eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2010
vol. 35
 
Share:
Share:

Review paper
Immunotoxic, genotoxic and carcinogenic effects of cyanotoxins

Anna Sierosławska

Centr Eur J Immunol 2010; 35 (2): 105-110
Online publish date: 2010/05/10
Article file
- 10_Immunotoxic.pdf  [0.07 MB]
Get citation
 
 
Introduction
Cyanotoxins, produced and released into the water during and after water blooms, pose a growing problem for human and animal health. Exposure to cyanotoxins may occur via several routes, e.g. by consuming contaminated water, food or dietary supplements, during recreational activities or by inhaling toxin-containing aerosols [1, 2]. Their toxic effects on vertebrates include different symptoms and are based on various mechanisms.
According to the target organ, cyanotoxins are divided into hepatotoxins (microcystins, nodularins, cylindro-spermopsins), neurotoxins (anatoxin-a, anatoxin-a(s), saxitoxins), cytotoxins and irritating toxins, also influencing the gastrointestinal system (lyngbiatoxins, aplysiatoxins, lipopolysaccharides) [3]. The above mentioned directions of toxic impact are clearly seen after acute exposure. However, the chronic exposure to low cyanotoxin levels brings also other effects, of not fully understood mechanisms and sometimes difficult to predict. That possibility should be taken into consideration in toxicity assessment, especially in case of the toxins of long persistence in water, such as microcystins (MCs).
This review attempts to sum up information concerning conditions and mechanisms of immunotoxic, genotoxic and carcinogenic activity of cyanotoxins. While MCs are the most studied cyanobacterial toxins, presented herein data in a large extend concerns that group of substances.

Immunotoxic effects of cyanotoxins
There is a growing evidence, that cyanotoxins may evoke immunotoxic effects. This is of great concern taking into consideration, that the disorders of immune functions may induce many negative changes in organism functioning, including carcinogenesis. Immunomodulatory potency of cyanotoxins seems to be of dualistic way, inducing both immunostimulatory and immunosuppressive responses. Comprehensive reviews of the results of studies on the cyanotoxin immunomodulatory potency are given in [4-6]. In this paper only selected effects, connected with non-specific immunity will be discussed, in the respect to their hypothetical mechanisms.
Phagocytosis, being the fundamental immune response, is of special interest in immunotoxicity assessment. The process involves cytoskeleton rearrangements, followed by secondary activities, such as superoxide production and inflammatory cytokine release, controlled by different signaling pathways [7]. The main mechanism of toxic activity of such cyanotoxins, as MCs or nodularins, is the inhibition of serine/threonine protein phosphatase 1 and 2A (PP1 and PP2A), leading to the hyperphosphorylation of cytosolic and cytoskeletal proteins [8, 9]. MCs are supposed to interfere with all of three main cytoskeleton protein components [9, 10].
As phagocytosis is an actin-dependent process [7], microcystin-induced collapse of microfilaments in the primary human and rat hepatocyte cultures [11, 12] and in other, non-hepatic cells [9], suggests that disruption of subtle remodeling of actin cytoskeleton in professional phagocytes exposed to the toxin may also be expected. Additionally, increased phosphorylation of the actin-associated proteins, such as vinculin or talin, may result in alternations in microfilaments binding to a cell membrane [citations in 9]. Actin cytoskeleton changes induced by MC-LR in all studied cell types are preceded by reorganization of intermediate filaments and microtubules, which suggests that these two latter ones are at least partially the cause of the observed microfilament collapse [9]. Toivola et al. [13] found, that MC-induced hyperphosphorylation of cytokeratin 18 and 8, the basic proteins of intermediate filaments, leads to their disruption. It is also suggested, that MCs may disrupt the microtubule cytoskeletal structures by direct binding to the tubulin cysteine residues [8]. The tubulin synthesis is dependent on the feedback regulated by the protein monomer pool. MC-mediated microtubule polymer collapse increases the free tubulin pool and inhibits new tubulin generation, which in the authors opinion, in some extend explains the severe loss of microtubules, observed in the cells treated with microcystic cyanobacteria extract [8]. MC-LR, the most toxic isoform of MCs, has been also found to cause increased phosphorylation of cytoplasmic dynein [14], which may be reflected in impaired microtubule motor functions. Dynein, microtubule-associated protein, is engaged in retrograde phagosome transport towards lysosomes allowing their fusion and degradation of the internalized particles [15, 16]. Indeed, reduction in the recovery of endosomal/lysosomal membranes by MCs in hepatocytes exposed to MC-LR has been reported [14], but no data from studies on phagocytic cells is available.
