193
Views
12
CrossRef citations to date
0
Altmetric
Review Article

The lethal effects and determinants of microcystin-LR on heart: a mini review

ORCID Icon, , &
Pages 517-526 | Received 17 Oct 2019, Accepted 31 Dec 2019, Published online: 08 Jan 2020

References

  • Advani, R.H., et al., 2012. Cardiac toxicity associated with bevacizumab (Avastin) in combination with CHOP chemotherapy for peripheral T cell lymphoma in ECOG 2404 trial. Leukemia & lymphoma, 53 (4), 718–720.
  • Allen, B., et al., 2019. Slow-release doxorubicin pellets generate myocardial cardiotoxic changes in mice without significant systemic toxicity. Cardiovascular toxicology, 19 (5), 482–484.
  • Apopa, P., et al., 2018. PARP1 is up-regulated in non-small cell lung cancer tissues in the presence of the cyanobacterial toxin microcystin. Frontiers in microbiology, 9, 1757.
  • Awadalla, M., et al., 2018. Advanced imaging modalities to detect cardiotoxicity. Current problems in cancer, 42 (4), 386–396.
  • Bergamini, C., et al., 2019. Role of speckle tracking echocardiography in the evaluation of breast cancer patients undergoing chemotherapy: review and meta-analysis of the literature. Cardiovascular toxicology, 19 (6), 485–492.
  • Best, J.H., Eddy, F.B., and Codd, G.A., 2001. Effects of purified MC-LR and cell extracts of Microcystis strains PCC 7813 and CYA 43 on cardiac function in brown trout (Salmo trutta) alevins. Fish physiology and biochemistry, 24 (3), 171–178.
  • Boddi, M., et al., 2013. Nonalcoholic fatty liver in nondiabetic patients with acute coronary syndromes. European journal of clinical investigation, 43 (5), 429–438.
  • Cao, L., et al., 2019. Effects of microcystin-LR on the microstructure and inflammation-related factors of jejunum in mice. Toxins, 11 (9), 482.
  • Chaudhari, A., et al., 2017. Circadian clocks, diets and aging. Nutrition and healthy aging, 4 (2), 101–112.
  • Chen, L., et al., 2013. The interactive effects of cytoskeleton disruption and mitochondria dysfunction lead to reproductive toxicity induced by microcystin-LR. PloS One, 8 (1), e53949.
  • Chen, Q.M., et al., 2001. Molecular mechanisms of cardiac hypertrophy induced by toxicants. Cardiovascular toxicology, 1 (4), 267–283.
  • Chen, S., et al., 2017. iTRAQ-based quantitative proteomic analysis of Microcystis aeruginosa exposed to spiramycin at different nutrient levels. Aquatic toxicology, 185, 193–200.
  • Clark, S., et al., 2007. Hepatic gene expression changes in mice associated with prolonged sublethal microcystin exposure. Toxicologic pathology, 35 (4), 594–605.
  • D’Andrea, M.R., et al., 2010. Simultaneous PCNA and TUNEL labeling for testicular toxicity evaluation suggests that detection of apoptosis may be more sensitive than proliferation. Biotechnic & histochemistry, 85 (3), 195–204.
  • Day, B.J., and Lewis, W., 2004. Oxidative stress in NRTI-induced toxicity. Cardiovascular toxicology, 4 (3), 207–216.
  • Ding, W.X., and Ong, C.N., 2003. Role of oxidative stress and mitochondrial changes in cyanobacteria-induced apoptosis and hepatotoxicity. FEMS microbiology letters, 220 (1), 1–7.
  • Fischer, A., et al., 2008. Hematoxylin and eosin staining of tissue and cell sections. Cold spring harbor protocols, 2008, 4986.
  • Guo, R., et al., 2017. New mechanism of lipotoxicity in diabetic cardiomyopathy: deficiency of Endogenous H2S Production and ER stress. Mechanisms of ageing and development, 162, 46–52.
  • Gupta, N., et al., 2003. Comparative toxicity evaluation of cyanobacterial cyclic peptide toxin microcystin variants (LR, RR, YR) in mice. Toxicology, 188 (2–3), 285–296.
  • He, S., et al., 2014. Application of the CellDetect(R) staining technique in diagnosis of human cervical cancer. Gynecologic oncology, 132 (2), 383–388.
  • Iseki, Y., et al., 2018. A new method for evaluating tumor-infiltrating lymphocytes (TILs) in colorectal cancer using hematoxylin and eosin (H-E)-stained tumor sections. PloS One, 13 (4), e0192744.
  • Kujbida, P., 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 (7), 1274–1280.
  • Kyrylkova, K., et al., 2012. Detection of apoptosis by TUNEL assay. In: C. Kioussi, ed. Odontogenesis: Methods and protocols (Methods in Molecular Biology). Vol 887. Humana Press. Available from: doi:https://doi.org/10.1007/978-1-61779-860-3_5.
