Next Article in Journal
Screening Tests for the Rapid Detection of Diarrhetic Shellfish Toxins in Washington State
Next Article in Special Issue
Reporter Dyes Demonstrate Functional Expression of Multidrug Resistance Proteins in the Marine Flatworm Macrostomum lignano: The Sponge-Derived Dye Ageladine A Is Not a Substrate of These Transporters
Previous Article in Journal
Evaluation of the in Vitro Anti-Atherogenic Properties of Lipid Fractions of Olive Pomace, Olive Pomace Enriched Fish Feed and Gilthead Sea Bream (Sparus aurata) Fed with Olive Pomace Enriched Fish Feed
Previous Article in Special Issue
Advances in the Study of the Structures and Bioactivities of Metabolites Isolated from Mangrove-Derived Fungi in the South China Sea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microcystin-LR and Cylindrospermopsin Induced Alterations in Chromatin Organization of Plant Cells

Department of Botany, Faculty of Science and Technology, University of Debrecen, Debrecen H-4010, Egyetem tér 1, Hungary
*
Authors to whom correspondence should be addressed.
Mar. Drugs 2013, 11(10), 3689-3717; https://doi.org/10.3390/md11103689
Submission received: 17 June 2013 / Revised: 19 August 2013 / Accepted: 22 August 2013 / Published: 30 September 2013
(This article belongs to the Special Issue Cytogenetic and Molecular Effects of Marine Compounds)

Abstract

:
Cyanobacteria produce metabolites with diverse bioactivities, structures and pharmacological properties. The effects of microcystins (MCYs), a family of peptide type protein-phosphatase inhibitors and cylindrospermopsin (CYN), an alkaloid type of protein synthesis blocker will be discussed in this review. We are focusing mainly on cyanotoxin-induced changes of chromatin organization and their possible cellular mechanisms. The particularities of plant cells explain the importance of such studies. Preprophase bands (PPBs) are premitotic cytoskeletal structures important in the determination of plant cell division plane. Phragmoplasts are cytoskeletal structures involved in plant cytokinesis. Both cyanotoxins induce the formation of multipolar spindles and disrupted phragmoplasts, leading to abnormal sister chromatid segregation during mitosis. Thus, MCY and CYN are probably inducing alterations of chromosome number. MCY induces programmed cell death: chromatin condensation, nucleus fragmentation, necrosis, alterations of nuclease and protease enzyme activities and patterns. The above effects may be related to elevated reactive oxygen species (ROS) and/or disfunctioning of microtubule associated proteins. Specific effects: MCY-LR induces histone H3 hyperphosphorylation leading to incomplete chromatid segregation and the formation of micronuclei. CYN induces the formation of split or double PPB directly related to protein synthesis inhibition. Cyanotoxins are powerful tools in the study of plant cell organization.

1. Introduction

The marine and freshwater habitats are considered to be a source of potential drugs. To date approximately 16,000 natural products have been isolated from marine, freshwater organisms so it is not surprising that these organisms are a wonderful source of biologically active natural products with diverse chemistry and pharmacology too [1]. For example marine organisms produce some of the most cytotoxic compounds ever discovered, but the yields of these compounds are invariably so small that natural sources are unlikely to provide enough material for drug development studies [2].
Marine biotoxins have drawn worldwide attention because of their involvement in human intoxication and the socio-economic impacts brought by those incidents. To understand the chemistry and the mechanism of action of the toxins is important because it helps in developing the adequate countermeasures, such as detection, determination and therapeutic methods. In addition many of the toxins have been found to be useful tools for probing biological or pharmacological phenomena, like use of okadaic acid in protein phosphatase studies [3].
The unicellular life forms such as several algae and cyanobacteria, which can become abundant under favorable environmental conditions in waters, are readily collectable, culturable, and thus, have become good targets of many natural products researchers [4]. In freshwaters the most prominent unicellular organisms are the cyanobacteria which can multiply and can develop huge biomass called as blooms [5]. These prokaryotic organisms not only elaborate the common primary metabolites and pharmaceutically useful compounds but also produce toxic substances. The cyanobacterial toxins are a heterogeneous bioactive group with diverse chemistry [6].
Cyanobacterial toxins are classified as hepatotoxins (microcystins and nodularins), neurotoxins (anatoxins and paralytic shellfish toxins), cytotoxins (cylindrospermopsins) and dermotoxins [5]. Their production can be found in a diverse range of blue-green species. The most studied cyanobacterial toxins are the peptide type microcystins (MCYs) and most recently the alkaloid cylindrospermopsin (CYN). In addition there are also the two cyanotoxin groups, from which human injury has been clearly identified [7]. Although there are many results and cytogenetic studies relating the effects of these cyanobacterial metabolites especially in animal and human cells, for understanding the whole mechanism or applying as tools in pharmacology, it is important to know the effects in different organisms (animal, plant, fungi) at different levels (histology, cytology, molecular biology). Thus, cytogenetic studies may help the understanding of cyanobacterial toxicity in natural habitats.
We are focusing in this review on the two main cyanotoxin-MCY and CYN induced changes of chromatin organization and the possible cellular mechanisms of those alterations in plant cells.

2. Cyanotoxins-Microcystins and Cylindrospermopsins

2.1. Microcystins

MCYs are cyclic heptapeptides consisting of seven amino acids, including d-amino acids and two unusual amino acids namely, N-methyldehydroalanine (Mdha) and a hydrophobic d-amino acid, 3-amino-9-methoxy-2-6,8-trimethyl-10-phenyldeca-4,6-dienoic acid (Adda) [8]. More than 60 structural variants are described, with variation occurring mainly at the two l-amino acids along with alterations to side chains [6]. The hepatotoxic microcystins (MCY) are known to be produced by several bloom forming cyanobacterial genera including Microcystis, Anabaena, Planktothrix, Nostoc [5]. The toxic mechanism of MC is the inhibition of protein phosphatases of type 1 and 2A (PP1 and PP2A, [9]). Cysteine-273 of the catalytic subunit of PP-1 (Cysteine-266 of PP2A) binds covalently to the carbonyl group of Mdha of MCY but this is not a requirement for the inhibition [10,11]. It is the introduction of Adda into the hydrophobic groove at the catalytic site of protein phosphatase that renders it inactive [12]. The Adda part of the molecules seems crucial for the toxicity, the loss of Adda or synthetic MCY without Adda are consequently non-toxic [13]. Microcystins containing the naturally occurring Z enantiomer (geometrical isomer) of Adda or linearized MCY are less potent protein phosphatase inhibitors [14]. The IC50 of protein phosphatase inhibition by MCY-LR is (0.1–0.25 nM) in vitro. Liver damage is one of the most common phenomena in the intoxication of microcystins in animal organisms which starts with loss of membrane integrity, cytoskeletal disorganization, cellular disruption, lipid peroxidation, DNA damage, apoptosis, necrosis, and ultimately death by hemorrhagic shock [15].

2.2. Cylindrospermopsins

CYN is a low molecular weight (MW.415) tricyclic alkaloid with guanidino group linked at C7 to hydroxymethyl uracil. Because of the negatively charged sulfate group and the positively charged guanidino group, the molecule is a zwitterion and very soluble in polar solvents [16]. CYN was detected in eight different cyanobacterial species and several producers considered as invasive organisms [7,17]. There are two possible natural epimers at the hydroxyl bridge with same toxicity, cylindrospermopsin and 7-epicylindrospermopsin. CYN in Umezakia natans from Japan and in Aphanizomenon ovalisporum from Israel samples have the same epimeric form, which is of opposite orientation to the toxin of C. raciborskii from Australia [18]. The uracil group of the molecules seems crucial for its mechanism, because without this part the cylindrospermopsin molecule effectively lacks toxicity [16]. The mechanisms of cylindrospermopsin toxicity are under investigation. Uptake of the toxin is relatively rapid since complete and irreversible block of protein synthesis occurs after a 1 h exposure in vitro. Inhibition of protein synthesis occurs at the ribosome during the peptide chain elongation step [19]. Evaluating the human and animal exposures, the toxin causes damage to liver, kidneys, lungs, heart, stomach, adrenal glands, the vascular system, and the lymphatic system [20]. Recent data from primary hepatocytes show two routes of toxic action, a rapid route probably through toxicity of a cytochrome P450 (CYP450) oxidation product of the toxin [19]. Glutathione synthesis is also reduced via a CYP450-dependent mechanism [21]. The main genotoxic outcome appears to be DNA fragmentation [22,23], although loss of whole chromosomes has also been shown to occur [22].

3. Particularities of Plant Chromatin and Microtubule Organization

Plant cells have particularities not found in other eukaryotic cells. These include plastids, the presence of cellulose cell wall, storage vacuoles and plasmodesmata, the latter connecting neighboring cells in tissues (see [24] for an example). Not taking into account the lack of unequivocal evidence for the presence of intermediate filaments in plant cell nucleus, the basic structural features of plant chromatin are similar to all eukaryotic cells. Cytoskeletal elements are relevant to this review, since at least mitotic microtubules (MTs) and microfilaments (MFs) have a crucial role in chromatin organization and dynamics. The particularities of plant cytoskeleton are [25,26,27,28,29]: (i) The presence of preprophase bands (PPBs) at G2 phase and early mitosis containing MT and MF arrays and localized at the future site of cell division; (ii) The presence of endoplasmic microtubules (EMTs) playing a role in regulating PPB formation and stabilizing cell plate formation during cytokinesis; (iii) The presence of phragmoplasts at the end of cell division, that contain MTs and MFs as well and direct vesicle traffic to the site of new cell wall formation at the equatorial plane of dividing cells; (iv) The absence of centrioles and the presence of diffuse microtubule organization centre (MTOC) like structures that make MTs more dynamic than in animal cells. The nucleation of MTs is likely to happen in the cortical cytoplasm and nuclear envelope. Spindle formation starts from multiple MTOCs from both poles of dividing cells. The principles of chromatid/chromosome dynamics during meiosis and mitosis in plant cells are similar to the other eukaryotic cell types. However, they have several particularities in this respect-dyneins as cytoskeleton associated motor proteins involved in chromosome movement are absent here, meaning that chromatid/chromosome regulation machinery is not the same as for animal cells; (v) The preferential arrangement of MTs at the periphery of non-dividing (interphase or differentiated) cells, just beneath the plasma membrane (cortical MTs, CMTs). CMTs play an essential role in the regulation of cell elongation and shape and are probably involved in the positioning of nucleus at the onset of mitosis; (vi) The primary role of actin cytoskeleton in the delivery of organelles such as peroxisomes or mitochondria.
The organization of cytoskeletal elements is strongly dependent on protein phosphorylation/dephosphorylation and on the synthesis of certain regulatory proteins like microtubule associated proteins (MAPs) [30,31,32,33]. For the above reason, due to the relevant particularities of plant cells—with special emphasis on PPB, spindle and phragmoplast formation—it is worthwhile to review the effects of PP1 and PP2A inhibitory MCY and protein synthesis inhibitory CYN in those systems.
Concerning chromatin in non-mitotic plant cells, both the disturbance of protein phosphatase activities by MCYs and protein synthesis by CYN are likely to interfere with a wide range of intracellular events, leading to cell death. Programmed cell death (PCD) of plant cells bears several particularities, like: (i) It is not always accompanied by internucleosomal DNA cleavage (DNA laddering) (see [34], for an example); (ii) Due to the presence of cell wall, phagocytosis of apoptotic cells by neighbouring cells is not possible [35] and (iii) Subsequent PCD and necrosis in plant cells are more common, than in animal cells [36]. Cyanotoxin effects on plant PCD will be discussed in Section 5.2 and Section 6.2.