Despite the evident potency of MCs to induce changes in cytoskeleton, the results from the studies on the toxin effects on the process of phagocytosis are divergent. After 24 h incubation of human neutrophils with MC-LR concentrations up to 10 ng/ml, no effects on cell viability or phagocytosis have been found [17]. On the contrary, concentration-dependent suppression of phagocytic ability was seen in mice peritoneal macrophages after cell triggering with MC-LR at 1 – 1000 nmol/l for 4 h [18]. In the same experiment no effects were seen when mouse macrophage-like RAW264.7 cells were used. Chronic exposure of the early life stages of common carp on the cyanobacterial extract containing MCs by bath, as well as oral administration of MC-containing biomass to common and silver carp caused significant decrease of phagocytic activity in blood leucocytes [19, 20]. Decreased phagocytic index was also reported after injection of cyanobacterial extract containing 9.94 and 19.88 µg of MCs equivalents/kg mouse b.w. for 14 days, without any influence on percentage phagocytosis [21]. However, observed in the in vivo experiments changes in phagocytic ability may not be the results of direct toxin influence on phagocyte physiology, some indirect effects should also be considered.
Oppositely, there are reports suggesting toxin-induced non-specific activation of immune cells [22]. Stimulation of human and rat neutrophils after cell exposure on MC isoforms LA, YR and in the highest degree MC-LR, manifested as increased cell rolling and adherence, accompanied by higher L-selectin and b2-integrin expression and stimulated chemotaxis, was observed by Kujbida et al. [23, 24]. Toxins concentrations used were 1 – 1000 nM, exposure time not exceeded 60 min. Likewise, nodularin and in smaller extend MC-LR, occurred to stimulate the early spontaneous polymorphonuclear cell adhesion at the toxin concentrations up to 1 nM [25]. In our in vitro study rainbow trout phagocytes incubated for 35 min with MC-LR at 5 µg/ml revealed the increase of zymosan particle phagocytosis [26].
Except of the toxin influence on cell adherence and engulfing ability of phagocytes, also other stages of phagocytosis were reported to be interfered by the cyanotoxins. Elimination of invading microorganisms by professional phagocytes depends heavily on the generation of reactive oxygen species (ROS) during the process termed respiratory burst [27]. MCs are known to stimulate reactive oxygen production in different cell types, such as hepatocytes [10, 28] or lymphocytes [29]. In our study dose-dependent, dualistic effects of MC-LR on respiratory burst activity in phorbol myristate acetate triggered rainbow trout neutrophils were recorded [26]. It that study stimulation of reactive radicals production after cell incubation with the toxin at 1 and 5 µg/ml was recorded, while the higher used doses (10 and 20 µg/ml) diminished studied parameter. On the contrary, decreased spontaneous ROS generation in mice peritoneal macrophages after cell exposure on MC-LR was already noticeable at the concentrations starting from 10 nmol/l [18]. In turn, inhibitory effects of MC-LR at 10 µg/l on spontaneous and Staphylococcus aureus stimulated ROS production were observed in neutrophils isolated from patients receiving hemodialysis, but not from the healthy donors [17].
Observed increase of intracellular Ca2+ (iCa2+) levels in the cells exposed on MCs may be a part of mechanisms on which the toxins influence the phagocyte functions, however the way of affecting the iCa2+ homeostasis and its real consequences are not yet fully known [10, 24]. The role of calcium ions in phagocytosis is significant, among others in actin cytoskeleton depolymerization and remodeling, in mediating phagosome-lysosome fusion and also in ROS production [30]. MC-induced elevated iCa2+ content was reported in different cell types, including neutrophils and lymphocytes [10, 24, 29]. Toxin effects on ROS production seems to be mediated by surge of calcium ions [8, 29], but also other mechanisms are proposed, such as MC-induced increase of NADPH oxidase activity or up-regulation of pro-apoptotic proteins Bax and Bid [28, 30, 32].
