.; Mountfort, D.; Kaspar, H.; Dietrich, D.R. Toxin production in cyanobacterial mats from ponds on the McMurdo Ice Shelf, Antarctica. Toxicon 2000, 38, 1731–1748. [Google Scholar] [CrossRef] [PubMed]
  117. Kleinteich, J.; Hildebrand, F.; Wood, S.A.; Ciŕs, S.; Agha, R.; Quesada, A.; Pearce, D.A.; Convey, P.; Kpper, F.C.; Dietrich, D.R. Diversity of toxin and non-toxin containing cyanobacterial mats of meltwater ponds on the Antarctic Peninsula: A pyrosequencing approach. Antarct. Sci. 2014, 26, 521–532. [Google Scholar] [CrossRef]
  118. 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 R.F. Scott’s Discovery Expedition. Eur. J. Phycol. 2018, 53, 115–121. [Google Scholar] [CrossRef]
  119. Usman, A.S.; Merican, F.; Zaki, S.; Broady, P.; Convey, P.; Muangmai, N. Microcystin production by oscillatorialean cyanobacteria isolated from cryopreserved Antarctic mats. Harmful Algae 2022, 120, 102336. [Google Scholar] [CrossRef]
  120. Addico, G.N.D.; Hardege, J.D.; Kohoutek, J.; deGraft-Johnson, K.A.A.; Babica, P. Cyanobacteria and microcystin contamination in untreated and treated drinking water in Ghana. Adv. Oceanogr. Limnol. 2017, 8, 92–106. [Google Scholar] [CrossRef]
  121. Ballot, A.; Krienitz, L.; Kotut, K.; Wiegand, C.; Pflugmacher, S. Cyanobacteria and cyanobacterial toxins in the alkaline crater lakes Sonachi and Simbi, Kenya. Harmful Algae 2005, 4, 139–150. [Google Scholar] [CrossRef]
  122. Magonono, M.; Oberholster, P.J.; Shonhai, A.; Makumire, S.; Gumbo, J.R. The Presence of Toxic and Non-Toxic Cyanobacteria in the Sediments of the Limpopo River Basin: Implications for Human Health. Toxins 2018, 10, 269. [Google Scholar] [CrossRef]
  123. Mchau, G.J.; Machunda, R.; Kimanya, M.; Makule, E.; Gong, Y.Y.; Mpolya, E.; Meneely, J.P.; Elliott, C.T.; Greer, B. First Report of the Co-occurrence of Cylindrospermopsin, Nodularin and Microcystins in the Freshwaters of Lake Victoria, Tanzania. Expo. Health 2020, 13, 185–194. [Google Scholar] [CrossRef]
  124. Otoigo, L. Identification and quantification of Cyanotoxins in Lake and household water around Lake Victoria, Kenya. AfricArXiv Preprints 2020. [Google Scholar] [CrossRef]
  125. Olokotum, M.; Humbert, J.-F.; Quiblier, C.; Okello, W.; Semyalo, R.; Troussellier, M.; Marie, B.; Baumann, K.; Kurmayer, R.; Bernard, C. Characterization of Potential Threats from Cyanobacterial Toxins in Lake Victoria Embayments and during Water Treatment. Toxins 2022, 14, 664. [Google Scholar] [CrossRef]
  126. Mohamed, Z.A.; Alamri, S.; Hashem, M. The link between microcystin levels in groundwater and surface Nile water, and assessing their potential risk to human health. J. Contam. Hydrol. 2022, 244, 103921. [Google Scholar] [CrossRef] [PubMed]
  127. Addico, G.; Hardege, J.; Komarek, J.; Babica, P.; Graft-Johnson, K.D. Cyanobacteria species identified in the Weija and Kpong reservoirs, Ghana, and their implications for drinking water quality with respect to microcystin. Afr. J. Mar. Sci. 2006, 28, 451–456. [Google Scholar] [CrossRef]
  128. Zewde, T.; Kifle, D.; Johansen, J.; Demissie, T.; Hansen, J.; Tadesse, Z. Cyanobacterial abundance and microcystins in water, seston and fish tissues in Lake Hora-Arsedi (Ethiopia). Afr. J. Aquat. Sci. 2020, 45, 475–485. [Google Scholar] [CrossRef]
  129. Mohamed, Z.A.; Deyab, M.A.; Abou-Dobara, M.I.; El-Sayed, A.K.; El-Raghi, W.M. Occurrence of cyanobacteria and microcystin toxins in raw and treated waters of the Nile River, Egypt: Implication for water treatment and human health. Environ. Sci. Pollut. Res. 2015, 22, 11716–11727. [Google Scholar] [CrossRef] [PubMed]
  130. Mokoena, M.M.; Mukhola, M.S.; Mokoena, M.M.; Mukhola, M.S. Current Effects of Cyanobacteria Toxin in Water Sources and Containers in the Hartbeespoort Dam Area, South Africa. Int. J. Environ. Res. Public Health 2019, 16, 4468. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, Z.; Kong, F.; Zhang, M. Groundwater contamination by microcystin from toxic cyanobacteria blooms in Lake Chaohu, China. Environ. Monit. Assess. 2016, 188, 280. [Google Scholar] [CrossRef]
  132. Tamele, I.J.; Vasconcelos, V. Microcystin Incidence in the Drinking Water of Mozambique: Challenges for Public Health Protection. Toxins 2020, 12, 368. [Google Scholar] [CrossRef]
  133. Kulabhusan, P.K.; Campbell, K. Recent trends in the detection of freshwater cyanotoxins with a critical note on their occurrence in Asia. Trends Environ. Anal. Chem. 2021, 32, e00150. [Google Scholar] [CrossRef]
  134. Bashir, F.; Bashir, A.; Rajput, V.D.; Bouaïcha, N.; Fazili, K.M.; Adhikari, S.; Negi, Y.; Minkina, T.; Almalki, W.H.; Ganai, B.A. Microcystis sp. AE03 strain in Dal Lake harbors cylindrospermopsin and microcystin synthetase gene cluster. Front. Sustain. Food Syst. 2022, 6, 1036111. [Google Scholar] [CrossRef]
  135. Anahas, A.M.P.; Gayathri, M.; Muralitharan, G. Isolation and Characterization of Microcystin-Producing Microcystis ae. In Microbiological Research in Agroecosystem Management; Velu, R., Ed.; Springer: Pune, India, 2013; pp. 235–248. [Google Scholar]
  136. Batool, U.; Tromas, N.; Simon, D.F.; Sauvé, S.; Shapiro, B.J.; Ahmed, M. Snapshot of cyanobacterial toxins in Pakistani freshwater bodies. Environ. Sci. Pollut. Res. 2024, 31, 24648–24661. [Google Scholar] [CrossRef]
  137. Ishikawa, K.; Kumagai, M.; Vincent, W.F.; Tsujimura, S.; Nakahara, H. Transport and accumulation of bloom-forming cyanobacteria in a large, mid-latitude lake: The gyre-Microcystis hypothesis. Limnology 2002, 3, 87–96. [Google Scholar] [CrossRef]
  138. Watanabe, M.F.; Tsujimura, S.; Oishi, S.; Niki, T.; Namikoshi, M. Isolation and identification of homoanatoxin-a from a toxic strain of the cyanobacterium Raphidiopsis mediterranea Skuja isolated from Lake Biwa, Japan. Phycologia 2003, 42, 364–369. [Google Scholar] [CrossRef]
  139. Gurbuz, F.; Metcalf, J.S.; Karahan, A.G.; Codd, G.A. Analysis of dissolved microcystins in surface water samples from Kovada Lake, Turkey. Sci. Total Environ. 2009, 407, 4038–4046. [Google Scholar] [CrossRef] [PubMed]
  140. Swe, T.; Miles, C.O.; Cerasino, L.; Mjelde, M.; Kleiven, S.; Ballot, A. Microcystis, Raphidiopsis raciborskii and Dolichospermum smithii, toxin producing and non-toxigenic cyanobacteria in Yezin Dam, Myanmar. Limnologica 2021, 90, 125901. [Google Scholar] [CrossRef]
  141. Welker, M.; Chorus, I.; Fastner, J.; Khan, S.; Haque, M.M.; Islam, S.; Khan, N.H. Microcystins (cyanobacterial toxins) in surface waters of rural Bangladesh: Pilot study. J. Water Health 2005, 3, 325–337. [Google Scholar] [CrossRef] [PubMed]
  142. Joung, S.-H.; Oh, H.-M.; Ko, S.-R.; Ahn, C.-Y. Correlations between environmental factors and toxic and non-toxic Microcystis dynamics during bloom in Daechung Reservoir, Korea. Harmful Algae 2011, 10, 188–193. [Google Scholar] [CrossRef]
  143. Somdee, T.; Kaewkhiaw, K.; Somdee, A. Detection of toxic cyanobacteria and quantifi cation of microcystins in four recreational water reservoirs in Khon Kaen, Thailand. Asia-Pac. J. Sci. Technol. 2017, 18, 1–8. [Google Scholar]
  144. Trung, B.; Dao, T.-S.; Faassen, E.; Lürling, M.; Trung, B.; Dao, T.-S.; Faassen, E.; Lürling, M. Cyanobacterial Blooms and Microcystins in Southern Vietnam. Toxins 2018, 10, 471. [Google Scholar] [CrossRef] [PubMed]
  145. Te, S.H.; Gin, K.Y.-H. The dynamics of cyanobacteria and microcystin production in a tropical reservoir of Singapore. Harmful Algae 2011, 10, 319–329. [Google Scholar] [CrossRef]
  146. Tavakoli, Y.; Mohammadipanah, F.; Te, S.H.; You, L.; Gin, K.Y.-H. Biodiversity, phylogeny and toxin production profile of cyanobacterial strains isolated from lake Latyan in Iran. Harmful Algae 2021, 106, 102054. [Google Scholar] [CrossRef] [PubMed]
  147. Belykh, O.I.; Sorokovikova, E.G.; Tomberg, I.V.; Fedorova, G.A.; Kuzmin, A.V.; Krasnopeev, A.Y.; Suslova, M.Y.; Potapov, S.A.; Belykh, T.I.; Norovsuren, J.; et al. Water Quality, Toxicity and Diversity of Planktonic and Benthic Cyanobacteria in Pristine Ancient Lake Khubsugul (Hövsgöl), Mongolia. Toxins 2023, 15, 213. [Google Scholar] [CrossRef]
  148. Mohamed, Z.A.; Shehri, A.M.A. Microcystins in groundwater wells and their accumulation in vegetable plants irrigated with contaminated waters in Saudi Arabia. J. Hazard. Mater. 2009, 172, 310–315. [Google Scholar] [CrossRef]
  149. 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]
  150. Al-Tebrineh, J.; Merrick, C.; Ryan, D.; Humpage, A.; Bowling, L.; Neilan, B.A. Community Composition, Toxigenicity, and Environmental Conditions during a Cyanobacterial Bloom Occurring along 1,100 Kilometers of the Murray River. Appl. Environ. Microbiol. 2012, 78, 263–272. [Google Scholar] [CrossRef]
  151. Griffiths, D.J.; Saker, M.L. The Palm Island mystery disease 20 years on: A review of research on the cyanotoxin cylindrospermopsin. Environ. Toxicol. 2003, 18, 78–93. [Google Scholar] [CrossRef]
  152. Ryan, E.F.; Hamilton, D.P.; Barnes, G.E. Recent occurrence of Cylindrospermopsis raciborskii in Waikato lakes of New Zealand. N. Z. J. Mar. Freshw. Res. 2003, 37, 829–836. [Google Scholar] [CrossRef]