4. An Overview of the Effects of MCY and CYN in Plants

There are a significant number of papers dealing with the effects of cyanotoxins on plants (see reviews [15,17,37,38,39,40] for examples). These include alterations of growth, development, anatomy, cell structure and physiological/biochemical processes as summarized on Table 1. Many of the effects listed can be directly related to the PP1 and PP2A inhibitory effect of MCYs and protein synthesis inhibitory effect of CYN. Other changes may be non-specific that is, not related directly to the biochemical effects of cyanotoxins (see Table 1 and later sections). All those alterations raised the need for the study of plant cellular changes that could reveal fine mechanisms of toxin action. Furthermore, growth and morphological changes did help in the understanding of cellular effects studied later. For example growth inhibition and histological marks of cell death can be related to the induction of PCD and non-specific cell death (necrosis) in plant cells. The cytological effects—focusing on chromatin and cytoskeletal changes will be discussed in the context of all levels (from organism to enzymes) of cyanotoxin effects in the following sections.
Table 1. Microcystin and cylindrospermopsin induced alterations in some vascular plants.
Table 1. Microcystin and cylindrospermopsin induced alterations in some vascular plants.
Cyanotoxins used in the experimentsType of the cyanotoxin induced alterationsPlant taxonReferences
I. Growth alterations
MCY-LR, -RR, -LF, -FR, -LW, -YR in purified, -LR, -RR, -FR, -YR in extract, M. aeruginosa bloom samplesInhibited seed germination, growth/elongation of shoot, primary root, leaves, inhibited increase of frond number, fresh/dry weightBrassica napus, Ceratophyllum demersum, Lemna minor, L. gibba, L. japonica, Lens esculenta, Lepidium sativum, Lolium perenne, Malus pumila, Medicago sativa, Myriophyllum variifolium, Oryza sativa, Phragmites australis, Pisum sativum, Sinapis alba, Spirodela oligorrhiza, Triticum durum, Vallisneria natans, Vicia faba, Vicia faba inoculated with rhizobial strains, Wolffia arrhiza, Zea mays[38,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69]
MCY-RRDecreased cell viabilityTobacco BY-2 cells[70]
CYN (purified, crude extract)Inhibited seed germination, growth/elongation of whole plant, shoot and mainroot, inhibited increase of fresh weight of leavesBrassica oleracea var. sabellica, Brassica juncea, Lactuca sativa, Lemna minor, callus-derived Phragmites australis plantlets, Sinapis alba, Wolffia arrhiza[52,71,72,73,74,75]
CYN in crude extractsConcentration- and exposure time/exposed organ dependent stimulation or inhibition of growthHydrilla verticillata, Lactuca sativa, Oryza sativa, Phaseolus vulgaris, Pisum sativum, Solanum lycopersicum, Spirodela oligorrhiza[74,76,77,78]
CYNInhibited pollen germinationNicotiana tabacum[79]
II. Morphological/Developmental alterations
MCY-LR, -RR purified and in crude extractInhibited root elongation and altered primary root/lateral root formation, missing root hairs/crown root formation, radial expansion in roots, root coalescenceLens esculenta, Oryza sativa, Phaseolus vulgaris, Phragmites australis, Pisum sativum, Sinapis alba, Triticum durum, Vallisneria natans, Zea mays[43,46,51,52,56,58,60,69,80,81]
MCY-LR, -RR, -YR purified and in crude extractInhibited photomorphogenesis of cotyledons: Chlorotic and smaller cotyledons, missing trichomes of petioles, malformed, chlorotic fronds/leaves, inhibited shoot elongation, the seedlings lying horizontally on the paper bed. Stimulation of floweringBrassica napus, Lemna minor, Sinapis alba, Spinacia oleracea variants[45,51,52,53,82]
MCY-LRInhibited shoot and root formation, decrease of somatic embryo numberPhragmites australis, Solanum tuberosum tissue cultures[46,58]
CYNIncreased root number, inhibited elongation, radial expansion of rootsHydrilla verticillata, callus-derived Phragmites australis plantlets, Sinapis alba[69,72,77]
CYNInhibited photomorphogenesis of cotyledons: The chlorotic, smaller cotyledons were violet colored in consequence of high level of anthocyaninsSinapis alba seedlings[52]
CYN in crude extractProlonged (9 days) exposure induced decrease of water content, browning, green color lost, shrunk leavesOryza sativa[78]
III. Histological and cytological alterations
MCY-LRLignification in cell walls (in cortical parenchyma, endodermis and pericycle, with increased autofluorescence)Phragmites australis plantlets, Sinapis alba seedlingsSection 5.2; [58,69,80]
MCY-LRSwelling of cells and formation of a callus-like tissue (in main roots and at the transit between main root and hypocotyls in mustard, in rhizome and roots of reed)Phragmites australis plantlets, Sinapis alba seedlingsSection 5.2; [58,69,80]
MCY-LREarly aerenchyma formationPhragmites australis plantlets[58]
MCY-LRInhibition of formation of vascular cylinder, xylem differentiation (xylem area and number of vessel elements)Phaseolus vulgaris, Sinapis alba seedlingsSection 5.2; [60,69]
MCY-LR, -RR, -YR purified and in crude extractCell death by necrosis in cotyledon, shoot and root tissuesBrassica napus, Lemna minor, Phaseolus vulgaris seedlings, Phragmites australis, Solanum tuberosum tissue culture, Sinapis alba seedlingsSection 5.2; [43,46,51,52,53,55,58,69,80,83]
CYNLignification in cell walls was detected in some endodermis and pericycle cells at high CYN concentrationSinapis alba seedlings[69]
CYNFormation of callus-like tissue and necrosis in reed root cortex, cell swelling in pith tissue without necrosis in mustardPhragmites australis plantlets, Sinapis alba seedlings[69,72]
CYNInhibition of xylem differentiationSinapis alba seedlings[69]
IV. Physiology
MCY-LR purified and MCY-RR, -LR, -YR, -(H4)YR, -WR, and -FR in crude extractInhibition/alteration of photosynthesis, decreased chlorophyll, carotenoid content, altered chl a/chl b ratio, alterations in chlorophyll fluorescence parametersCeratophyllum demersum, Elodea canadensis, Lemna minor, L. gibba, Lens esculenta, Lolium perenne, Myriophyllum spicatum, Phaseolus vulgaris, Phragmites australis, Pisum sativum, Potamogeton sps., Sinapis alba, Solanum tuberosum tissue culture, Spinacia oleracea variants, Spirodela oligorrhiza, Triticum durum, Vicia faba inoculated with rhizobial strains, Zea mays[45,46,47,48,52,59,61,62,63,64,68,82,83,84,85]
MCY-LRAfter transient induction, inhibited anthocyanin accumulation in the cotyledonsSinapis alba[51,52]
MCY-LR, M. aeruginosa toxic cultureInhibition of medium, nutrient uptake/absorbtion rates of phosphorus and nitrogen, nitrogen assimilation;
Increase of mineral nutrients content in roots per fresh weight
Lens esculenta, Phaseolus vulgaris, Pisum sativum, Potamogeton sps., Triticum durum, Vicia faba inoculated withrhizobial strains, Zea mays[46,61,67,68,85]
MCY-LRDecreased water and protein contentCeratophyllum demersum[63]
CYN in crude extractDecreased chlorophyll content or/and changes in the chl- a/chl-b ratioHydrilla verticillata, Sinapis alba[52,77]
CYN purified and in crude extractSoluble protein content per unit fresh weight showed mild increases, especially in W. arrhiza, increases of tubulin content in reed rootsLemna minor, Phragmites australis plantlets, Wolffia arrhiza[72,73]
V. Enzymology
MCY-LR, MCY-RR, -LW, -LR, -LR in crude extractInhibition of protein-phosphatases PP1 and PP2A: in vitro inhibition of active forms of PP1 and PP2A in diluted seed extract (both PP1 and PP2A IC50: ~0.1 nM); in vivo inhibition of PP1 and PP2ABrassica napus seed extract, Medicago sativa, Phragmites australis, Sinapis alba [9,43,69,80,86]
MCY-LRInhibition of PP1 and PP2A, blocking of sucrose-inducible gene expression (mRNAs of β-amylase, sporamin, AGPase)Ipomoea batatas, transgenic Nicotiana tabacum[87]
MCY-LRInhibition of PP2A, the major sucrose-phosphate synthase (SPS) phosphatase blocking of the light-induced activation of SPS and decreasing sucrose biosynthesis and CO2 fixationSpinacia oleracea[88]
MCY-LRDisturbance of jasmonic acid (JA) signal transduction; abrogation of the response to JA (both the increase in the specific activity of acid phosphatase (AP) and the reduction in overall protein content shows opposite tendency)Nicotiana tabacum[89]
MCY-LR (purified and in extract)Alterations in activities of hydrolase enzymes: Changes in activity of constitutive acid phosphatase and RNase; induction of ssDNase activities; PCD associated changes of ssDNase and dsDNase activities in plant cellsSpirodela oligorrhiza Sinapis alba Phragmites australis[48,51,90]
MCY-LR, -LF, -LR in extractLipid peroxidation, increased α- and β-tocopherol concentration (as a lipid antioxidant)Arabidopsis thaliana cell suspension, Medicago sativa, Triticum aestivum [91,92,93]
MCY-LR, -RR, -LF, -LW, -WR both in purified and in crude cyanobacterial extracts Phenomena induced by oxidative stress: in vitro reaction of MCY-LR and extracted plant GST producing GSH-MCY-LR conjugate, identification of in vivo formed GSH-MCY-LR conjugate; formation of H2O2, other ROS, increase in phenolic compounds, phenylalanine ammonia lyase (PAL), polyphenol oxidase (PPO) activities, concentration of endogenous nitric oxide (NO); decrease/alterations in glutathione pool; reduced glutathione (GSH) and glutathione disulfide concentration; induction/alterations of oxidative stress enzyme activities: microsomal and cytosolic/soluble glutathione-S-transferase (mGST and sGST), -peroxidases (GPx), -glutathione reductase (GR), ascorbate peroxidase (APX, POD), superoxide dismutase (SOD), catalase (CAT)Arabidopsis thaliana cell suspension, Brassica napus, Brassica rapa, Ceratophyllum demersum, Elodea canadensis, Lemna gibba, L. minor, Lepidium sativum, Medicago sativa, Myriophyllum spicatum, Oryza sativa, Phaseolus vulgaris, Phragmites australis , Sinapis alba, Spinacia oleracea variants, tobacco BY-2 cell suspension, Triticum aestivum, Vicia faba inoculated with rhizobial strains, Vigna unguiculata species variants[38,50,52,53,55,59,62,65,68,70,82,91,92,94,95,9697,98,99,100]
MCY-LR in extractInhibited production of nitric oxide (NO), decreased auxin (IAA) concentration in rootsOryza sativa[81,101]
CYNAlteration in protein synthesis: CYN inhibited the eukaryotic protein synthesis apparatus with similar potency in plant and mammalian cell extracts, partial inhibition of protein production in germinating pollen tubeswheat germ extract, Nicotiana tabacum[79,102]
CYNSignificantly decreased PP1 and PP2A activities in extracts of CYN treated plants (CYN did not cause significant decrease in PP1 activity in vitro)Sinapis alba[69]
CYN purified and in crude extractProtease isoenzyme activity gels showed significant alterations in protease enzyme pattern and activities; crude extract induced an increase of total protease activity at pH 5 and pH 8, while purified CYN increased the activity only at lower concentration regimes (0.01—1 μg mL−1) Lemna minor, Wolffia arrhiza[73]
CYN purified and in crude extractInduction of oxidative stress enzyme activities: Increased GST, GPx activities; increased POD activity only at low (0.05 μg mL−1) concentration—transient effectOryza sativa, Sinapis alba[52,78]

