Microcystin-LR

Microcystin

Last updated at: Nov. 2, 2023

Recommended name
Microcystin-LR
Synonyms
Cyanoginosin-LR
Recommended acronym
MC-LR
Abbreviation

Progenitors

Name
Microcystis aeruginosa
Note
Botes, D.P., Wessels, P.L., Kruger, H., Runnegar, M.T.C., Santikarn, S., Smith, R.J., Barna, J.C.J., Williams, D.H., 1985. Structural studies on cyanoginosins-LR, -YR, -YA, and -YM, peptide toxins from Microcystis aeruginosa. J. Chem. Soc. Perkin Trans. I 1 (0), 2747–2748. Adams, W.H., Stone, J.P., Sylvester, B., Stoner, R.D., Slatkin, D.N., Tempel, N.R., Siegelman, H.W., 1988. Pathophysiology of cyanoginosin-LR: in vivo and in vitro studies. Toxicol Appl Pharmacol. 96(2):248-57. doi: 10.1016/0041-008x(88)90084-1. PMID: 3194914.
Name
Synechococcus
Note
Carmichael, W.W., Li, R., 2006. Cyanobacteria toxins in the Salton Sea. Aquat. Biosyst. 2, 5 (2006). https://doi.org/10.1186/1746-1448-2-5
Name
Microcystis
Note
Oudra, B., Loudiki, M., Sbyyaa, B., Sabour, B., Martins, R., Amori, A. and Vasconcelos, V., 2002. Detection and variation of microcystin contents of Microcystis blooms in eutrophic Lalla Takerkoust Lake, Morocco. Lakes & Reservoirs: Research & Management, 7: 35-44. https://doi.org/10.1046/j.1440-1770.2002.00165.x
Name
Roseofilum
Note
Richardson, L.L., Sekar, R., Myers, J.L., Gantar, M., Voss, J.D., Kaczmarsky, L., Remily, E.R., Boyer, G.L., Zimba, P.V., 2007. The presence of the cyanobacterial toxin microcystin in black band disease of corals, FEMS Microbiology Letters, 272(2), 182–187, https://doi.org/10.1111/j.1574-6968.2007.00751.x Casamatta, D., Stanić, D., Gantar, M., Richardson, L.L., 2012. Characterization of Roseofilum reptotaenium (Oscillatoriales, Cyanobacteria) gen. et sp. nov. isolated from Caribbean black band disease, Phycologia, 51:5, 489-499, DOI: 10.2216/11-10.1
Name
Aphanizomenon flosaquae
Note
Šulčius, S., Pilkaitytė, R., Mazur-Marzec, H., Kasperovičienė, J., Ezhova, E., Błaszczyk, A., Paškauskas, R., 2015. Increased risk of exposure to microcystins in the scum of the filamentous cyanobacterium Aphanizomenon flos-aquae accumulated on the western shoreline of the Curonian Lagoon. Mar Pollut Bull. 99(1-2):264-70. doi: 10.1016/j.marpolbul.2015.07.057. Epub 2015 Jul 30. PMID: 26234611.
Name
Dolichospermum
Note
Sivonen, K., Namikoshi, M., Evans, W.R., Carmichael, W.W., Sun, F., Rouhiainen, L., Luukkainen, R., Rinehart, K.L., 1992. Isolation and characterization of a variety of microcystins from seven strains of the cyanobacterial genus Anabaena. Appl Environ Microbiol. 58(8):2495-500. doi: 10.1128/aem.58.8.2495-2500.1992. PMID: 1514796; PMCID: PMC195810.
Name
Gloeotrichia echinulata
Note
Carey, C.C., Haney, J.F. and Cottingham, K.L., 2007. First report of microcystin-LR in the cyanobacterium Gloeotrichia echinulata. Environ. Toxicol., 22: 337-339. https://doi.org/10.1002/tox.20245
Name
Leptolyngbya
Note
Mahanil, K., Sensupa, A., Pekkoh, J., Tragoolpua, Y., Pumas, C., 2021. Application of phycobiliproteins from Leptolyngbya sp. KC45 for natural illuminated colourant beverages. J Appl Phycol 33, 3747–3760. https://doi.org/10.1007/s10811-021-02556-3
Name
Planktothrix agardhii
Note
Fujii, K., Sivonen, K., Naganawa, E., Harada, K.-I., 2000. Non-Toxic Peptides from Toxic Cyanobacteria, Oscillatoria agardhii. Tetrahedron 56 (5), 725-733. https://doi.org/10.1016/S0040-4020(99)01017-0
Name
Nostoc
Note
Oudra, B., Dadi-El Andaloussi, M., Vasconcelos, V.M., 2009. Identification and quantification of microcystins from a Nostoc muscorum bloom occurring in Oukaïmeden River (High-Atlas mountains of Marrakech, Morocco). Environ Monit Assess 149, 437–444. https://doi.org/10.1007/s10661-008-0220-y