  153. Wood, S.A.; Hamilton, D.P.; Paul, W.J.; Safi, K.A.; Williamson, W.M. New Zealand Guidelines for Cyanobacteria in Recreational Fresh Waters: Interim Guidelines; Ministry for the Environment and Ministry of Health: Wellington, New Zealand, 2009. [Google Scholar]
  154. Wood, S.A. Bloom Forming and Toxic Cyanobacteria in New Zealand Species Diversity and Distribution, Cyanotoxin Production and Accumulation of Microcystins in Selected Freshwater Organisms. Ph.D. Thesis, Victoria University of Wellington, Wellington, New Zealand, 2004. [Google Scholar] [CrossRef]
  155. Wood, S.A.; Stirling, D.J. First identification of the cylindrospermopsin-producing cyanobacterium Cylindrospermopsis raciborskii in New Zealand. N. Z. J. Mar. Freshw. Res. 2003, 37, 821–828. [Google Scholar] [CrossRef]
  156. Svirčev, Z.; Krstič, S.; Miladinov-Mikov, M.; Baltič, V.; Vidovič, M. Freshwater Cyanobacterial Blooms and Primary Liver Cancer Epidemiological Studies in Serbia. J. Environ. Sci. Health Part C 2009, 27, 36–55. [Google Scholar] [CrossRef] [PubMed]
  157. Cirés, S.; Wörmer, L.; Carrasco, D.; Quesada, A. Sedimentation Patterns of Toxin-Producing Microcystis Morphospecies in Freshwater Reservoirs. Toxins 2013, 5, 939–957. [Google Scholar] [CrossRef] [PubMed]
  158. Stefanelli, M.; Scardala, S.; Cabras, P.A.; Orrù, A.; Vichi, S.; Testai, E.; Funari, E.; Manganelli, M. Cyanobacterial dynamics and toxins concentrations in Lake Alto Flumendosa, Sardinia, Italy. Adv. Oceanogr. Limnol. 2017, 8, 71–86. [Google Scholar] [CrossRef]
  159. Kabziński, A.K.M.; Juszczak, R.; Miękoś, E.; Tarczynska, E.; Sivonen, K.; Rapala, J. The First Report about the Presence of Cyanobacterial Toxins in Polish Lakes. Pol. J. Environ. Stud. 2000, 9, 171–178. [Google Scholar]
  160. Jurczak, T.; Tarczynska, M.; Karlsson, K.; Meriluoto, J. Characterization and diversity of cyanobacterial hepatotoxins (Microcystins) in blooms from polish freshwaters identified by liquid chromatography-electrospray ionisation mass spectrometry. Chromatographia 2004, 59, 571–578. [Google Scholar] [CrossRef]
  161. Grabowska, M.; Pawlik-Skowrońska, B. Replacement of chroococcales and nostocales by oscillatoriales caused a significant increase in microcystin concentrations in a dam reservoir. Oceanol. Hydrobiol. Stud. 2009, 37, 23–33. [Google Scholar] [CrossRef]
  162. Jančula, D.; Straková, L.; Sadílek, J.; Maršálek, B.; Babica, P. Survey of cyanobacterial toxins in Czech water reservoirs—The first observation of neurotoxic saxitoxins. Environ. Sci. Pollut. Res. 2014, 21, 8006–8015. [Google Scholar] [CrossRef]
  163. Kobos, J.; Błaszczyk, A.; Hohlfeld, N.; Toruńska-Sitarz, A.; Krakowiak, A.; Hebel, A.; Sutryk, K.; Grabowska, M.; Toporowska, M.; Kokociński, M.; et al. Cyanobacteria and cyanotoxins in Polish freshwater bodies. Oceanol. Hydrobiol. Stud. 2014, 42, 358–378. [Google Scholar] [CrossRef]
  164. Czyżewska, W.; Piontek, M.; Łuszczyńska, K. The Occurrence of Potential Harmful Cyanobacteria and Cyanotoxins in the Obrzyca River (Poland), a Source of Drinking Water. Toxins 2020, 12, 284. [Google Scholar] [CrossRef] [PubMed]
  165. Mooney, K.M.; Hamilton, J.T.G.; Floyd, S.D.; Foy, R.H.; Elliott, C.T. Initial studies on the occurrence of cyanobacteria and microcystins in Irish lakes. Environ. Toxicol. 2011, 26, 566–570. [Google Scholar] [CrossRef] [PubMed]
  166. Pekar, H.; Westerberg, E.; Bruno, O.; Lääne, A.; Persson, K.M.; Sundström, L.F.; Thim, A.-M. Fast, rugged and sensitive ultra high pressure liquid chromatography tandem mass spectrometry method for analysis of cyanotoxins in raw water and drinking water—First findings of anatoxins, cylindrospermopsins and microcystin variants in Swedish source waters and infiltration ponds. J. Chromatogr. A 2016, 1429, 265–276. [Google Scholar] [CrossRef] [PubMed]
  167. Sorokovikova, E.; Tikhonova, I.; Naidanova, Y.; Belykh, O. Identification of microcystin producing cyanobacteria in the plankton of Lake Baikal and Irkutsk Reservoir. Limnol. Freshw. Biol. 2024, 8, 1101–1108. [Google Scholar] [CrossRef]
  168. Sysolyatina, M.A.; Sidelev, S.I.; Kutyavina , T.I. Identification of toxin-producing cyanobacteria in water reservoirs of Kirov region and assessment of their relationship with water toxicity to hydrobionts. Limnol. Freshw. Biol. 2024, 8, 1109–1114. [Google Scholar] [CrossRef]
  169. Chernova, E.; Sidelev, S.; Russkikh, I.; Korneva, L.; Solovyova, V.; Mineeva, N.; Stepanova, I.; Zhakovskaya, Z. Spatial distribution of cyanotoxins and ratios of microcystin to biomass indicators in the reservoirs of the Volga, Kama and Don Rivers, the European part of Russia. Limnologica 2020, 84, 125819. [Google Scholar] [CrossRef]
  170. Belykh, O.I.; Gladkikh, S.S.; Sorokovikova, E.G.; Tikhonova, I.V.; Potapov, S.A.; Fedorova, G.A. Microcystin-Producing Cyanobacteria in Water Reservoirs of Russia, Belarus and Ukraine. Chem. Sustain. Dev. 2013, 21, 347–361. [Google Scholar]
  171. Rzymski, P.; Horyn, O.; Budzyńska, A.; Jurczak, T.; Kokociński, M.; Niedzielski, P.; Klimaszyk, P.; Falfushynska, H. A report of Cylindrospermopsis raciborskii and other cyanobacteria in the water reservoirs of power plants in Ukraine. Environ. Sci. Pollut. Res. 2018, 25, 15245–15252. [Google Scholar] [CrossRef]
  172. Lorenzi, A.S.; Cordeiro-Araújo, M.K.; Chia, M.A.; Bittencourt-Oliveira, M.d.C. Cyanotoxin contamination of semiarid drinking water supply reservoirs. Environ. Earth Sci. 2018, 77, 595. [Google Scholar] [CrossRef]
  173. Oliveira, E.D.C.; Castelo-Branco, R.; Silva, L.; Silva, N.; Azevedo, J.; Vasconcelos, V.; Faustino, S.; Cunha, A. First Detection of Microcystin-LR in the Amazon River at the Drinking Water Treatment Plant of the Municipality of Macapá, Brazil. Toxins 2019, 11, 669. [Google Scholar] [CrossRef]
  174. Ruiz, M.; Galanti, L.; Ruibal, A.L.; Rodriguez, M.I.; Wunderlin, D.A.; Amé, M.V. First Report of Microcystins and Anatoxin-a Co-occurrence in San Roque Reservoir (Córdoba, Argentina). Water Air Soil Pollut. 2013, 224, 1–17. [Google Scholar] [CrossRef]
  175. Forastier, M.; Zalocar, Y.; Andrinolo, D.; Domitrovic, H.A. Occurrence and toxicity of Microcystis aeruginosa (Cyanobacteria) in the Paraná River, downstream of the Yacyretá dam (Argentina). Rev. Biol. Tropical 2016, 64, 203–211. [Google Scholar] [CrossRef]
  176. Caly, L.F.; Rodríguez, D.C.; Peñuela, G.A. Monitoring of cyanobacteria and cyanotoxins in a Colombian tropical reservoir. Environ. Sci. Pollut. Res. 2022, 29, 52775–52787. [Google Scholar] [CrossRef] [PubMed]
  177. Gutiérrez, J.E.; Gutiérrez-Hoyos, N.; Benedetti, J.S.G.; Vives, M.J.; Sivasubramanian, V. Clarification of cyanotoxins in El Guajaro Reservoir, Colombia using a microalgae-based consortium MPMC. J. Chem. Technol. Biotechnol. 2022, 97, 1468–1481. [Google Scholar] [CrossRef]
  178. Ballesteros, I.; Cruz, S.D.L.; Rojas, M.; Salazar, G.; Martínez-Fresneda, M.; Castillejo, P. Screening of cyanotoxin producing genes in Ecuadorian freshwater systems. Acta Limnol. Bras. 2022, 34, e24. [Google Scholar] [CrossRef]
  179. Cuellar-Martinez, T.; Ruiz-Fernández, A.C.; Alonso-Hernández, C.; Amaya-Monterrosa, O.; Quintanilla, R.; Carrillo-Ovalle, H.L.; Arbeláez, M.N.; Díaz-Asencio, L.; Méndez, S.M.; Vargas, M.; et al. Addressing the Problem of Harmful Algal Blooms in Latin America and the Caribbean- A Regional Network for Early Warning and Response. Front. Mar. Sci. 2018, 5, 409. [Google Scholar] [CrossRef]
  180. López-Cortés, D.J.; Núñez Vázquez, E.J.; Dorantes-Aranda, J.J.; Band-Schmidt, C.J.; Hernández-Sandoval, F.E.; Bustillos-Guzmán, J.J.; Leyva-Valencia, I.; Fernández-Herrera, L.J. The State of Knowledge of Harmful Algal Blooms of Margalefidinium polykrikoides (a.k.a. Cochlodinium polykrikoides) in Latin America. Front. Mar. Sci. 2019, 6, 463. [Google Scholar] [CrossRef]
  181. Sunesen, I.; Méndez, S.M.; Mancera-Pineda, J.E.; Bottein, M.-Y.D.; Enevoldsen, H. The Latin America and Caribbean HAB status report based on OBIS and HAEDAT maps and databases. Harmful Algae 2021, 102, 101920. [Google Scholar] [CrossRef]
  182. Garita-Alvarado, C.A.; Bojorge-García, M.G.; Cantoral-Uriza, E.A. Cyanotoxins bioaccumulation in freshwater ecosystems in Latin America: A review. Hidrobiológica 2023, 33, 353–365. [Google Scholar] [CrossRef]
  183. Aguilera, A.; Almanza, V.; Haakonsson, S.; Palacio, H.; Rodas, G.A.B.; Barros, M.U.G.; Capelo-Neto, J.; Urrutia, R.; Aubriot, L.; Bonilla, S. Cyanobacterial bloom monitoring and assessment in Latin America. Harmful Algae 2023, 125, 102429. [Google Scholar] [CrossRef]
  184. Zamyadi, A.; McQuaid, N.; Prévost, M.; Dorner, S. Monitoring of potentially toxic cyanobacteria using an online multi-probe in drinking water sources. J. Environ. Monit. 2012, 14, 579–588. [Google Scholar] [CrossRef] [PubMed]
  185. Miller, T.R.; Beversdorf, L.J.; Weirich, C.A.; Bartlett, S.L. Cyanobacterial Toxins of the Laurentian Great Lakes, Their Toxicological Effects, and Numerical Limits in Drinking Water. Mar. Drugs 2017, 15, 160. [Google Scholar] [CrossRef]