5. Effects of MCY on Plant Mitotic and Non-Mitotic Chromatin

5.1. Mitotic Chromatin

MCY-LR exerts a characteristic dose-dependent effect on the mitosis of animal cells. It stimulates transiently mitotic activity (at low doses of the toxin or at shorter exposure times). In contrast, at higher MCY-LR doses, mitosis is inhibited, that is correlated with the induction of PCD and necrosis [103,104]. This phenomenon was named as “dualistic response” and it is probably related to the following mechanism: At low concentrations, the partial inhibition of PP1 and PP2A induces the maintenance of phosphorylation state of certain mitogen activated protein kinases (MAPKs), thereby activates them, causing stimulation of entry to the M phase of cell cycle. A severe inhibition of PP1 and PP2A will induce a general disturbance of cell functioning that finally leads to cell death [104].
Similar observations have been made in plants. Stimulation of mitosis was observed at low MCY-LR concentrations in several model systems, while inhibition of mitotic activity occurs in long-term treated, non-synchronized root tip meristematic cells of Phragmites australis and Sinapis alba. The inhibition of mitosis could be correlated to the inhibition of growth. At a close examination of distinct mitotic phases, the toxin stimulates early as well as late mitosis at different doses at relatively long-term exposures of roots [58,69,80,105]. A characteristic effect could be observed in shoot tip meristems of Ceratophyllum demersum, where MCY-LR clearly blocked cells in prophase/prometaphase [63]. Further studies were focused on root meristems in order to elucidate the mechanisms of MCY-LR induced mitotic changes. In non-synchronized root cells treated at relatively long term with MCY-LR a total arrest cells in early mitosis was not observed, but at certain concentrations it increased early mitosis indices, indicating that it may either inhibit progression of mitosis or change the duration and inhibit exit from mitosis [80,105]. Interestingly, dose-dependent short-term (4–30 h) toxin exposures of hydroxylurea-synchronized cells of V. faba showed apparently opposite effects: At low doses (1 μg mL−1), MCY-LR did not increase significantly the rate of cell division, but increased late mitosis index and delayed metaphase-anaphase transition [105]. This effect on early-late mitosis transition was similar to that observed in Tradescantia stamen hair cells microinjected with the toxin [106]. Since disturbances in the onset of anaphase were observed in MCY-LR treated mammalian (CHO-K1) cells too [103], this effect of the toxin might be universal in eukaryotic cells. At higher dose (10 μg mL−1), there was a delayed entry into M phase, but the acceleration of cell cycle and overall stimulation of mitosis, with a relatively low rate of late mitosis in V. faba [105]. These effects show that depending on the doses used at short-term treatments, MCY-LR delays entry to mitosis or the transition of cells to late mitosis and exit from late mitosis but allows the onset of a new cell cycle. Thus, it does not block completely progression of mitosis. What could be the cause of this? Since the toxin exerted its effects at low exposure times, one may assume that those effects could be correlated directly to the inhibition of PP1 and PP2A and the consequent hyperphosphorylation of proteins involved in the regulation of late mitosis. In addition, protein phosphatase inhibition assay showed that mitotic alterations are correlated with decreases of PP1 and PP2A activities in P. australis roots exposed to MCY-LR for 10 days [80]. Thus, the functioning of specific proteins could be changed. Indeed to date, three phenomena could be detected in plant cells (i) alterations in the organization of mitotic MTs; (ii) histone H3 hyperphosphorylation; (iii) abnormal sister chromatid segregation, the appearance of lagging chromosomes and micronuclei at the end of mitosis. Below, we will analyze these phenomena in detail.
Several studies proved MCY-induced alterations of mitotic spindle formation and consequently, abnormal chromosome movement during animal and human cell division. Characteristic anomalies were monopolar, multipolar or disrupted spindles [103,107,108,109]. Similar spindle anomalies were observed in plant cells, too ([69,80,105]; Figure 1a,b of this study). These alterations are important in the context of chromatin dynamics during mitosis: all of them are correlated with abnormal sister chromatid segregation [103,110,111]. This was the situation in MCY-LR treated plant cells ([69,80,105]; Table 2 of this study). Since plant cells possess diffuse MTOCs and not centrioles, these similarities may indicate that besides MT nucleation, other regulatory mechanisms common to animal and plant cells may occur at MCY exposures. Moreover, the disturbance of phragmoplast organization was observed in MCY-LR treated cells that may be due to mechanisms similar to the formation of spindle anomalies [80,105]. MAPs are good candidates to such mechanisms: Their binding to MTs is involved in the regulation of correct MT assembly/disassembly, bundling and stability both in animal and plant cells [26,33,108]. The binding of several MAPs is of crucial importance for correct mitotic MT assembly and depends on their phosphorylation state. For example, proteins of the MAP65 family found in Arabidopsis thaliana and Nicotiana tabacum are not able of mitotic or non-mitotic MT binding, when they are phosphorylated, e.g., PP1 and PP2A do not dephosphorylate them [32,112]. AtMAP65-1 regulates mitotic spindle and phragmoplast assembly by promoting MT bundling. Its hyperphosphorylation leads to spindle destabilization and blocks phragmoplast expansion [32]. Therefore, the same post-translational modification of a MAP alters two types of mitotic MT organization. It should be noted that the Arabidopsis ple4 genotype that bears a mutation in the AtMAP65-3/PLEIADE gene is characterized by alteration of AtMAP65-1 binding to MTs and multinucleate cells in root meristems: this mutant shows a clear relationship between mitotic MT assembly and correct chromatid segregation [113]. We can conclude that phosphorylation dependent MAPs might be targets of MCY action in plant cells and could be related to the direct biochemical effects of the toxin (i.e., PP1 and PP2A inhibition) on mitotic and non-mitotic MT dynamics and consequent chromatin organization. However, such studies are still awaited.
Figure 1. Conventional fluorescence (gn) and confocal microscopy (af) images of characteristic chromatin and microtubule alterations induced by cyanotoxins in plant cells as revealed by histochemical and immunohistochemical methods (see [105] as an example for methods). Chromatin label is shown in blue, Ser10-phosphorylated histone H3 in green and microtubules in red. (a) Normal metaphase spindle from in vitro cultured control Phragmites australis root tip meristem; (b) P. australis root tip meristem cell treated with 10 μg mL−1 MCY-LR. Note abnormal bundling of microtubules and spindle disruption; (c) Prophase cell of a P. australis root tip with normal preprophase band (PPB); (d) Split PPB (arrow) of a P. australis cell treated with 10 μg mL−1 CYN; (e) Control Vicia faba root tip meristem cell labeled for phospho-histone H3 Ser10. Histone H3 is phosphorylated mainly at the pericentromeric regions of metaphase chromosomes; (f) V. faba cell treated with 20 μg mL−1 MCY-LR. Note histone H3 hyperphosphorylation both at pericentromeric regions and chromosome arms; (g) Nuclei of V. faba meristematic cells labeled with DAPI. Micronuclei occur only sporadically (arrow); (h) Abundance of micronuclei in V. faba meristem treated with 20 μg mL−1 MCY-LR (arrows); (i) Nuclei of Sinapis alba cells from root elongation zone labeled with DAPI. No micronuclei or nucleus fragmentation can be detected; (j) Fragmented nucleus of a Sinapis alba cell from root elongation zone treated with 1 μg mL−1 MCY-LR; (k) Control P. australis cells from root elongation zone, with normally oriented cortical microtubules (CMTs); (l) P.australis root cells treated with 20 µg mL−1 MCY-LR showing CMT depolymerization and inhibition of cell elongation; (m,n) P.australis root cells treated with 10 μg mL−1 CYN, showing a cell with decrease of MT density (m) and a cell with CMT reorientation, inhibition of elongation and stimulation of radial expansion of cells at the transition of meristematic-elongation zone (n). Scalebars: 15 μm (a,b), 10 µm (c,d,gj), 5 µm (e,f); 30 µm (k,l), 25 µm (m,n). Micrographs taken by D. Beyer (a,b,e,f), C. Máthé (gn) and J. Roszik (c,d).
Figure 1. Conventional fluorescence (gn) and confocal microscopy (af) images of characteristic chromatin and microtubule alterations induced by cyanotoxins in plant cells as revealed by histochemical and immunohistochemical methods (see [105] as an example for methods). Chromatin label is shown in blue, Ser10-phosphorylated histone H3 in green and microtubules in red. (a) Normal metaphase spindle from in vitro cultured control Phragmites australis root tip meristem; (b) P. australis root tip meristem cell treated with 10 μg mL−1 MCY-LR. Note abnormal bundling of microtubules and spindle disruption; (c) Prophase cell of a P. australis root tip with normal preprophase band (PPB); (d) Split PPB (arrow) of a P. australis cell treated with 10 μg mL−1 CYN; (e) Control Vicia faba root tip meristem cell labeled for phospho-histone H3 Ser10. Histone H3 is phosphorylated mainly at the pericentromeric regions of metaphase chromosomes; (f) V. faba cell treated with 20 μg mL−1 MCY-LR. Note histone H3 hyperphosphorylation both at pericentromeric regions and chromosome arms; (g) Nuclei of V. faba meristematic cells labeled with DAPI. Micronuclei occur only sporadically (arrow); (h) Abundance of micronuclei in V. faba meristem treated with 20 μg mL−1 MCY-LR (arrows); (i) Nuclei of Sinapis alba cells from root elongation zone labeled with DAPI. No micronuclei or nucleus fragmentation can be detected; (j) Fragmented nucleus of a Sinapis alba cell from root elongation zone treated with 1 μg mL−1 MCY-LR; (k) Control P. australis cells from root elongation zone, with normally oriented cortical microtubules (CMTs); (l) P.australis root cells treated with 20 µg mL−1 MCY-LR showing CMT depolymerization and inhibition of cell elongation; (m,n) P.australis root cells treated with 10 μg mL−1 CYN, showing a cell with decrease of MT density (m) and a cell with CMT reorientation, inhibition of elongation and stimulation of radial expansion of cells at the transition of meristematic-elongation zone (n). Scalebars: 15 μm (a,b), 10 µm (c,d,gj), 5 µm (e,f); 30 µm (k,l), 25 µm (m,n). Micrographs taken by D. Beyer (a,b,e,f), C. Máthé (gn) and J. Roszik (c,d).
Marinedrugs 11 03689 g001
Table 2. A survey of the effects of MCY and CYN on plant chromatin organization.
Table 2. A survey of the effects of MCY and CYN on plant chromatin organization.
CyanotoxinPlant materialEffectMechanisms elucidated or probably involvedReferences
1.Mitotic chromatin
MCY-LR, MCY-RRTradescantia virginiana stamen hair cellsIncrease of metaphase transit time, temporary delay of sister chromatid segregationInhibition of serine-threonine protein phosphatases (type 1 and 2A)[106]
MCY-LRRoot tip meristems of Phragmites australis, Sinapis albaDualistic response: Mitotic activity increases at low, decreases at high cyanotoxin concentrationsInhibition of serine-threonine protein phosphatases (type 1 and 2A)[69,80]
MCY-LRRoot tip meristems of P. australis, Vicia fabaTransient increase of early and late mitotic activityInhibition of serine-threonine protein phosphatases (type 1 and 2A)[80,105]
MCY-LRShoot tip meristems of Ceratophyllum demersumArrest of mitosis in prophase/prometaphaseBlocking of MT dynamics at early mitosis; inhibition of serine-threonine protein phosphatases (type 1 and 2A)[63]
MCY-LRRoot tip meristems of V. fabaAcceleration of cell cycle at exposure to high (10 μg mL−1) toxin concentrationInhibition of serine- threonine protein phosphatases (type 1 and 2A)[105]
MCY-LR, MCY-XRRoot tip meristems of P. australis, S. alba, V. faba, Allium cepaDelay of metaphase/anaphase transition, incomplete sister chromatid segregation, the formation of micronucleiDisruption of mitotic MT structures; inhibition of serine-threonine protein phosphatases (type 1 and 2A); * Hyperphosphorylation of histone H3 at Ser10[69,80,105,114]
CYNRoot tip meristems of P. australisAlteration of early mitotic activity (increase of prophase/prometaphase, decrease of metaphase indices)* Alteration of PPB development, probably due to protein synthesis inhibition[72]
CYNRoot tip meristems of P. australis, S. albaAlteration of sister chromatid segregationDisruption of mitotic MT structures[72]
2. Non-mitotic chromatin
MCY-RRTobacco BY-2 cellsPerinuclear chromatin marginalizationOxidative stress (ROS generation)[115,116]
MCY-LR, MCY-RRTobacco BY-2 cells, Vallisneria natans mesophyll cells, P. australis root tipsChromatin condensationOxidative stress, induction of SSP nuclease activities[90,115,117118]
MCY-RR, MCY-LRTobacco BY-2 cells, S. alba rootsNuclear fragmentationOxidative stressFigure 1j and Figure 2g-j, [115]
MCYs (cyanobacterial extract)Oryza sativa seedlingsDNA fragmentation (smearing)Probably oxidative stress[119]
* Specific effect, probably due to the direct biochemical action of the given cyanotoxin.
An early proof showing that MCY-LR interferes with histone H3 phosphorylation at Ser10, came from the study of maize meiocytes [120]. Later on it turned out that in relation to mitotic chromatin dynamics, one of the most important post-translational modifications of histone H3 and its counterpart, centromeric histone H3 (CENH3, CENPA) is phosphorylation at specific Ser/Thr residues at their N-terminal regions [121,122]. These types of histone modifications are detected most commonly in mitotic and meiotic plant cells: during mitosis, phosphorylation starts in prophase, culminates in pericentromeric and centromeric regions of metaphase chromosomes and decreases gradually at late mitosis to be practically undetectable at the end of cytokinesis [121,122]. They are important for correct chromatin condensation and cohesion at prophase-metaphase and sister chromatid segregation at anaphase in all eukaryotic cells [123,124]. As an inhibitor of PP1 and PP2A, MCY-LR was expected to induce histone H3 hyperphosphorylation during plant mitosis. Indeed, in V. faba root tips the toxin induced high levels of phosphorylation not only at the pericentromeric regions, but in chromosome arms as well. This was accompanied by chromosome hypercondensation in metaphase and the formation of lagging chromosomes leading to the formation of micronuclei (Figure 1e–h; [105]). Therefore, sister chromatid segregation anomalies induced by MCY are related not only to the formation of aberrant mitotic MT structures, but on direct modifications at the chromatin level, too. Histone H3 hyperphosphorylation could be a suitable marker for the effects of MCY at the chromatin level. Whether the toxin induces this histone modification directly through the inhibition of PP1 and PP2A, or the modification is the result of hyperphosphorylation of histone-regulatory proteins active during the M phase of cell cycle, should be subject of future research. For example, the hyperphosphorylation of certain Aurora kinases induces their activation, that is, their capacity of histone phosphorylation [125]. Therefore one may assume that MCY-LR, as a phosphatase inhibitor induces histone H3 hyperphosphorylation not only by the direct inhibition of its dephosphorylation, but through hyperactivation of such kinases as well in V. faba. It should be noted however, that Aurora kinases with important roles in H3 phosphorylation are localized in centromeric regions of plant chromosomes [126,127]. Since we observed that MCY-LR induces H3 hyperphosphorylation both in pericentromeric regions and chromosome arms, it is unlikely that the toxin acts solely through this mechanism in V. faba: direct inhibition of H3 dephosphorylation may be more important in this respect.
Microfilaments—the other component of plant cell cytoskeleton-play an important role in cell division and the determination of cell shape/movements in eukaryotic cells [27,128]. The alteration of protein dephosphorylation by other drugs than MCY induces alterations in F-actin organization in guard cells and soybean cultured cells [129,130]. MCY-LR does induce MF disruptions in animal cells and this was related to programmed cell death or necrosis [107,109,131]. To our best knowledge, similar studies on plant cells are lacking, so this issue needs to be studied.

