11 research outputs found

    In Vitro Cytotoxicity Assays Of Solid Lipid Nanoparticles In Epithelial And Dermal Cells

    Get PDF
    In recent years, the interest in nanostructured systems to drug delivery has increased because they offer several advantages over conventional dosage forms. Solid Lipid Nanoparticles (SLN) have been highlighted among these systems because they have advantages such as high physical stability, protection against drug degradation and ease of scale-up and manufacturing, without using organic solvent. The aim of this work was to evaluate the potential of SLN, by in vitro cytotoxicity assays, for dermal drug delivery. SLN of three different lipids were prepared by hot high pressure homogenization and the cytotoxicity was assessed by 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyl tetrazolium bromide (MTT) test in mouse 3T3 fibroblasts and human HaCaT keratinocytes. SLN showed no cytotoxic potential suggesting a great potential for dermal application.3041Mishra, B., Patel, B.B., Tiwari, S., (2010) Nanomed.-Nanotechnol. Biol. Med., 6, p. 9Mehnert, W., Mäder, K., (2001) Adv. Drug Deliv. Rev., 47, p. 165Pardeike, J., Hommoss, A., Müller, R.H., (2009) Int. J. Pharm., 366, p. 170Lewinski, N., Colvin, V., Drezek, R., (2008) Small, 4, p. 26Schöler, N., Hahn, H., Müller, R.H., Liesenfeld, O., (2002) Int. J. Pharm., 231, p. 167Müller, R.H., Maassen, S., Schwarz, C., Mehnert, W., (1997) J. Control. Release, 47, p. 261Shöler, N., Hahn, H., Müller, R.H., Liesenfeld, O., (2002) Int. J. Pharm., 231, p. 167Weyenberg, W., Filev, P., Plas, D.V., Vandervoort, J., Smet, K.D., Sollie, P., Ludwig, A., (2007) Int. J. Pharm., 337, p. 291Kristl, J., Teskac, K., Milek, M., Rascan, I.M., (2008) Toxicol. Appl.Pharmacol., 232, p. 218Mosmann, T., (1983) J. Immunol. Methods, 65, p. 55Marcato, P.D., Caverzan, J., Rossi-Bergmann, B., Pinto, E.F., MacHado, D., Silva, R.A., Justos, G.Z., Durán, N., (2011) J. Nanosci. Nanotechnol

    Catalytic Antibodies: Expanding The Scope Of Enzymatic Catalysis [anticorpos Catalíticos: Expandindo O Alcance Da Catálise Enzimática]

    No full text
    The vast binding repertoire of the immune system has been exploited for the generation of tailor-made selective catalysts. Since the first reports of chemical reactions catalyzed by antibodies were published, research in this field, which borders chemistry and biology, has been rapidly established and a number of catalytic antibodies that carry out a wide range of reactions, have been developed. Recent advances have led to antibodies that catalyse complex, multi-step reactions and difficult chemical transformations, as well as reactions that do not have an organic equivalent at all. Current research in this field has been devoted to practical applications of this technology.2118698Davies, D.R., Chacko, S., (1993) Acc. Chem. Res., 26, p. 421Male, D., Cooke, A., Owen, M., Trowsdale, J., Champion, B., (1996) Advanced Immunology, , MosbyLondonPauling, L., (1948) Am. Sci., 36, p. 519Jencks, W., (1969) Catalysis in Chemistry and Enzymology, , McGraw-HillNew YorkRaso, V., Stollar, B.D., (1975) Biochemistry, 14, p. 584Raso, V., Stollar, B.D., (1975) Biochemistry, 14, p. 591Köhler, G., Milstein, C., (1975) Nature, 256, p. 5517Köhler, G., Houe, S.C., Milstein, C., (1976) Eur. J. Immunol., 6, p. 292Köhler, G., Milstein, C., (1976) Eur. J. Immunol., 6, p. 511Pollack, S.J., Jacobs, J.W., Schultz, P.G., (1986) Science, 234, p. 1570Tramontano, A., Janda, K.D., Lerner, R.A., (1986) Science, 234, p. 1566Schultz, P., (1989) Angew. Chem. Int. Ed. Engl., 28, p. 1283Lerner, R.A., Benkovic, S.J., Schultz, P.G., (1991) Science, 252, p. 659Powell, M.J., Hansen, D.E., (1989) Protein Eng., 3, p. 69Krafft, G.A., Wang, G.T., (1989) Ann. Rep. Med. Chem., 25, p. 299Schultz, P.G., (1989) Acc. Chem. Res., 22, p. 287Shokat, K.M., Schultz, P.G., (1990) Ann. Rev. Immunol., 8, p. 335Hilvert, D., Hill, K.W., (1991) Meth. Enzimol., 203, p. 352Suckling, C.J., (1991) Biochem. Soc. Trans, p. 216Schultz, P.G., Lerner, R.A., (1993) Acc. Chem. Res., 26, p. 391Stewart, J.D., Liotta, L.J., Benkovic, S.J., (1993) Acc. Chem. Res., 26, p. 396Stewart, J.D., Benkovic, S.J., (1993) Chem. Soc. Rev., p. 213Leumann, C., (1993) Angew. Chem. Int. Ed. Engl., 32, p. 1291Zouali, M., Hansen, D.E., (1994) Trends Biotechnol., 12, p. 73Blackburn, G.M., Wentworth, P., (1994) Chem. Ind., p. 338Hilvert, D., (1994) Citrr. Opin. Struct. Biol., 4, p. 612Janda, K.D., (1994) Pure Appl. Chem., 66, p. 703Scanlan, T.S., (1995) Ann. Rev. Med. Chem., 30, p. 255Hsieh-Wilson, L.C., Xiang, X.-D., Schultz, P.G., (1996) Acc. Chem. Res., 29, p. 164Schultz, P.G., (1988) Science, 240, p. 426Hilvert, D., (1992) Pure Appl. Chem., 64, p. 1103Hilvert, D., (1993) Acc. Chem. Res., 26, p. 552Stewart, J.D., Krebs, J.F., Siuzdak, G., Berdis, A.J., Smithrud, D.B., Benkovic, S.J., (1994) Proc. Natl. Acad. Sci. USA, 91, p. 7404Siuzdak, G., Krebs, J.F., Benkovic, S.J., Dyson, H.J., (1994) J. Am. Chem. Soc., 116, p. 7937Jacobsen, S.R., Schultz, P.G., (1995) Curr. Opin. Struct. Biol., 5, p. 818Miyashita, H., Hara, T., Tanimura, R., Tanaka, F., Kikuchi, M., Fujii, I., (1994) Proc. Natl. Acad. Sci. USA, 91, p. 6045Seiler, F.R., Gronski, P., Kurrle, R., Luben, G., Hartbus, H.P., Ax, W., Bosslet, K., Schwick, H.G., (1985) Angew. Chem. Int. Ed. Engl., 24, p. 139Tawfik, D.S., Green, B.S., Chap, R., Sela, M., Eshhar, Z., (1993) Proc. Natl. Acad. Sci. USA, 90, p. 373MacBeath, G., Hilvert, D., (1994) J. Am. Chem. Soc., 116, p. 6101Tawfik, D.S., Zemel, R.R., Arad-Yellin, R., Green, B.S., Eshhar, Z., (1990) Biochemistry, 29, p. 9916Thorn, S.N., Daniels, R.G., Auditor, M.-T.M., Hilvert, D., (1995) Nature, 373, p. 228Pollack, S.J., Hsium, P., Schultz, P.G., (1989) J. Am. Chem. Soc., 111, p. 5961Matayoshi, E.D., Wang, G.T., Krafft, G.A., Erickson, J.W., (1990) Science, 247, p. 954Cummings, R.T., Krafft, G.A., (1988) Tetrahedron Lett., 29, p. 65Huse, W.D., Sastry, L., Iverson, S.A., Kang, A.S., Alting-Mees, M., Burton, D.R., Benkovic, S.J., Lerner, R.A., (1989) Science, 246, p. 1275McCafferty, J., Griffiths, A.D., Winter, G., Criswell, D.J., (1990) Nature, 348, p. 552Kang, A.S., Barbas, C.F., Janda, K.D., Benkovic, S.J., Lerner, R.A., (1991) Proc. Natl. Acad. Sci. USA, 88, p. 4363Marks, J.D., Hoogenboom, H.R., Bonnert, T.P., McCafferty, J., Griffiths, A.D., Winter, G., (1991) J. Mot. Biol., 222, p. 581Barbas, C.F., III, Kang, A.S., Lerner, R.A., Benkovic, S.J., (1991) Proc. Natl. Acad. Sci. USA, 88, p. 7978Barbas, C.F., III, Bain, J.D., Hoekstra, D.M., Lerner, R.A., (1992) Proc. Natl. Acad. Sci. USA, 89, p. 4457Lerner, R.A., Kang, A.S., Bain, J.D., Burton, D.R., Barbas, C.F., III, (1992) Science, 258, p. 1313Gram, H., Marconi, L.A., Barbas, C.F., III, Collet, T.A., Lerner, R.A., Kang, A.S., (1992) Proc. Natl. Acad. Sci. USA, 89, p. 3576Collet, T.A., Roben, P., O'Kennedy, R., Barbas, C.F., III, Burton, D.R., Lerner, R.A., (1992) Proc. Natl. Acad. Sci. USA, 89, p. 10026Barbas, C.F., III, Rosenblum, J.S., Lerner, R.A., (1993) Proc. Natl. Acad. Sci. USA, 90, p. 6385Barbas, C.F., III, Amberg, W., Simoncsits, A., Jones, T.M., Lerner, R.A., (1993) Gene, 137, p. 57Griffiths, A.D., Williams, S.C., Hartley, O., Tomlinson, I.M., Waterhouse, P., Crosby, W.L., Kontermann, R.E., Winter, G., (1994) EMBO J., 13, p. 3245Burton, D.R., (1993) Acc. Chem. Res., 26, p. 405Posner, B., Smiley, J., Lee, I., Benkovic, S.J., (1994) Trends Biochem. Sci., 19, p. 145Sastry, L., Alting-Mees, M., Huse, W.D., Short, J.M., Sorge, J.A., Hay, B.N., Janda, K.N., Lerner, R.A., (1989) Proc. Natl. Acad. Sci. USA, 86, p. 5728Winter, G., Milstein, C., (1991) Nature, 349, p. 293Hoogenboom, H.R., Griffiths, A.D., Johnson, K.S., Criswell, D.J., Hudson, P., Winter, G., (1991) Nitcl. Acids Res., 19, p. 4133Smith, G.P., (1985) Science, 228, p. 1315Burton, D.R., Barbas, C.F., III, (1994) Adv. Immunol., 57, p. 191Janda, K.D., Lo, C.