Vector Species

Name
Chanodichthys erythropterus
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120
Name
Coilia nasus
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120
Name
Procambarus clarkii
Note
Chen, J., Xie, P., 2005. Tissue distributions and seasonal dynamics of the hepatotoxic microcystins-LR and -RR in two freshwater shrimps, Palaemon modestus and Macrobrachium nipponensis, from a large shallow, eutrophic lake of the subtropical China. Toxicon. 45(5):615-25. doi: 10.1016/j.toxicon.2005.01.003. PMID: 15777958.
Name
Mytilus trossulus
Note
Preece, E.P., Moore, B.C., Hardy, F.J., Deobald, L.A., 2015. First detection of microcystin in Puget Sound, Washington, mussels (Mytilus trossulus), Lake and Reservoir Management, 31:1, 50-54, DOI: 10.1080/10402381.2014.998398
Name
Lymnaea stagnalis
Note
Zurawell, R.W., Kotak, B.G., Prepas, E.E., 1999. Influence of lake trophic status on the occurrence of microcystin-LR in the tissue of pulmonate snails. Freshwater Biology, 42: 707-718. https://doi.org/10.1046/j.1365-2427.1999.00499.x Kotak, B.G., Zurawell, R.W., Prepas, E.E., Holmes, C.F.B., 1996. Microcystin-LR concentration in aquatic food web compartments from lakes of varying trophic status. Canadian Journal of Fisheries and Aquatic Sciences. 53(9): 1974-1985. https://doi.org/10.1139/f96-135
Name
Carassius auratus
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120 Jia, J., Luo, W., Lu, Y., Giesy, J.P., 2014. Bioaccumulation of microcystins (MCs) in four fish species from Lake Taihu, China: assessment of risks to humans. Sci Total Environ. 15;487:224-32. doi: 10.1016/j.scitotenv.2014.04.037. Epub 2014 May 3. PMID: 24784747.
Name
Hypophthalmichthys molitrix
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120 Jia, J., Luo, W., Lu, Y., Giesy, J.P., 2014. Bioaccumulation of microcystins (MCs) in four fish species from Lake Taihu, China: assessment of risks to humans. Sci Total Environ. 15;487:224-32. doi: 10.1016/j.scitotenv.2014.04.037. Epub 2014 May 3. PMID: 24784747.
Name
Parabramis pekinensis
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120
Name
Cyprinus carpio
Note
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y. and Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120
Name
Hypophthalmichthys nobilis
Note
Jia, J., Luo, W., Lu, Y., Giesy, J.P., 2014. Bioaccumulation of microcystins (MCs) in four fish species from Lake Taihu, China: assessment of risks to humans. Sci Total Environ. 15;487:224-32. doi: 10.1016/j.scitotenv.2014.04.037. Epub 2014 May 3. PMID: 24784747.
Name
Cyprinus carpio
Note
Jia, J., Luo, W., Lu, Y., Giesy, J.P., 2014. Bioaccumulation of microcystins (MCs) in four fish species from Lake Taihu, China: assessment of risks to humans. Sci Total Environ. 15;487:224-32. doi: 10.1016/j.scitotenv.2014.04.037. Epub 2014 May 3. PMID: 24784747. Singh, S., Asthana, R.K., 2014. Assessment of Microcystin Concentration in Carp and Catfish: A Case Study from Lakshmikund Pond, Varanasi, India. Bull Environ Contam Toxicol 92, 687–692. https://doi.org/10.1007/s00128-014-1277-7
Name
Clarias batrachus
Note
Singh, S., Asthana, R.K., 2014. Assessment of Microcystin Concentration in Carp and Catfish: A Case Study from Lakshmikund Pond, Varanasi, India. Bull Environ Contam Toxicol 92, 687–692. https://doi.org/10.1007/s00128-014-1277-7
Name
Odontesthes bonariensis
Note
Ame, M.V., Galanti, L.N., Menone, M.L., Gerpe, M.S., Moreno, V.J., Wunderlin, D.A., 2010. Microcystin–LR, –RR, –YR and –LA in water samples and fishes from a shallow lake in Argentina. Harmful Algae 9(1), 66-73. https://doi.org/10.1016/j.hal.2009.08.001
Name
Oreochromis niloticus
Note
Semyalo, R., Rohrlack, T., Naggawa, C., Nyakairu, G.W., 2010. Microcystin concentrations in Nile tilapia (Oreochromis niloticus) caught from Murchison Bay, Lake Victoria and Lake Mburo: Uganda. Hydrobiologia 638, 235–244. https://doi.org/10.1007/s10750-009-0047-8
Name
Penaeus vannamei
Note
Zimba, P.V., Camus, A., Allen, E.H., Burkholder, J.M., 2006. Co-occurrence of white shrimp, Litopenaeus vannamei, mortalities and microcystin toxin in a southeastern USA shrimp facility. Aquaculture 261(3), 1048-1055. https://doi.org/10.1016/j.aquaculture.2006.08.037
Name
Ictalurus
Note
Zimba, P.V., Khoo, L., Gaunt, P.S., Brittain, S., Carmichael, W.W., 2001. Confirmation of catfish, Ictalurus punctatus (Rafinesque), mortality from Microcystis toxins. Journal of Fish Diseases, 24: 41-47. https://doi.org/10.1046/j.1365-2761.2001.00273.x
Name
Corbicula
Note
Chen, J., Xie, P., 2008. Accumulation of hepatotoxic microcystins in freshwater mussels, aquatic insect larvae and oligochaetes in a large, shallow eutrophic lake (Lake Chaohu) of subtropical China. Fresenius Environmental Bulletin 17(7A), 849-854. https://hero.epa.gov/hero/index.cfm/reference/details/reference_id/7449317
Name
Macrobrachium
Note
Chen, J., Xie, P., 2005. Tissue distributions and seasonal dynamics of the hepatotoxic microcystins-LR and -RR in two freshwater shrimps, Palaemon modestus and Macrobrachium nipponensis, from a large shallow, eutrophic lake of the subtropical China. Toxicon. 45(5):615-25. doi: 10.1016/j.toxicon.2005.01.003. PMID: 15777958.