  • Lankoff, A., et al., 2006. Inhibition of nucleotide excision repair (NER) by microcystin-LR in CHO-K1 cells. Toxicon, 48 (8), 957–965.
  • La-Salete, R., et al., 2008. Mitochondria a key role in microcystin-LR kidney intoxication. Journal of applied toxicology, 28 (1), 55–62.
  • Li, G., et al., 2008. Microcystin-induced variations in transcription of GSTs in an omnivorous freshwater fish, goldfish. Aquatic toxicology, 88 (1), 75–80.
  • Li, Y., et al., 2016. Microcystin-LR induces mitochondria-mediated apoptosis in human bronchial epithelial cells. Experimental and therapeutic medicine, 12 (2):633–640.
  • Liu, H., et al., 2018a. Oxidative stress mediates microcystin-LR-induced endoplasmic reticulum stress and autophagy in KK-1 cells and C57BL/6 mice ovaries. Frontiers in physiology, 9, 1058.
  • Liu, P., et al., 2018b. Isolation, molecular identification, and characterization of a unique toxic cyanobacterium Microcystis sp. found in Hunan Province, China. Journal of toxicology and environmental health, part A, 81 (21), 1142–1149.
  • Liu, Y., et al., 2002. The toxic ffects of microcystin-LR on embryo-larval and juvenile development of loach, Misguruns mizolepis Gunthe. Toxicon, 40 (4), 395–399.
  • Ma, J., et al., 2018. Chronic exposure of nanomolar MC-LR caused oxidative stress and inflammatory responses in HepG2 cells. Chemosphere, 192, 305–317.
  • Martins, N., et al., 2019. Microcystin – LR exposure causes cardiorespiratory impairments and tissue oxidative damage in trahira, Hoplias malabaricus. Ecotoxicology and environmental safety, 173, 436–443.
  • Massey, I.Y., et al., 2018a. Exposure routes and health effects of microcystins on animals and humans: amini-review. Toxicon, 151, 156–162.
  • Massey, I.Y., Zhang, X., and Yang, F., 2018b. Importance of bacterial biodegradation and detoxification processes of microcystins for environmental health. Journal of toxicology and environmental health, part B, 21 (6):357–369.
  • McLellan, N.L., and Manderville, R.A., 2017. Toxic mechanisms of microcystins in mammals. Toxicology research, 6 (4), 391–405.
  • Milutinović, A., et al., 2003. Nephrotoxic effects of chronic administration of microcystins -LR and -YR. Toxicon, 42 (3), 281–288.
  • Milutinovic, A., et al., 2006. Microcystin-LR induces alterations in heart muscle. Folia biologica, 51, 116–118.
  • Omidi, A., Esterhuizen-Londt, M., and Pflugmacher, S., 2018. Still challenging: the ecological function of the cyanobacterial toxin microcystin – what we know so far. Toxin reviews , 37 (2), 87–105.
  • Papadimitriou, T., et al., 2012. Assessment of microcystin distribution and biomagnification in tissues of aquatic food web compartments from a shallow lake and evaluation of potential risks to public health. Ecotoxicology, 21 (4), 1155–1166.
  • Pearson, L., et al., 2010. On the chemistry, toxicology and genetics of the cyanobacterial toxins, microcystin, nodularin, saxitoxin and cylindrospermopsin. Marine drugs, 8 (5), 1650–1680.
  • Phaniendra, A., Jestadi, D.B., and Periyasamy, L., 2015. Free radicals: properties, sources, targets, and their implication in various diseases. Indian journal of clinical biochemistry, 30 (1), 11–26.
  • Pinheiro, C., et al., 2016. The interactive effects of microcystin-LR and cylindrospermopsin on the growth rate of the freshwater algae Chlorella vulgaris. Ecotoxicology, 25 (4), 745–758.
  • Qi, M., et al., 2016. Microcystin-LR induced developmental toxicity and apoptosis in zebrafish (Danio rerio) larvae by activation of ER stress response. Chemosphere, 157, 166–173.
  • Qiu, T., et al., 2009. The profound effects of microcystin on cardiac antioxidant enzymes, mitochondrial function and cardiac toxicity in rat. Toxicology, 257 (1–2), 86–94.
  • Rode, H. D., and Eisel, D. F., 2004. Apoptosis, cell death, and cell proliferation. 3rd ed. London: Roche Applied Science.
  • Ross, C., et al., 2019. Mesohaline conditions represent the threshold for oxidative stress, cell death and toxin release in the cyanobacterium Microcystis aeruginosa. Aquatic toxicology, 206, 203–211.
  • Ross, D.L., Gerald, W.P., and David, R.F., 1995. Hemodynamic and calorimetric changes induced by microcystin-LR in the rat. Journal of applied toxicology, 15, 303–311.
  • Saraf, S.R., et al., 2018. Effects of Microcystis on development of early life stage Japanese medaka (Oryzias latipes): comparative toxicity of natural blooms, cultured Microcystis and microcystin-LR. Aquatic toxicology, 194, 18–26.