-H.L., Li, T., Barbas, C.F., III, Wirsching, P., Lerner, R.A., (1994) Proc. Natl. Acad. Sci. USA, 91, p. 2532Jacobs, J., Schultz, P.G., (1987) J. Am. Chem. Soc., 109, p. 2174Seebach, D., (1990) Angew. Chem. Int. Ed. Engl., 29, p. 1320Janda, K.D., Benkovic, S.J., Lerner, R.A., (1989) Science, 244, p. 437Tramontano, A., Ammann, A., Lerner, R.A., (1988) J. Am. Chem. Soc., 110, p. 2282Carter, P., Wells, J.A., (1988) Nature, 332, p. 564Padlan, E., (1986) J. Mol. Biol., 190, p. 593Durfor, C.N., Bolin, R.J., Sugasawara, R.J., Massey, R.J., Jacobs, J.W., Schultz, P.G., (1988) J. Am. Chem. Soc., 110, p. 8713Janda, K.D., Ashley, J.A., Jones, T.M., McLeod, D.A., Schloeder, D.M., Weinhouse, M.I., (1990) J. Am. Chem. Soc., 112, p. 8886Okahata, Y., Yamaguchi, M., Tanaka, F., Fujii, I., (1995) Tetrahedron Lett., 51, p. 7673Janda, K.D., Schloeder, D., Benkovic, S.J., Lerner, R.A., (1988) Science, 241, p. 1188Iverson, B.L., Lerner, R.A., (1989) Science, 243, p. 1184Martin, M.T., Angeles, T.S., Sugasawara, R., Aman, N.I., Napper, A.D., Darsley, M.J., Sanchez, R.I., Titmas, R.C., (1994) J. Am. Cliem. Soc., 116, p. 6508Baldwin, E., Schultz, P.G., (1989) Science, 245, p. 1104Stewart, J.D., Roberts, V.A., Thomas, N.R., Getsoff, E.D., Benkovic, S.J., (1994) Biochemistry, 33, p. 1994Roberts, V.A., Stewart, J., Benkovic, S.J., Getzoff, E.D., (1994) J. Mol. Biol., 235, p. 1098Guo, J., Huang, W., Scanlan, T.S., (1994) J. Am. Chem. Soc., 116, p. 6062Zhou, G.W., Guo, J., Huang, W., Fletterick, R.J., Scanlan, T.S., (1994) Science, 265, p. 1059Guo, J., Huang, W., Zhou, G.W., Fletterick, R.J., Scalan, T.S., (1995) Proc. Natl. Acad. Sci. USA, 92, p. 1694Wade, H., Scanlan, T.S., (1996) J. Am. Chem. Soc., 118, p. 6510Charbonnier, J.-B., Carpenter, E., Gigant, B., Golinelli-Pimpaneau, B., Eshhar, Z., Green, B.S., Knossow, M., (1995) Proc. Natl. Acad. Sci. USA, 92, p. 11721Shokat, K.M., Leumann, C.J., Sugasawara, R., Schultz, P.G., (1989) Nature, 338, p. 269Shokat, K.M., Uno, T., Schultz, P.G., (1994) J. Am. Chem. Soc., 116, p. 2261Uno, T., Schultz, P.G., (1992) J. Am. Chem. Soc., 114, p. 6573Reymond, J.-L., Janda, K.D., Lerner, R.A., (1992) J. Am. Chem. Soc., 114, p. 2257Reymond, J.-L., Jahangiri, G.K., Stoudt, C., Lerner, R.A., (1993) J. Am. Chem. Soc., 115, p. 3909Jahangiri, G.K., Reymond, J.-L., (1994) J. Am. Chem. Soc., 116, p. 11264Jackson, D.Y., Schultz, P.G., (1991) J. Am. Chem. Soc., 113, p. 2319Li, T., Janda, K.D., Ashley, J.A., Lerner, R.A., (1994) Science, 264, p. 1289Li, T., Hilton, S., Janda, K.D., (1995) J. Am. Chem. Soc., 117, p. 3308Li, T., Janda, K.D., Lerner, R.A., (1996) Nature, 379, p. 326Napper, A.D., Benkovic, S.J., Tramontano, A., Lerner, R.A., (1987) Science, 237, p. 1041Jackson, D.Y., Jacobs, J.W., Sugasawara, R., Reich, S.H., Bartlett, P., Schultz, P.G., (1988) J. Am. Chem. Soc., 110, p. 4841Hilvert, D., Hill, K.W., Nared, K.D., Auditor, M.-T.M., (1989) J. Am. Chem. Soc., 111, p. 9261Braisted, A., Schultz, P.G., (1990) J. Am. Chem. Soc., 112, p. 7430Wirsching, P., Ashley, J.A., Benkovic, S.J., Janda, K.D., Lerner, R.A., (1991) Science, 252, p. 680Janda, K.D., Lerner, R.A., Tramontano, A., (1988) J. Am. Chem. Soc., 110, p. 4835Cochran, A.G., Sugasawara, R., Schultz, P.G., (1988) J. Am. Chem. Soc., 110, p. 7888Na, J., Houk, K.N., Hilvert, D., (1996) J. Am. Chem. Soc., 118, p. 6462Gray, J.V., Golinelli-Pimpaneau, B., Knowles, J.R., (1990) Biochemistry, 29, p. 376Bowdish, K., Tang, Y., Hicks, J.B., Hilvert, D., (1991) J. Biol. Chem., 266, p. 11901Shin, J.A., Hilvert, D., (1994) Bioorg. Med. Chem. Lett., 4, p. 2945Campbell, A.P., Tarasow, T.M., Massefski, W., Wright, P.E., Hilvert, D., (1993) Proc. Natl. Am. Sci. USA, 90, p. 8663Haynes, M.R., Stura, E.A., Hilvert, D., Wilson, I.A., (1994) Science, 263, p. 646Jackson, D.Y., Liang, M.N., Bartlett, P.A., Schultz, P.G., (1992) Angew. Chem. Int. Ed. Engl., 31, p. 182Wiest, O., Houk, K.N., (1995) J. Am. Chem. Soc., 117, p. 11628Wiest, O., Houk, K.N., (1994) J. Org. Chem., 59, p. 7582Davidson, M.M., Gould, I.R., Hillier, I., (1995) J. Chem. Soc. Chem. Commun., p. 63Oikawa, H., Katayama, K., Suzuki, Y., Ichihara, A., (1995) J. Chem. Soc. Chem. Commun., p. 1321Reymond, J.-L., Reber, J.-L., Lerner, R.A., (1994) Angew. Chem, Int. Ed. Engl., 33, p. 475Sinha, S.C., Keinan, E.J., (1995) J. Am. Clwm. Soc., 117, p. 3653Bahr, N., Giiller, R., Reymond, J.-L., Lerner, R.A., (1996) J. Am. Chem. Soc., 118, p. 3550Reymond, J.-L., Chen, Y., (1995) Tetrahedron Lett., 36, p. 2575Reymond, J.-L., Chen, Y., (1995) J. Org. Chem., 60, p. 6970Koch, T., Reymond, J.-L., Lerner, R.A., (1995) J. Am. Chem. Soc., 117, p. 9383Pollack, S.J., Nakayama, G.R., Schultz, P.G., (1988) Science, 242, p. 1038Pollack, S.J., Schultz, P.G., (1989) J. Am. Chem. Soc., 111, p. 1929Janjic, N., Tramontano, A., (1989) J. Am. Chem. Soc., 111, p. 9109Hsieh, L.C., Stephans, J.C., Schultz, P.G., (1994) J. Am. Chem. Soc., 116, p. 2167Koch, A., Reymond, J.-L., Lerner, R.A., (1994) J. Am. Chem. Soc., 116, p. 803Hsieh, L.C., Yonkovich, S., Kochersperger, L., Schultz, P.G., (1993) Science, 260, p. 337Kitazume, T., Lin, J.T., Yamamoto, T., Yamazaki, T., (1991) J. Am. Chem. Soc., 113, p. 8573Iwabuchi, Y., Miyashita, H., Tanimura, R., Kinoshita, K., Kikuchi, M., Fujii, I., (1994) J. Am. Chem. Soc., 116, p. 771Shabat, D., Ltzhaky, H., Reymond, J.-L., Keinan, E., (1995) Nature, 374, p. 143Gouverneur, V.E., Houk, K.N., De Pacual-Teresa, B., Beno, B., Janda, K.D., Lerner, R.A., (1993) Science, 262, p. 204Murata, M., Naoki, H., Matsunaga, S., Satake, M., Yasumoto, T., (1994) J. Am. Chem. Soc., 116, p. 7098Janda, K.D., Shevlin, C.G., Lerner, R.A., (1995) J. Am. Chem. Soc., 117, p. 2659Janda, K.D., Shevlin, C.G., Lerner, R.A., (1993) Science, 259, p. 490Cravatt, B.F., Ashley, J.A., Janda, K.D., Boger, D.L., Lerner, R.A., (1994) J. Am. Chem. Soc., 116, p. 6013Jacobsen, J.R., Prudent, J.R., Kochersperger, L., Yonkovich, S., Schultz, P.G., (1992) Science, 256, p. 365Jacobsen, J.R., Schultz, P.G., (1994) Proc. Natl. Acad. Sci. USA, 91, p. 5888Uno, T., Gong, B., Schultz, P.G., (1994) J. Am. Chem. Soc., 116 (1), p. 145Braisted, A.C., Schultz, P.G., (1994) J. Am. Chem. Soc., 116, p. 2211Yli-Kauhaluoma, J.T., Ashley, J.S., Lo, C.-H., Tucker, L., Wolfe, M.M., Janda, K.D., (1995) J. Am. Chem. Soc, 117, p. 7041Fujii, I., Tanaka, F., Miyashita, H., Tanimura, R., Kinoshita, K., (1995) J. Am. Chem. Soc., 117, p. 6199Lerner, R.A., Benkovic, S.J., (1988) BioEssays, 9, p. 107Green, B.S., Tawfik, D.S., (1989) Trends' Bioteclmol, 7, p. 304Bagshawc, K.D., (1989) Br. J. Cancer, 60, p. 275Lerner, R.A., Benkovic, S.J., (1990) Chemtracts-Org. Chem., 3, p. 1Schultz, P.G., Lerner, R.A., Benkovic, S.J., (1990) Chem. Eng. News, 68, p. 26Landry, D.W., Zhao, K., Yang, G.X.-Q., Glickman, M., Georgiadis, T.M., (1993) Science, 259, p. 1899Miyashita, H., Karaki, Y., Kikuchi, M., Fujii, I., (1993) Proc. Natl. Acad. Sci. USA, 90, p. 5337Smiley, J.A., Benkovic, S.J., (1995) J. Am. Chem. Soc., 117, p. 3877Yang, G., Chun, J., Arakawa-Uramoto, H., Wang, X., Gawinowicz, M.A., Zhao, K., Landry, D.W., (1996) J. Am. Chem. Soc., 118, p. 5881Wentworth, P., Datta, A., Blakey, D., Boyle, T., Partridge, L.J., Blackburn, G.M., (1996) Proc. Natl. Acad. Sci. USA, 93, p. 79

    Coagulation And Cancer Therapy: The Potential Of Natural Compounds

    No full text
    In the past few years, it has become clear that the processes of cancer metastasis and invasion are highly dependent on components of the blood coagulation cascade. Metastasis is critically dependent on the formation of new blood vessels and the role of many blood-clotting factors in tumor angiogenesis has been extensively studied. Angiogenesis constitutes an important point in the control of cancer progression and in recent years much attention has been paid to the development of antiangiogenic agents. Natural compounds constitute a promising alternative for application in antiangiogenesis therapy due to their multiple mechanisms of action. Moreover, they can be used as templates for the production of analogues with enhanced activity. In this review, the potential of natural compounds to target blood coagulation cascades and to prevent tumor growth will be highlighted in view of their underlying mechanisms. © 2005 Bentham Science Publishers Ltd.66461469Daly, M.E., Makris, A., Reed, M., Lewis, C.E., Hemostatic regulators of tumor angiogenesis, a source of antiangiogenic agents for cancer treatment? (2003) J. Natl. Cancer Inst., 95, pp. 1660-1673Stenina, O.I., Plow, E.F., MET orchestrates cancer and blood coagulation (2005) Nat. Med., 11, pp. 376-377Hanahan, D., Folkman, J., Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis (1996) Cell, 86, pp. 353-364Turner, H.E., Harris, A.L., Melmed, S., Wass, J.A.H., Angiogenesis in endocrine tumors (2003) Endocrine Rev., 24, pp. 600-632Bocci, G., Nicolaou, K.C., Kerbel, R.S., Protracted low-dose effects on human endothelial cell proliferation and survival in vitro reveal a selective antiangiogenic window for various chemotherapeutic drugs (2002) Cancer Res., 62, pp. 6938-6943Rickles, F.R., Patierno, S., Fernandez, P.M., Tissue factor, thrombin, and cancer (2003) Chest, 124, pp. 58S-68SVersteeg, H.H., Peppelenbosch, M.P., Spek, C.A., Tissue factor signal transduction in angiogenesis (2003) Carcinogenesis, 24, pp. 1009-1013Price, G.C., Thompson, S.A., Kam, P.C., Tissue factor and tissue factor pathway inhibitor (2004) Anaesthesia, 59, pp. 483-492Sawyer, T.K., Cancer metastasis therapeutic targets and drug discovery, emerging small-molecule protein kinase inhibitors (2004) Expert