Non-specific activation of immune system, observed in cyanotoxin-exposed patients [22] could be explained by stimulation of proinflammatory cytokine release, which in turn is often concomitant with oxidative stress. Indeed, dose-dependent induction of interleukin 1 (IL-1) and TNF- production was found in mouse macrophages stimulated in vitro with MC-LR at up to 1 µg/ml [33]. Incubation of human neutrophils with MC-LA, YR and LR at 1 or 1000 nM for 24 h increased the production of another proinflammatory mediator, IL-8 [34]. Additionally, these toxins stimulated cytokine-induced neutrophil chemo­attractant (CINC)-2 release from rat neutrophils [34]. On the contrary, the other authors [35, 36] reported decrease of mRNA levels of IL-1 and TNF- in mice treated with MC-LR-containing cyanobacterial bloom extracts and in mouse macrophages induced with pure MC-LR.
As stated above, there is a lot of discrepancies observed in the toxin effects on phagocytic cells. Some of them may be at least partially explained by different experimental conditions, especially used toxin concentrations and durations of exposure. Cellular uptake of MCs is mediated by multi-specific organic anion transporting peptides (rodent Oatps; human OATPs) [30], of which multiple types are expressed at high level on hepatocytes, enabling relatively quick influx of the toxin and faster effect exerting in the liver cells. The way MCs enters the other cell types, including phagocytes, is not fully clear, probably in some extend also via Oatps/OATPS [37]. Anyway, occurrence of the toxin influence on the non-hepatic cell morphology/functions is prolonged in time, compared to the effects on hepatocytes [9]. As phagocytes play an important role in regulation of cancer development and spontaneous tumorigenesis [38, 39], the immunomodulatory activity of the toxins should also be considered in relation to the observed carcinogenic effects of cyanotoxins.

Carcinogenic and genotoxic effects of cyanotoxins
Public concern about the carcinogenic potency of cyanotoxins arose when the correlation between incidences of primary liver and colorectal cancers and the presence of toxic cyanobacteria or their toxins in drinking sources has been suggested [40-44]. In most cases MC-LR was suspected to be responsible for these effects, however the laboratory studies on the toxin ability to induce cancers give sometimes ambiguous information.
The mechanisms underlying carcinogenic activity of MC-LR are probably divergent and realized on both genotoxic and epigenetic pathways [45]. It has been established, that during chronic exposure, MC-LR may act as the tumor inducer, as was found in the studies on mice given i.p. the toxin at the sublethal dose (20 µg/kg), five times per week over 28 weeks [46]. In that study neoplastic nodules were observed in mice livers without the use of any initiator. Similar findings were also reported in the study by Sano et al. [47], where the toxin was i.p. injected to mice for 14 months, causing the dose-dependent increase in hepatic adenomas and adenocarcinomas.
MC-LR is also supposed to be a potent liver tumor promoter. Nishiwaki-Matsushima et al. [48] found, that MC-LR administered i.p. at the dose of 10 µg/kg twice per week for 8 weeks to rats previously treated with tumor inducer, diethylnitrosamine, increased the number and percentage area of positive foci for the placental form of glutathione S-transferase, the sensitive marker of cancer initiation, in rat liver. The authors did not record such effects in the non-induced animals, which suggests that the duration of exposure may decide of the way of carcinogenic activity of MC-LR. Agreeable results were achieved by Charbonneau et al. [49] with the use of the similar experimental layout. Additionally, in that study carcinogenic effects were found also after oral uptake of the toxin at the dose of 80 µg/kg for 7 weeks. The results obtained by Lian et al. [41] confirmed tumor-promoting potency of MC-LR rather than initiating, in the short-term experiments. Mice were initially treated with a single dose of aflatoxin B1 and then were given i.p. MC-LR or nodularin at the dose of 10 g/kg, once a week for 15 weeks. The other group of mice were treated with the cyanotoxins without previous induction. There was no increase in adenoma or carcinoma appearance in animals without pre-treatment, however a few altered hepatocellular foci were identified. On the contrary, in the aflatoxin-initiated animals both cyanotoxins intensified liver tumors development, nodularin in higher degree than MC-LR.