  186. US. EPA. The Great Lakes: Lake Erie; United States Environmental Protection Agency: Washington, DC, USA, 2023.
  187. Steffen, M.M.; Davis, T.W.; McKay, R.M.L.; Bullerjahn, G.S.; Krausfeldt, L.E.; Stough, J.M.A.; Neitzey, M.L.; Gilbert, N.E.; Boyer, G.L.; Johengen, T.H.; et al. Ecophysiological Examination of the Lake Erie Microcystis Bloom in 2014: Linkages between Biology and the Water Supply Shutdown of Toledo, OH. Environ. Sci. Technol. 2017, 51, 6745–6755. [Google Scholar] [CrossRef]
  188. Mercado Borrayo, B.M. Estudio sobre la remoción de cianobacterias y sus metabolitos en la Planta Potabilizadora “Los Berros” Sistema Cutzamala. Ph.D. Thesis, Universidad Nacional Autónoma de México: Mexico City, Mexico, 2007. [Google Scholar]
  189. Kim, H.R.; Kim, C.K.; Ahn, T.S.; Yoo, S.A.; Lee, D.H. Effects of Temperature and Light on Microcystin Synthetase Gene Transcription in Microcystis Aeruginosa. Key Eng. Mater. 2005, 277–279, 606–611. [Google Scholar] [CrossRef]
  190. Sevilla, E.; Martin-Luna, B.; Bes, M.T.; Fillat, M.F.; Peleato, M.L. An active photosynthetic electron transfer chain required for mcyD transcription and microcystin synthesis in Microcystis aeruginosa PCC7806. Ecotoxicology 2011, 21, 811–819. [Google Scholar] [CrossRef] [PubMed]
  191. Kaplan, A.; Harel, M.; Kaplan-Levy, R.N.; Hadas, O.; Sukenik, A.; Dittmann, E. Frontiers | The Languages Spoken in the Water Body (or the Biological Role of Cyanobacterial Toxins). Front. Microbiol. 2012, 3, 138. [Google Scholar] [CrossRef] [PubMed]
  192. Dai, R.; Wang, P.; Jia, P.; Zhang, Y.; Chu, X.; Wang, Y. A review on factors affecting microcystins production by algae in aquatic environments. World J. Microbiol. Biotechnol. 2016, 32, 51. [Google Scholar] [CrossRef] [PubMed]
  193. Mohanty, B.; Majedi, S.M.; Pavagadhi, S.; Te, S.H.; Boo, C.Y.; Gin, K.Y.-H.; Swarup, S. Effects of Light and Temperature on the Metabolic Profiling of Two Habitat-Dependent Bloom-Forming Cyanobacteria. Metabolites 2022, 12, 406. [Google Scholar] [CrossRef]
  194. Hobson, P.; Fallowfield, H.J. Effect of irradiance, temperature and salinity on growth and toxin production by Nodularia spumigena. Hydrobiologia 2003, 493, 7–15. [Google Scholar] [CrossRef]
  195. Bui, T.; Dao, T.-S.; Vo, T.-G.; Lürling, M. Warming Affects Growth Rates and Microcystin Production in Tropical Bloom-Forming Microcystis Strains. Toxins 2018, 10, 123. [Google Scholar] [CrossRef]
  196. Stark, G.F.; Martin, R.M.; Smith, L.E.; Wei, B.; Hellweger, F.L.; Bullerjahn, G.S.; McKay, R.M.L.; Boyer, G.L.; Wilhelm, S.W. Microcystin aids in cold temperature acclimation: Differences between a toxic Microcystis wildtype and non-toxic mutant. Harmful Algae 2023, 129, 102531. [Google Scholar] [CrossRef] [PubMed]
  197. Wejnerowski, Ł.; Dulić, T.; Akter, S.; Font-Nájera, A.; Rybak, M.; Kamiński, O.; Czerepska, A.; Dziuba, M.K.; Jurczak, T.; Meriluoto, J.; et al. Community Structure and Toxicity Potential of Cyanobacteria during Summer and Winter in a Temperate-Zone Lake Susceptible to Phytoplankton Blooms. Toxins 2024, 16, 357. [Google Scholar] [CrossRef] [PubMed]
  198. Wejnerowski, Ł.; Rzymski, P.; Kokociński, M.; Meriluoto, J. The structure and toxicity of winter cyanobacterial bloom in a eutrophic lake of the temperate zone. Ecotoxicology 2018, 27, 752–760. [Google Scholar] [CrossRef]
  199. Wang, Q.; Niu, Y.; Xie, P.; Chen, J.; Ma, Z.; Tao, M.; Qi, M.; Wu, L.; Guo, L. Factors Affecting Temporal and Spatial Variations of Microcystins in Gonghu Bay of Lake Taihu, with Potential Risk of Microcystin Contamination to Human Health. Sci. World J. 2010, 10, 1795–1809. [Google Scholar] [CrossRef] [PubMed]
  200. 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] [PubMed]
  201. Pimentel, J.S.M.; Giani, A. Microcystin Production and Regulation under Nutrient Stress Conditions in Toxic Microcystis Strains. Appl. Environ. Microbiol. 2014, 80, 5836–5843. [Google Scholar] [CrossRef] [PubMed]
  202. Brookes, J.D.; Carey, C.C. Resilience to Blooms. Science 2011, 334, 46–47. [Google Scholar] [CrossRef] [PubMed]
  203. MacKeigan, P.W.; Zastepa, A.; Taranu, Z.E.; Westrick, J.A.; Liang, A.; Pick, F.R.; Beisner, B.E.; Gregory-Eaves, I. Microcystin concentrations and congener composition in relation to environmental variables across 440 north-temperate and boreal lakes. Sci. Total Environ. 2023, 884, 163811. [Google Scholar] [CrossRef] [PubMed]
  204. Jablonska, M.; Cerasino, L.; Boscaini, A.; Capelli, C.; Greco, C.; Klemenčič, A.K.; Mischke, U.; Salmaso, N.; Kurmayer, R. Distribution of toxigenic cyanobacteria in Alpine lakes and rivers as revealed by molecular screening. Water Res. 2024, 258, 121783. [Google Scholar] [CrossRef]
  205. Li, X.; Tikhonova, I.V.; Potapov, S.A.; Krasnopeev, A.Y.; Zhuchenko, N.A.; Niao, X.; Wang, L.; Sorokovikova, E.G.; Wang, W.; Belykh, O.I. World’s largest oligotrophic Lake Baikal: Concerns about cyanobacterial blooms and potential microcystin producers along the littoral zone. Harmful Algae 2025, 144, 102841. [Google Scholar] [CrossRef]
  206. Reinl, K.L.; Sterner, R.W.; Lafrancois, B.M.; Brovold, S. Fluvial seeding of cyanobacterial blooms in oligotrophic Lake Superior. Harmful Algae 2020, 100, 101941. [Google Scholar] [CrossRef]
  207. Cizmas, L.; Sharma, V.K.; Gray, C.M.; McDonald, T.J. Pharmaceuticals and personal care products in waters: Occurrence, toxicity, and risk. Environ. Chem. Lett. 2015, 13, 381–394. [Google Scholar] [CrossRef]
  208. Tijani, J.O.; Fatoba, O.O.; Petrik, L.F. A Review of Pharmaceuticals and Endocrine-Disrupting Compounds: Sources, Effects, Removal, and Detections. Water Air Soil Pollut. 2013, 224, 1770. [Google Scholar] [CrossRef]
  209. Edwards, C.; Lawton, L.A. Chapter 4 Bioremediation of Cyanotoxins. Adv. Appl. Microbiol. 2009, 67, 109–129. [Google Scholar] [CrossRef] [PubMed]
  210. Dziga, D.; Nybom, S.; Gagala, I.; Wasylewski, M. Biological Treatment for the Destruction of Cyanotoxins. In Water Treatment for Purification from Cyanobacteria and Cyanotoxins; Hiskia, A.E., Triantis, T.M., Antoniou, M.G., Kaloudis, T., Dionysiou, D.D., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2020; pp. 117–153. [Google Scholar]
  211. Ho, L.; Hoefel, D.; Saint, C.P.; Newcombe, G. Isolation and identification of a novel microcystin-degrading bacterium from a biological sand filter. Water Res. 2007, 41, 4685–4695. [Google Scholar] [CrossRef] [PubMed]
  212. Mohamed, Z.A.; Hashem, M.; Alamri, S.; Campos, A.; Vasconcelos, V. Fungal biodegradation and removal of cyanobacteria and microcystins: Potential applications and research needs. Environ. Sci. Pollut. Res. 2021, 28, 37041–37050. [Google Scholar] [CrossRef] [PubMed]
  213. Boopathi, T.; Ki, J.-S. Impact of Environmental Factors on the Regulation of Cyanotoxin Production. Toxins 2014, 6, 1951–1978. [Google Scholar] [CrossRef]
  214. Dalu, T.; Wasserman, R.J. Cyanobacteria dynamics in a small tropical reservoir: Understanding spatio-temporal variability and influence of environmental variables. Sci. Total Environ. 2018, 643, 835–841. [Google Scholar] [CrossRef]
  215. Martínez-Jerónimo, F.; Antuna-González, P.d.C.; Hernández-Zamora, M.; Martínez-Jerónimo, L.; Munoz, G.; Simon, D.F.; Sauvé, S. Year-long monitoring of phytoplankton community, toxigenic cyanobacteria, and total microcystins in a eutrophic tropical dam supplying the Mexico megacity. Front. Environ. Sci. 2022, 10, 984365. [Google Scholar] [CrossRef]
  216. Peng, L.; Tang, Q.; Gu, J.; Lei, L.; Chen, W.; Song, L. Seasonal variation of microcystins and their accumulation in fish in two large shallow lakes of China. Ecotoxicology 2020, 29, 790–800. [Google Scholar] [CrossRef]
  217. Filatova, D.; Picardo, M.; Núñez, O.; Farré, M. Analysis, levels and seasonal variation of cyanotoxins in freshwater ecosystems. Trends Environ. Anal. Chem. 2020, 26, e00091. [Google Scholar] [CrossRef]
  218. Melo, D.S.; Gontijo, E.S.J.; Frascareli, D.; Simonetti, V.C.; Machado, L.S.; Barth, J.A.C.; Moschini-Carlos, V.; Pompêo, M.L.; Rosa, A.H.; Friese, K. Self-Organizing Maps for Evaluation of Biogeochemical Processes and Temporal Variations in Water Quality of Subtropical Reservoirs. Water Resour. Res. 2019, 55, 10268–10281. [Google Scholar] [CrossRef]
  219. Visser, P.M.; Verspagen, J.M.H.; Sandrini, G.; Stal, L.J.; Matthijs, H.C.P.; Davis, T.W.; Paerl, H.W.; Huisman, J. How rising CO2 and global warming may stimulate harmful cyanobacterial blooms. Harmful Algae 2016, 54, 145–159. [Google Scholar] [CrossRef] [PubMed]
  220. dos Santos Machado, L.; Dörr, F.; Dörr, F.A.; Frascareli, D.; Melo, D.S.; Gontijo, E.S.J.; Friese, K.; Pinto, E.; Rosa, A.H.; Pompêo, M.M.; et al. Permanent occurrence of Raphidiopsis raciborskii and cyanotoxins in a subtropical reservoir polluted by domestic effluents (Itupararanga reservoir, São Paulo, Brazil). Environ. Sci. Pollut. Res. 2021, 29, 18653–18664. [Google Scholar] [CrossRef] [PubMed]
  221. Vione, D.; Rosario-Ortiz, F.L. Foreseen Effects of Climate-Impacted Scenarios on the Photochemical Fate of Selected Cyanotoxins in Surface Freshwaters. Environ. Sci. Technol. 2021, 55, 10928–10934. [Google Scholar] [CrossRef] [PubMed]
  222. Manning, S.R.; Nobles, D.R. Impact of global warming on water toxicity: Cyanotoxins. Curr. Opin. Food Sci. 2017, 18, 14–20. [Google Scholar] [CrossRef]
  223. Zepernick, B.N.; Wilhelm, S.W.; Bullerjahn, G.S.; Paerl, H.W. Climate change and the aquatic continuum: A cyanobacterial comeback story. Environ. Microbiol. Rep. 2023, 15, 3–12. [Google Scholar] [CrossRef]
  224. Mantzouki, E.; Lürling, M.; Fastner, J.; Domis, L.D.S.; Wilk-Woźniak, E.; Koreivienė, J.; Seelen, L.; Teurlincx, S.; Verstijnen, Y.; Krztoń, W.; et al. Temperature Effects Explain Continental Scale Distribution of Cyanobacterial Toxins. Toxins 2018, 10, 156. [Google Scholar] [CrossRef] [PubMed]
  225. Van de Waal, D.B.; Gsell, A.S.; Harris, T.; Paerl, H.W.; de Senerpont Domis, L.N.; Huisman, J. Hot summers raise public awareness of toxic cyanobacterial blooms. Water Res. 2024, 249, 120817. [Google Scholar] [CrossRef]
  226. Paerl, H.W.; Barnard, M.A. Mitigating the global expansion of harmful cyanobacterial blooms: Moving targets in a human- and climatically-altered world. Harmful Algae 2020, 96, 101845. [Google Scholar] [CrossRef]
  227. Free, G.; Bresciani, M.; Pinardi, M.; Peters, S.; Laanen, M.; Padula, R.; Cingolani, A.; Charavgis, F.; Giardino, C. Shorter blooms expected with longer warm periods under climate change: An example from a shallow meso-eutrophic Mediterranean lake. Hydrobiologia 2022, 849, 3963–3978. [Google Scholar] [CrossRef]
  228. Chorus, I.; Fastner, J.; Welker, M. Cyanobacteria and Cyanotoxins in a Changing Environment: Concepts, Controversies, Challenges. Water 2021, 13, 2463. [Google Scholar] [CrossRef]
  229. Amórtegui, J.C.E.; Pekar, H.; Retrato, M.D.C.; Persson, M.; Karlson, B.; Bergquist, J.; Zuberovic-Muratovic, A.; España Amórtegui, J.C.; Pekar, H.; Retrato, M.D.C.; et al. LC-MS/MS Analysis of Cyanotoxins in Bivalve Mollusks—Method Development, Validation and First Evidence of Occurrence of Nodularin in Mussels (Mytilus edulis) and Oysters (Magallana gigas) from the West Coast of Sweden. Toxins 2023, 15, 329. [Google Scholar] [CrossRef] [PubMed]
  230. Almuhtaram, H.; Kibuye, F.A.; Ajjampur, S.; Glover, C.M.; Hofmann, R.; Gaget, V.; Owen, C.; Wert, E.C.; Zamyadi, A. State of knowledge on early warning tools for cyanobacteria detection. Ecol. Indic. 2021, 133, 108442. [Google Scholar] [CrossRef]