References

Carey, C.C., 2007
Carey, C.C., Haney, J.F., Cottingham, K.L., 2007. First report of microcystin-LR in the cyanobacterium Gloeotrichia echinulata. Environ. Toxicol., 22: 337-339. https://doi.org/10.1002/tox.20245
Mahanil, K., 2021
Mahanil, K., Sensupa, A., Pekkoh, J., Tragoolpua, Y., Pumas, C., 2021. Application of phycobiliproteins from Leptolyngbya sp. KC45 for natural illuminated colourant beverages. J Appl Phycol 33, 3747–3760. https://doi.org/10.1007/s10811-021-02556-3
Fujii, K., 2000
Fujii, K., Sivonen, K., Naganawa, E., Harada, K.-I., 2000. Non-Toxic Peptides from Toxic Cyanobacteria, Oscillatoria agardhii. Tetrahedron 56 (5), 725-733. https://doi.org/10.1016/S0040-4020(99)01017-0
Oudra, B., 2009
Oudra, B., Dadi-El Andaloussi, M., Vasconcelos, V.M., 2009. Identification and quantification of microcystins from a Nostoc muscorum bloom occurring in Oukaïmeden River (High-Atlas mountains of Marrakech, Morocco). Environ Monit Assess 149, 437–444. https://doi.org/10.1007/s10661-008-0220-y
Jia, J., 2014
Jia, J., Luo, W., Lu, Y., Giesy, J.P., 2014. Bioaccumulation of microcystins (MCs) in four fish species from Lake Taihu, China: assessment of risks to humans. Sci Total Environ. 15;487:224-32. doi: 10.1016/j.scitotenv.2014.04.037. Epub 2014 May 3. PMID: 24784747.
Xie, L., 2005
Xie, L., Xie, P., Guo, L., Li, L., Miyabara, Y., Park, H.-D., 2005. Organ distribution and bioaccumulation of microcystins in freshwater fish at different trophic levels from the eutrophic Lake Chaohu, China. Environ. Toxicol., 20: 293-300. https://doi.org/10.1002/tox.20120
Botes, D.P., 1985
Botes, D.P., Wessels, P.L., Kruger, H., Runnegar, M.T.C., Santikarn, S., Smith, R.J., Barna, J.C.J., Williams, D.H., 1985. Structural studies on cyanoginosins-LR, -YR, -YA, and -YM, peptide toxins from Microcystis aeruginosa. J. Chem. Soc. Perkin Trans. I 1 (0), 2747–2748. doi: 10.1016/0041-008x(88)90084-1. PMID: 3194914.
Carmichael, W.W., 2006
Carmichael, W.W., Li, R., 2006. Cyanobacteria toxins in the Salton Sea. Aquat. Biosyst. 2, 5 (2006). https://doi.org/10.1186/1746-1448-2-5
Adams, W.H., 1988
Adams, W.H., Stone, J.P., Sylvester, B., Stoner, R.D., Slatkin, D.N., Tempel, N.R., Siegelman, H.W., 1988. Pathophysiology of cyanoginosin-LR: in vivo and in vitro studies. Toxicol Appl Pharmacol. 96(2):248-57. doi: 10.1016/0041-008x(88)90084-1. PMID: 3194914.
Richardson, L.L., 2007
Richardson, L.L., Sekar, R., Myers, J.L., Gantar, M., Voss, J.D., Kaczmarsky, L., Remily, E.R., Boyer, G.L., Zimba, P.V., 2007. The presence of the cyanobacterial toxin microcystin in black band disease of corals, FEMS Microbiology Letters, 272(2), 182–187, https://doi.org/10.1111/j.1574-6968.2007.00751.x
Singh, S., 2014
Singh, S., Asthana, R.K., 2014. Assessment of Microcystin Concentration in Carp and Catfish: A Case Study from Lakshmikund Pond, Varanasi, India. Bull Environ Contam Toxicol 92, 687–692. https://doi.org/10.1007/s00128-014-1277-7
Ame, M.V., 2010
Ame, M.V., Galanti, L.N., Menone, M.L., Gerpe, M.S., Moreno, V.J., Wunderlin, D.A., 2010. Microcystin–LR, –RR, –YR and –LA in water samples and fishes from a shallow lake in Argentina. Harmful Algae 9(1), 66-73. https://doi.org/10.1016/j.hal.2009.08.001
Semyalo, R., 2010
Semyalo, R., Rohrlack, T., Naggawa, C., Nyakairu, G.W., 2010. Microcystin concentrations in Nile tilapia (Oreochromis niloticus) caught from Murchison Bay, Lake Victoria and Lake Mburo: Uganda. Hydrobiologia 638, 235–244. https://doi.org/10.1007/s10750-009-0047-8
Zimba, P.V., 2006
Zimba, P.V., Camus, A., Allen, E.H., Burkholder, J.M., 2006. Co-occurrence of white shrimp, Litopenaeus vannamei, mortalities and microcystin toxin in a southeastern USA shrimp facility. Aquaculture 261(3), 1048-1055. https://doi.org/10.1016/j.aquaculture.2006.08.037
Zimba, P.V., 2001