Opin. Investig. Drugs, 13, pp. 1-19Herbst, R.S., Onn, A., Sandler, A., Angiogenesis and lung cancer, prognostic and therapeutic implications (2005) J. Clin. Oncol., 23, pp. 3243-3256Kerbel, R., Folkman, J., Clinical translation of angiogenesis inhibitors (2002) Nat. Rev. Cancer, 2, pp. 727-739Ma, L., Perini, R., McKnight, W., Dicay, M., Klein, A., Hollenberg, M.D., Wallace, J.L., Proteinase-activated receptors 1 and 4 counter-regulate endostatin and VEGF release from human platelets (2005) Proc. Natl. Acad. Sci. USA, 102, pp. 216-220Peppelenbosch, M.P., Versteeg, H.H., Cell biology of tissue factor, an unusual member of the cytokine receptor family (2001) Trends Cardiovasc. Med., 11, pp. 335-339Versteeg, H.H., Spek, C.A., Peppelenbosch, M.P., Richel, D.J., Tissue factor and cancer metastasis, the role of intracellular and extracellular signaling pathways (2004) Mol. Med., 10, pp. 6-11Guan, M., Jin, J., Su, B., Liu, W.W., Lu, Y., Tissue factor expression and angiogenesis in human glioma (2002) Clin. Biochem., 35, pp. 321-325Contrino, J., Hair, G., Kreutzer, D.L., Rickles, F.R., In situ detection of tissue factor in vascular endothelial cells, correlation with the malignant phenotype of human breast disease (1996) Nat. Med., 2, pp. 209-215Hair, G.A., Padula, S., Zeff, R., Schmeizl, M., Contrino, J., Kreutzer, D.L., de Moerloose, P., Rickles, F.R., Tissue factor expression in human leukemic cells (1996) Leuk. Res., 20, pp. 1-11Koomagi, R., Volm, M., Tissue-factor expression in human non-small-cell lung carcinoma measured by immunohistochemistry, correlation between tissue factor and angiogenesis (1998) Int. J. Cancer, 79, pp. 19-22Seto, S., Onodera, H., Kaido, T., Yoshikawa, A., Ishigami, S., Arii, S., Imamura, M., Tissue factor expression in human colorectal carcinoma, correlation with hepatic metastasis and impact on prognosis (2000) Cancer, 88, pp. 295-301Ueda, C., Hirohata, Y., Kihara, Y., Nakamura, H., Abe, S., Akahane, K., Okamoto, K., Otsuki, M., Pancreatic cancer complicated by disseminated intravascular coagulation associated with production of tissue factor (2001) Gastroenterol., 36, pp. 848-850Dupuy, E., Habib, A., Lebret, M., Yang, R., Levy-Toledano, S., Tobelem, G., Thrombin induces angiogenesis and vascular endothelial growth factor expression in human endothelial cells, possible relevance to HIF-1alpha (2003) J. Thromb. Haemost., 1, pp. 1096-1102Tsopanoglou, N.E., Maragoudakis, M.E., Role of thrombin in angiogenesis and tumor progression (2004) Semin. Thromb. Hemost., 30, pp. 63-69Hu, L., Lee, M., Campbell, W., Perez-Soler, R., Karpatkin, S., Role of endogenous thrombin in tumor implantation, seeding, and spontaneous metastasis (2004) Blood, 104, pp. 2746-2751Justo, G.Z., Durán, N., Queiroz, M.L., Myelopoietic response in tumour-bearing mice by an aggregated polymer isolated from Aspergillus oryzae (2000) Eur. J. Pharmacol., 388, pp. 219-226Beretz, A., Cazenave, J.P., Old and new natural products as the source of modern antithrombotic drugs (1991) Planta Med., 57, pp. S68-S72Freire, A.G., Melo, P.S., Haun, M., Durán, N., Aoyama, H., Ferreira, C.V., Cytotoxic effect of the diterpene lactone dehydrocrotonin from Croton cajucara on human promyelocytic leukemia cells (2003) Planta Med., 69, pp. 67-69Freire, A.C., de Assis, C.F., Frick, A.O., da Silva Melo, P., Haun, M., Aoyama, H., Durán, N., Ferreira, C.V., Influence of protein phosphatase inhibitors on HL60 cells death induction by dehydrocrotonin (2003) Leuk. Res., 27, pp. 823-829Ferreira, C.V., Bos, C.L., Versteeg, H.H., Justo, G.Z., Durán, N., Peppelenbosch, M.P., Molecular mechanism of Violacein-mediated human leukemia cell death (2004) Blood, 104, pp. 1459-1464Justo, G.Z., Durán, N., Queiroz, M.L., Natural killer cell activity, lymphocyte proliferation, and cytokine profile in tumor-bearing mice treated with MAPA. A magnesium aggregated polymer from Aspergillus oryzae (2003) Immunopharmacol. Immunotoxicol., 25, pp. 305-319Justo, G.Z., Silva, M.R., Queiroz, M.L., Effects of the green algae Chlorella vulgaris on the response of the host hematopoietic system to intraperitoneal Ehrlich ascites tumor transplantation in mice (2001) Immunopharmacol. Immunotoxicol., 23, pp. 119-132Melo, P.S., Justo, G.Z., de Azevedo, M.B., Durán, N., Haun, M., Violacein and its beta-cyclodextrin complexes induce apoptosis and differentiation in HL60 cells (2003) Toxicology, 186, pp. 217-225Kinghorn, A.D., Su, B.N., Jang, D.S., Chang, L.C., Lee, D., Gu, J.Q., Carcache-Blanco, E.J., Pezzuto, J.M., Natural inhibitors of carcinogenesis (2004) Planta Med., 70, pp. 691-705Tsuda, H., Ohshima, Y., Nomoto, H., Fujita, K., Matsuda, E., Iigo, M., Takasuka, N., Moore, M.A., Cancer prevention by natural compounds (2004) Drug Metab. Pharmacokinet., 19, pp. 245-263Melo, P.S., Justo, G.Z., Durán, N., Haun, M., Natural killer cell activity and anti-tumour effects of dehydrocrotonin and its synthetic derivatives (2004) Eur. J. Pharmacol., 487, pp. 47-54Mann, J., Natural products in cancer chemotherapy, past, present and future (2002) Nat. Rev., 2, pp. 143-147Magee, P.J., McGlynn, H., Rowland, I.R., Differential effects of isoflavones and lignans on invasiveness of NWA-MB-231 breast cancer cells in vitro (2004) Cancer Lett., 208, pp. 35-41Cao, Y., Cao, R., Brakenhielm, E., Antiangiogenic mechanisms of diet-derived polyphenols (2002) J. Nutr. Biochem., 13, pp. 380-390Ikeda, K., Sakai, Y., Nagase, H., Inhibitory effect of magnolol on tumour metastasis in mice (2003) Phytother. Res., 17, pp. 933-937Kimura, Y., New anticancer agents, in vitro and in vivo evaluation of the antitumor and antimetastatic actions of various compounds isolated from medicinal plants (2005) In Vivo, 19, pp. 37-60Oak, M.H., Chataigneau, M., Keravis, T., Chataigneau, T., Beretz, A., Andriantsitohaina, R., Stoclet, J.C., Schini-Kerth, V.B., Red wine polyphenolic compounds inhibit vascular endothelial growth factor expression in vascular smooth muscle cells by preventing the activation of the p38 mitogen-activated protein kinase pathway (2003) Arterioscler. Thromb. Vasc. Biol., 23, pp. 1001-1007Oak, M.H., El Bedoui, J., Schini-Kerth, V.B., Antiangiogenic properties of natural polyphenols from red wine and green tea (2005) J. Nutr. Biochem., 16, pp. 1-8Gorlach, A., Diebold, I., Schini-Kerth, V.B., Berchner-Pfannschmidt, U., Roth, U., Brandes, R.P., Kietzmann, T., Busse, R., Thrombin activates the hypoxia-inducible factor-1 signaling pathway in vascular smooth muscle cells, Role of the p22(phox)-containing NADPH oxidase (2001) Circ. Res., 89, pp. 47-54Herkert, O., Djordjevic, T., BelAiba, R.S., Gorlach, A., Insights into the redox control of blood coagulation, role of vascular NADPH oxidase-derived reactive oxygen species in the thrombogenic cycle (2005) Antioxid. Redox. Signal., 6, pp. 765-776Djordjevic, T., Pogrebniak, A., BelAiba, R.S., Bonello, S., Wotzlaw, C., Acker, H., Hess, J., Gerlach, A., The expression of the NADPH oxidase subunit p22phox is regulated by a redox-sensitive pathway in endothelial cells (2005) Free Radic. Biol. Med., 38, pp. 616-630Khan, S.G., Katiyar, S.K., Agarwal, R., Mukhtar, H., Enhancement of antioxidant and phase II enzymes by oral feeding of green tea polyphenols in drinking water to SKH-1 hairless mice, possible role in cancer chemoprevention (1992) Cancer Res., 52, pp. 4050-4052Hanasaki, Y., Ogawa, S., Fukui, S., The correlation between active oxygens scavenging and antioxidative effects of flavonoids (1994) Free Radic. Biol. Med., 16, pp. 845-850Lin, J.K., Chen, P.C., Ho, C.T., Lin-Shiau, S.Y., Inhibition of xanthine oxidase and suppression of intracellular reactive oxygen species in HL-60 cells by theaflavin-3, 3′-digallate, (-)-epigallocatechin-3-gallate, and propyl gallate (2000) J. Agric. Food Chem., 48, pp. 2736-2743Liu, J., Chang, S.K., Wiesenborn, D., Antioxidant properties of soybean isoflavone extract and tofu in vitro and in vivo (2005) J. Agric. Food Chem., 53, pp. 2333-2340Moskaug, J.O., Carlsen, H., Myhrstad, M.C., Blomhoff, R., Polyphenols and glutathione synthesis regulation (2005) Am. J. Clin. Nutr., 81, pp. 277S-283SMasuda, M., Suzui, M., Lim, J.T., Deguchi, A., Soh, J.W., Weinstein, I.B., Epigallocatechin-3-gallate decreases VEGF production in head and neck and breast carcinoma cells by inhibiting EGFR-related pathways of signal transduction (2002) J. Exp. Ther. Oncol., 2, pp. 350-359Shimizu, M., Deguchi, A., Lim, J.T., Moriwaki, H., Kopelovich, L., Weinstein, I.B., (-)-Epigallocatechin gallate and polyphenon E inhibit growth and activation of the epidermal growth factor receptor and human epidermal growth factor receptor-2 signaling pathways in human colon cancer cells (2005) Clin. Cancer Res., 11, pp. 