Artículo

Estamos trabajando para incorporar este artículo al repositorio
Consulte el artículo en la página del editor
Consulte la política de Acceso Abierto del editor

Abstract:

Accumulation and toxicity of cyanobacterial toxins, particularly microcystin-LR (MCLR) have been extensively studied in fish and aquatic invertebrates. However, MCLR excretion mechanisms, which could reduce this toxin's effects, have received little attention. The Patagonian silverside, Odontesthes hatcheri, is an omnivorous-planktivorous edible fish, which has been shown to digest cyanobacterial cells absorbing MCLR and eliminating the toxin within 48 h without suffering significant toxic effects. We studied the effects of MCLR on glycoconjugate composition and the possible role of multidrug resistance associated proteins (Abcc) in MCLR export from the cells in O. hatcheri intestine. We treated O. hatcheri with 5 μg MCLR g−1 body mass administered with the food. Twenty four hours later, the intestines of treated and control fish were processed for lectin-histochemistry using concanavalin A (ConA), Triticum vulgaris agglutinin (WGA), and Dolichos biflorus agglutinin (DBA). MCLR affected the distribution of glycoconjugates by augmenting the proportion of ConA-positive at the expense of WGA-positive cells. We studied MCLR effects on the transport of the Abcc-like substrates 2,4-dinitrophenyl-S-glutathione (DNP-SG) and calcein in ex vivo intestine preparations (everted and no-everted sacs and strips). In treated preparations, CDNB together with MCLR (113 μg MCLR g−1 intestine, equivalent to 1.14 μmol L−1 when applied in the bath) or the Abcc inhibitor, MK571 was applied for one hour, during which DNP-SG was measured in the bath every 10 min in order to calculate mass-specific DNP-SG transport rate. MCLR significantly inhibited DNP-SG transport (p < 0.05), especially in middle intestine (47 and 24%, for luminal and serosal transport, respectively). In middle intestine strips, MCLR and MK571inhibited DNP-SG transport in a concentration dependent fashion (IC50 3.3 and 0.6 μmol L−1, respectively). In middle intestine strips incubated with calcein-AM (0.25 μmol L−1), calcein efflux was inhibited by MCLR (2.3 μmol L−1) and MK571 (3 μmol L−1) by 38 and 27%, respectively (p < 0.05). Finally, middle intestine segments were incubated with different concentrations of MCLR applied alone or together with 3 μM MK571. After one hour, protein phosphatase 1 (PP1) activity, the main target of MCLR, was measured. 2.5 μM MCLR did not produce any significant effect, while the same amount plus MK571 inhibited PP1 activity (p < 0.05). This effect was similar to that of 5 μM MCLR. Our results suggest that in O. hatcheri enterocytes MCLR is conjugated with GSH via GST and then exported to the intestinal lumen through Abcc-like transporters. This mechanism would protect the cell from MCLR toxicity, limiting toxin transport into the blood, which is probably mediated by basolateral Abccs. From an ecotoxicological point of view, elimination of MCLR through this mechanism would reduce the amount of toxin available for trophic transference. © 2016

Registro:

Documento: Artículo
Título:Alterations in the intestine of Patagonian silverside (Odontesthes hatcheri) exposed to microcystin-LR: Changes in the glycosylation pattern of the intestinal wall and inhibition of multidrug resistance proteins efflux activity
Autor:Bieczynski, F.; Torres, W.D.C.; Painefilu, J.C.; Castro, J.M.; Bianchi, V.A.; Frontera, J.L.; Paz, D.A.; González, C.; Martín, A.; Villanueva, S.S.M.; Luquet, C.M.