5.2. Non-Mitotic Chromatin

It has been known for a long time that MCY induces PCD/apoptosis or necrosis in animal cells (see [109,132] for examples). In an early study, nuclear fragmentation induced by MCY-LR was detected in different rat cell types [133]. It should be noted that such nuclear fragmentation/micronucleus formation could not be detected in MCY-LR treated young mouse erythrocytes or human lymphocytes [134].
MCY-LR induces the formation of micronuclei not only by the formation of lagging chromosomes, but in non-mitotic plant cells as well. Altered sister chromatid segregation is characterized by the formation of one micronucleus per cell (Figure 1g,h of this study). In non-dividing cells, the formation of multiple micronuclei and chromatin fragmentation could be observed in several studies ([115]; Figure 1i,j and Figure 2g–j of this study). This phenomenon is generally attributed to PCD [135,136] and shows the possible role of MCY in its induction in plants. Indeed, there is a significant number of studies showing the subsequent events leading to nuclear fragmentation in MCY treated plant cells. In general, firstly chromatin condensation and perinuclear chromatin marginalization occurs, followed by nuclear and finally, DNA fragmentation [90,115,116,117,118,137]. MCY induced internucleosomal DNA fragmentation (DNA laddering) has been observed in several rat and human cell types [137,138]. In contrast, this could not be detected in MCY treated plant cells undergoing PCD. Instead, a non-specific DNA fragmentation (smearing) was observed on agarose gels [119]. Therefore, MCY induced PCD or apoptosis, a type of PCD might develop by partially different mechanisms in plant and animal cells.
As we mentioned before Section 4, MCYs are able of inducing necrosis in a significant number of plant species/systems. Necrosis is a “non-specific”/not programmed cell death type characterized by general disruption of cell structure in eukaryotes and in particular, plant cells [118]. PCD/apoptosis and necrosis can be induced by different MCY-RR concentrations in tobacco BY-2 cells [117] and a similar finding has been made by Jámbrik et al. [90] in MCY-LR treated P. australis roots. However, the clear separation of apoptotic and necrotic effects of microcystins needs further study: for MCY-LR treated mustard roots, it seems that the same cell undergoes PCD, followed by necrosis: nuclear fragmentation occurs firstly and subsequent disappearance of DNA from cells is observed (Figure 2b,f). In relation to this, it is known that PCD/apoptosis and necrosis are frequently subsequent events in plants [36]. Although in several cases chromatin marginalization was thought to be essential for the initial steps of PCD, when P. australis roots were treated with high MCY-LR doses, chromatin marginalization could be associated to necrosis and not PCD [90]. Indeed, this type of chromatin change can be associated to initial steps of necrosis of animal cells [139].
What are the possible mechanisms of cell death induction by MCY in plants? Several studies in animal and plant cells indicate that MCYs induce the elevation of ROS and the activities of enzymes involved in oxygen radical scavenging ([52,70,91,98,99,140]; Table 1 of this study). MCYs are able of binding GSH, thereby of forming glutathione conjugates by enzymatic or non-enzymatic mechanisms in a variety of organisms including plants, invertebrates and fish [47,94]. This is a detoxifying mechanism, but it induces the reduction of GSH and general glutathione pool in cells, contributing to oxidative stress [94,98]. As a general mechanism, the elevation of ROS induces DNA damage. If repair mechanisms fail, increases in the activities of nucleases and proteases will lead either to chromatin condensation and fragmentation and finally, PCD, or to necrosis [141,142]. It is believed that MCYs induce such processes. ROS elevation was linked to DNA damage and/or nuclear degradation in animal (see [143] for an example) and in plant cells [115,116]. The cyanotoxin induced increase of protease and nuclease activities as well as increases in peroxidase activities involved in oxygen radical scavenging have been demonstrated in several terrestrial and aquatic model plant systems [51,52,73,90].
Figure 2. MCY-LR induces callus formation (swollen cells), PCD and necrosis in the rhizodermis and cortex of S. alba primary roots. (a) Control root collar; (b) Same tissues stained with DAPI; (c) Cross-Section of main root-hypocotyl transition zone of seedlings treated with 20 μg mL−1 MCY-LR, showing the formation of a callus-like tissue (CA), necrosis (NT) and intensive lignification of endodermis and stele as shown by phloroglucin-HCl staining (arrow); (d) High autofluorescence of inner root tissues (arrow) induced by 20 μg mL−1 MCY-LR exposure: autofluorescence fades away in necrotic tissue (NT); (e) 20 μg mL−1 MCY-LR induces necrosis of rhizodermis and adjacent tissues (NT); (f) Same tissues as in (e) stained with DAPI, nuclei are absent in necrotic cells (NT), and the fragmentation of nuclei can be observed in cells neighboring necrotic tissue (arrow); (gj) Nuclei from the root tip meristem-elongation zone transition; (g) control; (hj) Treatment with 5 μg mL−1 MCY-LR: nuclear blebbing (h,i) leading to the fragmentation of chromatin (j); Scalebars: 80 μm (a,b,e,f), 200 μm (c,d) and 15 μm (gj). Micrographs taken by M. M-Hamvas (a,c,e), C. Máthé (b,d,fj).
Figure 2. MCY-LR induces callus formation (swollen cells), PCD and necrosis in the rhizodermis and cortex of S. alba primary roots. (a) Control root collar; (b) Same tissues stained with DAPI; (c) Cross-Section of main root-hypocotyl transition zone of seedlings treated with 20 μg mL−1 MCY-LR, showing the formation of a callus-like tissue (CA), necrosis (NT) and intensive lignification of endodermis and stele as shown by phloroglucin-HCl staining (arrow); (d) High autofluorescence of inner root tissues (arrow) induced by 20 μg mL−1 MCY-LR exposure: autofluorescence fades away in necrotic tissue (NT); (e) 20 μg mL−1 MCY-LR induces necrosis of rhizodermis and adjacent tissues (NT); (f) Same tissues as in (e) stained with DAPI, nuclei are absent in necrotic cells (NT), and the fragmentation of nuclei can be observed in cells neighboring necrotic tissue (arrow); (gj) Nuclei from the root tip meristem-elongation zone transition; (g) control; (hj) Treatment with 5 μg mL−1 MCY-LR: nuclear blebbing (h,i) leading to the fragmentation of chromatin (j); Scalebars: 80 μm (a,b,e,f), 200 μm (c,d) and 15 μm (gj). Micrographs taken by M. M-Hamvas (a,c,e), C. Máthé (b,d,fj).
Marinedrugs 11 03689 g002
An alternative mechanism to MCY induced ROS elevation is related to the phosphatase inhibitory effect of cyanotoxin. According to this, the inhibition of protein phosphatases will influence signal transduction pathways that alter stress related gene expression, leading to decrease of reducing power and increase of ROS [144]. Although protein phosphatase inhibition in general can induce directly ROS elevation [145], there is not enough evidence for such particular effect of MCY in plants. It is worth mentioning that in animal cells, MCY-LR can induce the inhibition of dephosphorylation of proteins involved in ROS formation [146].
In addition to chromatin disorganization, PCD and necrosis is associated with MT destabilization [147]. Indeed, MCY-LR does induce CMT depolymerization (Phragmites australis root tips) or reorientation (Ceratophyllum demersum shoot tips). Besides cell death, these processes are correlated to changes of cell shape and alterations in the morphology of axial organs (Figure 1k,l; [63,80]).

6. Effects of CYN on Plant Mitotic and Non-Mitotic Chromatin

6.1. Mitotic Chromatin

In animal cells, CYN induces a general inhibition of mitotic activity [148]. Moreover, it induces micronucleus formation via chromosome loss in human WIL2-NS lymphoblastoid cell line [22]. Micronucleus formation was observed both in dividing and non-dividing human Caco-2 and HepaRG cell lines, too [149]. Concerning plants, CYN induces transient increase of mitotic activity of P. australis root tip meristematic cells, but this is not observable in S. alba [69,72]. Therefore, a “dualistic response” or mitotic stimulation cannot be generalized yet for plants, as for MCY (see Section 5.1). However as for MCY, CYN induces spindle disruptions in both plant systems and disorganization of phragmoplasts in P. australis. These changes are accompanied with disturbances in sister chromatid segregation during anaphase [69,72]. The mechanisms laying behind these phenomena are largely unknown, but two hypotheses can be drawn out: (i) The inhibition of MAP synthesis, as CYN is a protein synthesis inhibitor; (ii) Since CYN inhibits PP1 and PP2A in a non-specific way [69,150], it may influence the functioning of phosphorylation-dependent MAPs, as MCY.
CYN induces the formation of double or split PPBs at least in P. australis roots ([72]; Figure 1c,d of this study). It remains to be established whether CYN induced PPB alterations are general in plants. Alterations in PPB assembly can be attributed to the protein synthesis inhibitory effect of the cyanotoxin, since other inhibitors, like cycloheximide have similar effects on plant cells [30]. In addition, disturbances of auxin transport (disfunctions of auxin transport proteins) induce the formation of abnormal PPBs, leading to changes in cell division plane [28]. PPB is believed to regulate cell division plane and in consequence, plant morphogenesis [26,30]; therefore, anomalies in its development may contribute to CYN induced changes in plant growth and morphogenesis (see Table 1). The possible mechanism of altered PPB formation is thought to be disturbance of MAP formation [72].

6.2. Non-Mitotic Chromatin

There is a significant number of studies on animal and human cells indicating that CYN induces PCD [17]. These include nuclear fragmentation/micronucleus formation in non-mitotic cells [149,151] and DNA degradation as shown by electrophoresis of purified DNA and the Comet assay [142,151,152]. The elevation of ROS and its possible role in the development of DNA damage and PCD/apoptosis has also been shown in fish as well as human cell lines [153,154]. However a recent study has shown that although CYN induces ROS elevation, it is not correlated with DNA damage and PCD in HepG2 cells [155]. Therefore, the correlation of CYN induced ROS elevation and DNA degradation is still controversial for animal and human systems. In animal cells (CHO K1 cells), CYN induces cytoskeletal reorganization, thereby inhibiting growth and inducing PCD and necrosis [109,156].
To date, there are no published data on the effects of CYN on chromatin organization in differentiating plant cells. However, several studies indicate that the cyanotoxin may induce PCD in plant cells. These include: (i) Increases in several protease isoenzyme activities in the aquatic plants Lemna minor and Wolffia arrhiza [73]; (ii) Increasing of enzyme activities that indicate the elevation of ROS and oxidative stress: GST, glutathione peroxidase in rice roots, peroxidase in Sinapis alba seedlings [52,78]; (iii) CYN reduces MT density and induces MT reorientation in Phragmites australis root cells in the meristematic and elongation zone of roots that inhibit growth, alter root morphology and is probably related to cell death. Reduction of MT density is not caused by the inhibition of tubulin synthesis, since CYN increases the production of this protein. Rather, it can be correlated to the synthesis and functioning of MAPs involved in the regulation of cytoskeletal stability (Figure 1k,m,n; [72]).
It is worth mentioning that the cyanotoxin induces the formation of necrotic-like tissue in P. australis roots, but not in S. alba seedlings [69,72]. Other plant systems should be studied in this respect in order to reveal a general rule for higher plants concerning induction of cell death.