2735-2746Inokuchi, H., Hirokane, H., Tsuzuki, T., Nakagawa, K., Igarashi, M., Miyzawa, T., Anti-angiogenic activity of tocotrienol (2003) Biosci. Biotechnol. Biochem., 67, pp. 1623-1627Miyazawa, T., Inokuchi, H., Hirokane, H., Tsuzuki, T., Nakagawa, K., Igarashi, M., Anti-angiogenic potential of tocotrienol in vitro (2004) Biochemistry (Moscow), 69, pp. 67-69Miyazawa, T., Tsuyoshi, T., Nakagawa, K., Igarashi, M., Antiangiogenic potency of vitamin E (2004) Ann. N. Y. Acad. Sci., 1031, pp. 401-404Liu, Z., Schwimer, J., Liu, D., Greenway, F.L., Anthony, C.T., Woltering, E.A., Black raspberry extract and fractions contain angiogenesis inhibitors (2005) J. Agric. Food Chem., 53, pp. 3909-3915Roy, S., Khama, S., Alessio, H.M., Vider, J., Bagchi, D., Bagchi, M., Sen, C.K., Anti-angiogenic property of edible berries (2002) Free Radic. Res., 36, pp. 1023-1031Masso-Welch, P.A., Zangani, D., Ip, C., Vaughan, M.M., Shoemaker, S.F., McGee, S.O., Ip, M.M., Isomers of conjugated linoleic acid differ in their effects on angiogenesis and survival of mouse mammary adipose vasculature (2004) J. Nutr., 134, pp. 299-307Kim, K.H., Park, H.S., Dietary supplementation of conjugated linoleic acid reduces colon tumor incidence in DMH-treated rats by increasing apoptosis with modulation of biomarkers (2003) Nutrition, 19, pp. 772-777Ip, M.M., Masso-Welch, P.A., Ip, C., Prevention of mammary cancer with conjugated linoleic acid, role of the stroma and the epithelium (2003) J. Mammary Gland Biol. Neoplasia, 8, pp. 103-118Sekine, K., Watanabe, E., Nakamura, J., Takasuka, N., Kim, D.J., Asamoto, M., Krutovskikh, V., Tsuda, H., Inhibition of azoxymethane-initiated colon tumor by bovine lactoferrin administration in F344 rats (1997) Jpn. J. Cancer Res., 88, pp. 523-526Shimamura, M., Yamamoto, Y., Ashino, H., Oikawa, T., Hazato, T., Tsuda, H., Iigo, M., Bovine lactoferrin inhibits tumor-induced angiogenesis (2004) Int. J. Cancer, 111, pp. 111-116Varadhachary, A., Wolf, J.S., Petrak, K., O'Malley, Jr., Spadaro, M., Curcio, C., Forni, G., Pericle, F., Oral lactoferrin inhibits growth of established tumors and potentiates conventional chemotherapy (2004) Int. J. Cancer, 111, pp. 398-403Ribatti, D., Ponzoni, M., Antiangiogenic strategies in neuroblastoma (2005) Cancer Treat. Rev., 31, pp. 27-34Kini, A.R., Peterson, L.A., Tallman, M.S., Lingen, M.W., Angiogenesis in acute promyelocytic leukemia, induction by vascular endothelial growth factor and inhibition by all-trans retinoic acid (2001) Blood, 97, pp. 3919-3924Liss, C., Fekete, M.J., Hasina, R., Lingen, M.W., Retinoic acid modulates the ability of macrophages to participate in the induction of the angiogenic phenotype in head and neck squamous cell carcinoma (2002) Int. J. Cancer, 100, pp. 283-289Pal, S., Iruela-Arispe, M.L., Harvey, V.S., Zeng, H., Nagy, J.A., Dvorak, H.F., Mukhopadhyay, D., Retinoic acid selectively inhibits the vascular permeabilizing effect of VPF/VEGF, an early step in the angiogenic cascade (2000) Microvasc. Res., 60, pp. 112-120Ahmed, S.I., Thomas, A.L., Steward, W.P., Vascular endothelial growth factor (VEGF) inhibition by small molecules (2004) J. Chemother., 16 (SUPPL. 4), pp. 59-63Stahl, P., Kissau, L., Mazitschek, R., Giannis, A., Waldmann, H., Natural products derived receptor tyrosine kinase inhibitors, identification of IGF1R, Tie-2, and VEGFR-3 inhibitors (2002) Angew. Chem. Int. Ed., 41, pp. 1174-1178Visse, R., Nagase, H., Matrix metalloproteinases and tissue inhibitors of metalloproteinases, structure, function, and biochemistry (2003) Circ. Res., 92, pp. 827-839Nguyen, M., Arkell, J., Jackson, C.J., Human endothelial gelatinases and angiogenesis (2001) Int. J. Biochem. Cell Biol., 33, pp. 960-970Ko, H.M., Park, Y.M., Jung, B., Kim, H.A., Choi, J.H., Park, S.J., Lee, H.K., Im, S.Y., Involvement of matrix metalloproteinase-9 in platelet-activating factor-induced angiogenesis (2005) FEBS Lett., 579, pp. 2369-2375Tang, Y., Nakada, M.T., Kesavan, P., McCabe, F., Millar, H., Rafferty, P., Bugelski, P., Yan, L., Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases (2005) Cancer Res., 65, pp. 3193-3199Bjorklund, M., Koivunen, E., Gelatinase-mediated migration and invasion of cancer cells (2005) Biochim. Biophys. Acta, 1755, pp. 37-69Huang, X., Chen, S., Xu, L., Liu, Y., Deb, D.K., Platanias, L.C., Bergan, R.C., Genistein inhibits p38 map kinase activation, matrix metalloproteinase type 2: And cell invasion in human prostate epithelial cells (2005) Cancer Res., 65, pp. 3470-3478Oak, M.H., El Bedoui, J., Anglard, P., Schini-Kerth, V.B., Red wine polyphenolic compounds strongly inhibit pro-matrix metalloproteinase-2 expression and its activation in response to thrombin via direct inhibition of membrane type 1-matrix metalloproteinase in vascular smooth muscle cells (2004) Circulation, 110, pp. 1861-1867Kondo, T., Ohta, T., Igura, K., Hara, Y., Kaji, K., Tea catechins inhibit angiogenesis in vitro, measured by human endothelial cell growth, migration and tube formation, through inhibition of VEGF receptor binding (2002) Cancer Lett., 180, pp. 139-144Demeule, M., Brossard, M., Page, M., Gingras, D., Beliveau, R., Matrix metalloproteinase inhibition by green tea catechins (2000) Biochim. Biophys. Acta, 1478, pp. 51-60Brakenhielm, E., Cao, R., Cao, Y., Suppression of angiogenesis, tumor growth, and wound healing by resveratrol, a natural compound in red wine and grapes (2001) FASEB J., 15, pp. 1798-1800Bawadi, H.A., Antunes, T.M., Shih, F., Losso, J.N., In vitro inhibition of the activation of Pro-matrix Metalloproteinase 1 (ProMMP-1) and Pro-matrix metalloproteinase 9 (Pro-MMP-9) by rice and soybean Bowman-Birk inhibitors (2004) J. Agric. Food Chem., 52, pp. 4730-4736Kobayashi, H., Suzuki, M., Kanayama, N., Terao, T., A soybean Kunitz trypsin inhibitor suppresses ovarian cancer cell invasion by blocking urokinase upregulation (2004) Clin. Exp. Metastasis, 21, pp. 159-166Kobayashi, H., Fukuda, Y., Yoshida, R., Kanada, Y., Nishiyama, S., Suzuki, M., Kanayama, N., Terao, T., Suppressing effects of dietary supplementation of soybean trypsin inhibitor on spontaneous, experimental and peritoneal disseminated metastasis in mouse model (2004) Int. J. Cancer, 112, pp. 519-524Fotsis, T., Pepper, M.S., Aktas, E., Breit, S., Rasku, S., Adlercreutz, H., Wahala, K., Schweigerer, L., Flavonoids, dietary-derived inhibitors of cell proliferation and in vitro angiogenesis (1997) Cancer Res., 57, pp. 2916-2921Paper, D.H., Natural products as angiogenesis inhibitors (1998) Planta Med., 64, pp. 686-695Moon, S.K., Cho, G.O., Jung, S.Y., Gal, S.W., Kwon, T.K., Lee, Y.C., Madamanchi, N.R., Kim, C.H., Quercetin exerts multiple inhibitory effects on vascular smooth muscle cells, role of ERK1/2, cell-cycle regulation, and matrix metalloproteinase-9 (2003) Biochem. Biophys. Res. Commun., 301, pp. 1069-1078Hsieh, T.C., Juan, G., Darzynkiewicz, Z., Wu, J.M., Resveratrol increases nitric oxide synthase, induces accumulation of p53 and p21(WAF1/CIP1), and suppresses cultured bovine pulmonary artery endothelial cell proliferation by perturbing progression through S and G2 (1999) Cancer Res., 59, pp. 2596-2601Favot, L., Martin, S., Keravis, T., Andriantsitohaina, R., Lugnier, C., Involvement of cyclin-dependent pathway in the inhibitory effect of delphinidin on angiogenesis (2003) Cardiovasc. Res., 59, pp. 479-487Martin, S., Favot, L., Matz, R., Lugnier, C., Andriantsitohaina, R., Delphinidin inhibits endothelial cell proliferation and cell cycle progression through a transient activation of ERK-1/-2 (2003) Biochem. Pharmacol., 65, pp. 669-675Yoo, H.G., Shin, B.A., Park, J.C., Kim, H.S., Kim, W.J., Chay, K.O., Ahn, B.W., Jung, Y.D., Induction of apoptosis by the green tea flavonol (-)-epigallocatechin-3-gallate in human endothelial ECV 304 cells (2002) Anticancer Res., 22, pp. 3373-3378Kojima-Yuasa, A., Hua, J.J., Kennedy, D.O., Matsui-Yuasa, I., Green tea extract inhibits angiogenesis of human umbilical vein endothelial cells through reduction of expression of VEGF receptors (2003) Life Sci., 73, pp. 1299-1313Iijima, K., Yoshizumi, M., Hashimoto, M., Akishita, M., Kozaki, K., Ako, J., Watanabe, T., Ouchi, Y., Red wine polyphenols inhibit vascular smooth muscle cell migration through two distinct signaling pathways (2002) Circulation, 105, pp. 2404-2410Ellouali, M., Boisson-Vidal, C., Jozefonvicz, J., Effect and interaction of focans with cells (1994) Colloids and Surfaces. B, Biointerfaces, 2, pp. 305-314Soeda, S., Ishida, S., Shimeno, S., Nagamatsu, A., An inhibition effect of oversulfated fucoidan on invasion through reconstituted basement membrane by murine Lewis lung carcinoma (1994) Jpn. J. Cancer Res., 85, pp. 1144-1150Rocha, H.A., Franco, C.R., Trindade, E.S., Carvalho, L.C., Veiga, S.S., Leite, E.L., Dietrich, C.P., Nader, H.B., A