  231. Oberholster, P.J.; Botha, A.-M.; Cloete, T.E. Toxic cyanobacterial blooms in a shallow, artificially mixed urban lake in Colorado, USA. Lakes Reserv. Res. Manag. 2006, 11, 111–123. [Google Scholar] [CrossRef]
  232. Vogiazi, V.; Cruz, A.d.l.; Mishra, S.; Shanov, V.; Heineman, W.R.; Dionysiou, D.D. A Comprehensive Review: Development of Electrochemical Biosensors for Detection of Cyanotoxins in Freshwater. ACS Sens. 2019, 4, 1151–1173. [Google Scholar] [CrossRef] [PubMed]
  233. Massey, I.Y.; Yang, F.; Ding, Z.; Yang, S.; Guo, J.; Tezi, C.; Al-Osman, M.; Kamegni, R.B.; Zeng, W. Exposure routes and health effects of microcystins on animals and humans: A mini-review. Toxicon 2018, 151, 156–162. [Google Scholar] [CrossRef]
  234. Haande, S.; Ballot, A.; Rohrlack, T.; Fastner, J.; Wiedner, C.; Edvardsen, B. Diversity of Microcystis aeruginosa isolates (Chroococcales, Cyanobacteria) from East-African water bodies. Arch. Microbiol. 2007, 188, 15–25. [Google Scholar] [CrossRef]
  235. Bertani, P.; Lu, W. Cyanobacterial toxin biosensors for environmental monitoring and protection. Med. Nov. Technol. Devices 2021, 10, 100059. [Google Scholar] [CrossRef]
  236. Shi, Y.; Guo, C.; Sun, Y.; Liu, Z.; Xu, F.; Zhang, Y.; Wen, Z.; Li, Z. Interaction between DNA and Microcystin-LR Studied by Spectra Analysis and Atomic Force Microscopy. Biomacromolecules 2011, 12, 797–803. [Google Scholar] [CrossRef]
  237. Bilibana, M.P.; Williams, A.R.; Rassie, C.; Sunday, C.E.; Makelane, H.; Wilson, L.; Ntshongontshi, N.; Jijana, A.N.; Masikini, M.; Baker, P.G.L.; et al. Electrochemical Aptatoxisensor Responses on Nanocomposites Containing Electro-Deposited Silver Nanoparticles on Poly(Propyleneimine) Dendrimer for the Detection of Microcystin-LR in Freshwater. Sensors 2016, 16, 1901. [Google Scholar] [CrossRef] [PubMed]
  238. McLellan, N.L.; Manderville, R.A. Toxic mechanisms of microcystins in mammals. Toxicol. Res. 2017, 6, 391–405. [Google Scholar] [CrossRef] [PubMed]
  239. 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]
  240. Svirčev, Z.; Chen, L.; Sántha, K.; Drobac Backović, D.; Šušak, S.; Vulin, A.; Palanački Malešević, T.; Codd, G.A.; Meriluoto, J.; Svirčev, Z.; et al. A review and assessment of cyanobacterial toxins as cardiovascular health hazards. Arch. Toxicol. 2022, 96, 2829–2863. [Google Scholar] [CrossRef] [PubMed]
  241. Kumari, M.; Gupta, S.K. Occurrence and Exposure to Trihalomethanes in Drinking Water: A Systematic Review and Meta-analysis. Expo. Health 2022, 14, 915–939. [Google Scholar] [CrossRef]
  242. Mutoti, M.I.; Edokpayi, J.N.; Mutileni, N.; Durowoju, O.S.; Munyai, F.L. Cyanotoxins in groundwater; occurrence, potential sources, health impacts and knowledge gap for public health. Toxicon 2023, 226, 107077. [Google Scholar] [CrossRef]
  243. Drobac, D.; Tokodi, N.; Simeunović, J.; Baltić, V.; Stanić, D.; Svirčev, Z. Human Exposure to Cyanotoxins and their Effects on Health. Arch. Ind. Hyg. Toxicol. 2013, 64, 305–315. [Google Scholar] [CrossRef]
  244. Annadotter, H.; Cronberg, G.; Lawton, L.; Hansson, H.-B.; Göthe, J.; Skulberg, O.M.; Cronberg, G.; Lawton, L.; Hansson, H.B.; Skulberg, O. An extensive outbreak of gastroenteritis associated with the toxic cyanobacterium (Oscillatoriales, Cyanophyceae) in Scania, South Sweden. In Cyanotoxins—Occurrence, Causes, Consequences; Chorus, I., Ed.; Springer: Berlin, Germany, 2001; pp. 200–208. [Google Scholar]
  245. Svirčev, Z.; Drobac, D.; Tokodi, N.; Mijović, B.; Codd, G.A.; Meriluoto, J. Toxicology of microcystins with reference to cases of human intoxications and epidemiological investigations of exposures to cyanobacteria and cyanotoxins. Arch. Toxicol. 2017, 91, 621–650. [Google Scholar] [CrossRef]
  246. Ohtani, I.; Moore, R.E.; Runnegar, M.T.C. Cylindrospermopsin: A potent hepatotoxin from the blue-green alga Cylindrospermopsis raciborskii. J. Am. Chem. Soc. 2002, 114, 7941–7942. [Google Scholar] [CrossRef]
  247. Hoeger, S.J.; Hitzfeld, B.C.; Dietrich, D.R. Occurrence and elimination of cyanobacterial toxins in drinking water treatment plants. Toxicol. Appl. Pharmacol. 2005, 203, 231–242. [Google Scholar] [CrossRef]
  248. Nielsen, M.C.; Jiang, S.C. Can cyanotoxins penetrate human skin during water recreation to cause negative health effects? Harmful Algae 2020, 98, 101872. [Google Scholar] [CrossRef] [PubMed]
  249. Osborne, N.J.; Shaw, G.R.; Webb, P.M. Health effects of recreational exposure to Moreton Bay, Australia waters during a Lyngbya majuscula bloom. Environ. Int. 2007, 33, 309–314. [Google Scholar] [CrossRef] [PubMed]
  250. Niamien-Ebrottie, J.E.; Bhattacharyya, S.; Deep, P.R.; Nayak, B. Cyanobacteria and cyanotoxins in the World: Review. Int. J. Appl. Res. Vet. Med. 2015, 1, 563–569. [Google Scholar]
  251. Azevedo, S.M.F.O.; Carmichael, W.W.; Jochimsen, E.M.; Rinehart, K.L.; Lau, S.; Shaw, G.R.; Eaglesham, G.K. Human intoxication by microcystins during renal dialysis treatment in Caruaru—Brazil. Toxicology 2002, 181–182, 441–446. [Google Scholar] [CrossRef] [PubMed]
  252. Pouria, S.; Andrade, A.d.; Barbosa, J.; Cavalcanti, R.; Barreto, V.; Ward, C.; Preiser, W.; Poon, G.K.; Neild, G.; Codd, G. Fatal microcystin intoxication in haemodialysis unit in Caruaru, Brazil. Lancet 1998, 352, 21–26. [Google Scholar] [CrossRef] [PubMed]
  253. 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.; et al. 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]
  254. 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]
  255. Wiśniewska, K.; Śliwińska-Wilczewska, S.; Savoie, M.; Lewandowska, A.U. Quantitative and qualitative variability of airborne cyanobacteria and microalgae and their toxins in the coastal zone of the Baltic Sea. Sci. Total Environ. 2022, 826, 154152. [Google Scholar] [CrossRef] [PubMed]
  256. Labohá, P.; Sychrová, E.; Brózman, O.; Sovadinová, I.; Bláhová, L.; Prokeš, R.; Ondráček, J.; Babica, P. Cyanobacteria, cyanotoxins and lipopolysaccharides in aerosols from inland freshwater bodies and their effects on human bronchial cells. Environ. Toxicol. Pharmacol. 2023, 98, 104073. [Google Scholar] [CrossRef]
  257. Murby, A.L.; Haney, J.F. Field and laboratory methods to monitor lake aerosols for cyanobacteria and microcystins. Aerobiologia 2015, 32, 395–403. [Google Scholar] [CrossRef]
  258. 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] [PubMed]
  259. Picanço, M.R.; Soares, R.M.; Cagido, V.R.; Azevedo, S.M.F.O.; Rocco, P.R.M.; Zin, W.A. Toxicity of a cyanobacterial extract containing microcystins to mouse lungs. Braz. J. Med. Biol. Res. 2004, 37, 1225–1229. [Google Scholar] [CrossRef] [PubMed]
  260. Soares, R.M.; Cagido, V.R.; Ferraro, R.B.; Meyer-Fernandes, J.R.; Rocco, P.R.M.; Zin, W.A.; Azevedo, S.M.F.O. Effects of microcystin-LR on mouse lungs. Toxicon 2007, 50, 330–338. [Google Scholar] [CrossRef] [PubMed]
  261. Carmichael, W.W.; Boyer, G.L. Health impacts from cyanobacteria harmful algae blooms: Implications for the North American Great Lakes. Harmful Algae 2016, 54, 194–212. [Google Scholar] [CrossRef] [PubMed]
  262. Rzymski, P.; Poniedziałek, B.; Karczewski, J. Gastroenteritis and liver carcinogenesis induced by cyanobacterial toxins. Gastroenterol. Pol. 2011, 18, 159–162. [Google Scholar]
  263. Zilberg, B. Gastroenteritis in Salisbury European children—A five-year study. Cent. Afr. J. Med. 1996, 12, 164–168. [Google Scholar]
  264. Wu, J.-X.; Huang, H.; Yang, L.; Zhang, X.-F.; Zhang, S.-S.; Liu, H.-H.; Wang, Y.-Q.; Yuan, L.; Cheng, X.-M.; Zhuang, D.-G.; et al. Gastrointestinal toxicity induced by microcystins. World J. Clin. Cases 2018, 6, 344–354. [Google Scholar] [CrossRef]
  265. Backer, L.C.; McNeel, S.V.; Barber, T.; Kirkpatrick, B.; Williams, C.; Irvin, M.; Zhou, Y.; Johnson, T.B.; Nierenberg, K.; Aubel, M.; et al. Recreational exposure to microcystins during algal blooms in two California lakes. Toxicon 2010, 55, 909–921. [Google Scholar] [CrossRef]
  266. Stewart, I.; Webb, P.M.; Schluter, P.J.; Fleming, L.E.; Burns, J.W.; Gantar, M.; Backer, L.C.; Shaw, G.R. Epidemiology of recreational exposure to freshwater cyanobacteria—An international prospective cohort study. BMC Public Health 2006, 6, 93. [Google Scholar] [CrossRef]
  267. Zheng, C.; Zeng, H.; Lin, H.; Wang, J.; Feng, X.; Qiu, Z.; Chen, J.a.; Luo, J.; Luo, Y.; Huang, Y.; et al. Serum microcystin levels positively linked with risk of hepatocellular carcinoma: A case-control study in southwest China. Hepatology 2017, 66, 1519–1528. [Google Scholar] [CrossRef]
  268. IARC. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Ingested Nitrate and Nitrite, and Cyanobacterial Peptide toxins; International Agency for Research on Cancer: Lyon, France, 2010. [Google Scholar]
  269. Gu, S.; Jiang, M.; Zhang, B. Microcystin-LR in Primary Liver Cancers: An Overview. Toxins 2022, 14, 715. [Google Scholar] [CrossRef] [PubMed]
  270. Malta, J.F.; Nardocci, A.C.; Razzolini, M.T.P.; Diniz, V.; Cunha, D.G.F. Exposure to microcystin-LR in tropical reservoirs for water supply poses high risks for children and adults. Environ. Monit. Assess. 2022, 194, 253. [Google Scholar] [CrossRef] [PubMed]
  271. Zanchett, G.; Oliveira-Filho, E.C. Cyanobacteria and Cyanotoxins: From Impacts on Aquatic Ecosystems and Human Health to Anticarcinogenic Effects. Toxins 2013, 5, 1896–1917. [Google Scholar] [CrossRef] [PubMed]
  272. Mulvenna, V.; Dale, K.; Priestly, B.; Mueller, U.; Humpage, A.; Shaw, G.; Allinson, G.; Falconer, I. Health Risk Assessment for Cyanobacterial Toxins in Seafood. Int. J. Environ. Res. Public Health 2012, 9, 807–820. [Google Scholar] [CrossRef]
  273. Arman, T.; Clarke, J.D. Microcystin Toxicokinetics, Molecular Toxicology, and Pathophysiology in Preclinical Rodent Models and Humans. Toxins 2021, 13, 537. [Google Scholar] [CrossRef]
  274. 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]
  275. Greer, B.; Maul, R.; Campbell, K.; Elliott, C.T. Detection of freshwater cyanotoxins and measurement of masked microcystins in tilapia from Southeast Asian aquaculture farms. Anal. Bioanal. Chem. 2017, 409, 4057–4069. [Google Scholar] [CrossRef]
  276. Merwe, D.v.d.; Sebbag, L.; Nietfeld, J.C.; Aubel, M.T.; Foss, A.; Carney, E. Investigation of a Microcystis aeruginosa cyanobacterial freshwater harmful algal bloom associated with acute microcystin toxicosis in a dog. J. Vet. Diagn. Investig. 2012, 24, 679–687. [Google Scholar] [CrossRef] [PubMed]
  277. Gorham, T.; Root, E.D.; Jia, Y.; Shum, C.K.; Lee, J. Relationship between cyanobacterial bloom impacted drinking water sources and hepatocellular carcinoma incidence rates. Harmful Algae 2020, 95, 101801. [Google Scholar] [CrossRef]
  278. Dietrich, D.R.; Fischer, A.; Michel, C.; Hoeger, S. Toxin mixture in cyanobacterial blooms—A critical comparison o. In Cyanobacterial Harmful Algal Blooms: State of the Science and Research Needs; Hudnell, K.H., Ed.; Springer: New York, NY, USA, 2008; pp. 885–912. [Google Scholar]
  279. Henri, J.; Huguet, A.; Delmas, J.-M.; Besson, A.; Sanders, P.; Fessard, V. Low in vitro permeability of the cyanotoxin microcystin-LR across a Caco-2 monolayer: With identification of the limiting factors using modelling. Toxicon 2014, 91, 5–14. [Google Scholar] [CrossRef]
  280. Henri, J.; Lanceleur, R.; Delmas, J.-M.; Fessard, V.; Huguet, A. Permeability of the Cyanotoxin Microcystin-RR across a Caco-2 Cells Monolayer. Toxins 2021, 13, 178. [Google Scholar] [CrossRef] [PubMed]