Zimba, P.V., Khoo, L., Gaunt, P.S., Brittain, S., Carmichael, W.W., 2001. Confirmation of catfish, Ictalurus punctatus (Rafinesque), mortality from Microcystis toxins. Journal of Fish Diseases, 24: 41-47. https://doi.org/10.1046/j.1365-2761.2001.00273.x
Chen, J., 2008
Chen, J., Xie, P., 2008. Accumulation of hepatotoxic microcystins in freshwater mussels, aquatic insect larvae and oligochaetes in a large, shallow eutrophic lake (Lake Chaohu) of subtropical China. Fresenius Environmental Bulletin 17(7A), 849-854. https://hero.epa.gov/hero/index.cfm/reference/details/reference_id/7449317
Chen, J., 2005
Chen, J., Xie, P., 2005. Tissue distributions and seasonal dynamics of the hepatotoxic microcystins-LR and -RR in two freshwater shrimps, Palaemon modestus and Macrobrachium nipponensis, from a large shallow, eutrophic lake of the subtropical China. Toxicon. 45(5):615-25. doi: 10.1016/j.toxicon.2005.01.003. PMID: 15777958.
Preece, E.P., 2015
Preece, E.P., Moore, B.C., Hardy, F.J., Deobald, L.A., 2015. First detection of microcystin in Puget Sound, Washington, mussels (Mytilus trossulus), Lake and Reservoir Management, 31:1, 50-54, DOI: 10.1080/10402381.2014.998398
Zurawell, R.W., 1999
Zurawell, R.W., Kotak, B.G., Prepas, E.E., 1999. Influence of lake trophic status on the occurrence of microcystin-LR in the tissue of pulmonate snails. Freshwater Biology, 42: 707-718. https://doi.org/10.1046/j.1365-2427.1999.00499.x
Kotak, B.G., 1996
Kotak, B.G., Zurawell, R.W., Prepas, E.E., Holmes, C.F.B., 1996. Microcystin-LR concentration in aquatic food web compartments from lakes of varying trophic status. Canadian Journal of Fisheries and Aquatic Sciences. 53(9): 1974-1985. https://doi.org/10.1139/f96-135
Oudra, B., 2002
Oudra, B., Loudiki, M., Sbyyaa, B., Sabour, B., Martins, R., Amori, A., Vasconcelos, V., 2002. Detection and variation of microcystin contents of Microcystis blooms in eutrophic Lalla Takerkoust Lake, Morocco. Lakes & Reservoirs: Research & Management, 7: 35-44. https://doi.org/10.1046/j.1440-1770.2002.00165.x
Casamatta, D., 2012
Casamatta, D., Stanić, D., Gantar, M., Richardson, L.L., 2012. Characterization of Roseofilum reptotaenium (Oscillatoriales, Cyanobacteria) gen. et sp. nov. isolated from Caribbean black band disease, Phycologia, 51:5, 489-499, DOI: 10.2216/11-10.1
Šulčius, S., 2015
Šulčius, S., Pilkaitytė, R., Mazur-Marzec, H., Kasperovičienė, J., Ezhova, E., Błaszczyk, A., Paškauskas, R., 2015. Increased risk of exposure to microcystins in the scum of the filamentous cyanobacterium Aphanizomenon flos-aquae accumulated on the western shoreline of the Curonian Lagoon. Mar Pollut Bull. 99(1-2):264-70. doi: 10.1016/j.marpolbul.2015.07.057. Epub 2015 Jul 30. PMID: 26234611.
Sivonen, K., 1992
Sivonen, K., Namikoshi, M., Evans, W.R., Carmichael, W.W., Sun, F., Rouhiainen, L., Luukkainen, R., Rinehart, K.L., 1992. Isolation and characterization of a variety of microcystins from seven strains of the cyanobacterial genus Anabaena. Appl Environ Microbiol. 58(8):2495-500. doi: 10.1128/aem.58.8.2495-2500.1992. PMID: 1514796; PMCID: PMC195810.
Structure
Formula
C49H74N10O12
Exact mono-isotopic mass
994.54877
Molfile
n/a
Alternative molfiles
n/a
SMILES
O=C(N(C)C(C(N[C@@H]1C)=O)=C)CC[C@H](C(O)=O)NC([C@@H](C)[C@H](/C=C/C(C)=C/[C@H](C)[C@@H](OC)CC2=CC=CC=C2)NC([C@H](CCCNC(N)=N)NC([C@@H](C)[C@H](C(O)=O)NC([C@@H](NC1=O)CC(C)C)=O)=O)=O)=O
Alternative SMILES
n/a
InChi key
ZYZCGGRZINLQBL-GWRQVWKTSA-N
Alternative InChi keys
n/a
InChi
InChI=1S/C49H74N10O12/c1-26(2)23-37-46(66)58-40(48(69)70)30(6)42(62)55-35(17-14-22-52-49(50)51)45(65)54-34(19-18-27(3)24-28(4)38(71-10)25-33-15-12-11-13-16-33)29(5)41(61)56-36(47(67)68)20-21-39(60)59(9)32(8)44(64)53-31(7)43(63)57-37/h11-13,15-16,18-19,24,26,28-31,34-38,40H,8,14,17,20-23,25H2,1-7,9-10H3,(H,53,64)(H,54,65)(H,55,62)(H,56,61)(H,57,63)(H,58,66)(H,67,68)(H,69,70)(H4,50,51,52)/b19-18+,27-24+/t28-,29-,30-,31+,34-,35-,36+,37-,38-,40+/m0/s1
Alternative InChis
Spectra available
True
Chem files