  • Saraste, A., and Pulkki, K., 2000. Morphologic and biochemical hallmarks of apoptosis. Cardiovascular research, 45 (3), 528–537.
  • Sawaya, H., et al., 2011. Early detection and prediction of cardiotoxicity in chemotherapy-treated patients. The American journal of cardiology, 107 (9), 1375–1380.
  • Sedmak, B., et al., 2009. The biological role of cyclic hepatotoxic and non-hepatotoxic cyanopeptides and its ecological consequences. In: E. B. Santos, ed. Ecotoxicology research developments. 169–300.
  • Shi, Y., et al., 2015. Oxidative stress and histopathological alterations in liver of Cyprinus carpio L. induced by intraperitoneal injection of microcystin-LR. Ecotoxicology, 24, 511–519.
  • Shuai, Y., et al., 2017. Characterization of microcystin-induced dualistic toxic effects on primary rat hepatocytes. Journal of environmental pathology, toxicology and oncology, 36 (1), 15–27.
  • Su, Y., et al., 2018. Mutual promotion of apoptosis and autophagy in prepubertal rat testes induced by joint exposure of bisphenol A and nonylphenol. Environmental pollution, 243, 693–702.
  • Suput, D., et al., 2010. Cardiotoxic injury caused by chronic administration of microcystin-YR. Folia biologica, 56 (1), 14–18.
  • Tang, Y., et al., 2018. Environmental risks of ZnO nanoparticle exposure on Microcystis aeruginosa: toxic effects and environmental feedback. Aquatic toxicology, 204, 19–26.
  • Uttara, B., et al., 2009. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Current neuropharmacology, 7 (1), 65–74.
  • Wang, Q., et al., 2019. Microcystin-LR induces angiodysplasia and vascular dysfunction through promoting cell apoptosis by the mitochondrial signaling pathway. Chemosphere, 218, 438–448.
  • Wang, Q., et al., 2008. Distribution of microcystins in various organs (heart, liver, intestine, gonad, brain, kidney and lung) of Wistar rat via intravenous injection. Toxicon, 52 (6), 721–727.
  • Wang, X., et al., 2018. The differential effects of microcystin-LR on mitochondrial DNA in the hippocampus and cerebral cortex. Environmental pollution, 240, 68–76.
  • Wei, J., et al., 2019. Simultaneous Microcystis algicidal and microcystin synthesis inhibition by a red pigment prodigiosin. Environmental pollution, 256, 113444.
  • Xie, J., et al., 2019. Enantiomeric environmental behavior, oxidative stress and toxin release of harmful cyanobacteria Microcystis aeruginosa in response to napropamide and acetochlor. Environmental pollution, 246, 728–733.
  • Xu, Y.Q., et al., 2012. Diurnal variation of hepatic antioxidant gene expression in mice. PloS One, 7, e44237–e44237.
  • Young, F.M., et al., 2005. Immunogold localisation of microcystins in cryosectioned cells of Microcystis. Journal of structural biology, 151 (2), 208–214.
  • Zegura, B., Lah, T., and Filipic, M., 2006. Alteration of intracellular GSH levels and its role in microcystin-LR-induced DNA damage in human hepatoma HepG2 cells. Mutation research/genetic toxicology and environmental mutagenesis, 611 (1–2), 25–33.
  • Zhang, C., et al., 2019a. Identification and characterization of a novel indigenous algicidal bacterium Chryseobacterium species against Microcystis aeruginosa. Journal of toxicology and environmental health, part A, 82 (15), 845–853.
  • Zhang, H., et al., 2007. Influence of intracellular Ca2+, mitochondria membrane potential, reactive oxygen species, and intracellular ATP on the mechanism of microcystin-LR induced apoptosis in Carassius auratus lymphocytes in vitro. Environmental toxicology, 22 (6), 559–564.
  • Zhang, Y., et al., 2019. Microcystin-LR disturbs testicular development of giant freshwater prawn Macrobrachium rosenbergii. Chemosphere, 222, 584–592.
  • Zhang, Z., et al., 2002. The acute toxic effects of microcystin LR in SD rats. Zhonghua Yu Fang Yi Xue Za Zhi, 36 (5), 295–297.
  • Zhao, Y., et al., 2008. In vivo studies on the toxic effects of microcystins on mitochondrial electron transport chain and ion regulation in liver and heart of rabbit. Comparative biochemistry and physiology part C: toxicology & pharmacology, 148, 204–210.
  • Zhou, M., Tu, W., and Xu, J., 2015. Mechanisms of microcystin-LR-induced cytoskeletal disruption in animal cells. Toxicon, 101, 92–100.
  • Zi, J., et al., 2018. Cyanobacteria blooms induce embryonic heart failure in an endangered fish species. Aquatic toxicology, 194, 78–85.
  • Zong, W., et al., 2017. Molecular mechanism for the regulation of microcystin toxicity to protein phosphatase 1 by glutathione conjugation pathway. BioMed research international, 2017, 1–10.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.