Filiación:Laboratorio de Ecotoxicología Acuática, INIBIOMA – (CONICET-UNCo), CEAN- Ruta 61 km 3, Paraje San Cabao, Junín de los Andes, Neuquén, 8371, Argentina
CEAN- Ruta 61 km 3, Paraje San Cabao, Junín de los Andes, Neuquén, 8371, Argentina
Instituto de Fisiología Experimental, IFISE-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 570, Rosario, Santa Fe 2000, Argentina
Laboratorio de Biología del Desarrollo, IFIBYNE-CONICET, Universidad Nacional de Buenos Aires, Pabellón II, Ciudad Universitaria, Ciudad Autónoma de Buenos Aires, 1428, Argentina
Dirección Técnica y de Desarrollo Tecnológico, Agua y Saneamientos Argentinos, Av. Figueroa Alcorta 6081, Ciudad Autónoma de Buenos Aires, 1425, Argentina
Palabras clave:Abcc transporters; Cyanotoxin; Glycoconjugates; Intestinal sacs; Lectin-histochemistry; Multixenobiotic resistance; Odontesthes hatcheri; 2,4 dinitrophenyl s glutathione; agglutinin; calcein; concanavalin A; glutathione derivative; glycoconjugate; lectin; microcystin LR; multidrug resistance protein; phosphoprotein phosphatase 1; unclassified drug; verlukast; dolichos biflorus agglutinin; fluorescein derivative; glutathione; microcystin; microcystin LR; multidrug resistance protein; plant lectin; propionic acid derivative; quinoline derivative; water pollutant; cyanobacterium; drug resistance; ecotoxicology; fish; inhibition; phytotoxicity; protein; toxin; trophic structure; animal experiment; animal tissue; Article; Atherinopsidae; body mass; chemical composition; concentration response; controlled study; Dolichos; Dolichos biflorus; enzyme activity; female; glycosylation; histochemistry; intestine cell; intestine wall; male; nonhuman; Odontesthes hatcheri; priority journal; protein transport; Triticum vulgaris; wheat; animal; drug effects; fluorescence microscopy; glycosylation; intestine mucosa; metabolism; Smegmamorpha; toxicity; transport at the cellular level; water pollutant; Argentina elongata; Cyanobacteria; Dolichos biflorus; Invertebrata; Odontesthes hatcheri; Triticum aestivum; Animals; Biological Transport; Concanavalin A; Fluoresceins; Glutathione; Glycosylation; Intestinal Mucosa; Microcystins; Microscopy, Fluorescence; Multidrug Resistance-Associated Proteins; Plant Lectins; Propionates; Quinolines; Smegmamorpha; Water Pollutants, Chemical
Año:2016
Volumen:178
Página de inicio:106
Página de fin:117
DOI: http://dx.doi.org/10.1016/j.aquatox.2016.07.016
Título revista:Aquatic Toxicology
Título revista abreviado:Aquat. Toxicol.
ISSN:0166445X
CODEN:AQTOD
CAS:calcein, 1461-15-0; concanavalin A, 11028-71-0; microcystin LR, 101043-37-2; multidrug resistance protein, 149200-37-3, 208997-77-7; verlukast, 115104-28-4; glutathione, 70-18-8; Concanavalin A; cyanoginosin LR; dolichos biflorus agglutinin; Fluoresceins; fluorexon; Glutathione; Microcystins; Multidrug Resistance-Associated Proteins; Plant Lectins; Propionates; Quinolines; verlukast; Water Pollutants, Chemical
Registro:https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_0166445X_v178_n_p106_Bieczynski

Referencias:

  • Amado, L.L., Monserrat, J.M., Oxidative stress generation by microcystins in aquatic animals: why and how (2010) Environ. Int., 36, pp. 226-235