While the toxin main target in the cell is PP1 and PP2A, enzymes of pleiotropic activity able to directly regulate the activities of multiple protein kinase cascades, their inhibition leads to the hyperphosphorylation of a number of proteins. In the consequence deregulation of cell-cycle control, uncontrolled cell proliferation, as well as the inhibition of proteins involved in DNA repair occurs [45, 50-53]. One of the MC-LR targets undergoing hyperphosphorylation is nuclear protein p53, an important tumor suppressor which contributes to the repair of genotoxic damage, stops proliferation and induces apoptosis of impaired cells [54, 55]. That is of special importance, taking under consideration that in the same time MC-LR is able to cause oxidative stress resulting in DNA damage in the exposed cells [30, 56]. The mechanisms of DNA repair such as the nucleotide excision repair (NER) and the DNA double strand break repair by the nonhomologous end joining, in which p53 is engaged [57, 58], are known to be impaired by MC [30, 45].
One of the consequences of oxidative stress in the cell is the c-Jun N-terminal kinases (JNK) pathway activation, which is also attributed to MC-LR activity [59]. It has been found, that elevated activation of JNK plays a major role in promoting tumorigenesis of primary human brain tumors [60].
Genotoxic activity of MC-LR, manifested as increased micronucleus frequency, was determined in human-hamster hybrid AL cell line after 30-day treatment with the toxin at 0.1 µg/ml [61]. Similarly, increase in micronucleus frequency in human lymphoblastoid cell line TK6 after MC-LR intoxication was observed by Zhan et al. [62], but the authors used much higher toxin concentration (80 µg/ml) with short duration of exposure (24 h). Moreover, the authors found increase in mutation frequency at the thymidine kinase (TK) locus. Most of the TK mutants induced by MC-LR were the result of loss of heterozygosity, which is known to be an important genetic event in tumorigenesis and has a common occurrence in a variety of human tumors. Confirming results were also obtained when cyanobacterial extract containing MCs was used [63]. In that study genotoxicity was reported in both bacteria model, with the use of SOS chromotest and human lymphocyte model, with the use of comet assay. Moreover, the mutagenic effects of MC-LR, determined as ouabain resistance (Ouar), mutations induced in various mammalian cell types by many known mutagens, were found in cultured human RSa cells, with the highest frequency at 15 µg/ml. Base substitution mutations at K-ras codon 12 in genomic DNA were also found in the cells exposed to 7.5-15 µg/ml of the toxin for 6 days [64].
On the contrary, Lankoff et al. [51] reported, that MC-LR had no effect on the frequency of chromosome aberrations in human lymphocytes. Moreover, the authors suggested, that observed in their study microcystin-LR-induced DNA damage might be rather connected with early stages of apoptosis due to toxin cytotoxicity, not genotoxicity. A lack of mutagenic effects of MC-LR containing extract was found with the use of Ames test and SOS umu test [65].
An excellent critical summary of research on MCs genotoxic effects are given in Annex to the SKLM opinion “Microcystins in Algae Products Used as a Food Supplement” [66].
Much fewer data is available for the other cyanotoxin suspected to be carcinogenic, cylindrospermopsin (CYN). The toxin works by inhibition of protein synthesis and its toxicity results from cyn metabolic activation [22, 67]. As the structure of CYN includes sulphate, guanidine and uracil groups, it has been suggested that CYN may act on nucleic acids and exert carcinogenic effects on exposed organisms [68].