References

Carmichael, W.W., 1988
Carmichael, W.W., Beasley, V., Bunner, D.L., Eloff, J.N., Falconer, I., Gorham, P., Harada, K., Krishnamurthy, T., Yu, M.J., Moore, R.E., Rinehart, K., Runnegar, M., Skulberg, O.M., Watanabe, M., 1988. Naming of cyclic heptapeptide toxins of cyanobacteria (blue-green algae). Toxicon. 1988;26(11):971-3. doi: 10.1016/0041-0101(88)90195-x. PMID: 3149803.
Botes, D.P., 1985
Botes, D.P., Wessels, P.L., Kruger, H., Runnegar, M.T.C., Santikarn, S., Smith, R.J., Barna, J.C.J., Williams, D.H., 1985. Structural studies on cyanoginosins-LR, -YR, -YA, and -YM, peptide toxins from Microcystis aeruginosa. J. Chem. Soc. Perkin Trans. I 1 (0), 2747–2748.
Certified
True
Certified links
Non certified reference material
True

Chemical analysis

Research
True
Standardized
Unknown
Validated
True
Official
EPA document # EPA/600/R-17/344

Structure recognition assays

Research
True
Standardized
Unknown
Validated
True
Official
n/a

Functional assays

Research
True
Standardized
Unknown
Validated
Unknown
Official
n/a

Animal assays

Research
True
Standardized
Unknown
Validated
Unknown
Official
n/a

References

Shoemaker, J.A., 2017
Shoemaker, J.A., Tettenhorst, D.R., de la Cruz, A., 2017. Single Laboratory Validated Method for Determination of Microcystins and Nodularin in Ambient Freshwaters by Solid Phase Extraction and Liquid Chromatography/Tandem Mass Spectrometry (LC/MS/MS), U.S. Environmental Protection Agency, EPA document # EPA/600/R-17/344.
Zaffiro, A., 2016
Zaffiro, A., Rosenblum, L., Wendelken, S.C., 2016. Method 546: Determination of Total Microcystins and Nodularins in Drinking Water and Ambient Water by Adda Enzyme-Linked Immunosorbent Assay. U.S. Environmental Protection Agency.
Bostan, H.B., 2018
Bostan, H.B., Taghdisi, S.M., Taghdisi, S.M., Demertzis, N., Rezaee, R., Panahi, Y., Tsatsakis, A.M., Karimi, G., 2018. Determination of microcystin-LR, employing aptasensors. Biosens Bioelectron. 15;119:110-118. doi: 10.1016/j.bios.2018.08.003. Epub 2018 Aug 8. PMID: 30121422.
Yakes, B.J., 2015
Yakes, B.J., Handy, S.M., Kanyuck, K.M., DeGrasse, S.L., 2015. Improved screening of microcystin genes and toxins in blue-green algal dietary supplements with PCR and a surface plasmon resonance biosensor. Harmful Algae 47, 9-16. https://doi.org/10.1016/j.hal.2015.05.001
Long, F., 2009
Long, F., Shi, H.C., He, M., Sheng, J.W., Wang, J.F., 2009. Sensitive and rapid chemiluminescence enzyme immunoassay for microcystin-LR in water samples. Analytica Chimica Acta 649(1), 123-127. https://doi.org/10.1016/j.aca.2009.07.026
Ma, W., 2009
Ma, W., Chen, W., Qiao, R., Liu, C., Yang, C., Li, Z., Xu, D., Peng, C, Jin, Z., Xu, C., Zhu, S., Wang, L., 2009. Rapid and sensitive detection of microcystin by immunosensor based on nuclear magnetic resonance. Biosensors and Bioelectronics 25(1), 240-243. https://doi.org/10.1016/j.bios.2009.06.020
Sassolas, A., 2011
Sassolas, A., Catanante, G., Fournier, D., Marty, J.L., 2011. Development of a colorimetric inhibition assay for microcystin-LR detection: comparison of the sensitivity of different protein phosphatases. Talanta. 85(5):2498-503. doi: 10.1016/j.talanta.2011.07.101. Epub 2011 Aug 5. PMID: 21962674.
Niu, W., 2012
Niu, W., He, E., Wu, Q., Zhou, W., Zhang, Y., Huang, B., Zhao, X., 2012. Use of fluorescent europium chelates as labels for detection of microcystin-LR in Taihu Lake, China. Journal of Rare Earths 30(9), 941-946. https://doi.org/10.1016/S1002-0721(12)60158-6
Murphy, C., 2015
Murphy, C., Stack, E., Krivelo, S., McPartlin, D.A., Byrne, B., Greef, C., Lochhead, M.J., Husar, G., Devlin, S., Elliott, C.T., O'Kennedy, R.J., 2015. Detection of the cyanobacterial toxin, microcystin-LR, using a novel recombinant antibody-based optical-planar waveguide platform. Biosens Bioelectron. 67:708-14. doi: 10.1016/j.bios.2014.10.039. Epub 2014 Oct 23. PMID: 25459059.
Neumannm, A.-C., 2015
Neumannm, A.-C., Wang, X., Niessner, R., Knopp, D., 2015. Determination of microcystin-LR in surface water by a magnetic bead-based colorimetric immunoassay using antibody-conjugated gold nanoparticles. Anal. Methods, 2016,8, 57-63. https://doi.org/10.1039/C5AY02164B
Masango, M., 2008
Masango, M., Myburgh, J., Botha, C., Labuschagne, L., Naicker, D., 2008, A comparison of in vivo and in vitro assays to assess the toxicity of algal blooms. Water Research 42, 3241-3248.
Lawton, L.A., 1994
Lawton, L.A., Edward, C., Codd, G.A., Extraction and high-performance liquid chromatographic method for the determination of microcystins in raw and treated Waters. Analyst 119, 1525-1530.
Turner, A.D., 2018
Turner, A.D., Waack, J., Lewis, A., Edwards, C., Lawton, L. 2018. Development and single-laboratory validation of a UHPLC-MS/MS method for quantitation of microcystins and nodularin in natural water, cyanobacteria, shellfish and algal supplement tablet powder. J. Chromatogr. B 1074-1075, 111-123.
Wang, C.C., 2016
Wang, C.C., Petty, E.E., Smith, C.M. 2016. Rapid and efficient analysis of microcystins, nodularin, cylindrospermopsin, and anatoxin-a in drinking water by LC tandem MS, J. AOAC Int. 99, 1565-1571.
Altaner, S., 2019
Altaner, S., Puddick, J., Fessard, V., Feurstein, D., Zemskov, I., Dietrich, D.R. 2019. Simultaneous Detection of 14 Microcystin Congeners from Tissue Samples Using UPLC- ESI-MS/MS and Two Dierent Deuterated Synthetic Microcystins as Internal Standards. Toxins 11, 388.
Yang, R., 2018
Yang, R., Song, D., Fang, S., Liu, Y., Zhou, X., Long, F., Zhu, A., 2018. Development of novel portable and reusable fiber optical chemiluminescent biosensor and its application for sensitive detection of microcystin-LR. Biosensors and Bioelectronics 121(15), 27-33. https://doi.org/10.1016/j.bios.2018.08.062
Aguete, E.C., 2003
Aguete, E.C., Gago-Martínez, A., Leão, J.M., Rodríguez-Vázquez, J.A., Menàrd, C., Lawrence, J.F., 2003. HPLC and HPCE analysis of microcystins RR, LR and YR present in cyanobacteria and water by using immunoaffinity extraction. Talanta. 59(4):697-705. doi: 10.1016/S0039-9140(02)00610-0. PMID: 18968958.
Liu, Y., 2019
Liu, Y., Ji., J., Cui, F., Sun, J., Wu, H., Pi, F., Zhang, Y., Sun, X., 2019. Development of a two-step immunochromatographic assay for microcystin-LR based on fluorescent microspheres. Food Control 95, 34-40. https://doi.org/10.1016/j.foodcont.2018.07.036
Pang, P., 2018
Pang, P., Teng, X., Chen, M., Zhang, Y., Wang, H., Yang, C., Yang, W., Barrow, C.J., 2018. Ultrasensitive enzyme-free electrochemical immunosensor for microcystin-LR using molybdenum disulfide/gold nanoclusters nanocomposites as platform and Au@Pt core-shell nanoparticles as signal enhancer. Sensors and Actuators B: Chemical 266, 400-407. https://doi.org/10.1016/j.snb.2018.03.154
Zhang, G., 2018
Zhang, G., Li, C., Wu, S., Zhang, Q., 2018. Label-free aptamer-based detection of microcystin-LR using a microcantilever array biosensor. Sensors and Actuators B: Chemical 260, 42-47. https://doi.org/10.1016/j.snb.2017.12.112
Ward, C.J., 1997
Ward, C.J., a,  Beattie, K.A.,  Lee, E.Y.C., Codd, G.A., 1997. Colorimetric protein phosphatase inhibition assay of laboratory strains and natural blooms of cyanobacteria: comparisons with high-performance liquid chromatographic analysis for microcystins. FEMS Microbiology Letters 153, 465-473.
Samdal, I.A., 2014
Samdal, I.A., Ballot, A., Løvberg, K.E., Miles, C.O., 2014, Multihapten approach leading to a sensitive ELISA withbroad cross-reactivity to microcystins and nodularin. Environ. Sci. Technol. 2014, 48, 14, 8035–8043
Manubolu, M., 2018
Manubolu, M., Lee, J., Riedl, K.M., Kua, Z., Collart, L.P., Ludsin, S.A. 2018. Optimization of extraction methods for quantification of microcystin-LR and microcystin-RR in fish, vegetable, and soil matrices using UPLC–MS/MS. Harmful Algae 76, 47-57.
Regulatory status
False

References

World Health Organization, 2020
World Health Organization 2020, Cyanobacterial toxins: microcystins, Background document for development of WHO Guidelines for drinking-water quality and Guidelines for safe recreational water environments.
US EPA, 2015
United States Environmental Protection Agency, Drinking Water Health Advisory for the Cyanobacterial Microcystin Toxins. EPA-820R15100.
Human toxic syndrome(s)
n/a
Organ system toxicity
Hepatotoxicity
Risk assessment
Unknown
Molecular targets known
True
Molecular targets
Protein phosphatase 2A, Protein phosphatase 1
Toxic to aquatic animals
True
TEF available
True