  • Amé, M.V., Baroni, M.V., Galanti, L.N., Bocco, J.L., Wunderlin, D.A., Effects of microcystin-LR on the expression of P-glycoprotein in Jenynsia multidentata (2009) Chemosphere, 74, pp. 1179-1186
  • Atencio, L., Moreno, I., Jos, A., Pichardo, S., Moyano, R., Blanco, A., Cameán, A.M., Dose-dependent antioxidant responses and pathological changes in tenta (Tinca tinca) after acute oral exposure to Microcystis under laboratory conditions (2008) Toxicon, 52 (1), pp. 1-12
  • Azevedo, S.M.F.O., Evans, W.R., Carmichael, W.W., Namikoshi, M., First report of microcystins from a Brazilian isolate of the cyanobacterium Microcystis aeruginosa (1994) J. Appl. Phycol., 6, pp. 261-264
  • Bard, S.M., Multixenobiotic resistance as a cellular defense mechanism in aquatic organisms (2000) Aquat. Toxicol., 48, pp. 357-389
  • Barltrop, D., Brueton, M.J., Chapter 8: the gastrointestinal tract and short-term toxicity tests (1990) Short-Term Toxicity Tests for Non-genotoxic Effects, , P.J. Bourdeau John Wiley and Sons Ltd 99–110 pp
  • Bieczynski, F., Bianchi, V.A., Luquet, C.M., Accumulation and biochemicaleffects of microcystin-LR on the Patagonian pejerrey (Odontesthes hatcheri) fed with the toxic cyanobacteria Microcystis aeruginosa (2013) Fish. Physiol. Biochem., 39 (5), pp. 1309-1321
  • Bieczynski, F., De Anna, J.S., Pirez, M., Brena, B.M., Villanueva, S.S.M., Luquet, C.M., Cellular transport of microcystin-LR in rainbow trout (Oncorhynchus mykiss) across the intestinal wall: possible involvement of multidrug resistance-associated proteins (2014) Aquat. Toxicol., 154, pp. 97-106
  • Bieczynski, F., Cellular transport mechanisms, acumulation kinetics and toxic effects of microcystin-LR in Patagonian freshwater fish (2015) PhD Thesis, , National University of Comahue Argentina
  • Boaru, D.A., Dragos¸, N., Schirmer, K., Microcystin-LR induced cellular effects inmammalian and fish primary hepatocyte cultures and cell lines: a comparative study (2006) Toxicology, 218, pp. 134-148
  • Bradford, M.M., A rapid and sensitive method for the quantification of microgram quantities of protein utilizing the principle of protein dye binding (1976) Anal. Biochem., 72, pp. 248-254
  • Burkhardt-Holm, P., Lectin histochemistry of rainbow trout (Oncorhynchus mykiss) gill and skin (1997) Histochem. J., 29 (11-12), pp. 893-899
  • Bury, N.R., Newlands, A.D., Eddy, F.B., Codd, G.A., In vivo and in vitro intestinal transport of 3H-microcystin-LR, a cyanobacterial toxin, in rainbow trout (Oncorhynchus mykiss) (1998) Aquat. Toxicol., 42, pp. 139-148
  • Campos, A., Vasconcelos, V., Molecular mechanisms of microcystin ioxicity in animal cells (2010) Int. J. Mol. Sci., 11, pp. 268-287
  • Carmichael, W.W., An, J., Using an enzyme linked immunosorbent assay (ELISA) and a protein phosphatase inhibition assay (PPIA) for the detection of microcystins and nodularins (1999) Nat. Toxins, 7, pp. 377-385
  • Carmichael, W.W., Cyanobacteria secondary metabolites- the cyanotoxins (1992) J. Appl. Bacteriol., 72, pp. 445-459
  • Chan, L.M.S., Lowes, S., Hirst, B.H., The ABCs of drug transport in intestine and liver: efflux proteins limiting drug absorption and bioavailability (2004) Eur. J. Pharm. Sci., 21, pp. 25-51