Shen et al. demonstrated that the toxin given i.p. to mice in a single dose of 0.2 mg/kg caused DNA strand breaks which might be one of the key mechanisms for CYN genotoxicity [69]. Moreover, covalent binding of CYN or its metabolites to DNA in mice, as well as early activation of P53 genes in human cell lines were observed [70, 71]. Cytogenetic damage in WIL2-NS lymphoblastoid cell line was also reported by Humpage et al. [72]. The authors suggest two mechanisms of CYN activity, one at the level of the DNA, inducing strand breaks and the other, at the level of kinetochore/spindle function, inducing loss of whole chromosomes. On the contrary, no clastogenic effects were detected after CHO-K1 cells exposure at 0.05-2 µg/ml, with or without metabolic activation of the toxin [73]. No increase in cancer was found in P53def transgenic mice after oral administration of chlorinated purified CYN or toxic cell-free extract for 90 and 170 days [74]. In that study similar effects were also found when MC-LR, and another cyanotoxin, saxitoxin, were used. In conclusion, the information on genotoxic/carcinogenic potency of CYN is incomplete and rather preliminary, so at the current state of knowledge it is difficult to predict such toxin activity.

Conclusions
Cyanotoxins, having diverse chemical properties, constitute a group of highly active compounds, potent to exert varied biological effects, including cancer. However, on the basis of the available literature it can be seen, that only one cyanotoxin, MC-LR, has been more extensively studied on the effects being the main problems of that review. In case of the other ones, data is very scared. Moreover, there is no studies on the possible connection between observed immunomodulating activity of MCs or other cyanotoxins and tumor cases. The need of investigation on that area seems to be obvious.

References
1. Rellan S, Osswald J, Saker M et al. (2009): First detection of anatoxin-a in human and animal dietary supplements containing cyanobacteria. Food Chem Toxicol 47: 2189-2195.
2. Stewart I, Webb PM, Schluter PJ et al. (2006): Recreational and occupational field exposure to freshwater cyanobacteria – a review of anecdotal and case reports, epidemiological studies and the challenges for epidemiologic assessment. Environ Health, doi: 10.1186/1476-069X-5-6.
3. Briand JF, Jacquet S, Bernard C et al. (2003): Health hazards for terrestrial vertebrates from toxic cyanobacteria in surface water ecosystems. Vet Res 34: 361-377.
4. Rymuszka A, Sierosławska A (2009): The immunotoxic and nephrotoxic influence of cyanotoxins to vertebrates. Centr Eur J Immunol 34: 129-136.
5. Rymuszka A, Sierosławska A (2008): Immunotoxic potential of cyanotoxins on the immune system of fish. Centr Eur J Immunol 33: 150-152.
6. Sierosławska A, Rymuszka A (2009): Cyanohepatotoxins influence on the neuroendocrine and immune systems in fish – a short review. Neuroendocrinol Lett 30 (Suppl 1): 13-16.
7. May RC and Machesky LM (2001): Phagocytosis and the actin cytoskeleton. J Cell Sci 114: 1061-1077.
8. Ding WX, Shen HM, Ong CN (2000): Microcystic cyanobacteria extract induces cytoskeletal disruption and intracellular glutathione alteration in hepatocytes. Environ Health Perspect 108: 605-609.
9. Wickstrom ML, Khan SA, Haschek WM et al. (1995): Alterations in microtubules, Intermediate filaments, and microfilaments induced by microcystin-LR in cultured cells. Toxicol Pathol 23: 326-337.
10. Gehringer MM (2004): Microcystin-LR and okadaic acid-induced cellular effects: a dualistic response. FEBS Lett 557: 1-8.
11. Batista T, de Sousa G, Suput JS et al. (2003): Microcystin-LR causes the collapse of actin filaments in primary human hepatocytes. Aquatic Toxicol 65: 85-91.
12. Ghosh S, Khan SA, Wickstrom M et al. (1995): Effects of microcystin-LR on actin and the actin-associated proteins a-actinin and talin in hepatocytes. Nat Toxins 3: 405-414.
13. Toivola DM and Eriksson JE (1999): Toxins affecting cell signalling and alteration of cytoskeletal structure. Toxicol In Vitro 13: 521-530.
14. Runnegar M, Wei X, Hamm-Alvarez SF (1999): Increased protein phosphorylation of cytoplasmic dynein results in impaired motor function. Biochem J 346: 1-6.