  281. Nishiwaki, R.; Ohta, T.; Sueoka, E.; Suganuma, M.; Harada, K.-I.; Watanabe, M.F.; Fujiki, H. Two significant aspects of microcystin-LR: Specific binding and liver specificity. Cancer Lett. 1994, 83, 283–289. [Google Scholar] [CrossRef] [PubMed]
  282. Kemp, A.; John, J. Microcystins associated with Microcystis dominated blooms in the Southwest wetlands, Western Australia. Environ. Toxicol. 2006, 21, 125–130. [Google Scholar] [CrossRef] [PubMed]
  283. Graham, J.L.; Loftin, K.A.; Meyer, M.T.; Ziegler, A.C. Cyanotoxin Mixtures and Taste-and-Odor Compounds in Cyanobacterial Blooms from the Midwestern United States. Environ. Sci. Technol. 2010, 44, 7361–7368. [Google Scholar] [CrossRef]
  284. 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]
  285. Sabart, M.; Pobel, D.; Briand, E.; Combourieu, B.; Salençon, M.J.; Humbert, J.F.; Latour, D. Spatiotemporal Variations in Microcystin Concentrations and in the Proportions of Microcystin-Producing Cells in Several Microcystis aeruginosa Populations. Appl. Environ. Microbiol. 2010, 76, 4750–4759. [Google Scholar] [CrossRef]
  286. Srivastava, A.; Choi, G.-G.; Ahn, C.-Y.; Oh, H.-M.; Ravi, A.K.; Asthana, R.K. Dynamics of microcystin production and quantification of potentially toxigenic Microcystis sp. using real-time PCR. Water Res. 2012, 46, 817–827. [Google Scholar] [CrossRef]
  287. Kotak, B.G.; Zurawell, R.W. Cyanobacterial toxins in Canadian freshwaters: A review. Lake Reserv. Manag. 2007, 23, 109–122. [Google Scholar] [CrossRef]
  288. Watson, S.B.; Zastepa, A.; Boyer, G.L.; Matthews, E. Algal bloom response and risk management: On-site response tools. Toxicon 2017, 129, 144–152. [Google Scholar] [CrossRef]
  289. Boyer, G.L. The occurrence of cyanobacterial toxins in New York lakes: Lessons from the MERHAB-Lower Great Lakes program. Lake Reserv. Manag. 2007, 23, 153–160. [Google Scholar] [CrossRef]
  290. Jacoby, J.M.; Kann, J. The occurrence and response to toxic cyanobacteria in the Pacific Northwest, North America. Lake and Reservoir Management 2007, 23, 123–143. [Google Scholar] [CrossRef]
  291. Wang, Q.; Xie, P.; Chen, J.; Liang, G. Distribution of microcystins in various organs (heart, liver, intestine, gonad, brain, kidney and lung) of Wistar rat via intravenous injection. Toxicon 2008, 52, 721–727. [Google Scholar] [CrossRef] [PubMed]
  292. Robinson, N.A.; Pace, J.G.; Matson, C.F.; Miura, G.A.; Lawrence, W.B. Tissue distribution, excretion and hepatic biotransformation of microcystin-LR in mice. J. Pharmacol. Exp. Ther. 1991, 256, 176–178. [Google Scholar] [CrossRef] [PubMed]
  293. Falconer, I.; Buckley, T.; Runnegar, M. Biological half-life, organ distribution and excretion of 125-I-labelled toxic peptide from the blue-green alga Microcystis aeruginosa. Aust. J. Biol. Sci. 1986, 39, 17–21. [Google Scholar] [CrossRef]
  294. Clarke, J.D.; Dzierlenga, A.; Arman, T.; Toth, E.; Li, H.; Lynch, K.D.; Tian, D.-D.; Goedken, M.; Paine, M.F.; Cherrington, N. Nonalcoholic fatty liver disease alters microcystin-LR toxicokinetics and acute toxicity. Toxicon 2019, 162, 1–8. [Google Scholar] [CrossRef] [PubMed]
  295. Guo, X.; Chen, L.; Chen, J.; Xie, P.; Li, S.; He, J.; Li, W.; Fan, H.; Yu, D.; Zeng, C. Quantitatively evaluating detoxification of the hepatotoxic microcystin-LR through the glutathione (GSH) pathway in SD rats. Environ. Sci. Pollut. Res. 2015, 22, 19273–19284. [Google Scholar] [CrossRef]
  296. Fischer, W.J.; Altheimer, S.; Cattori, V.; Meier, P.J.; Dietrich, D.R.; Hagenbuch, B. Organic anion transporting polypeptides expressed in liver and brain mediate uptake of microcystin. Toxicol. Appl. Pharmacol. 2005, 203, 257–263. [Google Scholar] [CrossRef] [PubMed]
  297. Ma, X.; Shang, X.; Qin, X.; Lu, J.; Liu, M.; Wang, X. Characterization of organic anion transporting polypeptide 1b2 knockout rats generated by CRISPR/Cas9: A novel model for drug transport and hyperbilirubinemia disease. Acta Pharm. Sin. B 2020, 10, 850–860. [Google Scholar] [CrossRef]
  298. Kaur, G.; Fahrner, R.; Wittmann, V.; Stieger, B.; Dietrich, D.R. Human MRP2 exports MC-LR but not the glutathione conjugate. Chem.-Biol. Interact. 2019, 311, 108761. [Google Scholar] [CrossRef]
  299. Zhang, W.; Liang, G.; Wu, L.; Tuo, X.; Wang, W.; Chen, J.; Xie, P. Why mammals more susceptible to the hepatotoxic microcystins than fish: Evidences from plasma and albumin protein binding through equilibrium dialysis. Ecotoxicology 2013, 22, 1012–1019. [Google Scholar] [CrossRef]
  300. Feurstein, D.; Holst, K.; Fischer, A.; Dietrich, D.R. Oatp-associated uptake and toxicity of microcystins in primary murine whole brain cells. Toxicol. Appl. Pharmacol. 2009, 234, 247–255. [Google Scholar] [CrossRef] [PubMed]
  301. Schmidt, J.R.; Wilhelm, S.W.; Boyer, G.L. The Fate of Microcystins in the Environment and Challenges for Monitoring. Toxins 2014, 6, 3354–3387. [Google Scholar] [CrossRef]
  302. Kondo, F.; Ikai, Y.; Oka, H.; Ishikawa, N.; Watanabe, M.F.; Watanabe, M.; Harada, K.-I.; Suzuki, M. Separation and identification of microcystins in cyanobacteria by frit-fast atom bombardment liquid chromatography/mass spectrometry. Toxicon 1992, 30, 227–237. [Google Scholar] [CrossRef]
  303. Pflugmacher, S.; Wiegand, C.; Oberemm, A.; Beattie, K.A.; Krause, E.; Codd, G.A.; Steinberg, C.E.W. Identification of an enzymatically formed glutathione conjugate of the cyanobacterial hepatotoxin microcystin-LR: The first step of detoxication. Biochim. Biophys. Acta (BBA) Gen. Subj. 1998, 1425, 527–533. [Google Scholar] [CrossRef]
  304. Buratti, F.M.; Scardala, S.; Funari, E.; Testai, E. Human Glutathione Transferases Catalyzing the Conjugation of the Hepatoxin Microcystin-LR. Chem. Res. Toxicol. 2011, 24, 926–933. [Google Scholar] [CrossRef]
  305. He, J.; Chen, J.; Wu, L.; Li, G.; Xie, P. Metabolic Response to Oral Microcystin-LR Exposure in the Rat by NMR-Based Metabonomic Study. J. Proteome Res. 2012, 11, 5934–5946. [Google Scholar] [CrossRef] [PubMed]
  306. Li, S.; Chen, J.; Xie, P.; Guo, X.; Fan, H.; Yu, D.; Zeng, C.; Chen, L. The role of glutathione detoxification pathway in MCLR-induced hepatotoxicity in SD rats. Environ. Toxicol. 2015, 30, 1470–1480. [Google Scholar] [CrossRef] [PubMed]
  307. Metcalf, J.S.; Beattie, K.A.; Pflugmacher, S.; Codd, G.A. Immuno-crossreactivity and toxicity assessment of conjugation products of the cyanobacterial toxin, microcystin-LR. FEMS Microbiol. Lett. 2000, 189, 155–158. [Google Scholar] [CrossRef]
  308. Arman, T.; Lynch, K.D.; Goedken, M.; Clarke, J.D. Sub-chronic microcystin-LR renal toxicity in rats fed a high fat/high cholesterol diet. Chemosphere 2021, 269, 128773. [Google Scholar] [CrossRef] [PubMed]
  309. Pace, J.G.; Robinson, N.A.; Miura, G.A.; Matson, C.F.; Geisbert, T.W.; White, J.D. Toxicity and kinetics of [3H]microcystin-LR in isolated perfused rat livers. Toxicol. Appl. Pharmacol. 1991, 107, 391–401. [Google Scholar] [CrossRef] [PubMed]
  310. Botha, N.; Venter, M.v.d.; Downing, T.G.; Shephard, E.G.; Gehringer, M.M. The effect of intraperitoneally administered microcystin-LR on the gastrointestinal tract of Balb/c mice. Toxicon 2004, 43, 251–254. [Google Scholar] [CrossRef] [PubMed]
  311. Chen, L.; Chen, J.; Zhang, X.; Xie, P. A review of reproductive toxicity of microcystins. J. Hazard. Mater. 2016, 301, 381–399. [Google Scholar] [CrossRef] [PubMed]
  312. Buratti, F.M.; Manganelli, M.; Vichi, S.; Stefanelli, M.; Scardala, S.; Testai, E.; Funari, E. Cyanotoxins: Producing organisms, occurrence, toxicity, mechanism of action and human health toxicological risk evaluation. Arch. Toxicol. 2017, 91, 1049–1130. [Google Scholar] [CrossRef] [PubMed]
  313. Ahari, H.; Nowruzi, B.; Anvar, A.A.; Porzani, S.J. The Toxicity Testing of Cyanobacterial Toxins In vivo and In vitro by Mouse Bioassay: A Review. Mini-Rev. Med. Chem. 2022, 22, 1131–1151. [Google Scholar] [CrossRef]
  314. Aranda, Y.N.; Bhatt, P.; Ates, N.; Engel, B.A.; Simsek, H. Cyanophage-cyanobacterial interactions for sustainable aquatic environment. Environ. Res. 2023, 229, 115728. [Google Scholar] [CrossRef]
  315. Gupta, N.; Pant, S.C.; Vijayaraghavan, R.; Rao, P.V.L. Comparative toxicity evaluation of cyanobacterial cyclic peptide toxin microcystin variants (LR, RR, YR) in mice. Toxicology 2003, 188, 285–296. [Google Scholar] [CrossRef]
  316. Žegura, B.; Štraser, A.; Filipič, M. Genotoxicity and potential carcinogenicity of cyanobacterial toxins—A review. Mutat. Res./Rev. Mutat. Res. 2011, 727, 16–41. [Google Scholar] [CrossRef] [PubMed]
  317. Poniedziałek, B.; Rzymski, P.; Karczewski, J. The role of the enzymatic antioxidant system in cylindrospermopsin-induced toxicity in human lymphocytes. Toxicol. Vitr. 2015, 29, 926–932. [Google Scholar] [CrossRef] [PubMed]
  318. Mello, F.D.; Braidy, N.; Marçal, H.; Guillemin, G.; Nabavi, S.M.; Neilan, B.A. Mechanisms and Effects Posed by Neurotoxic Products of Cyanobacteria/Microbial Eukaryotes/Dinoflagellates in Algae Blooms: A Review. Neurotox. Res. 2017, 33, 153–167. [Google Scholar] [CrossRef]
  319. Colas, S.; Marie, B.; Lance, E.; Quiblier, C.; Tricoire-Leignel, H.; Mattei, C. Anatoxin-a: Overview on a harmful cyanobacterial neurotoxin from the environmental scale to the molecular target. Environ. Res. 2021, 193, 110590. [Google Scholar] [CrossRef]
  320. Smith, J.L.; Boyer, G.L.; Zimba, P.V. A review of cyanobacterial odorous and bioactive metabolites: Impacts and management alternatives in aquaculture. Aquaculture 2008, 280, 5–20. [Google Scholar] [CrossRef]
  321. Du, X.; Liu, H.; Yuan, L.; Wang, Y.; Ma, Y.; Wang, R.; Chen, X.; Losiewicz, M.D.; Guo, H.; Zhang, H. The Diversity of Cyanobacterial Toxins on Structural Characterization, Distribution and Identification: A Systematic Review. Toxins 2019, 11, 530. [Google Scholar] [CrossRef] [PubMed]
  322. Zong, W.; Wang, X.; Du, Y.; Zhang, S.; Zhang, Y.; Teng, Y. Molecular Mechanism for the Regulation of Microcystin Toxicity to Protein Phosphatase 1 by Glutathione Conjugation Pathway. BioMed Res. Int. 2017, 2017, 9676504. [Google Scholar] [CrossRef] [PubMed]
  323. Zhang, J.; Wang, J.; Wang, X.; Liu, S.; Zhou, L.; Liu, X. Evaluation of Microplastics and Microcystin-LR Effect for Asian Clams (Corbicula fluminea) by a Metabolomics Approach. Mar. Biotechnol. 2023, 25, 763–777. [Google Scholar] [CrossRef]
  324. Liu, J.; Wang, H.; Wang, B.; Chen, T.; Wang, X.; Huang, P.; Xu, L.; Guo, Z. Microcystin-LR promotes proliferation by activating Akt/S6K1 pathway and disordering apoptosis and cell cycle associated proteins phosphorylation in HL7702 cells. Toxicol. Lett. 2016, 240, 214–225. [Google Scholar] [CrossRef] [PubMed]
  325. Liu, J.; Wang, B.; Huang, P.; Wang, H.; Xu, K.; Wang, X.; Xu, L.; Guo, Z. Microcystin-LR promotes cell proliferation in the mice liver by activating Akt and p38/ERK/JNK cascades. Chemosphere 2016, 163, 14–21. [Google Scholar] [CrossRef] [PubMed]
  326. Toivola, D.M.; Eriksson, J.E.; Brautigan, D.L. Identification of protein phosphatase 2A as the primary target for microcystin-LR in rat liver homogenates. FEBS Lett. 1994, 344, 175–180. [Google Scholar] [CrossRef] [PubMed]
  327. Runnegar, M.T.; Kong, S.; Berndt, N. Protein phosphatase inhibition and in vivo hepatotoxicity of microcystins. Am. J. Physiol.-Gastrointest. Liver Physiol. 1993, 265, G224–G230. [Google Scholar] [CrossRef]
  328. Chen, Y.-M.; Lee, T.-H.; Lee, S.-J.; Huang, H.-B.; Huang, R.; Chou, H.-N. Comparison of protein phosphatase inhibition activities and mouse toxicities of microcystins. Toxicon 2006, 47, 742–746. [Google Scholar] [CrossRef]