  • Codd, G.A., Morrison, L.F., Metcalf, J.S., Cyanobacterial toxins: risk management for health protection (2005) Toxicol. Appl. Pharmacol., 203, pp. 264-272
  • Contardo-Jara, V., Pflugmacher, S., Wiegand, C., Multi-xenobiotic-resistance a possible explanation for the insensitivity of bivalves towards cyanobacterial toxins (2008) Toxicon, 52, pp. 936-943
  • Díaz, A.O., García, A.M., Devincenti, C.V., Goldemberg, A.L., Morphological and histochemical characterization of the mucosa of the digestive tract in Engraulis anchoita (Hubbs and Marini, 1935) (2003) Anat. Histol. Embryol., 32, pp. 341-346
  • Díaz, A.O., García, A.M., Goldemberg, A.L., Glycoconjugates in the branchial mucous cells of Cynoscion guatucupa (cuvier, 1830) (Pisces: Sciaenidae) (2005) Scientia Marina, 69, pp. 545-553
  • Díaz, A.O., García, A.M., Goldemberg, A.L., Glycoconjugates in the mucosa of the digestive tract of Cynoscion guatucupa: a histochemical study (2008) Acta Histochem., 110, pp. 76-85
  • Díaz, A.O., García, A.M., Escalante, A.H., Goldemberg, A.L., Glycoproteins histochemistry of the gills of Odontesthes bonariensis (Teleostei, Atherinopsidae) (2010) J. Fish Biol., 77, pp. 1665-1673
  • Díaz, A.O., Tano de la Hoz, M.F., García, A.M., Escalante, A.H., Goldemberg, A.L., Characterization of glycoconjugates in the pharyngeal cavity and the oesophagus of Odontesthes bonariensis by lectins (2012) Biotemas, 25 (4), pp. 157-163
  • Dörr, F.A., Pinto, E., Soares, R.M., Azevedo, S.M.F.O., Microcystins in SouthAmerican aquatic ecosystems: occurrence, toxicity and toxicological assays (2010) Toxicon, 56, pp. 1247-1256
  • Dietrich, D., Hoeger, S., Guidance values for microcystins in water and cyanobacterial supplement products (blue-green algal supplements): a reasonable or misguided approach? (2005) Toxicolo. Appl. Pharmacol., 203, pp. 273-289
  • Domeneghini, C., Pannelli Straini, R., Veggetti, A., Gut glycoconjugates in Sparus aurata L. (Pisces: Teleostei). A comparative histochemical study in larval and adult ages (1998) Histol. Histopathol., 13, pp. 359-372
  • Echenique, R., Aguilera, A., Chapter III: Floraciones de Cyanobacteria toxígenas en la República Argentina: antecedentes (2009) Cianobacterias y Cianotoxinas: Identificación, Toxicología, Monitoreo y Evaluación de riesgo., p. 238. , In: Giannuzzi, L., (Ed.), Moglia editors, Corrientes, 1st ed.. ISBN 978-987-05-5749-4
  • Eriksson, J.E., Toivola, D., Meriluoto, J.A., Karaki, H., Han, Y.G., Hartshorne, D., Hepatocyte deformation induced by cyanobacterial toxins reflects inhibition of protein phosphatases (1990) Biochem. Biophys. Res. Commun., 173, pp. 1347-1353
  • Evers, R., Kool, M., Smith, A.J., van Deemter, L., de Haas, M., Borst, P., Inhibitory effect of the reversal agents V-104, GF120918 and Pluronic L61 on MDR1 Pgp-MRP1- and MRP2-mediated transport (2000) Cancer Res. Campaign, 83 (3), pp. 366-374
  • Falconer, I.R., Yeung, D.S., Cytoskeletal changes in hepatocytes induced by Microcystis toxins and their relation to hyperphosphorylation of cell proteins (1992) Chem. Biol. Interact., 81, pp. 181-196