fucan from the brown seaweed Spatoglossum schroederi inhibits Chinese hamster ovary cell adhesion to several extracellular matrix proteins (2001) Braz. J. Med. Biol. Res., 34, pp. 621-626Harimaya, Y., Koizumi, K., Andoh, T., Nojima, H., Kuraishi, Y., Saiki, I., Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells (2002) Cancer Lett., 187, pp. 121-127Ingber, D., Fujita, T., Kishimoto, S., Sudo, K., Kanamaru, T., Brem, H., Folkman, J., Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumour growth (1990) Nature, 348, pp. 555-557Kusaka, M., Sudo, K., Fujita, T., Marui, S., Itoh, F., Ingber, D., Folkman, J., Potent anti-angiogenic action of AGM-1470, comparison to the fumagillin parent (1991) Biochem. Biophys. Res. Commun., 74, pp. 1070-1076Maiti, T.K., Chatterjee, J., Dasgupta, S., Effect of green tea polyphenols on angiogenesis induced by an angiogenin-like protein (2003) Biochem. Biophys. Res. Commun., 308, pp. 64-67Kimura, Y., Okuda, H., Resveratrol isolated from Polygonum cuspidatum root prevents tumor growth and metastasis to lung and tumor-induced neovascularization in Lewis lung carcinoma-bearing mice (2001) J. Nutr., 131, pp. 1844-1849Su, S.J., Yeh, T.M., Chuang, W.J., Ho, C.L., Chang, K.L., Cheng, H.L., Liu, H.S., Chow, N.H., The novel targets for anti-angiogenesis of genistein on human cancer cells (2005) Biochem. Pharmacol., 69, pp. 307-318Bagchi, D., Sen, C.K., Bagchi, M., Atalay, M., Anti-angiogenic, antioxidant, and anti-carcinogenic properties of a novel anthocyanin-rich berry extract formula (2004) Biochemistry (Moscow), 69, pp. 75-80Huang, C., Huang, Y., Li, J., Hu, W., Aziz, R., Tang, M.S., Sun, N., Stoner, G.D., Inhibition of benzo[α]pyrene diolepoxide-induced transactivation of activated protein 1 and nuclear factor κB by black raspberry extracts (2002) Cancer Res., 62, pp. 6857-6863Seeram,

    Sensitized Photooxygenation And Peroxidase-catalyzed Inactivation Of Xanthine Oxidase - Evidence Of Cysteine Damage By Singlet Oxygen

    No full text
    Xanthine oxidase (XO) has been investigated for its decreased activity in several cancerous tissues and constitutive generation of reactive oxygen species (ROS) in vivo seems to contribute significantly to its inactivation. Singlet oxygen (1O2) production has been suggested to be relevant when considering folic acid metabolism by cancer cells. Thus, the susceptibility of XO to inactivation by 1O2 generated either by the bioenergized systems folic acid/peroxidase/GSH/Mn2+/O2 and malonaldehyde/peroxidase/Mn2+/O2 or by methylene blue (MB) or eosin-sensitized photooxygenation was studied. Our results showed that other ROS were also responsible for XO inactivation when MB was used. In contrast, eosin produced almost exclusively 1O2. Kinetic studies of XO oxidation in the malonaldehyde/peroxidase system showed that histidine (His) is a competitive inhibitor with respect to XO. A similar result was observed in the eosin-photosensitized process, suggesting the involvement of 1O2 in both processes. In addition, an efficient quenching of XO oxidation by guanosine in the folic acid/peroxidase system was observed. Amino acid analysis revealed that cysteine (Cys) is more affected than other XO amino acids also prone to oxidation such as tyrosine (Tyr), methionine (Met) and His. These results indicate that 1O2 may cause oxidative damage to the Cys residues of XO, with loss of enzyme activity. Alteration of the flavin prosthetic site is hypothesized.322145154Floyd, R.A., (1990) FASEB J., 4, pp. 2587-2597Sun, Y., (1990) Free Rad. Biol. Med., 8, pp. 583-599Josephy, P.D., (1996) Mutagenesis, 11, pp. 3-7Maeda, H., Akaike, T., (1998) Biochemistry, 63, pp. 854-865Szatrowski, T.P., Nathan, C.F., (1991) Cancer Res., 51, pp. 794-798Durak, I., Beduk, Y., Kavutcu, M., Suzer, O., Yaman, O., Ozturk, H.S., Canbolat, O., Ulutepe, S., (1997) Cancer Invest., 15, pp. 212-216Prajda, N., Weber, G., (1975) FEBS Lett., 59, pp. 245-249Rhoads, A.R., Morris, H.P., (1977) J. Natl. Cancer Res., 59, pp. 905-910Weber, G., Kizaki, H., Tzeng, D., Shiotani, T., Olah, E., (1978) Life Sci., 23, pp. 729-736Alsabti, E., (1980) Neoplasma, 27, pp. 95-99Prajda, N., Donohue, J.P., Weber, G., (1981) Life Sci., 29, pp. 853-860Weber, G., Burt, M.E., Jackson, R.C., Prajda, N., Lui, M.S., Takeda, E., (1983) Cancer Res., 43, pp. 1019-1023Komada, H., Kise, Y., Nakagawa, M., Yamamura, M., Hioki, K., Yamamoto, M., (1990) Cancer Res., 50, pp. 2418-2422Pence, B.C., Taylor, M.F., (1990) J. Invest. Dermatol., 95, pp. 213-216Cameron, G.S., Pence, B.C., (1992) J. Invest. Dermatol., 99, pp. 189-192Ikeda, T., Shimokata, K., Daikoku, T., Fukatsu, T., Tsutsui, Y., Nishiyama, Y., (1992) Arch. Virol., 127, pp. 11-24Durak, I., Ormeci, N., Akyol, O., Canbolat, O., Kavutcu, M., Bulbul, M., (1994) Dig. Dis. Sci., 39, pp. 721-728Vaidya, S.M., Kamlakar, P.I., Kamble, S.M., (1998) Indian J. Med. Sci., 52, pp. 244-247Prajda, N., Morris, H.P., Weber, G., (1976) Cancer Res., 36, pp. 4639-4646Kim, H.S., Minard, P., Legoy, M.D., Thomas, D., (1986) Biochem. J., 233, pp. 493-497Michaeli, A., Feitelson, J., (1994) Photochem. Photobiol., 59, pp. 284-289Hille, R., (1998) J. Biol. Inorg. Chem., 3, pp. 559-560Xia, M., Dempski, R., Hille, R., (1999) J. Biol. Chem., 274, pp. 3323-3330Durán, N., (1982) Chemical and Biochemical Generation of Excited States, p. 345. , Adam, W. and Cilento, G. (Eds.), Academic Press, New YorkInnocentini, L.H., Durán, N., (1982) Brazil. J. Med. Biol. Res., 15, pp. 11-16Nishino, T., Tsushima, K., (1986) J. Biol. Chem., 261, pp. 11242-11246Johnson, D.B., Coughlan, M.P., (1978) Biotechnol. Bioeng., 20, pp. 1085-1095Coughlan, M.P., Johnson, D.B., (1979) Biochem. Soc: Trans., 7, pp. 18-21Michaeli, A., Grimes, H., Coughlan, M.P., (1979) Intern. J. Biochem., 10, pp. 317-319Gisler, G.G., Diaz, J., Durán, N., (1982) Physiol. Chem. Phys., 14, pp. 335-342Gisler, G.C., Diaz, J., Durán, N., (1983) Arq. Biol. Tecnol (Brazil), 26, pp. 345-352Durán, N., Gisler, G.C., Diaz, J., (1984) Physiol. Chem. Phys. Med. NMR, 16, pp. 325-331Shiota, T., Disraely, M.N., McCann, M.P., (1964) J. Biol. Chem., 239, pp. 2259-2266Thijssen, H.H.W., (1978) Anal. Biochem., 54, pp. 609-611Haining, J.L., Legan, J.S., (1967) Anal. Biochem., 21, pp. 337-340McHale, A., Grimes, H., Coughlan, M.P., (1979) Int. J. Biochem., 10, pp. 317-319Durá, N., Makita, Y., Innocentini, L.H., (1979) Biochem. Biophys. Res. Commun., 88, pp. 642-648Vidigal-Martinelli, C.C.C., Zinner, K., Kachar, B., Durán, N., Cilento, G., (1979) FEBS Lett., 108, pp. 266-268Innocentini, L.H., De Toledo, S.M., Durán, N., (1980) An. Acad. Brasil. Cienc., 52, pp. 691-693Bray, R.C., Malmstrom, B.G., (1964) Biochem. J., 93, pp. 633-634Nilsson, R., Merckel, P.B., Kearns, D.R., (1972) Photochem. Photobiol., 16, pp. 109-116Foote, C.S., (1978) Singlet Oxygen-Reactions with Organic Compounds and Polymers, p. 135. , Ranby, B. and Rabek, J.F. (Eds.). Wiley Intersc. Publ., LondonFoote, C.S., (1986) Free Radicals in Biology, 2, p. 85. , Pryor, W.A. (Ed.), Academic Press, New YorkDiMascio, P., Medeiros, M.H.G., Bechara, E.J.H., Catalani, L.H., (1995) Ciência e Cultura, 47, pp. 297-311Tanielian, C., (1986) Biochimie, 68, pp. 797-806Cadet, I., Teoule, R., (1978) Photochem. Photobiol., 28, pp. 661-667Ravanat, J.-L., Berger, M., Buchko, G.W., Benard, J.-F., Van Lier, J., Cadet, J., (1991) Chim. Phys., 88, pp. 1069-1076Devasagayam, T.P.A., Steenken, S., Obendorf, M.S.W., Schulz, W.A., Sies, H., (1991) Biochemistry, 25, pp. 6283-6289Foote, C.S., (1979) Singlet Oxygen, p. 139. , Wasserman, H.H. and Murray, R.W. (Eds.), Academic Press, New YorkKepka, A.G., Grossweiner, L.I., (1973) Photochem. Photobiol., 18, pp. 46-61Durán, N., Farias-Furtado, S.T., Faljoni-Alario, A., Campa, A., Brunet, J.E., Freer, J., (1984) J. Photochem., 25, pp. 285-294Justo, G.Z., Innocentini-Mei, L.H., Durán, N., (1990) An. Acad. Brasil. Ciênc., 62, pp. 31-38Spector, T., Ferone, R., (1984) J. Biol. Chem., 259, pp. 10784-10786Robinson, K., Pilot, T.F., Meany, J.E., (1990) Physiol. Chem. Phys. Med NMR, 22, pp. 95-103Haberland, A., Schultz, A.K., Schimke, I., (1992) Biochem. Pharmacol., 43, pp. 2117-2120Augusto, O., Cilento, G., Jung, J., Song, P.S., (1978) Biochem. Biophys. Res. Comm., 83, pp. 963-969Stea, B., Backlund, P.S., Berkey, P.B., Cho, A.K., Halpern, B.C., Halpern, R.M., Smith, R.A., (1978) Cancer Res., 38, pp. 2378-2384Nichols, M.B., Low, P.S., (1986) Arch. Biochem. Biophys., 250, pp. 488-497Fukahori, M., Ichimori, K., Ishida, H., Nakagawa, H., Okino, H., (1994) Free Radic. Res., 21, pp. 203-212Silva, E., Ugarte, R., Andrade, A., Edwards, A.M., (1994) J. Photochem. Photobiol. B: Biol., 23, pp. 43-48Anderson, R.F., Hille, R., Patel, K.B., (1995) Int. J. Rad Biol., 68, pp. 535-541Houston, M., Chumley, P., Radi, R., Rubbo, H., Freeman, B.A., (1998) Arch. Biochem. Biophys., 355, pp. 1-8Durán, N., Haun, M., De Toledo, S.M., Cilento, G., Silva, E., (1983) Photochem. Photobiol, 37, pp. 247-250Durán, N., Cadenas, E., (1987) Res. Chem. Intermediates, 8, pp. 147-187Cilento, G., (1995) Ciência e Cultura, 47, pp. 312-32