15. Oda H, Stockert RJ, Collins C et al. (1995). Interaction of the microtubule cytoskeleton with endocytic vesicles and cytoplasmic dynein in cultured rat hepatocytes. J Biol Chem 270: 15242-15249.
16. Habermann A, Schroer TA, Griffiths G et al. (2001): Immunolocalization of cytoplasmic dynein and dynactin subunits in cultured macrophages: enrichment on early endocytic organelles. J Cell Sci 114: 229–240.
17. Goncalves EAP, Dalboni MA, Peres AT et al. (2006): Effect of microcystin on leukocyte viability and function Toxicon 47: 774-779.
18. Yan L, Yan L, Ping-Ping S (2007): Effect of microcystin-LR on phagocytic function and reactive oxygen species level of mouse macrophages. Chin J Pharmacol Toxicol 21:55-58.
19. Palikova M, Navratil S, Krejci R et al. (2004): Outcomes of repeated exposure of the carp (Cyprinus carpio L.) to cyanobacteria extract. Acta Vet Brno 73: 259-265.
20. Palikova M, Kovaru F, Navratil S et al. (1998): The effects of pure microcystin LR and biomass of blue-green algae on selected immunological indices of carp (Cyprinus carpio L.) and silver carp (Hypophthalmichthys molitrix Val.). Acta Vet Brno 67: 265-272.
21. Shen PP, Zhao SW, Zheng WJ et al. (2003): Effects of cyanobacteria bloom extract on some parameters of immune function in mice. Toxicol Lett 143: 27-36.
22. Hudnell HK, ed. (2008): Cyanobacterial harmful algal blooms: State of the sciences and research needs. Springer Sciences and Business Media, LLC.
23. Kujbida P, Hatanaka E, Campa A et al. (2006): Effects of microcystins on human polymorphonuclear leukocytes. Biochem Bioph Res Commun 341: 273-277.
24. Kujbida P, Hatanaka E, Vinolo MAR et al. (2009): Microcystins -LA, -YR, and -LR action on neutrophil migration. Biochem Biophys Res Commun 382: 9-14
25. Hernandez M, Macia M, Padilla C et al. (2000): Modulation of human polymorphonuclear leukocyte adherence by cyanopeptide toxins. Environ Res 84: 64-68.
26. Sieroslawska A, Rymuszka A, Bownik A et al. (2007): The influence of microcystin-LR on flsh phagocytic cells. Hum Exp Toxicol 26: 1-5.
27. Babior BM (1984): The respiratory burst of phagocytes. J Clin Invest 73: 599-601.
28. Nong Q, Komatsu M, Izumo K et al. (2007): Involvement of reactive oxygen species in Microcystin-LR-induced cytogenotoxicity. Free Radic Res 41: 1326-1337.
29. Zhang H, Zhang J, Chen Y et al. (2007): Influence of intracellular Ca(2+), mitochondria membrane potential, reactive oxygen species, and intracellular ATP on the mechanism of microcystin-LR induced apoptosis in Carassius auratus lymphocytes in vitro. Environ Toxicol 22: 559-564.
30. Rosales C (ed.). (2005): Molecular mechanisms of phagocytosis.
31. Campos A, Vasconcelos V (2010): Molecular mechanisms of microcystin toxicity in animal cells. Int J Mol Sci 11: 268-287.
32. Weng D, Lu Y, Wei Y et al. (2007): The role of ROS in microcystin-LR-induced hepatocyte apoptosis and liver injury in mice. Toxicology 232: 15-23.
33. Rocha MFG, Sidrim JJC, Soares AM et al. (2000): Supernatants from macrophages stimulated with microcystin-LR induce electrogenic intestinal response in rabbit ileum. Pharmacol Toxicol 87: 46-51.
34. Kujbida P, Hatanaka E, Campa A et al. (2008): Analysis of chemokines and reactive oxygen species formation by rat and human neutrophils induced by microcystin-LA, -YR and -LR. Toxicon 51: 1274-1280.
35. Shi Q, Cui J, Zhang J et al. (2004): Expression modulation of multiple cytokines in vivo by cyanobacteria blooms extract from Taihu Lake, China. Toxicon 44: 871-879.