  • Fardel, O., Jigorel, E., Le Vee, M., Payen, L., Physiological, pharmacological and clinical features of the multidrug resistance protein 2 (2005) Biomed. Pharmacother., 59, pp. 104-114
  • Ferrão-Filho, A.S., Kozlowsky-Suzuki, B., Cyanotoxins: bioaccumulation and effects on aquatic animals (2011) Mar. Drugs, 9, pp. 2729-2772
  • Ferreira, M., Costa, J., Reis-Henriques, M.A., ABC transporters in fish species: a review (2014) Frontiers Physiol., 5 (266), pp. 1-12
  • Fischer, W.J., Dietrich, D.R., Pathological and biochemical characterization of microcystin-induced hepatopancreas and kidney damage in carp (Cyprinus carpio) (2000) Toxicol. Appl. Pharmacol., 164, pp. 73-81
  • Fischer, W.J., Altheimer, S., Cattori, V., Meier, P.J., Dietrich, D.R., Hagenbuch, B., Organic anion transporting polypeptides expressed in liver and brain mediate uptake of microcystin (2005) Toxicol. Appl. Pharmacol., 203, pp. 257-263
  • Fischer, S., Lončar, J., Zaja, R., Schnell, S., Schirmer, K., Smital, T., Luckenbach, T., Constitutive mRNA expression and protein activity levels of nine ABC efflux transporters in seven permanent cell lines derived from different tissues of rainbow trout (Oncorhynchus mykiss) (2011) Aquat. Toxicol., 101, pp. 438-446
  • Gekeler, V., Ise, W., Sanders, K.H., Ulrich, W.R., Beck, J., The leukotriene LTD4 receptor antagonist MK571 specifically modulates mrp associated multidrug resistance (1995) Biochem. Biophys. Res. Commun., 208 (1), pp. 345-352
  • Gotoh, Y., Suzuki, H., Kinoshita, S., Hirohashi, T., Kato, Y., Sugiyama, Y., Involvement of an organic anion transporter (canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2) in gastrointestinal secretion of glutathione conjugates in rats (2000) J. Pharmacol. Exp. Ther., 292, pp. 433-439
  • Gottesman, M.M., Pastan, I., Biochemistry of multidrug resistance mediated by the multidrug transporter (1993) Annu. Rev. Biochem, 62, pp. 385-427
  • Honkanen, R.E., Zwiller, J., Moore, R.E., Daily, S.L., Khatra, B.S., Dukelow, M., Boynton, A.L., Characterization of microcystin-LR, a potent inhibitor of type 1 and type 2A protein phosphatases (1990) J. Biol. Chem., 265, pp. 19401-19404
  • Hooser, S.B., Beasley, V.R., Lovell, R.A., Carmichael, W.W., Haschek, W.M., Toxicity of microcystin-LR, a cyclic heptapeptide hepatotoxin from Microcystis aeruginosa, to rats and mice (1989) Vet. Pathol., 26, pp. 246-252
  • Horn, M.H., Gawlicka, A.K., German, D.P., Logothetis, E.A., Cavanagh, J.W., Boyle, K.S., Structure and function of the stomachless digestive system in three related species of New World silverside fishes (Atherinopsidae) representing herbivory, omnivory, and carnivory (2006) Mar. Biol., 149, pp. 1237-1245
  • Hualde, J.P., Torres, W.D.C., Moreno, P., Ferrada, M., Demicheli, M.A., Molinari, L.J., Luquet, C.M., Growth and feeding of patagonian pejerrey Odontesthes hatcheri reared in net cages (2011) Aquacult. Res., 42, pp. 754-763
  • Ito, E., Kondo, F., Harada, K., First report on the distribution of orally administered microcystin-LR in mouse tissue using an immunostaining method (2000) Toxicon, 38, pp. 37-48