7. Conclusions

The effects of MCY and CYN on chromatin organization in plants are summarized on Table 2 and Figure 3. MCY induces anomalies at the chromatin level in mitotic plant cells, namely it alters sister chromatid segregation leading to the formation of lagging chromosomes which will turn into micronuclei as they decondense at the end of mitosis. To date, two main mechanisms have been elucidated in relation to this: (i) Abnormal organization of mitotic MTs, including disruption of mitotic spindles and phragmoplasts, and the formation of monopolar/multipolar spindles; (ii) Hyperphosphorylation of histone H3. These processes can be correlated directly to the protein phosphatase inhibitory effect of the toxin. Concerning non-dividing cells, MCY induces changes characteristic for the onset of PCD and/or apoptosis: Chromatin marginalization and condensation, nuclear fragmentation, DNA degradation. This is probably related to the elevation of ROS and increases of SSP nuclease and protease activities.
Concerning CYN, it induces spindle-phragmoplast disruption and abnormal PPB formation, probably due to its protein synthesis inhibitory effects in mitotic plant cells. To date, there is no direct evidence of CYN induced PCD in plant cells, but there are several indirect proofs e.g., modulation of protease activities and increases in ROS and oxygen radical scavenging enzymes-that raise the possibility of inducing this phenomenon. CYN-induced necrosis has been proven in in vitro grown P. australis plantlets only.
A generalized diagram for the effects of MCY on plant chromatin is shown on Figure 3. It should be noted that concerning CYN, there is still little knowledge in plants concerning this topic.
As proposed in Section 5 and Section 6, the effects of cyanotoxins on plant chromatin organization raise many future questions and research directions. These will contribute to a better understanding of the effects of MCY and CYN on plant cells and will serve as tools for a general understanding of plant cell functioning.
Figure 3. Proposed network for the effects of MCY on plant chromatin organization and functioning. Continuous arrows represent pathways already elucidated, while dashed arrows or question marks represent cellular processes or pathways that need further proof. Color code: red—mitotic cells, blue—non-mitotic cells, purple—all cell types. Abbreviations: APC/C-anaphase promoting complex-cyclosome; CMT-cortical microtubules; GSH-reduced glutathione; MAP-microtubule associated protein; MAPK-mitogen activated protein kinase; MT-microtubule; PCD-programmed cell death; PP1-protein phosphatase of type 1; PP2A-protein phosphatase of type 2A; ROS-reactive oxygen species.
Figure 3. Proposed network for the effects of MCY on plant chromatin organization and functioning. Continuous arrows represent pathways already elucidated, while dashed arrows or question marks represent cellular processes or pathways that need further proof. Color code: red—mitotic cells, blue—non-mitotic cells, purple—all cell types. Abbreviations: APC/C-anaphase promoting complex-cyclosome; CMT-cortical microtubules; GSH-reduced glutathione; MAP-microtubule associated protein; MAPK-mitogen activated protein kinase; MT-microtubule; PCD-programmed cell death; PP1-protein phosphatase of type 1; PP2A-protein phosphatase of type 2A; ROS-reactive oxygen species.
Marinedrugs 11 03689 g003