  329. Ito, E.; Takai, A.; Kondo, F.; Masui, H.; Imanishi, S.; Harada, K.-i. Comparison of protein phosphatase inhibitory activity and apparent toxicity of microcystins and related compounds. Toxicon 2002, 40, 1017–1025. [Google Scholar] [CrossRef]
  330. Botha, N.; Gehringer, M.M.; Downing, T.G.; Venter, M.v.d.; 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] [PubMed]
  331. Ding, W.-X.; Nam Ong, C. Role of oxidative stress and mitochondrial changes in cyanobacteria-induced apoptosis and hepatotoxicity. FEMS Microbiol. Lett. 2003, 220, 1–7. [Google Scholar] [CrossRef] [PubMed]
  332. Lad, A.; Hunyadi, J.; Connolly, J.; Breidenbach, J.D.; Khalaf, F.K.; Dube, P.; Zhang, S.; Kleinhenz, A.L.; Baliu-Rodriguez, D.; Isailovic, D.; et al. Antioxidant Therapy Significantly Attenuates Hepatotoxicity following Low Dose Exposure to Microcystin-LR in a Murine Model of Diet-Induced Non-Alcoholic Fatty Liver Disease. Antioxidants 2022, 11, 1625. [Google Scholar] [CrossRef] [PubMed]
  333. Wang, X.; Ying, F.; Chen, Y.; Han, X. Microcystin (-LR) affects hormones level of male mice by damaging hypothalamic-pituitary system. Toxicon 2012, 59, 205–214. [Google Scholar] [CrossRef]
  334. Ding, W.-X.; Shen, H.-M.; Ong, C.-N. Calpain Activation after Mitochondrial Permeability Transition in Microcystin-Induced Cell Death in Rat Hepatocytes. Biochem. Biophys. Res. Commun. 2002, 291, 321–331. [Google Scholar] [CrossRef] [PubMed]
  335. Menezes, C.; Alverca, E.; Dias, E.; Sam-Bento, F.; Pereira, P. Involvement of endoplasmic reticulum and autophagy in microcystin-LR toxicity in Vero-E6 and HepG2 cell lines. Toxicol. Vitr. 2013, 27, 138–148. [Google Scholar] [CrossRef]
  336. Moreno, I.; Pichardo, S.; Jos, A.; Gómez-Amores, L.; Mate, A.; Vazquez, C.M.; Cameán, A.M. Antioxidant enzyme activity and lipid peroxidation in liver and kidney of rats exposed to microcystin-LR administered intraperitoneally. Toxicon 2005, 45, 395–402. [Google Scholar] [CrossRef]
  337. Wei, Y.; Weng, D.; Li, F.; Zou, X.; Young, D.O.; Ji, J.; Shen, P. Involvement of JNK regulation in oxidative stress-mediated murine liver injury by microcystin-LR. Apoptosis 2008, 13, 1031–1042. [Google Scholar] [CrossRef]
  338. Weng, D.; Lu, Y.; Wei, Y.; Liu, Y.; Shen, P. The role of ROS in microcystin-LR-induced hepatocyte apoptosis and liver injury in mice. Toxicology 2007, 232, 15–23. [Google Scholar] [CrossRef]
  339. Falconer, I.R.; Dornbusch, M.; Moran, G.; Yeung, S.K. Effect of the cyanobacterial (blue-green algal) toxins from Microcystis aeruginosa on isolated enterocytes from the chicken small intestine. Toxicon 1992, 30, 790–793. [Google Scholar] [CrossRef]
  340. Ding, W.-X.; Shen, H.-M.; Ong, C.-N. Pivotal Role of Mitochondrial Ca2+ in Microcystin-Induced Mitochondrial Permeability Transition in Rat Hepatocytes. Biochem. Biophys. Res. Commun. 2001, 285, 1155–1161. [Google Scholar] [CrossRef] [PubMed]
  341. Clark, S.P.; Ryan, T.P.; Searfoss, G.H.; Davis, M.A.; Hooser, S.B. Chronic Microcystin Exposure Induces Hepatocyte Proliferation with Increased Expression of Mitotic and Cyclin-associated Genes in P53-deficient Mice. Toxicol. Pathol. 2008, 36, 190–203. [Google Scholar] [CrossRef] [PubMed]
  342. Casas-Rodriguez, A.; Cameán, A.M.; Jos, A. Potential Endocrine Disruption of Cyanobacterial Toxins, Microcystins and Cylindrospermopsin: A Review. Toxins 2022, 14, 882. [Google Scholar] [CrossRef] [PubMed]
  343. Hataley, E.K.; Shahmohamadloo, R.S.; Almirall, X.O.; Harrison, A.L.; Rochman, C.M.; Zou, S.; Orihel, D.M. Experimental Evidence from the Field that Naturally Weathered Microplastics Accumulate Cyanobacterial Toxins in Eutrophic Lakes. Environ. Toxicol. Chem. 2022, 41, 3017–3028. [Google Scholar] [CrossRef] [PubMed]
  344. Moura, D.S.; Pestana, C.J.; Moffat, C.F.; Hui, J.; Irvine, J.T.S.; Edwards, C.; Lawton, L.A. Adsorption of cyanotoxins on polypropylene and polyethylene terephthalate: Microplastics as vector of eight microcystin analogues. Environ. Pollut. 2022, 303, 119135. [Google Scholar] [CrossRef]
  345. Feng, S.; Deng, S.; Tang, Y.; Liu, Y.; Yang, Y.; Xu, S.; Tang, P.; Lu, Y.; Duan, Y.; Wei, J.; et al. Microcystin-LR Combined with Cadmium Exposures and the Risk of Chronic Kidney Disease: A Case–Control Study in Central China. Environ. Sci. Technol. 2022, 56, 15818–15827. [Google Scholar] [CrossRef]
  346. Lacagnina, S. The Developmental Origins of Health and Disease (DOHaD). Am. J. Lifestyle Med. 2019, 14, 47–50. [Google Scholar] [CrossRef]
  347. Marumure, J.; Simbanegavi, T.T.; Makuvara, Z.; Karidzagundi, R.; Alufasi, R.; Goredema, M.; Gufe, C.; Chaukura, N.; Halabowski, D.; Gwenzi, W. Emerging organic contaminants in drinking water systems: Human intake, emerging health risks, and future research directions. Chemosphere 2024, 356, 141699. [Google Scholar] [CrossRef]
  348. Lin, X.; Xu, J.; Keller, A.A.; He, L.; Gu, Y.; Zheng, W.; Sun, D.; Lu, Z.; Huang, J.; Huang, X.; et al. Occurrence and risk assessment of emerging contaminants in a water reclamation and ecological reuse project. Sci. Total Environ. 2020, 744, 140977. [Google Scholar] [CrossRef]
  349. Jensen, J.; Mesman, M.; Bierkens, J.; Loibner, A.; Rutgers, M.; Bogolte, T.; Celis, R.; Dirven-van Breemen, E.M.; Erlacher, E.; Ehlers, C.; et al. Ecological Risk Assessment of Contaminated Land-DECISION Support for Site Specific Investigations; Rijksinstituut voor Volksgezondheid en Milieu RIVM: Bilthoven, The Netherlands, 2006; pp. 1–136. [Google Scholar]
  350. Gwenzi, W.; Muhoyi, E.; Mukura, T.J. Health risk assessment and mitigation of emerging contaminants: A call for an integrated approach. In Emerging Contaminants in the Terrestrial-Aquatic-Atmosphere Continuum: Occurrence, Health Risks, and Mitigation, Gwenzi, W., Ed.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 325–342. [Google Scholar]
  351. Damikouka, I.; Katsiri, A.; Tzia, C. Application of HACCP principles in drinking water treatment. Desalination 2007, 210, 138–145. [Google Scholar] [CrossRef]
  352. Tsitsifli, S.; Tsoukalas, D.S. Water Safety Plans and HACCP implementation in water utilities around the world: Benefits, drawbacks and critical success factors. Environ. Sci. Pollut. Res. 2019, 28, 18837–18849. [Google Scholar] [CrossRef] [PubMed]
  353. Ma, M.; Liu, R.; Liu, H.; Qu, J. Mn(VII)–Fe(II) pre-treatment for Microcystis aeruginosa removal by Al coagulation: Simultaneous enhanced cyanobacterium removal and residual coagulant control. Water Res. 2014, 65, 73–84. [Google Scholar] [CrossRef] [PubMed]
  354. Rodríguez, E.; Onstad, G.D.; Kull, T.P.J.; Metcalf, J.S.; Acero, J.L.; Gunten, U.v. Oxidative elimination of cyanotoxins: Comparison of ozone, chlorine, chlorine dioxide and permanganate. Water Res. 2007, 41, 3381–3393. [Google Scholar] [CrossRef] [PubMed]
  355. Chen, J.; Qu, R.; Pan, X.; Wang, Z. Oxidative degradation of triclosan by potassium permanganate: Kinetics, degradation products, reaction mechanism, and toxicity evaluation. Water Res. 2016, 103, 215–223. [Google Scholar] [CrossRef] [PubMed]
  356. Banker, R.; Carmeli, S.; Werman, M.; Teltsch, B.; Porat, R.; Sukenik, A. Uracil Moiety is Required for Toxicity of the Cyanobacterial Hepatotoxin Cylindrospermopsin. J. Toxicol. Environ. Health Part A 2001, 62, 281–288. [Google Scholar] [CrossRef]
  357. Falconer, I.R.; Humpage, A.R.; 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]
  358. Ho, L.; Tanis-Plant, P.; Kayal, N.; Slyman, N.; Newcombe, G. Optimising water treatment practices for the removal of Anabaena circinalis and its associated metabolites, geosmin and saxitoxins. J. Water Health 2009, 7, 544–556. [Google Scholar] [CrossRef] [PubMed]
  359. Laszakovits, J.R.; MacKay, A.A. Removal of cyanotoxins by potassium permanganate: Incorporating competition from natural water constituents. Water Res. 2019, 155, 86–95. [Google Scholar] [CrossRef] [PubMed]
  360. Schneider, M.; Bláha, L. Advanced oxidation processes for the removal of cyanobacterial toxins from drinking water. Environ. Sci. Eur. 2020, 32, 94. [Google Scholar] [CrossRef]
  361. Reitzel, K.; Hansen, J.; Andersen, F.Ø.; Hansen, K.S.; Jensen, H.S. Lake Restoration by Dosing Aluminum Relative to Mobile Phosphorus in the Sediment. Environ. Sci. Technol. 2005, 39, 4134–4140. [Google Scholar] [CrossRef]
  362. Kibuye, F.A.; Zamyadi, A.; Wert, E.C. A critical review on operation and performance of source water control strategies for cyanobacterial blooms: Part I-chemical control methods. Harmful Algae 2021, 109, 102099. [Google Scholar] [CrossRef] [PubMed]
  363. Fan, G.; Hong, L.; Luo, J.; You, Y.; Zhang, J.; Hua, P.; Du, B.; Zhan, J.; Ning, R.; Bao, M. Photocatalytic inactivation of harmful algae and degradation of cyanotoxins microcystin-LR using GO-based Z-scheme nanocatalysts under visible light. Chem. Eng. J. 2020, 392, 123767. [Google Scholar] [CrossRef]
  364. Fan, J.; Ho, L.; Hobson, P.; Daly, R.; Brookes, J.; Fan, J.; Ho, L.; Hobson, P.; Daly, R.; Brookes, J. Application of Various Oxidants for Cyanobacteria Control and Cyanotoxin Removal in Wastewater Treatment. J. Environ. Eng. 2014, 140, 04014022. [Google Scholar] [CrossRef]
  365. Newcombe, G.; Ho, L.; Neto, J.C. Controlling cyanotoxin occurrence. In Toxic Cyanobacteria in Water; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  366. Deas, M.L.; Tanaka, S.K.; Limanto, E.; Miao, E. Pilot Testing of. Environmentally-Safe Algaecide on Copco reservoir Water—2011 Study Results; California for PacifiCorp: Portland, OR, USA, 2012. [Google Scholar]
  367. Deas, M.L.; Vaughn, J.C.; Tanaka, S.K. Algaecide Pilot Study: Copco Reservoir; California for PacifiCorp: Portland, OR, USA, 2009. [Google Scholar]
  368. Qian, H.; Hu, B.; Yu, S.; Pan, X.; Wu, T.; Fu, Z. The Effects of Hydrogen Peroxide on the Circadian Rhythms of Microcystis aeruginosa. PLoS ONE 2012, 7, e33347. [Google Scholar] [CrossRef] [PubMed]
  369. Zhu, X.; Kong, H.; Gao, Y.; Wu, M.; Kong, F. Low concentrations of polycyclic aromatic hydrocarbons promote the growth of Microcystis aeruginosa. J. Hazard. Mater. 2012, 237–238, 371–375. [Google Scholar] [CrossRef]
  370. Sandrini, G.; Piel, T.; Xu, T.; White, E.; Qin, H.; Slot, P.C.; Huisman, J.; Visser, P.M. Sensitivity to hydrogen peroxide of the bloom-forming cyanobacterium Microcystis PCC 7806 depends on nutrient availability. Harmful Algae 2020, 99, 101916. [Google Scholar] [CrossRef] [PubMed]
  371. Krippner, R.; Staples, J. Suspected Allergy to Artemether—Lumefantrine Treatment of Malaria. J. Travel Med. 2003, 10, 303–305. [Google Scholar] [CrossRef]
  372. Mahapatra, A.D.; Bhowmik, P.; Banerjee, A.; Das, A.; Ojha, D.; Chattopadhyay, D. Ethnomedicinal Wisdom: An Approach for Antiviral Drug Development. In New Look to Phytomedicine, Khan, M.S.A., Ahmad, I., Chattopadhyay, D., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 35–61. [Google Scholar]
  373. Kim, J.K.; Kim, K.H.; Shin, Y.C.; Jang, B.-H.; Ko, S.-G. Utilization of traditional medicine in primary health care in low- and middle-income countries: A systematic review. Health Policy Plan. 2020, 35, 1070–1083. [Google Scholar] [CrossRef]
  374. Li, Z.; Zhu, X.; Wu, Z.; Sun, T.; Tong, Y. Recent Advances in Cyanotoxin Synthesis and Applications: A Comprehensive Review. Microorganisms 2023, 11, 2636. [Google Scholar] [CrossRef]
Figure 1. Conceptual summary of the nature and occurrence of cyanotoxins and their health risks and future research needs.