Risk assessment References

Foxall, T.L., 1981
Foxall, T.L., Sasner, J.J. (1981). Effects of a Hepatic Toxin from the Cyanophyte Microcystis Aeruginosa . In: Carmichael, W.W. (eds) The Water Environment. Environmental Science Research. Springer, Boston, MA. https://doi.org/10.1007/978-1-4613-3267-1_26
Liu, W., 2016
Liu, W., Chen, C., Chen, L., Wang, L., Li, J., Chen, Y., Jin, J., Kawan, A., Zhang, X. Sex-dependent effects of microcystin-LR on hypothalamic-pituitary-gonad axis and gametogenesis of adult zebrafish. Sci Rep 6, 22819 (2016). https://doi.org/10.1038/srep22819
Liu, W., 2019
Liu, W., Zhan, C., Zhang, T., Zhang, X. Microcystin-LR influences the in vitro oocyte maturation of zebrafish by activating the MAPK pathway. Aquat Toxicol. 2019 Oct;215:105261. doi: 10.1016/j.aquatox.2019.105261. Epub 2019 Jul 25. PMID: 31419757.
Liu, H., 2018
Liu, H., Zhang, X., Zhang, S., Huang, H., Wu, J., Wang, Y., Yuan, L., Liu, C., Zeng, X., Cheng, X., Zhuang, D., Zhang, H. Oxidative Stress Mediates Microcystin-LR-Induced Endoplasmic Reticulum Stress and Autophagy in KK-1 Cells and C57BL/6 Mice Ovaries. Front Physiol. 2018 Aug 6;9:1058. doi: 10.3389/fphys.2018.01058. PMID: 30131715; PMCID: PMC6090159.
Zegura, B., 2004
Zegura, B., Lah, T.T., Filipic, M., 2004. The role of reactive oxygen species in microcystin-LR-induced DNA damage. Toxicology. 200(1):59-68. doi: 10.1016/j.tox.2004.03.006. PMID: 15158564.
Zegura, B., 2006
Zegura, B., Lah, T.T., Filipic, M., 2006. Alteration of intracellular GSH levels and its role in microcystin-LR-induced DNA damage in human hepatoma HepG2 cells. Mutat Res. 611(1-2):25-33. doi: 10.1016/j.mrgentox.2006.06.038. Epub 2006 Sep 22. PMID: 16996296.
Komatsu, M., 2007
Komatsu, M., Furukawa, T., Ikeda, R., Takumi, S., Nong, Q., Aoyama, K., Akiyama, S.-I., Keppler, D., Takeuchi, T., 2007. Involvement of Mitogen-Activated Protein Kinase Signaling Pathways in Microcystin-LR–Induced Apoptosis after its Selective Uptake Mediated by OATP1B1 and OATP1B3, Toxicological Sciences 97 (2), 407–416, https://doi.org/10.1093/toxsci/kfm054
Nishiwaki-Matsushima, R., 1991
Nishiwaki-Matsushima, R., Nishiwaki, S., Ohta, T., Yoshizawa, S., Suganuma, M., Harada, K.-i., Watanabe, M.F., Fujiki, H., 1991. Structure-Function Relationships of Microcystins, Liver Tumor Promoters, in Interaction with Protein Phosphatase. Japanese Journal of Cancer Research, 82: 993-996. https://doi.org/10.1111/j.1349-7006.1991.tb01933.x
Beattie, K.A., 2003
Beattie, K.A., Ressler, J., Wiegand, C., Krause, E., Codd, G.A., Steinberg, C.E., Pflugmacher, S., 2003. Comparative effects and metabolism of two microcystins and nodularin in the brine shrimp Artemia salina. Aquat Toxicol. 62(3):219-26. doi: 10.1016/s0166-445x(02)00091-7. PMID: 12560170.
Ding, W.X., 2001
Ding, W.X., Shen, H.M., Ong, C.N., 2001. Pivotal role of mitochondrial Ca(2+) in microcystin-induced mitochondrial permeability transition in rat hepatocytes. Biochem Biophys Res Commun. 285(5):1155-61. doi: 10.1006/bbrc.2001.5309. PMID: 11478775.
Ding, W.X., 2001
Ding, W.X., Shen, H.M., Ong, C.N., 2001. Critical role of reactive oxygen species formation in microcystin-induced cytoskeleton disruption in primary cultured hepatocytes. J Toxicol Environ Health A. 64(6):507-19. doi: 10.1080/152873901753215966. PMID: 11732701.
Campos, A., 2010
Campos, A., Vasconcelos, V., 2010. Molecular Mechanisms of Microcystin Toxicity in Animal Cells. Int. J. Mol. Sci. 11, 268-287. https://doi.org/10.3390/ijms11010268
Mikhailov, A., 2003
Mikhailov, A., Härmälä-Braskén, A.S., Hellman, J., Meriluoto, J., Eriksson, J.E., 2003. Identification of ATP-synthase as a novel intracellular target for microcystin-LR. Chem Biol Interact. 142(3):223-37. doi: 10.1016/s0009-2797(02)00075-3. PMID: 12453662.
Chen, T., 2006
Chen, T., Cui, J., Liang, Y., Xin, X., Owen Young, D., Chen, C., Shen, P., 2006. Identification of human liver mitochondrial aldehyde dehydrogenase as a potential target for microcystin-LR. Toxicology. 2006 Mar 1;220(1):71-80. doi: 10.1016/j.tox.2005.12.001. Epub 2006 Jan 18. PMID: 16413648.
MacKintosh, R.W., 1995
MacKintosh, R.W., Dalby, K.N., Campbell, D.G., Cohen, P.T., Cohen, P., MacKintosh, C., 1995. The cyanobacterial toxin microcystin binds covalently to cysteine-273 on protein phosphatase 1. FEBS Lett. 1995 Sep 11;371(3):236-40. doi: 10.1016/0014-5793(95)00888-g. PMID: 7556599.
Kondo, F., 1996
Kondo, F., Matsumoto, H., Yamada, S., Ishikawa, N., Ito, E., Nagata, S., Ueno, Y., Suzuki, M., Harada, K., 1996. Detection and identification of metabolites of microcystins formed in vivo in mouse and rat livers. Chem Res Toxicol. 1996 Dec;9(8):1355-9. doi: 10.1021/tx960085a. PMID: 8951240.
Pflugmacher, S., 1998
Pflugmacher, S., Wiegand, C., Oberemm, A., Beattie, K.A., Krause, E., Codd, G.A., Steinberg, C.E., 1998. Identification of an enzymatically formed glutathione conjugate of the cyanobacterial hepatotoxin microcystin-LR: the first step of detoxication. Biochim Biophys Acta. 1998 Nov 27;1425(3):527-33. doi: 10.1016/s0304-4165(98)00107-x. PMID: 9838216.
Pflugmacher, S., 2005
Pflugmacher, S., Wiegand, C., Werner, S., Schröder, H., Kankaanpää, H., 2005. Activity and substrate specificity of cytosolic and microsomal glutathione S-transferase in Australian black tiger prawns (Penaeus monodon) after exposure to cyanobacterial toxins. Environ Toxicol. 2005 Jun;20(3):301-7. doi: 10.1002/tox.20121. PMID: 15892065.