Acknowledgements

This work has been supported by Hungarian National Research Foundation Grants OTKA K81370, F046493, GVOP-3.2.1.-2004-04-0110/3.0, GVOP-TST 3.3.1-05/1-2005-05-0004/3.0. The work/publication is supported by the TÁMOP-4.2.2/B-10/1-2010-0024 project. The authors would like to thank Dániel Beyer for his useful comments on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nigrelli, R.F.; Stempien, M.F.; Ruggirei, G.D.; Liguori, V.R.; Cecil, J.T. Substances of potential biomedical importance from marine organisms. Fed. Proc. 1967, 26, 1197–1205. [Google Scholar]
  2. Bhatnagar, I.; Kim, S.-K. Immense essence of excellence: Marine microbial bioactive compounds. Mar. Drugs 2010, 8, 2673–2701. [Google Scholar] [CrossRef]
  3. Vasas, G.; Borbely, G.; Nanasi, P.; Nanasi, P.P. Alkaloids from Cyanobacteria with Diverse Powerful Bioactivities. Mini. Rev. Med. Chem. 2010, 10, 946–955. [Google Scholar] [CrossRef]
  4. De Pauw, N.; Persoone, G. Microalgae for Aquaculture. In Microalgal Biotechnology; Borowitzka, M.A., Borowitzka, L.J., Eds.; Cambridge University: Cambridge, UK, 1988; pp. 197–221. [Google Scholar]
  5. Carmichael, W.W. Cyanobacteria secondary metabolites—The cyanotoxins. J. Appl. Bacteriol. 1992, 72, 445–459. [Google Scholar] [CrossRef]
  6. Sivonen, K.K.; Jones, G. Cyanobacterial Toxins. In Toxic Cyanobacteria in Water; Chorus, I., Bartram, J., Eds.; Spon: London, UK, 1999; pp. 41–111. [Google Scholar]
  7. Moreira, C.; Azevedo, J.; Antunes, A.; Vasconcelos, V. Cylindrospermopsin: Occurrence, methods of detection and toxicology. J. Appl. Microbiol. 2012, 114, 605–620. [Google Scholar]
  8. Botes, D.P.; Wessels, P.L.; Kruger, H.; Runnegar, M.T.C.; Santikarn, S.; Smith, R.J.; Barna, J.C.J.; Williams, D.H. Structural studies on cyanoginosins-LR, YR, YA, and YM, peptide toxins from Microcystis aeruginosa. J. Chem. Soc. Perkin Trans. I 1985, 1, 2747–2748. [Google Scholar]
  9. MacKintosh, C.; Beattie, K.A.; Klumpp, S.; Cohen, P.; Codd, G.A. Cyanobacterial microcystin-LR is a potent and specific inhibitor of protein phosphatases 1 and 2A from both mammals and higher plants. FEBS Lett. 1990, 264, 187–192. [Google Scholar] [CrossRef]
  10. Zhang, L.; Zhao, S.; Deans Zirattu, S.; Lee, E.Y.C. Mutagenesis of the catalytic subunit of rabbit muscle protein phosphatase-1. Mol. Cell. Biochem. 1993, 127, 113–119. [Google Scholar]
  11. MacKintosh, R.W.; Dalby, K.N.; Campbell, D.G.; Cohen, P.T.W.; Cohen, P.; MacKintosh, C. The cyanobacterial toxin microcystin binds covalently to cysteine-273 on protein phosphatase 1. FEBS Lett. 1995, 371, 236–240. [Google Scholar] [CrossRef]
  12. Goldberg, J.; Huang, H.; Kwon, Y.; Greengard, P.; Nairn, A.C.; Kuriyan, J. Three dimensional structure of the catalytic subunit of protein serine/threonine phosphatase-1. Nature 1995, 376, 745–753. [Google Scholar] [CrossRef]
  13. Abdel-Rahman, S.; El Ayouty, Y.M.; Kamael, H.A. Characterization of heptapeptide toxins extracted from Microcystis aeruginosa (Egyptian isolate)—Comparison with some synthesized analogs. Int. J. Pept. Protein Res. 1993, 41, 1–7. [Google Scholar] [CrossRef]
  14. Bourne, D.G.; Jones, G.J.; Blakeley, R.L.; Jones, A.; Negri, A.P.; Riddles, P. Enzymatic pathway for the bacterial degradation of the cyanobacterial cyclic peptide toxin Microcystin-LR. Appl. Environ. Microbiol. 1996, 62, 4086–4094. [Google Scholar]
  15. Wiegand, C.; Pflugmacher, S. Ecotoxicological effects of selected cyanobacterial secondary metabolites: A short review. Toxicol. Appl. Pharmacol. 2005, 203, 201–218. [Google Scholar] [CrossRef]
  16. 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]
  17. Kinnear, S. Cylindrospermopsin: A decade of progress on bioaccumulation research. Mar. Drugs 2010, 8, 542–564. [Google Scholar] [CrossRef]
  18. Harada, K.I.; Ohtani, I.; Iwamoto, K.; Suzuki, M.; Watanabe, M.F.; Watanabe, M.; Terao, K. Isolation of cylindrospermopsin from a cyanobacterium Umezakia natans and its screening method. Toxicon 1994, 32, 73–84. [Google Scholar] [CrossRef]
  19. Froscio, S.M.; Humpage, A.R.; Burcham, P.C.; Falconer, I.R. Cylindrospermopsin-induced protein synthesis inhibition and its dissociation from acute toxicity in mouse hepatocytes. Environ. Toxicol. 2003, 18, 243–251. [Google Scholar] [CrossRef]
  20. Hawkins, P.R.; Runnegar, M.T.C.; Jackson, A.R.B.; Falconer, I.R. Severe hepatotoxicity caused by the tropical cyanobacterium (blue-green alga) Cylindrospermopsis raciborskii (Woloszynska) Seenaya and Subba Raju isolated from a domestic water supply reservoir. Appl. Environ. Microbiol. 1985, 50, 1292–1295. [Google Scholar]
  21. Runnegar, M.T.; Kong, S.M.; Zhong, Y.Z.; Lu, S.C. Inhibition of reduced glutathione synthesis by cyanobacterial alkaloid cylindrospermopsin in cultured hepatocytes. Biochem. Pharmacol. 1995, 49, 219–225. [Google Scholar] [CrossRef]
  22. Humpage, A.R.; Fenech, M.; Thomas, P.; Falconer, I.R. Micronucleus induction and chromosome loss in transformed human white cells indicate clastogenic and aneugenic action of the cyanobacterial toxin, cylindrospermopsin. Mutat. Res. 2000, 472, 155–161. [Google Scholar] [CrossRef]
  23. Humpage, A.R.; Fontaine, F.; Froscio, S.; Burcham, P.; Falconer, I.R. Cylindrospermopsin genotoxicity and cytotoxicity, role of cytochrome P-450 and oxidative stress. J. Toxicol. Environ. Health A 2005, 68, 739–753. [Google Scholar] [CrossRef]
  24. Mathur, J. The illuminated plant cell. Trends Plant Sci. 2007, 12, 506–513. [Google Scholar] [CrossRef]
  25. Goddard, R.H.; Wick, S.M.; Silflow, C.D.; Snustad, D.P. Microtubule components of the plant cell cytoskeleton. Plant Physiol. 1994, 104, 1–6. [Google Scholar]
  26. Baskin, T.I. The Cytoskeleton. In Biochemistry and Molecular Biology of Plants; Buchanan, B., Gruissem, W., Jones, R., Eds.; The American Society of Plant Biologists: Rockville, MD, USA, 2000; pp. 202–258. [Google Scholar]
  27. Mathur, J. Cell shape development in plants. Trends Plant Sci. 2004, 9, 583–590. [Google Scholar] [CrossRef]
  28. Dhonukshe, P.; Mathur, J.; Hülskamp, M.; Gadella, T.W.J. Microtubule plus-ends reveal essential links between intracellular polarization and localized modulation of endocytosis during division-plane establishment in plant cells. BMC Biol. 2005, 3. [Google Scholar] [CrossRef] [Green Version]
  29. Lee, Y.J.; Liu, B. Cytoskeletal Motor Proteins in Plant Cell Division. In Cell Division Control in Plants, Plant Cell Monographs 9; Verma, D.P.S., Hong, Z., Eds.; Springer: Berlin, Heidelberg, Germany, 2008; pp. 169–193. [Google Scholar]
  30. Mineyuki, T. The preprophase band of microtubules: Its function as a cytokinetic apparatus in higher plants. Int. Rev. Cytol. 1999, 187, 1–49. [Google Scholar] [CrossRef]
  31. Yokota, E.; Imamichi, N.; Tominaga, M.; Shimmen, T. Actin cytoskeleton is responsible for the change of cytoplasmic organization in root hair cells induced by a protein phosphatase inhibitor, calyculin A. Protoplasma 2000, 213, 184–193. [Google Scholar] [CrossRef]
  32. Smertenko, A.P.; Chang, H.-Y.; Sonobe, S.; Fenyk, S.I.; Weingartner, M.; Bögre, L.; Hussey, P.J. Control of the AtMAP65-1 interaction with microtubules through the cell cycle. J. Cell Sci. 2006, 119, 3227–3237. [Google Scholar] [CrossRef]
  33. Hamada, T. Microtubule associated proteins in higher plants. J. Plant Res. 2007, 120, 79–98. [Google Scholar] [CrossRef]
  34. Jones, A.M.; Dangl, J.L. Logjam at the Styx: Programmed cell death in plants. Trends Plant Sci. 1996, 1, 114–119. [Google Scholar] [CrossRef]
  35. Levine, A.; Pennell, R.I.; Alvarez, M.E.; Palmer, R.; Lamb, C. Calcium mediated apoptosis in a plant hypersensitive disease resistance response. Curr. Biol. 1996, 6, 427–437. [Google Scholar] [CrossRef]
  36. Kosslak, R.M.; Chamberlin, M.A.; Palmer, R.G.; Bowen, B.A. Programmed cell death in the root cortex of soybean root necrosis mutants. Plant J. 1997, 11, 729–745. [Google Scholar]
  37. White, S.H.; Duivenvoorden, L.J.; Fabbro, L.D. A decision-making framework for ecological impacts associated with the accumulation of cyanotoxins (cylindrospermopsin and microcystin). Lakes Reserv. Res. Manag. 2005, 10, 25–37. [Google Scholar] [CrossRef]
  38. Pflugmacher, S.; Jung, K.; Lundvall, L.; Neumann, S.; Peuthert, A. Effects of cyanobacterial toxins and cyanobacterial cell-free crude extract on germination of alfalfa (Medicago sativa) and induction of oxidative stress. Environ. Toxicol. Chem. 2006, 25, 2381–2387. [Google Scholar] [CrossRef]
  39. Babica, P.; Bláha, L.; Maršálek, B. Exploringthe natural role of microcystins—A reviewof effects on photoautotrophic organisms. J. Phycol. 2006, 42, 9–20. [Google Scholar] [CrossRef]
  40. Saqrane, S.; Oudra, B. CyanoHAB occurrence and water irrigation cyanotoxin contamination: Ecological impacts and potential health risks. Toxins 2009, 1, 113–122. [Google Scholar] [CrossRef]
  41. Yamasaki, S. Probable effects of algal bloom on the growth of Phragmites australis (Cav.) Trin. ex. Steud. J. Plant Res. 1993, 106, 113–120. [Google Scholar] [CrossRef]
  42. Kós, P.; Gorzó, G.; Surányi, G.; Borbély, G. Simple and efficient method for isolation and measurement of cyanobacterial hepatotoxins by plant tests (Sinapis alba L.). Anal. Biochem. 1995, 225, 49–53. [Google Scholar] [CrossRef]
  43. Kurki-Helasmo, K.; Meriluoto, J. Microcystin uptake inhibits growth and protein phosphatase activity in mustard (Sinapis alba L.) seedlings. Toxicon 1998, 36, 1921–1926. [Google Scholar] [CrossRef]
  44. Casanova, T.M.; Burch, D.M.; Brock, A.M.; Bond, M.P. Does toxic Microcystis aeruginosa affect aquatic plant establishment? Environ. Toxicol. 1999, 14, 97–109. [Google Scholar] [CrossRef]
  45. Weiss, J.; Liebert, H.P.; Braune, W. Influence of microcystin-RR on growth and photosynthetic capacity of the duckweed Lemna minor L. J. Appl. Bot. Angew. Bot. 2000, 74, 100–105. [Google Scholar]
  46. McElhiney, J.; Lawton, L.A.; Leifert, C. Investigations into the inhibitory effects of microcystins on plant growth, and the toxicity of plant tissues following exposure. Toxicon 2001, 39, 1411–1420. [Google Scholar] [CrossRef]
  47. Pflugmacher, S. Possible allelopathic effects of cyanotoxins, with reference to microcystin-LR, in aquatic ecosystems. Environ. Toxicol. 2002, 17, 407–413. [Google Scholar] [CrossRef]
  48. Romanowska-Duda, Z.; Tarczynska, M. The influance of microcystin-LR and hepatotoxic extract on the water plant Spirodela oligorrhiza. Environ. Toxicol. 2002, 17, 434–440. [Google Scholar] [CrossRef]
  49. Wiegand, C.; Peuthert, A.; Pflugmacher, S.; Carmeli, S. Effects of microcin SF608 and microcystin-LR, two cyanobacterial compounds produced by Microcystis sp., on aquatic organisms. Environ. Toxicol. 2002, 17, 400–406. [Google Scholar] [CrossRef]
  50. Gehringer, M.M.; Kewada, V.; Coates, N.; Downing, T.G. The use of Lepidium sativum in a plant bioassay system for the detection of microcystin-LR. Toxicon 2003, 41, 871–876. [Google Scholar] [CrossRef]
  51. M-Hamvas, M.; Máthé, C.; Papp, M.; Grigorszky, I.; Molnár, E.; Vasas, G.; Borbély, G. Microcystin-LR alters growth, anthocyanin content and single-stranded DNase enzyme activities in Sinapis alba L. seedlings. Aquat. Toxicol. 2003, 62, 1–9. [Google Scholar] [CrossRef]
  52. M.-Hamvas, M.; Máthé, C.; Vasas, G.; Jámbrik, K.; Papp, M.; Beyer, D.; Mészáros, I.; Borbély, G. Cylindrospermopsin and microcystin-LR alter the growth, development and peroxidase enzyme activity of white mustard (Sinapis alba L.) seedlings, a comparative analysis. Acta Biol. Hung. 2010, 61, 35–48. [Google Scholar] [CrossRef]
  53. Chen, J.; Song, L.; Dai, J.; Gan, N.; Liu, Z. Effects of microcystins on the growth and the activity of superoxide dismutase and peroxidase of rape (Brassica napus L.) and rice (Oryza sativa L.). Toxicon 2004, 43, 393–400. [Google Scholar] [CrossRef]
  54. Chen, J.; Dai, J.; Zhang, H.; Wang, C.; Zhou, G.; Han, Z.; Liu, Z. Bioaccumulation of microcystin and its oxidative stress in the apple (Malus pumila). Ecotoxicology 2010, 19, 796–803. [Google Scholar] [CrossRef]
  55. Mitrovic, S.M.; Allis, O.; Furey, A.; James, K.J. Bioaccumulation and harmful effects of microcystin-LR in the aquatic plants Lemna minor and Wolffia arrhiza and the filamentous alga Chladophora fracta. Ecotoxicol. Environ. Saf. 2005, 61, 345–352. [Google Scholar] [CrossRef]
  56. Yin, L.; Huang, J.; Li, D.; Liu, Y. Microcystin-RR uptake and its effects on the growth of submerged macrophyte Vallisneria natans (lour.) hara. Environ. Toxicol. 2005, 20, 308–313. [Google Scholar] [CrossRef]
  57. Jang, H.M.; Ha, K.; Takamura, N. Reciprocal allelopathic responses between toxic cyanobacteria (Microcystis aeruginosa) and duckweed (Lemna japonica). Toxicon 2007, 49, 727–733. [Google Scholar] [CrossRef]