Figure 1. Conceptual summary of the nature and occurrence of cyanotoxins and their health risks and future research needs.
Life 15 00825 g001
Figure 2. Chemical structures of (A) microcystin-LR, (B) cylindrospermopsin, (C) anatoxin-a, (D) anatoxin-a(S), and (E) saxitoxin (reproduced from [110] with permission from Elsevier).
Figure 2. Chemical structures of (A) microcystin-LR, (B) cylindrospermopsin, (C) anatoxin-a, (D) anatoxin-a(S), and (E) saxitoxin (reproduced from [110] with permission from Elsevier).
Life 15 00825 g002
Figure 3. Overview of human exposure pathways, toxicokinetic processes, and health risks of cyanotoxins in drinking water systems.
Figure 3. Overview of human exposure pathways, toxicokinetic processes, and health risks of cyanotoxins in drinking water systems.
Life 15 00825 g003
Table 1. The summary of conventional and emerging methods employed to detect cyanotoxins.
Table 1. The summary of conventional and emerging methods employed to detect cyanotoxins.
MethodKey
Capabilities
MeritsDemeritsReference
Conventional PCRQualitative presence/absence testHighly qualitativeLow sensitivity[28]
Electrochemical biosensors Analysis of low concentration of MCsHigh recovery rates and low detection limits can be integrated into smartphonesAffected by temperature, pH, and ion concentration[29]
ELISA Detects 100 MC congenersApplicable for field screeningNon-congener specific, low selectivity, cross-reactivity[30]
GC/FID or MSUses gas chromatographyInexpensive, sensitiveAffected by the co-occurrence of cyanobacterial species, requires equipment setup, and requires expertise[30]
HPCESeparates based on charge and molecular massFast, easily applicable for the separation of cyanotoxin variantsHighly expensive[31]
HPLC- (-MS, MS/MS, UV, PDA)Can be coupled with other techniques for reliable, reproducible resultsHigh congener
resolution
Low selectivity, problematic quantitation, and high sample matrix interference[32]
LC/ESI-MS/MSPrecisely and accurately identifies specific MC congenersLow matrix interferenceLimited standards[33]
LC/TOF/MSPrecisely and accurately identifies specific MC congenersMinimizes
matrix interference
Limited standards for congeners[33]
Microarrays High-throughput
gene expression studies,
applicable in monitoring and tracking
Efficient, quick, and simultaneous analysisHigh cost[34]
PPIA Specialized colourimetry
for MC detection
Low instrument requirementsCannot distinguish MC variants, low sensitivity[23]
qPCRDetects expression of toxin-encoding genesImproved sensitivityThe presence of toxin-encoding genes does not translate to transcription, translation, and expression[35]
Whole organism assay,
e.g., mouse bioassay
May employ microbes, invertebrates, vertebrates, cell cultures, plants, and plant extractsCan be calibrated against specific variants, highly qualitativeLow sensitivity, low specificity
(no toxin identity), and not suitable for routine detection
[36]
ELISA—Enzyme-Linked Immunosorbent Assay; ESI—Electrospray Ionization; FID—Flame Ionization Detector; GC—gas chromatography; HPCE—high-performance capillary electrophoresis; LC—liquid chromatography; MC—microcystin; MS—mass spectrometry; MS/MS—tandem mass spectrometry; qPCR—quantitative polymerase chain reaction; PCR—polymerase chain reaction; PDA—photodiode array; UV—ultraviolet; and TOF—time-of-flight.
Table 2. Human health risks, including reported toxicity mechanisms following exposure via drinking water systems.
Table 2. Human health risks, including reported toxicity mechanisms following exposure via drinking water systems.
CyanotoxinExposure routeToxicityMode of ActionReference
Microcystin (MC)Reservoir waterToxin covalently binds and inhibits catalytic subunits on protein PP1 and PP2A, causing hyper-phosphorylation of cellular microtubules, hence loss of cellular structure.MC-LR daily intake of 2.03 µg/L was associated with liver damage in children. Causes cellular apoptosis.[37]
MCWater used for dialysis treatmentsMCs disrupted the liver plate pattern. MCs are inhibitors of eukaryotic protein serine/threonine phosphatases 1 and 2A.Liver cell damage[38]
MCContact and swallowing MC-contaminated
reservoir water
Pulmonary intravascular formation of protein deposits and removal of platelets from circulation.Liver damage, pneumonia,
and pulmonary thrombosis. Death.
[39]
Microcystin-LR (MC-LR)InjectionIntrahepatic hemorrhaging.
MC-LR covalently binds to protein phosphatase 1/2A.
MC-LR dose-dependent hepatocellular hypertrophy, degradation
and necrosis. Hepatic inflammation. Fibrosis in the livers of rats.
[40]
MC-LR plus thioacetamide (TAA) (MC-LR/TAA)Drinking contaminated waterInhibits serine/threonine phosphatase activity, enhances proliferative activity, or activates HSCs to transform to a myofibroblast phenotype.Liver inflammation.[41]
SaxitoxinContact with water during recreational activitiesBlocking sodium channels in nerve axons. Inhibits the transfer of excitatory signals.Causes paralysis of the respiratory muscles and death by respiratory failure.[42]
Cylindrospermopsin (CYN)Contact and drinking
CYN-contaminated water
Causes oxidative stress in human neutrophils, eventually leading to lipid peroxidation and decreased cell survival.Apoptosis in human T-lymphocytes.[43]
CYNIntraperitoneal administrationCauses oxidative stress in human neutrophils, eventually leading to lipid peroxidation and decreased cell survival.Human hepatoblastoma and human colon adenocarcinoma cells.[44]
MCDrinking contaminated lake water and foodMC-LR leads to hepatic steatosis with molecular alterations in circadian rhythm regulation, lipid metabolic processes, and the cell cycle pathway.Liver damage and lipid metabolism dysfunction.
Long-term exposure leads to non-alcoholic fatty liver disease (NAFLD)
[45]
MCInjectionDamages diabetes genes and other proteins, e.g., Ppp3ca, Ide, Marcks, Pgk1, and Ndufs4.Increased risk of diabetes in humans. Reduction in blood insulin levels.[46]
MC-LRHuman hepatocellular carcinoma cell line (HepG2) was exposed to
solutions of MC-LR
Under normoxic conditions, MC-LR stimulates the proliferation of the HepG2 cell line, promoting liver tumor and liver cancer cell growth.
Under hypoxic conditions, MC-LR induced apoptosis in the HepG2 cell line by inducing prolonged oxidative stress.
Liver cancer[47]
MC-LROral intake of MC-LR-contaminated waterMC-LR causes alterations in TBARS, superoxide dismutase (SOD) activity, and glutathione content in the liver and intestine of mice.Exposure to 50 mg MC-LR/kg every 48 h generates significant damage to the liver and intestine.[48]
Table 3. General toxicity, including target organs and receptors of cyanotoxins.
Table 3. General toxicity, including target organs and receptors of cyanotoxins.
CyanotoxinReceptor Organ, Tissue or ProcessToxicityReference
Hepatotoxicity
Microcystin-LR (MC-LR) and dehydrobutyrine (Dhb)-containing MC
variant [Asp3, ADMAdda5, Dhb7] MC-HtyR isolated from Nostoc spp.
In vivo phosphoprotein phosphatase (PPP) family (PP1, PP2A, PPP4, and PPP5)
  • Dhb-containing MCs and MC-LR inhibited PP1, PP2A, PPP4, and PPP5.
  • MC-LR formed a covalent bond with a cysteine residue in the PPP, while Dhb-MC-HtyR did not form any covalent interaction with PP2A.
  • Nostocyclin, a cyclic peptide also isolated from Nostoc spp., was non-toxic with more than 500-fold less inhibitory effects on PP1, PP2A, PPP4, and PPP5.
  • Given that LD50 for MC-LR was lower than that of Dhb-containing MCs and nostocyclin, the inhibition of protein phosphatases seems to be the most harmful toxicity mechanism of MCs in vivo.
[49]
Microcystinss (MCLR, LA, LF, LW, LY)
with changed Z4 residues
Protein phosphatase 2A
  • MC (MCLR, LA, LF, LW, and LY) inhibition effects on PP2A were explored by a colorimetric protein phosphatase inhibition assay.
  • The inhibition sequence of MCs on PP2A was detected as follows: MC-LR > MC-LW > MC-LA > MC-LF > MC-LY.
[50]
MC-LR (MC-LR)
and Cylindrospermopsin (CYN)
Salmonella typhimurium, L5178Y Tk+/− cells, and Caco-2 cells
  • An in vitro battery was used to assess the mutagenicity and genotoxic potential of a mixture of MC-LR and CYN.
  • The in vitro mutagenicity and genotoxicity showed by CYN/MC-LR mixtures do not differ substantially from that observed for CYN tested individually.
  • The results indicate that cyanobacterial mixtures require a specific (geno)toxicity evaluation as their effects cannot be extrapolated from those of the individual cyanotoxins.
[51]
MC-LRMicrofilament depolarization and expression of microRNA-451a (miR-451a) in HL7702 liver cells
  • Data demonstrated that MC-LR increased microfilament depolarization, elevated phosphorylation levels of mitogen-activated protein kinase (MAPKERK1/2) and vasodilator-stimulated phosphoprotein (VASP), but lowered miR-451a RNA expression levels.
  • Data demonstrated that transfection with miR-451a may not be effective in the presence of MC-LR, as evidenced by the inability of excess microRNA to prevent toxin-induced inhibition of threonine protein phosphatases 1 (PP1) and 2A (PP2A) and microfilament reorganization in HL7702 cells.
[52]
Neurotoxicity
CYNThe skin of albino Californian rabbits
  • Edema and erythema were observed after 24 h following intradermal exposure to 0.2 mL of lyophilized Umezakia ovalisporum (formerly Aphanizomenon ovalisporum) extract, and skin irritation was rated moderate.