  • Ito, E., Takai, A., Kondo, F., Masui, H., Imanishi, S., Harada, K., Comparison ofprotein phosphatase inhibitory activity and apparent toxicity of microcystinsand related compounds (2002) Toxicon, 40 (7), pp. 1017-1025
  • Jos, A., Pichardo, S., Prieto, A.I., Repetto, G., Vázquez, C.M., Moreno, I., Cameán, A.M., Toxic cyanobacterial cells containing microcystins induce oxidative stress in exposed tilapia fish (Oreochromis sp.) under laboratory conditions (2005) Aquat. Toxicol., 72, pp. 261-271
  • Kim, J.J., Khan, W.I., Goblet cells and mucins: role in innate defense in enteric infections (2013) Pathogens, 2 (1), pp. 55-70
  • Klaassen, C.D., Aleksunes, L.M., Xenobiotic, bile acid, and cholesterol transporters: function and regulation (2010) Pharmacol. Rev., 62, pp. 1-96
  • Kondo, F., Matsumoto, H., Yamada, S., Ishikawa, N., Ito, E., Nagata, S., Ueno, Y., Harada, K., Detection and identification of metabolites of microcystinsformed in vivo in mouse and rat livers (1996) Chem. Res. Toxicol., 9, pp. 1355-1359
  • Kuiper-Goodman, T., Falconer, I., Fitzgerald, J., Human health aspects (1999) Toxic Cyanobacteria in Water: a Guide to Their Public Health Consequences, Monitoring and Management, , I. Chorus J. Bartram first ed. WHO New York 400 pp
  • Kurelec, B., The multixenobiotic resistance mechanism in aquatic organisms (1992) Crit. Rev. Toxicol., 22, pp. 23-43
  • Lemaitre, C., Orange, N., Saglio, P., Saint, N., Gagnon, J., Molle, G., Characterization and ion channel activities of novel antibacterial proteins from the skin mucosa of carp (Cyprinus carpio) (1996) Eur. J. Biochem., 240, pp. 143-149
  • Leslie, E.M., Deeley, R.G., Cole, S.P.C., Multidrug resistance proteins: role of P-glycoprotein, MRP1 MRP2, nd BCRP (ABCG2) in tissue defense (2005) Toxicol. Appl. Pharmacol., 204, pp. 216-237
  • Li, D., Jiao, J., Shatos, M.A., Hodges, R.R., Dartt, D.A., Effect of VIP on intracellular [Ca2+], extracellular regulated kinase 1/2, and secretion in cultured rat conjunctival goblet cells (2013) Invest. Ophthalmol. Visual Sci., 54 (4), pp. 2872-2884
  • Linden, S.K., Sutton, P., Karlsson, N.G., Korolik, V., McGuckin, M.A., Mucins in the mucosal barrier to infection (2008) Mucosal Immunol., 1 (3), pp. 183-197
  • Loretz, C.A., Electrophysiology of ion transport in teleost intestinal cells (1995) Cellular and Molecular Approaches to Fish Ionic Regulation, , C.H. Wood T.J. Shuttleworth Academic Press London, UK 1–352 pp
  • Lu, H., Choudhuri, S., Ogura, K., Csanaky, I.L., Lei, X., Cheng, X., Song, P.Z., Klaassen, C.D., Characterization of organic anion transporting polypeptide 1b2-nullmice: essential role in hepatic uptake/toxicity of phalloidin and microcystin-LR (2008) Toxicol. Sci., 103, pp. 35-45
  • Lu, X., Long, Y., Sun, R., Zhou, B., Lin, L., Zhong, S., Cui, Z., Zebrafish Abcb4 is a potential efflux transporter of microcystin-LR (2015) Comp. Biochem. Physiol. C, 167, pp. 35-42
  • Luckenbach, T., Fischer, S., Sturm, A., Current advances on ABC drug transporters in fish (2014) Comp. Biochem. Physiol. C, 165, pp. 28-52
  • 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 (1995) FEBS Lett., 371, pp. 236-240
  • Malbrouck, C., Kestemont, P., Effects of microcystins on fish (2006) Environ. Toxicol. Chem., 25 (1), pp. 72-86