  58. Máthé, C.; M-Hamvas, M.; Vasas, G.; Surányi, G.; Bácsi, I.; Beyer, D.; Tóth, S.; Tímár, M.; Borbély, G. Microcystin-LR, a cyanobacterial toxin, induces growth inhibition and histological alterations in common reed (Phragmites australis) plants regenerated from embryogenic calli. New Phytol. 2007, 176, 824–835. [Google Scholar] [CrossRef]
  59. Saqrane, S.; El ghazali, I.; Ouahid, Y.; El Hassni, M.; El Hadrami, I.; Bouarab, L.; del Campo, F.F.; Oudra, B.; Vasconcelos, V. Phytotoxic effects of cyanobacteria extract on the aquatic plant Lemna gibba: Microcystin accumulation, detoxication and oxidative stress induction. Aquat. Toxicol. 2007, 83, 284–294. [Google Scholar] [CrossRef]
  60. Saqrane, S.; El Ghazali, I.; Oudra, B.; Bouarab, L.; Vasconcelos, V. Effects of cyanobacteria producing microcystins on seed germination and seedling growth of several agricultural plants. J. Environ. Sci. Health Part B 2008, 43, 443–451. [Google Scholar]
  61. Saqrane, S.; Ouahid, Y.; El Ghazali, I.; Oudra, B.; Bouarab, L.; del Campo, F.F. Physiological changes in Triticum durum, Zea mays, Pisum sativum and Lens esculenta cultivars, caused by irrigation with water contaminated with microcystins: A laboratory experimental approach. Toxicon 2009, 53, 786–796. [Google Scholar] [CrossRef]
  62. Yi, D.; Yijun, Z.; Xue, B.; Zhihui, F.; Kai, C. Phytotoxic effects of cyanobacteria extract on Lemna minor and Myriophyllum spicatum phyto-tolerance and superoxide dismutase activity. Environ. Toxicol. 2009, 24, 304–308. [Google Scholar] [CrossRef]
  63. Szigeti, Z.M.; Jámbrik, K.; Roszik, J.; M-Hamvas, M.; Tándor, I.; Beyer, D.; Vasas, G.; Vereb, G.; Surányi, G.; Máthé, C. Cytoskeletal and developmental alterations in Ceratophyllum demersum induced by microcystin-LR, a cyanobacterial toxin. Aquat. Bot. 2010, 92, 179–184. [Google Scholar] [CrossRef]
  64. Bácsi, I.; Surányi, G.; Gonda, S.; Gyémánt, G.; Vasas, G. Observation of sward destruction caused by irrigation with toxic Microcystis morphospecies containing water in Southern Hungary. Bull. Environ. Contam. Toxicol. 2011, 86, 232–237. [Google Scholar] [CrossRef]
  65. El Khalloufi, F.; Oufdou, K.; Lahrouni, M.; El Ghazali, I.; Saqrane, S.; Vasconcelos, V.; Oudra, B. Allelopatic effects of cyanobacteria extracts containing microcystins on Medicago sativa-Rhizobia symbiosis. Ecotoxicol. Environ. Saf. 2011, 74, 431–438. [Google Scholar] [CrossRef]
  66. Kováts, N.; Ács, A.; Paulovits, G.; Vasas, G. Response of Lemna minor clones to microcystis toxicity. Appl. Ecol. Environ. Res. 2011, 9, 17–26. [Google Scholar]
  67. Lahrouni, M.; Oufdou, K.; Faghire, M.; Peix, A.; El Khalloufi, F.; Vasconcelos, V.; Oudra, B. Cyanobacterial extracts containing microcystins affect the growth, nodulation process and nitrogen uptake of faba bean (Vicia faba L., Fabaceae). Ecotoxicology 2012, 21, 681–687. [Google Scholar] [CrossRef]
  68. Lahrouni, M.; Oufdou, K.; El Khalloufi, F.; Baz, M.; Lafuente, A.; Dary, M.; Pajuelo, E.; Oudra, B. Physiological and biochemical defense reactions of Vicia faba L.-Rhizobium symbiosis face to chronic exposure to cyanobacterial bloom extract containing microcystins. Environ. Sci. Pollut. Res. 2013, 20, 5405–5415. [Google Scholar] [CrossRef]
  69. Máthé, C.; Vasas, G.; Borbély, G.; Erdődi, F.; Beyer, D.; Kiss, A.; Surányi, G.; Gonda, S.; Jámbrik, K.; M-Hamvas, M. Histological, cytological and biochemical alterations induced by microcystin-LR and cylindrospermopsin in white mustard (Sinapis alba L.) seedlings. Acta Biol. Hung. 2013, 64, 75–89. [Google Scholar]
  70. Yin, L.Y.; Huang, J.Q.; Huang, W.M.; Li, D.H.; Wang, G.H.; Liu, Y.D. Microcystin-RR-induced accumulation of reactive oxygen species and alteration of antioxidant systems in tobacco BY-2 cells. Toxicon 2005, 46, 507–512. [Google Scholar] [CrossRef]
  71. Vasas, G.; Gásprár, A.; Surányi, G.; Batta, G.; Gyémánt, G.; M-Hamvas, M.; Máthé, C.; Grigorszky, I.; Molnár, E.; Borbely, G. Capillary Electrophoretic assay and purification of cylindrospermopsin, a cyanobacterial toxin from Aphanizomenon ovalisporum by plant test (Blue-Green Sinapis Test). Anal. Biochem. 2002, 302, 95–103. [Google Scholar] [CrossRef]
  72. Beyer, D.; Surányi, G.; Vasas, G.; Roszik, J.; Erdődi, F.; M-Hamvas, M.; Bácsi, I.; Bátori, R.; Serfőző, Z.; Szigeti, Z.M.; Vereb, G.; Demeter, Z.; Gonda, S.; Máthé, C. Cylindrospermopsin induces alterations of root histology and microtubule organization in common reed (Phragmites australis) plantlets cultured in vitro. Toxicon 2009, 54, 440–449. [Google Scholar] [CrossRef]
  73. Jámbrik, K.; Máthé, C.; Vasas, G.; Bácsi, I.; Surányi, G.; Gonda, S.; Borbély, G.; M-Hamvas, M. Cylindrospermopsin inhibits growth and modulates protease activity in the aquatic plants Lemna minor L. and Wolffia arrhiza (L.) Horkel. Acta Biol. Hung. 2010, 61, 77–94. [Google Scholar] [CrossRef]
  74. Silva, P.; Vasconcelos, V. Allelopathic effect of Cylindrospermopsis raciborskii extracts on the germination and growth of several plant species. Chem. Ecol. 2010, 26, 263–271. [Google Scholar] [CrossRef]
  75. Kittler, K.; Schreiner, M.; Krumbein, A.; Manzei, S.; Koch, M.; Rohn, S.; Maul, R. Uptake of the cyanobacterial toxin cylindrospermopsin in Brassica vegetables. Food Chem. 2012, 133, 875–879. [Google Scholar] [CrossRef]
  76. Kinnear, S.H.W.; Duivenvoorden, L.J.; Fabbro, L.D. Growth and bioconcentration in Spirodella oligorrhiza following exposure to Cylindrospermopsis raciborskii whole cell extracts. Austr. J. Ecotoxicol. 2007, 13, 19–31. [Google Scholar]
  77. Kinnear, S.H.W.; Fabbro, L.D.; Duivenvoorden, L.J. Variable growth responses of water thyme (Hydrilla verticillata) to whole-cell extracts of Cylindrospermopsis raciborskii. Arch. Environ. Contam. Toxicol. 2008, 54, 187–194. [Google Scholar] [CrossRef]
  78. Prieto, A.; Campos, A.; Cameán, A.; Vasconcelos, V. Effects on growth and oxidative stress status of rice plants (Oryza sativa) exposed to two extracts of toxin-producing cyanobacteria (Aphanizomenon ovalisporum and Microcystis aeruginosa). Ecotoxicol. Environ. Saf. 2011, 74, 1973–1980. [Google Scholar] [CrossRef]
  79. Metcalf, J.S.; Barakate, A.; Codd, G.A. Inhibition of plant protein synthesis by the cyanobacterialhepatotoxin, cylindrospermopsin. FEMS Microbiol. Lett. 2004, 235, 125–129. [Google Scholar] [CrossRef]
  80. Máthé, C.; Beyer, D.; Erdődi, F.; Serfőző, Z.; Székvölgyi, L.; Vasas, G.; M-Hamvas, M.; Jámbrik, K.; Gonda, S.; Kiss, A.; Szigeti, Z.M.; Surányi, G. Microcystin-LR induces abnormal root development by altering microtubule organization in tissue-cultured common reed (Phragmites australis) plantlets. Aquat. Toxicol. 2009, 92, 122–130. [Google Scholar] [CrossRef]
  81. Chen, J.; Han, F.X.; Wang, F.; Zhang, H.Q.; Shi, Z.Q. Accumulation and phytotoxicity of microcystin-LR in rice (Oryza sativa). Ecotoxicol. Environ. Saf. 2012, 76, 193–199. [Google Scholar] [CrossRef]
  82. Pflugmacher, S.; Aulhorn, M.; Grimm, B. Influence of a cyanobacterial crude extract containingmicrocystin-LR on the physiology and antioxidative defence systems of different spinach variants. New Phytol. 2007, 175, 482–489. [Google Scholar] [CrossRef]
  83. Abe, T.; Lawson, T.; Weyers, J.D.B.; Codd, G.A. Microcystin-LR inhibits photosynthesis of Phaseolus vulgaris primary leaves: Implications for current spray irrigation practice. New Phytol. 1996, 133, 651–658. [Google Scholar] [CrossRef]
  84. Saqrane, S.; El Ghazali, I.; Oudra, B.; Bouarab, L.; Dekayir, S.; Mandi, L.; Ouazzani, N.; Vasconcelos, V.M. Detection of microcystin contamination by the measurement of the variability of the in vivo chlorophyll fluorescence in aquatic plant Lemna gibba. Toxicon 2009, 53, 9–14. [Google Scholar] [CrossRef]
  85. Zhang, S.H.; Cheng, S.P.; Wang, H.Q.; He, F.; Wu, Z.B. Allelopathic interactions between the Potamogeton spp. and toxic cyanobacteria (Microcystis aeruginosa). Allelopath. J. 2009, 23, 379–390. [Google Scholar]
  86. Peuthert, A.; Lawton, L.; Pflugmacher, S. In vivo influence of cyanobacterial toxins on enzyme activity and gene expression of protein phosphatases in alfalfa (Medicago sativa). Toxicon 2008, 52, 84–90. [Google Scholar] [CrossRef]
  87. Takeda, S.; Mano, S.; Ohto, M.; Nakamura, K. Inhibitors of protein phosphatases 1 and 2A block sugar-inducible gene expression in plants. Plant Physiol. 1994, 106, 567–574. [Google Scholar]
  88. Siegl, G.; MacKintosh, C.; Stittl, M. Sucrose-phosphate synthase is dephosphorylated by protein phosphatase 2A in spinach leaves. Evidence from the effects of okadaic acid and microcystin. FEBS Lett. 1990, 270, 198–202. [Google Scholar] [CrossRef]
  89. Kenton, P.; Mur, L.A.J.; Draper, J. A requirement for calcium and protein phosphatase in the jasmonate-induced increase in tobacco leaf acid phosphatase specific activity. J. Exp. Bot. 1999, 50, 1331–1341. [Google Scholar]
  90. Jámbrik, K.; Máthé, C.; Vasas, G.; Beyer, D.; Molnár, E.; Borbély, G.; M-Hamvas, M. Microcystin-LR induces chromatin alterations and modulates neutral single-strand-preferring nuclease activity in Phragmites australis. J. Plant Physiol. 2011, 168, 678–686. [Google Scholar] [CrossRef]
  91. Yin, L.; Huang, J.; Huang, W.; Li, D.; Liu, Y. Responses of antioxidant system in Arabidopsis thaliana suspension cells to the toxicity of microcystin-RR. Toxicon 2005, 46, 859–864. [Google Scholar] [CrossRef]
  92. Peuthert, A.; Chakrabarti, S.; Pflugmacher, S. Uptake of microcystins-LR and -LF (cyanobacterial toxins) in seedlings of several important agricultural plant species and the correlation with cellular damage (lipid peroxidation). Environ. Toxicol. 2007, 22, 436–442. [Google Scholar] [CrossRef]
  93. Peuthert, A.; Pflugmacher, S. In fluence of the cyanotoxin microcystin-LR on tocopherol in alfalfa seedlings (Medicago sativa). Toxicon 2010, 56, 411–417. [Google Scholar] [CrossRef]
  94. Pflugmacher, S.; Wiegand, C.; Oberemm, A.; Beattie, K.A.; Krause, E.; Codd, G.A.; Steinberg, C.E.W. Identification of enzymatically formed glutathione conjugate of the cyanobacterial hepatotoxin microcystin-LR: The first step of detoxification. Biochim. Biophys. Acta 1998, 1425, 527–533. [Google Scholar] [CrossRef]
  95. Pflugmacher, S.; Codd, G.A.; Steinberg, C.E.W. Effects of the cyanobacterial toxin microcystin-LR on detoxication emzymes in aquatic plants. Environ. Toxicol. 1999, 14, 111–115. [Google Scholar] [CrossRef]
  96. Pflugmacher, S.; Wiegand, C.; Beattie, K.A.; Krause, E.; Steinberg, C.E.W.; Codd, G.A. Uptake, effects, and metabolism of cyanobacterial toxins in the emergent reed plant Phragmites australis (CAV.) Trin. ex Steud. Environ. Toxicol. Chem. 2001, 20, 846–852. [Google Scholar]
  97. Pietsch, C.; Wiegand, C.; Amé, M.V.; Nicklisch, A.; Wunderlin, D.; Pflugmacher, S. The effects of a cyanobacterial crude extract on different aquatic organisms: Evidence for cyanobacterial toxin modulating factors. Environ. Toxicol. 2001, 16, 535–542. [Google Scholar] [CrossRef]
  98. Pflugmacher, S. Promotion of oxidative stress in the aquatic macrophyte Ceratophyllum demersum during biotransformation of the cyanobacterial toxin microcystin-LR. Aquat. Toxicol. 2004, 70, 169–178. [Google Scholar] [CrossRef]
  99. Chen, J.; Zhong, Y.M.; Zhang, H.Q.; Shi, Z.Q. Nitrate reductase-dependent nitric oxide production is involved in microcystin-LR-induced oxidative stress in Brassica rapa. Water Air Soil Pollut. 2012, 223, 4141–4152. [Google Scholar] [CrossRef]
  100. Pichardo, S.; Pflugmacher, S. Study of the antioxidant response of several bean variants to irrigation with water containing MC-LR and cyanobacterial crude extract. Environ. Toxicol. 2011, 26, 300–306. [Google Scholar] [CrossRef]
  101. Chen, J.; Zhang, H.Q.; Hu, L.B.; Shi, Z.Q. Microcystin-LR-induced phytotoxicity in rice crown root is associated with the cross-talk between auxin and nitric oxide. Chemosphere 2013, 93, 283–293. [Google Scholar] [CrossRef]
  102. Froscio, S.M.; Humpage, A.R.; Wickramasinghe, W.; Shaw, G.; Falconer, I.R. Interaction of the cyanobacterial toxin cylindrospermopsin with the eukaryotic protein synthesis system. Toxicon 2008, 51, 191–198. [Google Scholar] [CrossRef]
  103. Lankoff, A.; Banasik, A.; Obe, G.; Deperas, M.; Kuzminski, K.; Tarczynska, M.; Jurczak, T.; Wojcik, A. Effect of microcystin-LR and cyanobacterial extract from Polish reservoir of drinking water on cell cycle progression, mitotic spindle, and apoptosis in CHO-K1 cells. Toxicol. Appl. Pharmacol. 2003, 189, 204–213. [Google Scholar] [CrossRef]
  104. Gehringer, M.M. Microcystin-LR and okadaic acid-induced cellular effects: A dualistic response. FEBS Lett. 2004, 557, 1–8. [Google Scholar] [CrossRef]
  105. Beyer, D.; Tándor, I.; Kónya, Z.; Bátori, R.; Roszik, J.; Vereb, G.; Erdődi, F.; Vasas, G.; M-Hamvas, M.; Jambrovics, K.; Máthé, C. Microcystin-LR, a protein phosphatase inhibitor induces alterations in mitotic chromatin and microtubule organization leading to the formation of micronuclei in Vicia faba. Ann. Bot. 2012, 110, 797–808. [Google Scholar] [CrossRef]