[53]
CYNAbdominal skin of Balb/c mice exposed to 100 µg of
CYN mL−1 via topical application
  • Yellow/brown crusts, dried skin, blood, or serous fluid oozing from exposed skin and desquamation were observed.
  • Skin lesions were observed starting from the second induction day onwards.
  • Cell infiltration predominantly of the mononuclear cells, edema, inflammation, and thickening were noted on the ears after 24 h and 48 h of exposure.
  • At an exposure dose of 73 µg CYN/mL ear swelling was observed.
[54]
Cytotoxicity
Interactive cytotoxic effects of CYN and a pesticide (chlorpyrifos) Differentiated SH-SY5Y human neuronal (neuroblastoma) cells
  • A concentration-dependent decrease in viability was observed for the contaminants.
  • Interactive toxic effects induced signs of necrosis and apoptosis, but acetylcholinesterase activity appeared unaffected.
  • The results demonstrate the need to understand the interactive toxic effects of cyanotoxins and other health stressors.
[55]
Variants of cyanobacterial extracts (crude MC-containing, purified MC-containing, and non-MC-containing extracts)Cultured human lymphocytes
  • The effects of MC-producing Cyanobacteria on human lymphocyte culture and their potential for adverse human health effects were investigated.
  • The study indicated the highest cytotoxicity and genotoxicity in the purified extract containing MCs (MC-LR, MC-RR, and MC-YR).
  • No significant effect or clear relation between MC concentration in crude extracts and human lymphocyte mortality and DNA damage was observed.
  • This research supports the necessity of the application of additional tests for the determination of total threat from natural cyanobacterial blooms, apart from the determination of concentration and toxicity of the main group of cyanotoxins.
[56]
Dermatotoxicity
Extracts of M. aeruginosa (non-toxic strain), A. circinalis, N. spumigena M. aeruginosa (toxic strain), A. incerta, and C. raciborskiiSkin of human volunteers
  • The results showed that between 10% and 40% of both atopic and non-atopic individuals showed adverse reactions to at least one of the active patches of each of the six cyanobacterial species and concentrations investigated.
  • A small number of participants showed evidence of induced erythema via skin contact, but no dose-dependent or threshold behavior was evident.
[57]
Miscellaneous toxicity effects
MC-leucine arginine (MC-LR)DNA
  • The direct effect of MC-LR on DNA was explored by using spectral analysis and molecular biotechnology.
  • Spectra analysis and molecular biotechnology showed that there is no direct interaction between DNA and MC-LR
  • This study discovered that the effects of MC-LR on DNA originate mainly from the secondary effects of MC-LR rather than from the direct interaction between DNA and MC-LR.
[58]
MC-LRInteraction of MC-LR with proteins and DNA
  • Experiments were performed to explore the interaction of MC-LR and proteins.
  • The results indicated that there is binding of MCs to a wide range of proteins in vitro.
  • Under the conditions tested, MC-LR does not exhibit affinity for DNA.
[59]
Combination of serum MCs,
plasma As, and Cd
Human kidneys
  • The association between MCs, As, and chronic kidney disease (CKD) was investigated using conditional logistic regression.
  • The results showed that MCs and As were significantly associated with CKD risk.
  • Both MCs and As are independent risk factors for CKD, exerting a synergistic effect between them.
  • Combined exposure to MCs, As, and Cd concentration can increase the risk of CKD.
[60]
Table 4. Summary of the methods employed to remove cyanotoxins from drinking water systems in different world regions and their effects.
Table 4. Summary of the methods employed to remove cyanotoxins from drinking water systems in different world regions and their effects.
CyanotoxinReceptor Organ, Tissue, or ProcessEffectReference
Conventional methods
North AmericaCoagulation and
sand filtration
Reduced intracellular cyanotoxins by 59–97%, with most MC removal occuring during coagulation, filtration, and settling.[61]
EgyptSedimentation/
sand filtration
and coagulation
(flocculation)
Sedimentation, coagulation, and filtration removed 91–98.9% of MCs, but flocculation may release MCs. Chlorination partially degraded toxins, sometimes exceeding WHO limits.[62]
Egypt Coagulation,
sedimentation,
and flocculation
Removed various toxin-producing Cyanobacteria, but O. limnetica cells were only partially removed.[62]
ChinaCoagulation (flocculation)
with flocculants and
coagulants, including ferric chloride and aluminum salts
Removal of cyanotoxins by 90% from drinking water.[63]
CanadaClarification,
coagulation,
and filtration
Cyanotoxin-producing Aphanizomenon cells were poorly removed by coagulation. Pseudanabaena and Anabaena were efficiently removed from the water by filtration and clarification.[64]
CanadaCoagulation90% of intracellular MCs were removed. Removal of sludge after coagulation was recommended to avoid the re-release of MCs to water.[65]
EgyptRapid sand filtration and disinfection methodsRapid sand filtration of drinking water partially removed Microcystis and Pseudanabaena limnetica (formerly Oscillatoria limnetica) (ranging from 25 to 34%). O. limnetica cells were reported to resist disinfection, which is applied in the final stages of water treatment, and disinfection only reduced O. limnetica by between 25 and 66.7%.[66]
PortugalOzonation and coagulation2.5–7.9% and 6.8–11.7% of microcystins (MCs) were removed by coagulation before and after ozonation, respectively. [67]
ChinaFerrate
oxidation
Ferrate oxidation removed up to 98% of cyanotoxins, with efficiency influenced by contact time, dosage, and pH. A 30 min contact period at 40 mg/L ferrate and pH 6–10 yielded optimal results.[68]
ChinaChlorination
and UV application
Chlorine (0.2–2.0 mg/L) and ultraviolet (UV) treatment damaged up to 96.6% of M. aeruginosa cells and degraded microcystin-LR (MC-LR_ by 69–83%. Combined UV/chlorine treatment was most effective with removal percentages of MC-LR between 68.9 and 82.7% in 15 min.[69]
BelgiumDissolved
air flotation
Air flotation removed 40–80% of MCs, 90–100% of Anabaena, and 30% of Planktothrix cells.[70]
CanadaSand
and graphitized
sand filtration
Graphitized sand filtration reduced MC-LR to <0.61 μg/L (within WHO limits), outperforming standard sand filtration at a lower cost (CAD 160 vs. 6000).[71]
FranceRapid
and slow
sand filtration
Bacteria-assisted filtration removed up to 99% of algal cells and toxins, offering a low-cost potable water solution.[72]
South AustraliaBiological
sand filtration
MC-LR and microcystin-LA MC-LA were completely removed
from drinking water.
[73]
Membrane-based methods/Adsorption (carbon-based methods)
GermanyUltrafiltrationMembranes removed 98% of intracellular MCs and >96% anatoxin-a, eliminating both toxins and toxin-producing microbes.[74]
CanadaGranular activated carbon (GAC) and powdered activated
carbon (PAC) adsorbents
GAC reduced MC-LR from up to 47 to 1 μg/L; PAC (100 mg/L) removed 86%, lowering MC-LR from 22 to 3 μg/L.[75]
South AfricaPAC (tyre-basedTyre-based PAC achieved 100% MC-LR removal at pH 4, 10,000 mg/L carbon, in 34 min.[76]
SwitzerlandGravity-driven membrane (GDM)GDM systems removed M. aeruginosa and lowered cyanotoxins to <1 μg/L within 15 days.[77]
AustraliaMicrofiltration and ultrafiltrationMembranes removed >98% of intracellular M. aeruginosa cyanotoxins.[78]
ChinaNanomaterial
algacidal sheet
Algaecidal sheets removed 56–89% of M. aeruginosa and Anabaena within one day.[79]
AustraliaGACGAC removed up to 95% of anatoxin and extracellular MCs.[80]
BrazilGAC coupled with Fenton oxidation/flocculation/coagulation/sedimentationGAC columns adsorbed 4.15 μg/g MCs; combined Fenton oxidation and GAC effectively purified water to safe limits.[81]
GermanyUltrafiltrationMembranes removed 98% of MCs and >96% anatoxin-a, eliminating toxins and producers.[74]
CanadaGranular activated carbon (GAC) and powdered activated carbon (PAC) adsorbentsGAC decreased MC-LR to 1 μg/L;
PAC (100 mg/L) reduced MC-LR from 22 to 3 μg/L.
[75]
South AfricaPAC (tyre-based)Tyre-based PAC removed 100% MC-LR at optimal conditions (pH 4, 10,000 mg/L, 34 min).[76]
Advanced oxidation technologies for the degradation of cyanotoxins
CanadaPotassium permanganate oxidationUp to 95% of MCs were removed from drinking water.[65]
ChinaKMnO4 pre-oxidationMnO4 combined with sand filtration or Mn-oxidizing bacteria (e.g., Pseudomonas) enhanced the removal of MC-LR and BP-4 via transformation of Mn2+ to Mn oxides.[82]
China•OH oxidation•OH (1 mg/L) pre-treatment, alone or with 0.5 mg/L NaClO, inactivated blooms and maintained disinfection byproducts (DBPs) within Chinese drinking water safety limits.[83]
China•OH oxidationIn a 480 m3/day plant, •OH (0.88 mg/L) rapidly inactivated algae; combined with ClO2 and sand filtration, it met all Chinese water quality standards.[84]
GermanOzonationOzone (1.5 mg/L) degraded M. aeruginosa toxins; as little as 0.2 mg/L oxidized MC-LR below detection within seconds to minutes, reducing toxicity.[85]
AustraliaOzonationOzone (0.2 mg/L) degraded MC-LR below high-performance liquid chromatography (HPLC) detection limits within a short period of time (sec to min).[86]
United KingdomTiO2 and UVTiO2 and UV removed MCs in under 45 min.[87]
ChinaFerrate (VI) and UVFerrate (VI) (0.08–0.17 mmol/L) with UV degraded up to 100% of MC-LR.[88]
Biodegradation
EgyptBiodegradation
by Bacillus sp.
Up to 300 μg/L of CYN was biodegraded at rates from 1.25 to 50 μg/L/day.[89]
PolandBiodegradation
by Aeromonas sp.
CYN biodegradation by Aeromonas sp. was 100% at pH 7 and 25–30 °C but dropped to 0% at pH 11 and colder temperatures.[90]
New ZealandBiodegradation by
Sphingomonas bacteria
Sphingomonas species in a ceramic-supported bioreactor removed up to 100% of MCs.[91]
Methods based on plant coagulants
VietnamUse of Moringa oleifera
(drumstik tree)
Plant-based coagulants removed up to 97% of turbidity (15 g/L, pH 6.8) and 86–94% at pH 6.2.[92]
CanadaUse of cactusCactus species reduced turbidity by 92–98% and lowered pH from 8.89 to 6.[93]
MoroccoOpuntia ficus-indica
and Vicia faba
seed coagulants
At 0.5–1 g/L and pH 5, plant coagulants removed up to 85% of M. aeruginosa and about 80% of intracellular cyanotoxins.[94]
BrazilCoagulation using M. oleifera/flocculation/dissolved air flotationCombining plant-based methods effectively removed
M. aeruginosa from water.
[95]
BrazilM. oleifera coagulantsM. oleifera coagulants reduced turbidity, suspended matter, and chlorophyll-a by 40–60%.[96]
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

Marumure, J.; Gwenzi, W.; Makuvara, Z.; Simbanegavi, T.T.; Alufasi, R.; Goredema, M.; Gufe, C.; Karidzagundi, R.; Rzymski, P.; Halabowski, D. Global Occurrence of Cyanotoxins in Drinking Water Systems: Recent Advances, Human Health Risks, Mitigation, and Future Directions. Life 2025, 15, 825. https://doi.org/10.3390/life15050825

AMA Style

Marumure J, Gwenzi W, Makuvara Z, Simbanegavi TT, Alufasi R, Goredema M, Gufe C, Karidzagundi R, Rzymski P, Halabowski D. Global Occurrence of Cyanotoxins in Drinking Water Systems: Recent Advances, Human Health Risks, Mitigation, and Future Directions. Life. 2025; 15(5):825. https://doi.org/10.3390/life15050825

Chicago/Turabian Style

Marumure, Jerikias, Willis Gwenzi, Zakio Makuvara, Tinoziva T. Simbanegavi, Richwell Alufasi, Marvelous Goredema, Claudious Gufe, Rangarirayi Karidzagundi, Piotr Rzymski, and Dariusz Halabowski. 2025. "Global Occurrence of Cyanotoxins in Drinking Water Systems: Recent Advances, Human Health Risks, Mitigation, and Future Directions" Life 15, no. 5: 825. https://doi.org/10.3390/life15050825

APA Style

Marumure, J., Gwenzi, W., Makuvara, Z., Simbanegavi, T. T., Alufasi, R., Goredema, M., Gufe, C., Karidzagundi, R., Rzymski, P., & Halabowski, D. (2025). Global Occurrence of Cyanotoxins in Drinking Water Systems: Recent Advances, Human Health Risks, Mitigation, and Future Directions. Life, 15(5), 825. https://doi.org/10.3390/life15050825

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

Article Metrics

Back to TopTop