    Potential Applications Of Violacein: A Microbial Pigment

    No full text
    Violacein is a versatile pigment from a bacterium Chromobacterium violaceum that exhibits several biological activities and, at present, has gained increasing importance in industrial markets, such as in medicine, cosmetics, and textiles. In this mini-review, we aimed to describe violacein production and to explore its various biological properties in a pharmacological context, including its antioxidant, immunomodulatory, antitumoral, and antiparasitic activities. In addition, its use in the fields of cosmetics, textiles, food, toys, and insecticides has emerged as unusual potential areas of application to be discussed here. © Springer Science+Business Media, LLC 2011.21715241532Ahmetagic, A., Pemberton, J.M., Stable high level expression of the violacein indolocarbazole anti-tumour gene cluster and the Streptomyces lividans amy A gene in E. coli K12 (2010) Plasmid, 63, pp. 79-85Alves, O.L., Gimenez, I.F., De Azevedo, M.M.M., Durán, N., Melo, O.S., Pharmacological use of cyclodextrine-Au-Thiol-derivative/ hydrophobic compound nanoparticles as antitumoral, antibacterial, antiviral and/or antiparasites, its obtention process and formulation (2005), Brazilian Patent PIBr 0502657-1Andrighetti-Frohner, C.R., Antonio, R.V., Creczynski-Pasa, T.B., Barandi, C.R.M., Simões, C.M.O., Cytotoxicity and potential antiviral evaluation of violacein produced by Chromobacterium violaceum (2003) Mem Inst Oswaldo Cruz, 98, pp. 834-848Antonisamy, P., Ignacimuthu, S., Immunomodulatory, analgesic and antipyretic effects of violacein isolated from Chromobacterium violaceum (2010) Phytomedicine, 17, pp. 300-304Antonisamy, P., Kannan, P., Ignacimuthu, S., Anti-diarrhoeal and ulcer-protective effects of violacein isolated from Chromobacterium violaceum in Wistar rats (2009) Fundam Clin Pharmacol, 23, pp. 483-490Aoki, S., Nomura, T., Violacein-containing natural bactericides, their manufacture, and cosmetics containing them (1998), Jpn Kokai Tokkyo Koho JP 10139612Baek, S.H., Kang, H.S., Jang, I.H., Lee, J.S., Kim, S.Y., Baek, J.H., Kang, J.G., Ahn, J.M., (2007) Insecticide and Fungicide Containing Violacein, and Their Preparation Method, , Repub Korean Kongkae Taeho Kongbo KR 2007088150 ABromberg, N., Dreyfuss, J.L., Regatieri, C.V., Palladino, M.V., Durán, N., Nader, H.B., Haun, M., Justo, G.Z., Growth inhibition and proapoptotic activity of violacein in Ehrlich ascites tumor (2010) Chem Biol Interact, , doi:10.1016/j.cbi.2010.04.016Cao, W., Chen, W., Sun, S., Guo, P., Song, J., Tian, C., Cao, W., Tian, C., Investigating the antioxidant mechanism of violacein by density functional theory method. Investigating the antioxidant mechanism of violacein by density functional theory method (2007) J Mol Struct Theochem, 817, pp. 1-4Ftm, C., Justo, G.Z., Durán, N., Nogueira, P.A., Scp, L., The use of violacein in its free and encapsulated form in polymeric systems against malaria (2005), Brazilian Patent PIBr 056399-0De Azevedo Mbm, Alderete, J., Acs, L., Loh, W., Faljoni-Alario, A., Durán, N., Violacein/cyclodextrin inclusion complex by diffusion coefficient and circular dichroism measurements (2000) J Inclusion Phenom Mol Recognit Chem, 37, pp. 67-74De Azevedo, M.B.M., Alderete, J., Rodriguez, J.A., De Souza, A.O., Faljoni-Alario, A., Durán, N., Biological activities of new antitumoral of indole derivatives in a inclusion complex with cyclodextrin (2000) J Inclusion Phenom Mol Recognit Chem, 37, pp. 93-101De Azevedo, M.B.M., Justo, G.Z., Rettori, D., Rodriguez, J.A., Haun, M., Durán, N., Antioxidant activity of an inclusion complex of violacein and β-cyclodextrin (2000) Proc Intern Symp Control Rel Bioact Mater, 27, pp. 506-507De Azevedo, M.B.M., Melo, P.S., Almeida, A.B.A., Souza-Brito, A.R.M., Haun, M., Durán, N., Antiulcerogenic activity of violacein/ beta-cyclodextrin inclusion complexes and violacein (2000) Proc Intern Symp Control Rel Bioact Mater, 27, pp. 508-509De Carvalho, D.D., Costa, F.T.M., Duran, N., Haun, M., Cytotoxic activity of violacein in human colon cancer cells (2006) Toxicol in Vitro, 20, pp. 1514-1521De Moss, R.D., Violacein (1967) Antibiotics, 2, pp. 77-81De Moss, R.D., Happel, M.E., Nutritional requirements of Chromobacterium violaceum (1959) J Bacteriol, 77, pp. 137-141De Souza, A.O., Aily, D.C.G., Sato, D.N., Durán, N., In vitro violacein activity against Mycobacterium tuberculosis H37RA (1999) Rev Inst Adolfo Lutz, 58, pp. 59-62Durán, N., De Azevedo, M.M.M., Compound incorporated in a polymeric nanoparticle with pharmaceutical or a cosmetic, its preparation process and cosmetic or pharmaceutical formulations (2004), Brazilian Patent PIBr 0404306-5Durán, N., De Souza, A.O., Process of violacein application as antimycobacterial compound (2001), Brazilian Patent PIBr 0101346-7Durán, N., Faljoni-Alario, A., Bacterial chemistry-I. Studies of a potential phototherapeutic substance from Chromobacterium violaceum (1980) An Acad Bras Cienc, 52, pp. 297-302Durán, N., Haun, M., Trypanocide. State of art (1991) Mem Inst Oswaldo Cruz Suppl i, 86, pp. 29-30Durán, N., Haun, M., Production, obtention and purification process and antitumoral activity of 3-[1,2-dihydro-5-(5-hydroxy-1H-indol-3-yl)-2-oxo-3H-pyrrol-3-ylidene]-1,3- dihydro-2H-indol-2-one (1997), Brazilian Patent PIBr 9702918Durán, N., Menck, C.F.M., Chromobacterium violaceum: A Review of pharmacological and industrial perspectives (2001) Crit Rev Microbiol, 27, pp. 201-222Durán, N., Justo, G.Z., Melo, P.S., De Azevedo, M.B.M., Souza-Brito, A.R.M., Almeida, A.B.A., Haun, M., Evaluation of the antiulcerogenic activity of violacein and its modulation by the inclusion complexation with beta-cyclodextrin (2003) Can J Physiol Pharmacol, 81, pp. 387-396Durán, N., Erazo, S., Campos, V., Bacterial Chemistry-II. Antimicrobial photoproduct from pigment of Chromobacterium violaceum (1983) An Acad Bras Cienc, 55, pp. 231-234Durán, N., Campos, V., Riveros, R., Joyas, A., Pereira, M.F., Haun, M., Bacterial Chemistry-III. Preliminary studies on trypanosomal activities of Chromobacterium violaceum products (1989) An Acad Bras Cienc, 61, pp. 31-36Durán, N., De Azevedo, M.B.M., Alderete, J., Formulation process of cyclodextrin/violacein for using as antibacterial, antitumoral, antiviral and trypanocide (1998), Brazilian Patent PIBr 9801307Durán, N., Justo, G.Z., Melo, P.S., Martins Jr., D., Cordi, L., Violacein: Properties and biological activities (2007) Biotechnol Appl Biochem, 48, pp. 127-133Ferreira, C.V., Bos, C.L., Versreeg, H.H., Justo, G.Z., Durán, N., Peppelenbosch, M.P., Molecular mechanism of violaceinmediated human leukemia cell death (2004) Blood, 104, pp. 1459-1464Gimenez, I.F., Anazetti, M.C., Melo, P.S., Haun, M., De Azevedo, M.M.M., Durán, N., Alves, O.L., Cytotoxicity on V79 and HL60 cell lines by thiolated-beta-cyclodextrin- Au/violacein nanoparticles (2005) J Biomed Nanotechnol, 1, pp. 352-358Haun, M., Pereira, M.F., Hoffman, M.E., Riveros, R., Joyas, A., Campos, V., Durán, N., Bacterial Chemistry-VI: Biological activities and cyto-toxicity of 1, 3-dihydro-2H-Indol-2-one derivatives (1992) Biol Res, 25, pp. 21-25Kato, H., Hata, T., Shirata, A., Tsukamoto, T., Improvement of lightfastness of fibers or fiber products dyed with violacein or deoxyviolacein bluish purple dyes by treating the dyed fibers with thiourea (1999), Jpn Kokai Tokkyo Koho JP 11152687 AKidder, G.W., Stuart, C.A., Growth studies on ciliates. I. The role of bacteria in the growth and reproduction of Colpoda (1939) Physiol Zoo, 12, pp. 329-340Kodach, L.L., Bos, C.L., Durán, N., Peppelenbosh, M.P., Ferreira, C.V., Jch, H., Inhibition of Akt-mediated signal transduction in human colorectal cancer cells by violacein abrogates 5-fluorouracil chemoresistance (2006) Carcinogenesis, 27, pp. 508-516Konzen, M., De Marco, D., Cordova, C.A.S., Vieira, T.O., Antonio, R.V., Creczynski-Pasa, T.B., Antioxidant properties of violacein: Possible relation on its biological function (2006) Bioorg Med Chem, 14, pp. 8307-8313Laatsch, H., Thomson, R.H., Spectroscopic properties of violacein and related compound: Crystal structure of tetramethylviolacein (1984) J Chem Soc Perkin Trans, 2, pp. 1331-1339Leon, L.L., Miranda, C.C., De Souza, A.O., Durán, N., Antileishmanial activity of the violacein extracted from Chromobacterium violaceum (2001) J Antimicrobial