  • Meier-Abt, F., Hammann-Hanni, A., Stieger, B., Ballatori, N., Boyer, J.L., The organic anion transport polypeptide 1d1 (Oatp1d1) mediates hepatocellular uptake §of phalloidin and microcystin into skate liver (2007) Toxicol. Appl. Pharmacol., 218, pp. 274-279
  • Meyer, W., Luz, S., Schnapper, A., Lectin histochemical aspects of mucus function in the oesophagus of the reticulated Python (Python reticulates) (2009) Anat. Histol. Embryol., 38, pp. 316-318
  • Mottino, A.D., Hoffman, T., Jennes, L., Cao, J., Vore, M., 2001. Expression of multidrug resistance-associated protein 2 in small intestine from pregnant and postpartum rats. American Journal of Physiology (2001) Gastrointest. Liver Physiol., 280, pp. G1261-G
  • Othaz Brida, M.A., Agúndez, J.P., Gil, M.I., Labollita, H.A., Control de floraciones algales en los embalses de la Cuenca del Río Negro (2010) VI Congreso Argentino De Presas Y Aprovechamientos Hidroeléctricos, Neuquén, 3–6 noviembre, p. 2010
  • Pavagadhi, S., Balasubramanian, R., Toxicological evaluation of microcystins in aquatic fish species: current knowledge and future directions (2013) Aquat. Toxicol., 142-143, pp. 1-16
  • Pavlova, V., Georgieva, L., Paunova, T., Stoitsova, S., Nikolova, E., Carbohydrate localization in intestinal glycocalyx (2013) Sci. Technol., 3 (1), pp. 17-21
  • Pflugmacher, S., Wiegand, C., Oberemm, A., Beattie, K.A., Krause, E., Codd, G.A., Identification of an enzymatically formed glutathione conjugate of the cyanobacterial hepatoxin microcystin-LR: the first step of detoxication (1998) Biochim. Biophys. Acta, 1425, pp. 527-533
  • Puig, A., Estructura espacial y temporal de la taxocenosis de entomostracos (Crustacea) limnéticos en el embalse Ramos Mexía (provincia del Neuquén y de Río Negro, Argentina (1992) PhD Thesis, , University of Buenos Aires
  • Runnegar, M., Berndt, N., Kong, S.-M., Lee, E.Y.C., Zhang, L., In vivo and in vitro binding of microcystin to protein phosphatases 1 and 2A (1995) Biochem. Biophys. Res. Commun., 216 (1), pp. 162-169
  • Sahin, A., Tencalla, F.G., Dietrich, D.R., Naegeli, H., Biliary excretion of biochemically active cyanobacteria (blue-green algae) hepatotoxins in fish (1996) Toxicology, 106, pp. 123-130
  • Sivonen, K., Jones, G., Chapter 3: cyanobacterial toxins (1999) Toxic Cyanobacteria in Water: A Guide to Their Public Health Consequences, Monitoring and Management., , I. Chorus J. Bartram first ed. WHO New York, NY 12–23pp
  • Soares, R.M., Magalhães, V.F., Azevedo, S.M., Accumulation and depuration of microcystins (cyanobacteria hepatotoxins) in Tilapia rendalli (Cichlidae) under laboratory conditions (2004) Aquat. Toxicol., 70, pp. 1-10
  • Suprasert, A.T., Fujioka, T., Yamada, K., The histochemistry of glycoproteins in the colonic epithelium of the chicken (1987) Histochemistry, 86, pp. 491-497
  • Takano, M., Yumoto, R., Murakami, T., Expression and function of efflux drug transporters in the intestine (2006) Pharmacol. Ther., 109, pp. 137-161
  • Tencalla, F., Dietrich, D., Biochemical characterization of microcystin toxicity in rainbow trout (Oncorhynchus mykiss) (1997) Toxicon, 35, pp. 583-595