Wiegand, C., 2000
Wiegand, C., Pflugmacher, S., Oberemm, A., Meems, N., Beattie, K.A., Steinberg, C.E.W., Codd, G.A. (1999), Uptake and effects of microcystin-LR on detoxication enzymes of early life stages of the zebra fish (Danio rerio). Environ. Toxicol., 14: 89-95. https://doi.org/10.1002/(SICI)1522-7278(199902)14:1<89::AID-TOX12>3.0.CO;2-7
Zhang, Y., 2019
Zhang, Y., Zhuang, H., Yang, H., Xue, W., Wang, L., Wei, W. Microcystin-LR disturbs testicular development of giant freshwater prawn Macrobrachium rosenbergii. Chemosphere. 2019 May;222:584-592. doi: 10.1016/j.chemosphere.2019.01.146. Epub 2019 Jan 29. PMID: 30731378.
Zhou, Y., 2019
Zhou, Y., Sun, M., Tang, Y., Chen, Y., Zhu, C., Yang, Y., Wang, C., Yu, G., Tang, Z. Responses of the proteome in testis of mice exposed chronically to environmentally relevant concentrations of Microcystin-LR. Ecotoxicol Environ Saf. 2020 Jan 15;187:109824. doi: 10.1016/j.ecoenv.2019.109824. Epub 2019 Oct 22. PMID: 31654863.
Wang, X., 2016
Wang, X., Ding, J., Xiang, Z., Jiang, P., Du, J., Han, X. Microcystin-LR causes sexual hormone disturbance in male rat by targeting gonadotropin-releasing hormone neurons. Toxicon. 2016 Dec 1;123:45-55. doi: 10.1016/j.toxicon.2016.10.011. Epub 2016 Oct 20. PMID: 27773737.
Wu, J., 2019
Wu, J., Liu, H., Huang, H., Yuan, L., Liu, C., Wang, Y., Cheng, X., Zhuang, D., Xu, M., Chen, X., Losiewicz, M.D., Zhang, H. p53-Dependent pathway and the opening of mPTP mediate the apoptosis of co-cultured Sertoli-germ cells induced by microcystin-LR. Environ Toxicol. 2019 Oct;34(10):1074-1084. doi: 10.1002/tox.22808. Epub 2019 Jun 3. PMID: 31157505.
Chen, J., 2017
Chen, J., Meng, S., Xu, H., Zhang, Z., Wu, X. Effects of Microcystis on Hypothalamic-Pituitary-Gonadal-Liver Axis in Nile Tilapia (Oreochromis niloticus). Bull Environ Contam Toxicol. 2017 Apr;98(4):562-566. doi: 10.1007/s00128-017-2051-4. Epub 2017 Feb 24. PMID: 28236009.
Chen, Y., 2011
Chen, Y., Xu, J., Li, Y., Han, X. Decline of sperm quality and testicular function in male mice during chronic low-dose exposure to microcystin-LR. Reprod Toxicol. 2011 May;31(4):551-7. doi: 10.1016/j.reprotox.2011.02.006. Epub 2011 Feb 18. PMID: 21338672.
Huang, H., 2016
Huang, H., Liu, C., Fu, X., Zhang, S., Xin, Y., Li, Y., Xue, L., Cheng, X., Zhang, H. Microcystin-LR Induced Apoptosis in Rat Sertoli Cells via the Mitochondrial Caspase-Dependent Pathway: Role of Reactive Oxygen Species. Front Physiol. 2016 Sep 9;7:397. doi: 10.3389/fphys.2016.00397. PMID: 27667976; PMCID: PMC5016609.
Chen, Y., 2017
Chen, Y., Wang, J., Zhang, Q., Xiang, Z., Li, D., Han, X. Microcystin-leucine arginine exhibits immunomodulatory roles in testicular cells resulting in orchitis. Environ Pollut. 2017 Oct;229:964-975. doi: 10.1016/j.envpol.2017.07.081. Epub 2017 Jul 29. PMID: 28765008.
Chen, Y., 2017
Chen, Y., Wang, J., Pan., C., Li, D., Han, X. Microcystin-leucine-arginine causes blood-testis barrier disruption and degradation of occludin mediated by matrix metalloproteinase-8. Cell Mol Life Sci. 2018 Mar;75(6):1117-1132. doi: 10.1007/s00018-017-2687-6. Epub 2017 Oct 25. PMID: 29071384.
Masango, M.G., 2010
Masango, M.G., Myburgh, J.G., Labuschagne, L., Govender, D., Bengis, R.G., Naicker, D., 2010. ASSESSMENT OF MICROCYSTIS BLOOM TOXICITY ASSOCIATED WITH WILDLIFE MORTALITY IN THE KRUGER NATIONAL PARK, SOUTH AFRICA. J Wildl Dis 1 January 2010; 46 (1): 95–102. doi: https://doi.org/10.7589/0090-3558-46.1.95
Miller, M.A., 2010
Miller, M.A., Kudela, R.M., Mekebri, A., Crane, D., Oates, S.C., Tinker, M.T., Staedler, M., Miller, W.A., Toy-Choutka, S., Dominik, C., Hardin, D., Langlois, G., Murray, M., Ward, K., Jessup, D.A., 2010. Evidence for a novel marine harmful algal bloom: cyanotoxin (microcystin) transfer from land to sea otters. PLoS One. 2010 Sep 10;5(9):e12576. doi: 10.1371/journal.pone.0012576. PMID: 20844747; PMCID: PMC2936937.
Yuan, L., 2020
Yuan, L., Liu, H., Liu, X., Zhang, X., Wu, J., Wang, Y., Du, X., Wang, R., Ma, Y., Chen, X., Petlulu, P., Cheng, X., Zhuang, D., Guo, H., Zhang, H., 2020. Epigenetic modification of H3K4 and oxidative stress are involved in MC-LR-induced apoptosis in testicular cells of SD rats. Environ Toxicol. 2020 Feb;35(2):277-291. doi: 10.1002/tox.22865. Epub 2019 Nov 5. PMID: 31691492.
Wang, Y., 2020
Wang, Y., Liu, H., Liu, X., Zhang, X., Wu, J., Yuan, L., Du, X., Wang, R., Ma, Y., Chen, X., Cheng, X., Zhuang, D., Zhang, H., 2020. Histone acetylation plays an important role in MC-LR-induced apoptosis and cycle disorder in SD rat testicular cells. Chemosphere. 2020 Feb;241:125073. doi: 10.1016/j.chemosphere.2019.125073. Epub 2019 Oct 12. PMID: 31683423.
Pan, C., 2018
Pan, C., Chen, Y., Xu, T., Wang, J., Li, D., Han, X. Chronic exposure to microcystin-leucine-arginine promoted proliferation of prostate epithelial cells resulting in benign prostatic hyperplasia. Environ Pollut. 2018 Nov;242(Pt B):1535-1545. doi: 10.1016/j.envpol.2018.08.024. Epub 2018 Aug 17. PMID: 30145517.
Su, Y., 2016
Su, Y., Li, L., Hou, J., Wu, N., Lin, W., Li, G. Life-cycle exposure to microcystin-LR interferes with the reproductive endocrine system of male zebrafish. Aquat Toxicol. 2016 Jun;175:205-12. doi: 10.1016/j.aquatox.2016.03.018. Epub 2016 Mar 22. PMID: 27060240.
Meng, X., 2019