Chemother, 48, pp. 449-450Lichstein, H.C., Van De Sand, V.F., Violacein an antibiotic pigment produced by Chromobacterium violaceum (1945) J Infect Dis, 76, pp. 47-51Scp, L., Blanco, Y.C., Justo, G.Z., Nogueira, P.A., Rodrigues, F.L.S., Goelnitz, U., Wunderlich, G., Costa, F.T.M., Violacein extracted from Chromobacterium violaceum inhibits Plasmodium growth in vitro and in vivo (2009) Antimocrob Agents Chemother, 53, pp. 2149-2152Lu, Y., Wang, L., Xue, Y., Zhang, C., Xing, X.H., Lou, K., Zhang, Z., Su, Z., Production of violet pigment by a newly isolated psychrotrophic bacterium from a glacier in Xinjiang (2009) China. Biochem Eng J, 43, pp. 135-141Martins, D., Costa, F.T.M., Brocchi, M., Durán, N., Evaluation of the antibacterial activity of poly-(D,L-lactide-co- glycolide) nanoparticles containing violacein (2010) J Nanopart Res, , submittedMartins, D., Frungillo, L., Anazzetti, M.C., Melo, P.S., Durán, N., Antitumoral activity of L-ascorbic acid-poly-D,L-(lactide-co-glycolide) nanoparticles containing violacein (2010) Intern J Nanomed, 5, pp. 77-85Matz, C., Deines, P., Boenigk, J., Arndt, H., Eberl, L., Kjellberg, S., Jurgens, K., Impact of violacein-producing bacteria on survival and feeding of bacterivorous nanoflagellates (2004) Appl Environ Microbiol, 70, pp. 1593-1599May, G., Brummer, B., Ott, H., (1991) Purification of the Antiviral Compound Violacein from Cultures of Chromobacterium, , Ger Offen DE 3935066Meiring, U., Lanzendoerfer, G., Riedel, H., Kallmayer, V., Viala, S., Mocigemba, N., Schaefer, J., (2007) Cosmetic Preparations Containing Violacein As Dye, pp. A1. , Ger Offen DE 102005051869Melo, P.S., Maria, S.S., Vidal, B.C., Haun, M., Duran, N., Violacein cytotoxicity and induction of apoptosis in V79 cells (2000) Vitro Cell Dev Biol Anim, 36, pp. 539-543Melo, P.S., Justo, G.Z., De Azevedo, M.B.M., Durán, N., Haun, M., Violacein and its beta-cyclodextrin complexes induce apoptosis and differentiation in HL60 cells (2003) Toxicology, 186, pp. 217-225Melo, P.S., De Azevedo, M.M.M., Frungillo, L., Anazetti, M.C., Marcato, P.D., Durán, N., Nanocytotoxicity: Violacein and violaceinloaded poly (D,L-lactide-co- glycolide) nanoparticles acting on human leukemic cells (2009) J Biomed Nanotechnol, 5, pp. 192-201Mendes, A.S., De Carvalho, J.E., Duarte, M.C.T., Durán, N., Bruns, R.E., Optimized production process of violacein and deoxyviolacein by Chromobacterium violaceum by factorial design and response surface methodology (2001), Brazilian Patent PIBr 0100199-0Mendes, A.S., De Carvalho, J.E., Duarte, M.C.T., Durán, N., Bruns, R.E., Factorial design and response surface optimization of crude violacein for Chromobacterium violaceum production (2001) Biotechnol Lett, 23, pp. 1963-1969Naito, S., Shiga, I., Yamaguchi, N., Antimicrobial activity of violet pigment produced by Janthinobacterium lividum isolated from Yudemen (1986) Nippon Shokuhin Kogyo Gakkaishi, 33, pp. 759-763Nakamura, Y., Sawada, T., Morita, Y., Tamiya, E., Isolation of a psychrotrophic bacterium from the organic residue of a water tank keeping rainbow trout and antibacterial effect of violet pigment produced from the strain (2002) Biochem Eng J, 12, pp. 79-86Nakamura, Y., Asada, C., Sawada, T., Production of antibacterial violet pigment by psychrotropic bacterium RT102 strain (2003) Biotechnol Bioprocess Eng, 8, pp. 37-40Nomura, T., Natural dye violacein manufacture with Chromobacterium and Janthiobacterium (1994), Jpn Kokai Tokkyo Koho JP 06253864 APantanella, F., Berlutti, F., Passariello, C., Sarli, S., Morea, C., Schippa, S., Violacein and biofilm production in Janthinobacterium lividum (2007) J Appl Microbiol, 102, pp. 992-999Philip, D.S., Sarovich, D.S., Pemberton, J.M., PPSY: A vector for the stable cloning and expression of streptomycete single gene phenotypes in Escherichia coli (2008) Plasmid, 60, pp. 53-58Philip, D.S., Sarovich, D.S., Pemberton, J.M., Complete sequence and analysis of the stability functions of pPSX, a vector that allows stable cloning and expression of Streptomycete genes in Escherichia coli K12 (2009) Plasmid, 62, pp. 39-43Rettori, D., Durán, N., Stationary tray bioreactor for the bacterial metabolite production (1997), Brazilian Patent PIBr 9702986-6Rettori, D., Rodriguez, R.A., Durán, N., Chromobacterium violaceum under oxidative stress (1998) Rev Farm Bioquim Univ São Paulo, 34 (SUPPL. 1), pp. 169-169Riveros, R., Haun, M., Durán, N., Bacterial Chemistry-V: Effect of growth conditions on production of violacein by Chromobacterium violaceum BB-78 strain (1989) Brazilian J Med Biol Res, 22, pp. 569-577Saraiva, V.S., Marshall, J.-C., Cools-Lartigue, J., Burnier Jr., M.N., Cytotoxic effects of violacein in human uveal melanoma cell lines (2004) Melanoma Res, 14, pp. 421-424Sarovich, D.S., Pemberton, J.M., PPSX: A novel vector for the cloning and heterologous expression of antitumor antibiotic gene clusters (2007) Plasmid, 57, pp. 306-313Shirata, A., Tsukamoto, T., Yasui, H., Kato, H., Hayasaka, S., Kojima, A., Production of bluish-purple pigments by Janthinobacterium lividum isolated from the raw silk and dyeing with them (1997) Nippon Sanshigaku Zasshi, 66, pp. 377-385Shirata, A., Tsukamoto, T., Kato, H., Hata, T., Yasui, T., Kojima, A., Blue purple pigment manufacture with microorganism and applications of the pigment (1998), Jpn Kokai Tokkyo Koho JP 10113169 AShirata, A., Tsukamoto, T., Yasui, H., Hata, T., Hayasaka, S., Kojima, A., Kato, H., Isolation of bacteria producing bluish-purple pigment and use for dyeing (2000) Japan Agric Res Quart, 34, pp. 131-140Sousa, A.O., Egito, L.C.M., Medeiros, S.R.B., Agnez-Lima, L.F., Study on genotoxicity of violacein (2005) Genet Mol Biol, 28 (SUPPL.), p. 183Takii, T., Hamasaki, S., Hirano, K., Abe, C., Onozaki, K., Simple fibroblast-based assay to test the pyrazinamide susceptibility of Mycobacterium tuberculosis (2005) Antimicrob Agents Chemother, 49, pp. 804-807Tan, T.L., Montforts, F.P., Meyer, D., (2002) Microbiological Method for the Biosynthesis of Natural Blue-violet Colorants Violacein and Desoxyviolacein, , PCT Int. Appl. WO 2002050299 A2Tobie, W.C., The pigment of Bacillus violaceus I. The production, extraction, and purification of violacein (1934) J Bacteriol, 29, pp. 223-227Wang, H., Jiang, P., Lu, Y., Ruan, Z., Jiang, R., Xing, X.H., Lou, K., Wei, D., Optimization of culture conditions for violacein production by a new strain of Duganella sp. B2 (2009) Biochem Eng J, 44, pp. 119-124Yada, S., Wang, Y., Zou, Y., Nagasaki, K., Hosokawa, K., Osaka, I., Arakawa, R., Enomoto, K., Isolation and characterization of two groups of novel marine bacteria producing violacein (2008) Marine Biotechnol, 10, pp. 128-132Yang, L.H., Xiong, H., Lee, O.O., Qi, S.H., Qian, P.Y., Effect of agitation on violacein production in Pseudoalteromonas luteoviolacea isolated from a marine sponge (2007) Lett Appl Microbiol, 44, pp. 625-63

    Evaluation Of The Antiulcerogenic Activity Of Violacein And Its Modulation By The Inclusion Complexation With β-cyclodextrin

    No full text
    The effects of β-cyclodextrin (βCD) inclusion complexation on the ability of violacein to prevent gastric ulceration in mice were studied. Violacein-βCD inclusion complexes were prepared in 1:1 and 1:2 molar ratios and analysed by differential scanning calorimetry and powder X-ray diffractometry. Violacein previously administered orally at 10 mg/kg significantly reduced indomethacin-induced gastric lesions, as well as 100 mg/kg of cimetidine (positive control). However, βCD complexation in both molar ratios significantly potentiated the protective action of violacein. In the HCl-ethanol-induced gastric ulcer model, violacein and the 1:2 inclusion complex (10 mg/kg, p.o.) inhibited gastric damage by almost 85%, whereas a 63% reduction was observed for the positive control, lansoprazole, at 30 mg/kg. In contrast, treatment with the 1:1 inclusion complex resulted in almost total disappearance of the antiulcer activity in this model. No significant changes in stress-induced gastric injury were found. In addition, the 1:2 inclusion complex improved the antilipoperoxidant activity of violacein in rat liver cells exposed to t-butyl hydroperoxide, whereas the 1:1 complex was less active than violacein. In summary, the 1:2 βCD inclusion complex has gastroprotective properties similar to or higher than that of violacein. An increase in mucosal defensive mechanisms and protection against peroxidative damage might be involved.814387396Ahmed, M.O., El-Gibaly, I., Ahmed, S.M., Effect of cyclodextrins on the physicochemical properties and antimycotic activity of clotrimazole (1998) Int. J. Pharm., 171, pp. 111-121Al-Shabanah, O.A., Raza, M., Al-Harbi, M.M., Al-Bekairi, A.M., Al-Gharably, N.M., Qureshi, S., Effect of ninhydrin on the biochemical and histopathological changes induced by ethanol in gastric mucosa of rats (2000) Life Sci., 67, pp. 559-566Atay, S., Tarnawski, A.S., Dubois, A., Eicosanoids and the stomach (2000) Prostaglandins Other Lipid Mediat, 61, pp. 105-124Bibby, D.C., Davies, N.M., Tucker, I.G., Mechanisms by which cyclodextrins modify drug release from polymeric drug delivery systems (2000) Int. J. Pharm., 197, pp. 1-11Blandizzi, C., Natale, G., Gherardi, G., Lazzeri, G., Marveggio, C., Colucci, R., Carignani, D., Del Tacca, M., Acid-independent gastroprotective effects of lansoprazole in experimental mucosal injury (1999) Dig. Dis. Sci., 44, pp. 2039-2050Brunton, L.L., Fármacos para controle da acidez gástrica e tratamento de úlceras pepticas (1996) As Bases Farmacológicas da Terapêutica. 