  • Varki, A., Schauer, R., Chapter 14: sialic acids (2009) Essentials of Glycobiology, , A. Varki R.D. Cummings J.D. Esko H.H. Freeze P. Stanley C.R. Bertozzi G.W. Hart M.E. Etzler 2nd ed. Cold Spring Harbor Laboratory Press New York
  • Vila, I., Soto, D., Atherinidae (Pisces) of rapel reservoir, Chile.V erh (1981) Internat. Verein. Limnology, 21, pp. 1334-1338
  • Wiegand, C., Pflugmacher, S., Ecotoxicological effects of selected cyanobacterial secondary metabolites a short review (2005) Toxicol. Appl. Pharmacol., 203, pp. 201-218
  • Williams, D.E., Craig, M., Dawe, S.C., Kent, M.L., Holmes, C.F.B., Andersen, R.J., Evidence for a covalently bound form of microcystin-LR in salmon liver and dungeness crab larvae (1997) Chem. Res. Toxicol., 10, pp. 463-469
  • Xie, L., Xie, P., Ozawa, K., Honma, T., Yokoyama, A., Park, H.D., Dynamics of microcystins-LR and −RR in the phytoplanktivorous silver carp in a sub-chronic toxicity experiment (2004) Environ. Pollut., 127, pp. 431-439
  • Zaja, R., Munić, V., Sauerborn Klobučar, R., Ambriović-Ristov, A., Smital, T., Cloning and molecular characterization of apical efflux transporters (ABCB1 ABCB11 and ABCC2) in rainbow trout (Oncorhynchus mykiss) hepatocytes (2008) AquaticToxicology, 90, pp. 322-332
  • Zar, H.J., Biostatistical Analysis (1999), 4th ed. Prentice Hall New Jersey

Citas:

---------- APA ----------
Bieczynski, F., Torres, W.D.C., Painefilu, J.C., Castro, J.M., Bianchi, V.A., Frontera, J.L., Paz, D.A.,..., Luquet, C.M. (2016) . Alterations in the intestine of Patagonian silverside (Odontesthes hatcheri) exposed to microcystin-LR: Changes in the glycosylation pattern of the intestinal wall and inhibition of multidrug resistance proteins efflux activity. Aquatic Toxicology, 178, 106-117.
http://dx.doi.org/10.1016/j.aquatox.2016.07.016
---------- CHICAGO ----------
Bieczynski, F., Torres, W.D.C., Painefilu, J.C., Castro, J.M., Bianchi, V.A., Frontera, J.L., et al. "Alterations in the intestine of Patagonian silverside (Odontesthes hatcheri) exposed to microcystin-LR: Changes in the glycosylation pattern of the intestinal wall and inhibition of multidrug resistance proteins efflux activity" . Aquatic Toxicology 178 (2016) : 106-117.
http://dx.doi.org/10.1016/j.aquatox.2016.07.016
---------- MLA ----------
Bieczynski, F., Torres, W.D.C., Painefilu, J.C., Castro, J.M., Bianchi, V.A., Frontera, J.L., et al. "Alterations in the intestine of Patagonian silverside (Odontesthes hatcheri) exposed to microcystin-LR: Changes in the glycosylation pattern of the intestinal wall and inhibition of multidrug resistance proteins efflux activity" . Aquatic Toxicology, vol. 178, 2016, pp. 106-117.
http://dx.doi.org/10.1016/j.aquatox.2016.07.016
---------- VANCOUVER ----------
Bieczynski, F., Torres, W.D.C., Painefilu, J.C., Castro, J.M., Bianchi, V.A., Frontera, J.L., et al. Alterations in the intestine of Patagonian silverside (Odontesthes hatcheri) exposed to microcystin-LR: Changes in the glycosylation pattern of the intestinal wall and inhibition of multidrug resistance proteins efflux activity. Aquat. Toxicol. 2016;178:106-117.
http://dx.doi.org/10.1016/j.aquatox.2016.07.016