Meng, X., Peng, H., Ding, Y., Zhang, L., Yang, J., Han, X. A transcriptomic regulatory network among miRNAs, piRNAs, circRNAs, lncRNAs and mRNAs regulates microcystin-leucine arginine (MC-LR)-induced male reproductive toxicity. Sci Total Environ. 2019 Jun 1;667:563-577. doi: 10.1016/j.scitotenv.2019.02.393. Epub 2019 Feb 28. PMID: 30833255.
Li, Y., 2015
Li, Y., Zhang, M., Chen, P., Liu, R., Liang, G., Yin, L., Pu, Y. Effects of Microcystin-LR Exposure on Spermiogenesis in Nematode Caenorhabditis elegans. Int. J. Mol. Sci. 2015, 16, 22927-22937. https://doi.org/10.3390/ijms160922927
Adegoke, E.O., 2018
Adegoke, E.O., Wang, C., Machebe, N.S., Wang, X., Wang, H., Adeniran, S.O., Zhang, H., Zheng, P., Zhang, G. Microcystin-leucine arginine (MC-LR) induced inflammatory response in bovine sertoli cell via TLR4/NF-kB signaling pathway. Environ Toxicol Pharmacol. 2018 Oct;63:115-126. doi: 10.1016/j.etap.2018.09.001. Epub 2018 Sep 6. PMID: 30212741.
Hou, J., 2017
Hou, J., Su, Y., Lin, W., Guo, H., Xie, P., Chen, J., Gu, Z., Li, L. Microcystin-LR retards gonadal maturation through disrupting the growth hormone/insulin-like growth factors system in zebrafish. Ecotoxicol Environ Saf. 2017 May;139:27-35. doi: 10.1016/j.ecoenv.2017.01.025. PMID: 28109900.
Li, X., 2017
Li, X., Zhuang, X., Xu, T., Mao, M., Wang, C., Chen, Y., Han, X., Wu, J. Expression analysis of microRNAs and mRNAs in ovarian granulosa cells after microcystin-LR exposure. Toxicon. 2017 Apr;129:11-19. doi: 10.1016/j.toxicon.2017.01.022. Epub 2017 Feb 2. PMID: 28161121.
Wu, J., 2015
Wu, J., Yuan, M., Song, Y., Sun, F., Han, X. MC-LR Exposure Leads to Subfertility of Female Mice and Induces Oxidative Stress in Granulosa Cells. Toxins (Basel). 2015 Dec 2;7(12):5212-23. doi: 10.3390/toxins7124872. PMID: 26633508; PMCID: PMC4690122.
Zhang, S., 2016
Zhang, S., Liu, C., Li, Y., Imam, M.U., Huang, H., Liu, H., Xin, Y., Zhang, H. Novel Role of ER Stress and Autophagy in Microcystin-LR Induced Apoptosis in Chinese Hamster Ovary Cells. Front Physiol. 2016 Nov 8;7:527. doi: 10.3389/fphys.2016.00527. PMID: 27877136; PMCID: PMC5099254.
Zhan, C., 2019
Zhan, C., Zhang, F., Liu, W., Zhang, X. Microcystin-LR promotes zebrafish (Danio rerio) oocyte (in vivo) maturation by activating ERK1/2-MPF signaling pathways, and cAMP is involved in this process. Environ Pollut. 2020 Apr;259:113843. doi: 10.1016/j.envpol.2019.113843. Epub 2019 Dec 19. PMID: 31887595.
Dao, T.S., 2018
Dao, T.S., Vo, T.M., Wiegand, C., Bui, B.T., Dinh, K.V. Transgenerational effects of cyanobacterial toxins on a tropical micro-crustacean Daphnia lumholtzi across three generations. Environ Pollut. 2018 Dec;243(Pt B):791-799. doi: 10.1016/j.envpol.2018.09.055. Epub 2018 Sep 15. PMID: 30241003.
Liu, W., 2014
Liu, W., Qiao, Q., Chen, Y., Wu, K., Zhang, X. Microcystin-LR exposure to adult zebrafish (Danio rerio) leads to growth inhibition and immune dysfunction in F1 offspring, a parental transmission effect of toxicity. Aquat Toxicol. 2014 Oct;155:360-7. doi: 10.1016/j.aquatox.2014.07.011. Epub 2014 Jul 16. PMID: 25105566.
Liu, Y., 2002
Liu, Y., Song, L., Li, X., Liu, T. The toxic effects of microcystin-LR on embryo-larval and juvenile development of loach, Misguruns mizolepis Gunthe. Toxicon. 2002 Apr;40(4):395-9. doi: 10.1016/s0041-0101(01)00173-8. PMID: 11738232.
Qi, M., 2016
Qi, M., Dang, Y., Xu, Q., Yu, L., Liu, C., Yuan, Y., Wang, J. Microcystin-LR induced developmental toxicity and apoptosis in zebrafish (Danio rerio) larvae by activation of ER stress response. Chemosphere. 2016 Aug;157:166-73. doi: 10.1016/j.chemosphere.2016.05.038. Epub 2016 May 21. PMID: 27219292.
Wu, Q., 2017
Wu, Q., Yan, W., Cheng, H., Liu, C., Hung, T.C., Guo, X., Li, G. Parental transfer of microcystin-LR induced transgenerational effects of developmental neurotoxicity in zebrafish offspring. Environ Pollut. 2017 Dec;231(Pt 1):471-478. doi: 10.1016/j.envpol.2017.08.038. Epub 2017 Aug 29. PMID: 28837927.
Zeng, C., 2014
Zeng, C., Sun, H., Xie, P., Wang, J., Zhang, G., Chen, N., Yan, W., Li, G. The role of apoptosis in MCLR-induced developmental toxicity in zebrafish embryos. Aquat Toxicol. 2014 Apr;149:25-32. doi: 10.1016/j.aquatox.2014.01.021. Epub 2014 Feb 2. PMID: 24555956.
Zhao, S., 2020
Zhao, S., Zhong, S., Wang, F., Wang, H., Xu, D., Li, G. Microcystin-LR exposure decreased the fetal weight of mice by disturbance of placental development and ROS-mediated endoplasmic reticulum stress in the placenta. Environ Pollut. 2020 Jan;256:113362. doi: 10.1016/j.envpol.2019.113362. Epub 2019 Oct 11. PMID: 31672369.
Gérard, C., 2005
Gérard, C., Brient, L., Le Rouzic, B. (2005), Variation in the response of juvenile and adult gastropods (Lymnaea stagnalis) to cyanobacterial toxin (microcystin-LR). Environ. Toxicol., 20: 592-596. https://doi.org/10.1002/tox.20147
Chernoff, N., 2020
Chernoff, N., Hill, D., Lang, J., Schmid, J., Le, T., Farthing, A., Huang, H. The Comparative Toxicity of 10 Microcystin Congeners Administered Orally to Mice: Clinical Effects and Organ Toxicity. Toxins. 2020; 12(6):403. https://doi.org/10.3390/toxins12060403

TEF References

Wolf, H.U., 2002
Wolf, H.U., Frank, C. Toxicity assessment of cyanobacterial toxin mixtures. Environ Toxicol. 2002;17(4):395-9. doi: 10.1002/tox.10066. PMID: 12203962.
Haney, J.F., 2011
Haney, J.F., 2011. Peer Review of Cyanotoxin Toxicity Criteria and Health Based Water Concentrations to Protect Human Swimmers, Dogs and Cattle.
Notes
Wade Huang
Contact
Zhihong Wang
Contact
John Ramsdell
Contact
Christopher Miles
Contact