36. Chen T, Zhao X, Liu Y et al. (2004): Analysis of immunomodulating nitric oxide iNOS and cytokines mRNA in mouse macrophages induced by microcystin-LR. Toxicology 197: 67-77.
37. Feurstein D, Holst K, Fischer A et al. (2009): Oatp-associated uptake and toxicity of microcystins in primary murine whole brain cells. Toxicol Appl Pharmacol 234: 247-255.
38. Weitzman SA, Gordon LI (1990): Inflammation and cancer: role of phagocyte-generated oxidants in carcinogenesis. Blood 76: 655-663.
39. de Visser KE, Eichten A, Coussens LM (2006): Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 6: 24-37.
40. UenoY, Nagata S, Tsutsumi T et al. (1996): Detection of microcystins, a blue-green algal hepatotoxin, in drinking water sampled in Haimen and Fusui, endemic areas of primary liver cancer in China, by highly sensitive immunoassay. Carcinogenesis 17: 1317-1321.
41. Lian M, Liu Y, Yu SZ et al. (2006): Hepatitis B virus x gene and cyanobacterial toxins promote aflatoxin B1-induced hepatotumorigenesis in mice. World J Gastroenterol 12: 3065-3072.
42. Hernández JM, López-Rodas V, Costas E (2009): Microcystins from tap water could be a risk factor for liver and colorectal cancer: A risk intensified by global change. Med Hypotheses 72: 539-540.
43. Yu SZ (1995): Primary prevention of hepatocellular carcinoma. Gastroenterol Hepatol 10: 674-682.
44. Zhou L, Yu H, Hen K (2002): Relationship between microcystin in drinking water and colorectal cancer. Biomed Envir Sci 15: 166-171.
45. Lankoff A, Bialczyk J, Dziga et al. (2006): Inhibition of nucleotide excision repair (NER) by microcystin-LR in CHO-K1 cells. Toxicon 48: 957-965.
46. Ito E, Kondo F, Terao K et al. (1997): Neoplastic nodular formation in mouse liver induced by repeated intraperitoneal injections of microcystin-LR. Toxicon 35: 1453-1457.
47. Sano T, Takagi H, Sadakane K et al. (2004): Carcinogenic effects of microcystin-LR and Dhb-microcystin-LR on mice liver. Sixth International Conference on Toxic Cyanobacteria, Bergen, Norway, Abstract Book: 59.
48. Nishiwaki-Matsushima R, Ohta T, Nishiwaki S et al. (1992): Liver tumour promotion by the cyanobacterial cyclic peptid toxin microcystin-LR. J Cancer Res Clin Oncol 118: 420-424.
49. Charbonneau M, Hitzfeld BC, Dietrich DR (2004): Liver preneoplasia in rats following an initiation promotion protocol with diethylnitrosamine and microcystin-LR. Unpublished results cited in Dietrich D and Hoeger S (2005): Guidance values for microcystins in water and cyanobacterial supplement products (blue-green algal supplements): a reasonable or misguided approach? Toxic Appl Pharmacol 203: 273- 289.
50. Millward TA, Zolnierowicz S, Hemmings BA (1999): Regulation of protein kinase cascades by protein phosphatase 2A. TIBS 24: 186-191.
51. Lankoff A, Krzowski Ł, Głąb A et al. (2004): DNA damage and repair in human peripheral blood lymphocytes following treatment with microcystin-LR. Mut Res 559: 131-142.
52. Clark SP, Ryan TP, Searfoss GH et al. (2008): Chronic microcystin exposure induces hepatocyte proliferation with increased expression of mitotic and cyclin-associated genes in p53-deficient mice. Toxicol Pathol 36: 190-203.
53. Lankoff A, Bialczyk J, Dziga D et al. (2006): The repair of gamma-radiation-induced DNA damage is inhibited by microcystin-LR, the PP1 and PP2A phosphatase inhibitor, Mutagenesis 21: 83-90.
54. Guzman RE, Solter PF, Runnegar MT (2003): Inhibition of nuclear protein phosphatase activity in mouse hepatocytes by the cyanobacterial toxin microcystin-LR. Toxicon 41: 773-781.