  106. Wolniak, S.M.; Larsen, P.M. Changes in the metaphase transit times and the pattern of sister chromatid separation in stamen hair cells of Tradescantia after treatment with protein phosphatase inhibitors. J. Cell Sci. 1992, 102, 691–705. [Google Scholar]
  107. Toivola, D.M.; Eriksson, J.E. Toxins affecting cell signalling and alteration of cytoskeletal structure. Toxicol. Vitr. 1999, 13, 521–530. [Google Scholar] [CrossRef]
  108. Venoux, M.; Basbous, J.; Berthenet, C.; Prigent, C.; Fernandez, A.; Lamb, N.J.; Rouquier, S. ASAP is a novel substrate of the oncogenic mitotic kinase Aurora-A: Phosphorylation on Ser625 is essential to spindle formation and mitosis. Hum. Mol. Genet. 2008, 17, 215–224. [Google Scholar]
  109. Gácsi, M.; Antal, O.; Vasas, G.; Máthé, C.; Borbély, G.; Saker, M.L.; Győri, J.; Farkas, A.; Vehovszky, Á.; Bánfalvi, G. Comparative study of cyanotoxins affectiong cytoskeletal and chromatin structures in CHO-K1 cells. Toxicol. Vitr. 2009, 23, 710–718. [Google Scholar] [CrossRef]
  110. Ayaydin, F.; Vissi, E.; Mészáros, T.; Miskolczi, P.; Kovács, I.; Fehér, A.; Dombrádi, V.; Erdődi, F.; Gergely, P.; Dudits, D. Inhibition of serine/threonine-specific protein phosphatases causes premature activation of cdc2MsF kinase at G2/M transition and early microtubule organisation in alfalfa. Plant J. 2000, 23, 85–96. [Google Scholar] [CrossRef]
  111. Holy, J.M. Curcumin disrupts mitotic spindle structure and induces micronucleation in MCF-7 breast cancer cells. Mutat. Res. 2002, 518, 71–84. [Google Scholar] [CrossRef]
  112. Sasabe, M.; Soyano, T.; Takahashi, Y.; Sonobe, S.; Igarashi, H.; Itoh, T.J.; Hidaka, M.; Machida, Y. Phosphorylation of NtMAP65-1 by a MAP kinase down-regulates its activity of microtubule bundling and stimulates progression of cytokinesis of tobacco cells. Genes Dev. 2006, 20, 1004–1014. [Google Scholar] [CrossRef]
  113. Smertenko, A.P.; Chang, H.Y.; Wagner, V.; Kaloriti, D.; Fenyk, S.; Sonobe, S.; Lloyd, C.; Hauser, M.T.; Hussey, P.J. The Arabidopsis microtubule-associated protein AtMAP65-1: Molecular analysis of its microtubule bundling activity. Plant Cell 2004, 16, 2035–2047. [Google Scholar] [CrossRef]
  114. Laughinghouse, H.D., IV; Prá, D.; Silva-Stenico, M.E.; Rieger, A.; Dal-Souto Frescura, V.; Fiore, M.F.; Tedesco, S.B. Biomonitoring genotoxicity and cytotoxicity of Microcystis aeruginosa (Chroococcales, Cyanobacteria) using the Allium cepa test. 2012, 432, 180–188. [Google Scholar]
  115. Huang, W.; Xin, W.; Li, D.; Liu, Y. Microcystin-RR induced apoptosis in tobacco BY-2 suspension cells is mediated by reactive oxygen species and mitochondrial permeability transition pore status. Toxicol. Vitr. 2008, 22, 328–337. [Google Scholar] [CrossRef]
  116. Yin, L.Y.; Huang, J.Q.; Li, W.; Liu, Y.D. Microcystin-RR-induced apoptosis in tobacco BY-2 cells. Toxicon 2006, 48, 204–210. [Google Scholar] [CrossRef]
  117. Huang, W.; Xing, W.; Li, D.; Liu, D. Morphological and ultrastructural changes in tobacco BY-2 cells exposed to microcystin-RR. Chemosphere 2009, 76, 1006–1012. [Google Scholar] [CrossRef]
  118. Jiang, J.; Gu, X.; Song, R.; Wang, X.; Yang, L. Microcystin-LR induced oxidative stress and ultrastructural alterations in mesophyll cells of submerged macrophyte Vallisneria natans (Lour.) Hara. J. Hazard. Mater. 2011, 190, 188–196. [Google Scholar] [CrossRef]
  119. Chen, J.Z.; Ye, J.Y.; Zhang, H.Y.; Jiang, X.J.; Zhang, Y.X.; Liu, Z.L. Freshwater toxic cyanobacteria induced DNA damage in apple (Malus pumila), rape (Brassica napus) and rice (Oryza sativa). J. Hazard. Mater. 2011, 190, 240–244. [Google Scholar] [CrossRef]
  120. Kaszás, E.; Cande, W.Z. Phosphorylation of histone H3 is correlated with changes in the maintenance of sister chromatid cohesion during meiosis in maize, rather than the condensation of the chromatin. J. Cell Sci. 2000, 113, 3217–3226. [Google Scholar]
  121. Schroeder-Reiter, E.; Houben, A.; Wanner, G. Immunogold labeling of chromosomes for scanning electron microscopy: A closer look at phosphorylated histone H3 in mitotic metaphase chromosomes of Hordeum vulgare. Chromosome Res. 2003, 11, 585–596. [Google Scholar] [CrossRef]
  122. Zhang, X.; Li, X.; Marshall, J.B.; Zhong, C.X.; Dawe, R.K. Phosphoserine on maize CENTROMERIC HISTONE H3 and histone H3 demarcate the centromere and pericentromere during chromosome segregation. Plant Cell 2005, 17, 572–583. [Google Scholar] [CrossRef]
  123. Manzanero, S.; Rutten, T.; Kotseruba, V.; Houben, A. Alterations in the distribution of histone H3 phosphorylation in mitotic plant chromosomes in response to cold treatment and the protein phosphatase inhibitor cantharidin. Chromosome Res. 2002, 10, 467–476. [Google Scholar] [CrossRef]
  124. Pérez-Cadahía, B.; Drobic, B.; Davie, J.R. H3 phosphorylation: Dual role in mitosis and interphase. Biochem. Cell Biol. 2009, 87, 695–709. [Google Scholar] [CrossRef]
  125. Eyers, P.A.; Maller, J.L. Regulation of Xenopus Aurora A activation by TPX2. J. Biol. Chem. 2004, 279, 9008–9015. [Google Scholar] [CrossRef]
  126. Demidov, D.; VanDamme, D.; Geelen, D.; Blattner, F.R.; Houben, A. Identification and dynamics of two classes of Aurora-like kinases in Arabidopsis and other plants. Plant Cell 2005, 17, 836–848. [Google Scholar] [CrossRef] [Green Version]
  127. Kawabe, A.; Matsunaga, S.; Nakagawa, K.; Kurihara, D.; Yoneta, A.; Hasezawa, S.; Uchiyama, S.; Fukui, K. Characterization of plant Aurora kinases during mitosis. Plant Mol. Biol. 2005, 58, 1–13. [Google Scholar] [CrossRef]
  128. Yu, M.; Yuan, M.; Ren, H. Visualization of actin cytoskeletal dynamics during the cell cycle in tobacco (Nicotiana tabacum L. cv. Bright Yellow) cells. Biol. Cell 2006, 98, 295–306. [Google Scholar] [CrossRef]
  129. Hwang, J.U.; Lee, Y. Abscisic acid-induced actin reorganization in guard cells of dayflower is mediated by cytosooic calcium levels and by protein kinase and protein phosphatase activities. Plant Physiol. 2001, 125, 2120–2128. [Google Scholar] [CrossRef]
  130. Lee, S.; Park, J.; Lee, Y. Phosphatidic acid induces actin polymerization by activating protein kinases in soybean cells. Mol. Cells 2003, 15, 313–319. [Google Scholar]
  131. Falconer, I.R.; Yeung, D.S.K. Cytoskeletal changes in hepatocytes induced by Microcystis toxins and their relation to hyperphosphorylation of cell proteins. Chem. Biol. Interact. 1992, 81, 181–196. [Google Scholar] [CrossRef]
  132. Fladmark, K.E.; Serres, M.H.; Larsen, N.L.; Yasumoto, T.; Aune, T.; Døskeland, S.O. Sensitive detection of apoptogenic toxins in suspension cultures of rat and salmon hepatocytes. Toxicon 1998, 36, 1101–1114. [Google Scholar] [CrossRef]
  133. Khan, S.A.; Ghosh, S.; Wickstrom, M.; Miller, L.A.; Hess, R.; Haschek, W.M.; Beasley, V.R. Comparative pathology of microcystin-LR in cultured hepatocytes, fibroblasts, and renal epithelial cells. Nat. Toxins 1995, 3, 119–128. [Google Scholar] [CrossRef]
  134. Abramsson-Zetterberg, L.; Beckman Sundh, U.; Mattsson, R. Cyanobacterial extracts and microcystin-LR are inactive in the micronucleus assay in vivo and in vitro. Mutat. Res. 2010, 699, 5–10. [Google Scholar] [CrossRef]
  135. Michalakis, J.; Georgatos, D.; Romanos, J.; Koutala, H.; Georgoulias, V.; Tsifitis, D.; Theodoropoulos, P.A. Micromolar taxol, with or without hyperthermia, induces mitotic catastrophe and cell necrosis in HeLa cells. Cancer Chemother Pharm. 2005, 56, 615–622. [Google Scholar] [CrossRef]
  136. Lytvyn, D.I.; Venets, A.I.; Blume, Y.B. UV-B overexposure induces programmed cell death in a BY-2 tobacco cell line. Environ. Exp. Bot. 2010, 68, 51–57. [Google Scholar] [CrossRef]
  137. Mankiewicz, J.; Tarczynska, M.; Fladmark, K.E.; Døskeland, S.O.; Walter, Z.; Zalewski, M. Apoptotic effect of cyanobacterial extract on rat hepatocytes and human lymphocytes. Environ. Toxicol. 2001, 16, 225–233. [Google Scholar] [CrossRef]
  138. McDermott, C.M.; Nho, C.W.; Howard, W.; Holton, B. The cyanobacterial toxin, microcystin-LR, can induce apoptosis in a variety of cell types. Toxicon 1998, 36, 1981–1996. [Google Scholar] [CrossRef]
  139. Darzynkiewicz, Z.; Juan, G.; Li, X.; Gorczyca, W.; Murakami, T.; Traganos, F. Cytometry in cell necrobiology: Analysis of apoptosis and accidental cell death (necrosis). Cytometry 1997, 27, 1–20. [Google Scholar] [CrossRef]
  140. Ding, W.X.; Shen, H.M.; Ong, C.N. Critical role of reactive oxygen species and mitochondrial permeability transition in microcystin-induced rapid apoptosis in rat hepatocytes. Hepatology 2000, 32, 547–555. [Google Scholar] [CrossRef]
  141. Dangl, J.L.; Dietrich, R.A.; Thomas, H. Senescence and Programmed Cell Death. In Biochemistry and Molecular Biology of Plants; Buchanan, B., Gruissem, W., Jones, R., Eds.; The American Society of Plant Biologists: Rockville, MD, USA, 2000; pp. 1044–1100. [Google Scholar]
  142. Kusaka, K.; Tada, Y.; Shigemi, T.; Sakamoto, M.; Nakayashiki, H.; Tosa, Y.; Shigeyuki, M. Coordinate involvement of cysteine protease and nuclease in the executive phase of plant apoptosis. FEBS Lett. 2004, 578, 363–367. [Google Scholar] [CrossRef]
  143. Žegura, B.; Lah, T.T.; Filipič, M. The role of reactive oxygen species in microcystin-LR-induced DNA damage. Toxicology 2004, 200, 59–68. [Google Scholar] [CrossRef]
  144. Valério, E.; Chaves, S.; Tenreiro, R. Diversity and impact of prokaryotic toxins on aquatic environments: A review. Toxins 2010, 2, 2359–2410. [Google Scholar] [CrossRef]
  145. Ros Barceló, A. The generation of H2O2 in the xylem of Zinnia elegans is mediated by an NADPH-oxidase-like enzyme. Planta 1998, 207, 207–216. [Google Scholar] [CrossRef]
  146. Campos, A.; Vasconcelos, V. Molecular mechanisms of microcystin toxicity in animal cells. Review. Int. J. Mol. Sci. 2010, 11, 268–287. [Google Scholar] [CrossRef]
  147. Bagniewska-Zadworna, A. The root microtubule cytoskeleton and cell cycle analysis through desiccation of Brassica napus seedlings. Protoplasma 2008, 233, 177–185. [Google Scholar] [CrossRef]
  148. Lankoff, A.; Wojcik, A.; Lisowska, H.; Bialczyk, J.; Dziga, D.; Carmichael, W.W. No induction of structural chromosomal aberrations in cylindrospermopsin-treated CHO-K1 cells without and with metabolic activation. Toxicon 2007, 50, 1105–1115. [Google Scholar] [CrossRef]
  149. Bazin, E.; Mourot, A.; Humpage, A.R.; Fessard, V. Genotoxicity of a freshwater cyanotoxin, cylindrospermopsin, in two human cell lines: Caco-2 and HepaRG. Environ. Mol. Mutagen. 2010, 51, 251–259. [Google Scholar]
  150. Chong, M.W.K.; Wong, B.S.F.; Lam, P.K.S.; Shaw, G.R.; Seawright, A.A. Toxicity and uptake mechanism of cylindrospermopsin and lophyrotomin in primary rat hepatocytes. Toxicon 2002, 40, 205–211. [Google Scholar] [CrossRef]
  151. Štraser, A.; Filipič, M.; Žegura, B. Genotoxic effects of the cyanobacterial hepatotoxin cylindrospermopsin on the HepG2 cell line. Arch. Toxicol. 2011, 85, 1617–1626. [Google Scholar] [CrossRef]
  152. Shen, X.; Lam, P.K.S.; Shaw, G.R.; Wickramasinghe, W. Genotoxicity investigation of a cyanobacterial toxin, cylindrospermopsin. Toxicon 2002, 40, 1499–1501. [Google Scholar] [CrossRef]
  153. Gutiérrez-Praena, D.; Jos, Á.; Pichardo, S.; Cameán, A.M. Oxidative stress responses in tilapia (Oreochromis niloticus) exposed to a single dose of pure cylindrospermopsin under laboratory conditions: Influence of exposure route and time of sacrifice. Aquat. Toxicol. 2011, 105, 100–106. [Google Scholar] [CrossRef]
  154. Gutiérrez-Praena, D.; Pichardo, S.; Jos, Á.; Moreno, F.J.; Cameán, A.M. Biochemical and pathological toxic effects induced by the cyanotoxin Cylindrospermopsin on the human cell line Caco-2. Water Res. 2012, 46, 1566–1575. [Google Scholar] [CrossRef]
  155. Štraser, A.; Filipič, M.; Gorenc, I.; Žegura, B. The influence of cylindrospermopsin on oxidative DNA damage and apoptosis induction in HepG2 cells. Chemosphere 2013, 92, 24–30. [Google Scholar] [CrossRef]
  156. Fessard, V.; Bernard, C. Cell alterations but no DNA strand breaks induced in vitro by cylindrospermopsin in CHO K1 cells. Environ. Toxicol. 2003, 18, 353–359. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Máthé, C.; M-Hamvas, M.; Vasas, G. Microcystin-LR and Cylindrospermopsin Induced Alterations in Chromatin Organization of Plant Cells. Mar. Drugs 2013, 11, 3689-3717. https://doi.org/10.3390/md11103689

AMA Style

Máthé C, M-Hamvas M, Vasas G. Microcystin-LR and Cylindrospermopsin Induced Alterations in Chromatin Organization of Plant Cells. Marine Drugs. 2013; 11(10):3689-3717. https://doi.org/10.3390/md11103689

Chicago/Turabian Style

Máthé, Csaba, Márta M-Hamvas, and Gábor Vasas. 2013. "Microcystin-LR and Cylindrospermopsin Induced Alterations in Chromatin Organization of Plant Cells" Marine Drugs 11, no. 10: 3689-3717. https://doi.org/10.3390/md11103689

Article Metrics

Back to TopTop