55. Vousden KH and Lu X (2002): Live or let die: The cell’s response to p53. Nature 2: 594-604.
56. Zegura B, Lah TT, Filipic M (2004): The role of reactive oxygen species in microcystin-LR-induced DNA damage. Toxicology 200: 59-68.
57. Tang W, Willers H, Powell SN (1999): P53 directly enhances rejoining of DNA double-strand breaks with cohesive ends in g-irradiated mouse fibroblasts. Cancer Res 59: 2562-2565.
58. Smith ML and Seo YR (2002): p53 regulation of DNA excision repair pathways. Mutagenesis 17: 149-156.
59. Wei Y, Weng D, Li F et al. (2008): Involvement of JNK regulation in oxidative stress-mediated murine liver injury by microcystin-LR. Apoptosis 13: 1031-1042.
60. Antonyak MA, Kenyon LC, Godwin AK et al. (2002): Elevated JNK activation contributes to the pathogenesis of human brain tumors. Oncogene 21: 5038-5046.
61. Huang P and Xu A (2009): Genotoxic effects of microcystin-LR in mammalian cells. Proceedings of 3rd iCBBE 2009: 1-3.
62. Zhan L, Sakamoto H, Sakuraba M (2004): Genotoxicity of microcystin-LR in human lymphoblastoid TK6 cells. Mutat Res 557: 1-6.
63. Mankiewicz J, Walter Z, Tarczynska M et al. (2002): Genotoxicity of cyanobacterial extracts containing microcystins from Polish water reservoirs as determined by SOS chromotest and comet assay. Environ Toxicol 17: 341-350.
64. Suzuki H, Watanabe MF, Wu Y et al. (1998): Mutagenicity of microcystin-LR in human RSa cells. Int J Mol Med 2: 109-112.
65. Wu JY, Xu QJ, Gao G et al. (2006): Evaluating genotoxicity associated with microcystin-LR and its risk to source water safety in Meiliang Bay, Taihu Lake. Environ Toxicol 21: 250-255.
66. Engeli B (2007): Annex to the SKLM opinion “Microcystins in Algae Products Used as a Food Supplement” available at: http://www.dfg.de/download/pdf/dfg_im_profil/reden_stellungnahmen/2007/sklm_microcystine_07_annex_en.pdf
67. Froscio SM, Humpage AR, Burcham PC et al. (2003): Cylindrospermopsin-induced protein synthesis inhibition and its dissociation from acute toxicity in mouse hepatocytes. Environ Toxicol 18: 243-251.
68. Ohtani I, Moore RE, Runnegar MTC (1992): Cylindrospermopsin: a potent hepatotoxin from the blue-green alga Cylindrospermopsis raciborskii. J Am Chem Soc 114: 7941-7942.
69. Shen X, Lam PK, Shaw GR et al. (2002): Genotoxicity investigation of a cyanobacterial toxin, cylindrospermopsin. Toxicon 40: 1499-1501.
70. Shaw GR, Seawright AA, Moore MR et al. (2000): Cylindrospermopsin, a cyanobacterial alkaloid: evaluation of its toxicologic activity. Ther Drug Monit 22: 89-92.
71. Bain P, Shaw G, Patel B (2007): Induction of p53-regulated gene expression in human cell lines exposed to the cyanobacterial toxin cylindrospermopsin. J Toxicol Environ Health 70: 1687-1693.
72. Humpage AR, Fenech M, Thomas P et al. (2000): Micronucleus induction and chromosome loss in transformed human white cells indicate clastogenic and aneugenic action of the cyanobacterial toxin, cylindrospermopsin. Mutation Res 472: 155-161.
73. Lankoff A, Wojcik A, Lisowska H et al. (2007): No induction of structural chromosomal aberrations in cylindrospermopsin-treated CHO-K1 cells without and with metabolic activation. Toxicon 50: 1105-1115.
74. Senogles-Derhama PJ, Seawright A, Shaw G et al. (2003): Toxicological aspects of treatment to remove cyanobacterial toxins from drinking water determined using the heterozygous P53 transgenic mouse model. Toxicon 41: 979-988.
Copyright: © 2010 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.