9th Ed., pp. 662-673. , Edited by J.G. Hardman and L.E. Limbird. McGraw-Hill, México City, MéxicoChatjigakis, A.K., Donzé, C., Coleman, A.W., Solubility behavior of β-cyclodextrin in water/cosolvent mixtures (1992) Anal. Chem., 64, pp. 1632-1634Connors, K.A., The stability of cyclodextrin complexes in solution (1997) Chem. Rev., 97, pp. 1325-1358Croft, K.D., The chemistry and biological effects of flavonoids and phenolic acids (1998) Ann. N.Y. Acad. Sci., 854, pp. 435-442Das, D., Bandyopadhyay, D., Bhattacharjee, M., Banerjee, R., Hydroxyl radical is the major causative factor in stress-induced gastric ulceration (1997) Free Rad. Biol. Med., 23, pp. 8-18De Azevedo, M.B.M., Alderete, J.B., Lino, A.C.S., Loh, W., Faljoni-Alario, A., Durán, N., Violacein/β-cyclodextrin inclusion complex formation studied by measurements of diffusion coefficient and circular dichroism (2000) J. Incl. Phenom. Mol. Recognit. Chem., 37, pp. 67-74De Azevedo, M.B.M., Alderete, J., Rodriguez, J.A., Souza, A.O., Rettori, D., Torsoni, M.A., Faljoni-Alario, A., Durán, N., Biological activities of violacein a new antitumoral indole derivative in an inclusion complex with β-cyclodextrin (2000) J. Incl. Phenom. Mol. Recognit. Chem., 37, pp. 93-101Desai, J.K., Parmar, N.S., Gastric and duodenal antiulcer activity of sulpiride, a dopamine D 2 receptor antagonist, in rats (1994) Agents Action, 42, pp. 149-152Durán, N., Menck, C.F.M., Chromobacterium violaceum: A review of pharmacological and industrial perspectives (2001) Crit. Rev. Microbiol., 27, pp. 201-222Evangelista, S., Meli, A., Influence of antioxidants and radical scavengers on ethanol-induced gastric ulcers in the rat (1985) Gen. Pharmacol., 16, pp. 285-286Florence, A.T., Jani, P.U., Novel oral drug formulations. Their potential in modulating adverse effects (1994) Drug Saf., 10, pp. 233-266Glavin, G.B., Szabo, S., Experimental gastric mucosal injury: Laboratory models reveal mechanisms of pathogenesis and new therapeutic strategies (1992) FASEB J., 6, pp. 825-831Goa, K.L., Monk, J.P., Enprostil: A preliminary review of its pharmacodynamics and pharmacokinetic properties and therapeutic efficacy in the treatment of peptic ulcer disease (1987) Drugs, 3, pp. 539-559Gracioso, J.S., Vilegas, W., Hiruma-Lima, C.A., Souza Brito, A.R.M., Effects of tea from Turnera ulmifolia L. on mouse gastric mucosa support the Turneraceae as a new source of antiulcerogenic drugs (2002) Biol. Pharm. Bull., 25, pp. 487-491Gremse, D.A., Lansoprazole: Pharmacokinetics, pharmacodynamics and clinical uses (2001) Expert. Opin. Pharmacother., 2, pp. 1663-1670Grover, J.K., Vats, V., Proton pump inhibitors (1999) Trop. Gastroenterol., 20, pp. 16-28Guguen-Guillouzo, C., Guillouzo, A., (1986) Isolated and Culture Hepatocytes, pp. 1-12. , Edited by A. Guillouzo and C. Guguen-Guillouzo. Les Editions INSERM, Paris, and John Libbey Eurotext, LondonHalliwell, B., Gutteridge, J.M.C., (1999) Free Radicals in Biology and Medicine. 3rd Ed., pp. 1-936. , Clarendon Press, Oxford, U.KHayden, L.J., Thomas, G., West, G.B., Inhibitors of gastric lesions in the rat (1978) J. Pharmacol., 30, pp. 244-246Hiruma-Lima, C.A., Gracioso, J.S., Nunes, D.S., Souza Brito, A.R.M., Effects of an essential oil from the bark of Croton cajucara Benth. on experimental gastric ulcer models in rats and mice (1999) J. Pharm. Pharmacol., 51, pp. 341-346Hiruma-Lima, C.A., Gracioso, J.S., Rodriguez, J.A., Haun, M., Nunes, D.S., Souza Brito, A.R.M., Gastroprotective effect essential oil from Croton cajucara Benth. (Euphorbiaceae) (2000) J. Ethnopharmacol., 69, pp. 229-234Hiruma-Lima, C.A., Gracioso, J.S., Toma, W., Almeida, A.B.A., Paula, A.C.B., Brasil, D.S.B., Müller, A.H., Souza Brito, A.R.M., Gastroprotective effect of aparisthman, a diterpene isolated from Aparisthmium cordatum, on experimental gastric ulcer models in rats and mice (2001) Phytomedicine, 8, pp. 94-100Hiruma-Lima, C.A., Toma, W., Gracioso, J.S., Almeida, A.B.A., Batista, L.M., Magri, L., Paula, A.C.B., Souza Brito, A.R.M., Natural trans-crotonin: The antiulcerogenic effect of another diterpene isolated from the bark of Croton cajucara Benth (2002) Biol. Pharm. Bull., 25, pp. 452-456Horáková, L., Stolc, S., Antioxidant and pharmacodynamic effects of pyridoindole stobadine (1998) Gen. Pharmac., 30, pp. 627-638Ito, M., Inaguma, K., Suzuki, Y., Segami, T., Suzuki, Y., Healing-promoting action of zinc-cimetidine complex on acetic acid-induced gastric ulcers in rats (1995) Jpn. J. Pharmacol., 68, pp. 287-295Ito, M., Suzuki, Y., Ishihara, M., Suzuki, Y., Anti-ulcer effects of antioxidants: Effect of probucol (1998) Eur. J. Pharmacol., 354, pp. 189-196Joyeux, M., Rolland, A., Fleurentin, J., Mortier, F., Dorfman, P., Tert-butyl hydroperoxide-induced injury in isolated rat hepatocytes: A model for studying anti-hepatotoxic crude drugs (1990) Planta Med., 56, pp. 171-174Kaneko, T., Baba, N., Protective effect of flavonoids on endothelial cells against linoleic acid hydroperoxide-induced toxicity (1999) Biosci. Biotechnol. Biochem., 6, pp. 323-328Kovacs, T.O., Campbell, D., Haber, M., Rose, P., Jennings, D.E., Richter, J., Double-blind comparison of lansoprazole 15 mg, lansoprazole 30 mg, and placebo in maintenance of healed gastric ulcer (1998) Dig. Dis. Sci., 43, pp. 779-785Levine, R.J., A method for rapid production of stress ulcer in rats (1971) Peptic Ulcer, pp. 92-97. , Edited by C.J. Pfeiffer. Munksgaard, CopenhagenLewis, D.A., Hanson, P.J., (1991) Progress in Medicinal Chemistry, pp. 201-231. , Edited by G.P. Ellis and G.B. West. Elsevier Science Publishers, AmsterdamLien, E.J., Ren, S., Bui, H.H., Wang, R., Quantitative structure-activity relationship analysis of phenolic antioxidants (1999) Free Rad. Biol. Med., 26, pp. 285-294Ligumsky, M., Sestieri, M., Okon, M., Ginsburg, I., Antioxidants inhibit ethanol-induced gastric injury in the rat. Role of manganese, glycine, and carotene (1995) Scand. J. Gastroenterol., 30, pp. 854-860Lissi, E.A., Faure, M., Montoya, N., Videla, L.A., Reactivity of indole derivatives towards oxygenated radicals (1991) Free Radical Biol. Commun., 15, pp. 211-222Loftsson, T., Brewster, M.E., Pharmaceutical applications of cyclodextrins. 1. Drug solubilization and stabilization (1996) J. Pharm. Sciences, 85, pp. 1017-1025Malterud, K.E., Diep, O.H., Sund, R.B., C-methylated dihydrochalcones from Myrica gale L: Effects as antioxidants and as scavengers of 1,1-diphenyl-2-picrylhydrazyl (1996) Pharmacol. Toxicol., 78, pp. 111-116McGuigan, J.E., Peptic ulcer and gastritis (1991) Harrison's Principles of Internal Medicine. 12th Ed., pp. 1229-1243. , Edited by J.D. Wilson, E. Braun Wald, K.J. Isselbacher, R.G. Petersdorf, J.B. Martin, A.S. Fauchi, and R.K. Root. McGraw-Hill, New YorkMirossay, L., Kohút, A., Antiulcerogenic effect of pentacaine, chlorpromazine and stobadine on ethanol- and indomethacin-induced stomach lesions in rats (1991) Physiol. Pharmacol., 40, pp. 441-444Mirossay, L., Kohút, A., Effect of stobadine on indomethacin- and ethanol-induced stomach lesions and gastric secretion (1991) Physiol. Phramacol., 40, pp. 437-440Mizui, T., Doteuchi, M., Effect of polyamines on acidified ethanol-induced gastric lesion in rats (1983) Jpn. Pharmacol., 33, pp. 939-945Mizui, T., Doteuchi, M., Lipid peroxidation: A possible role in gastric damage induced by ethanol in rats (1986) Life Sci., 38, pp. 2163-2167Mizui, T., Sato, H., Hirose, F., Doteuchi, M., Effect of antioxidants drugs on gastric damage induced by ethanol in rats (1987) Life Sci., 41, pp. 755-763Mura, P., Faucci, M.T., Parrini, P.L., Furlanetto, S., Pinzauti, S., Influence of the preparation method on the physicochemical properties of ketoprofen-cyclodextrin binary systems (1999) Int. J. Pharm., 179, pp. 117-128Olfert, E.D., Cross, B.M., McWilliam, A.A., (1993) Guide to the Care and Use of Experimental Animals, 1, pp. 1-213. , Canadian Council on Animal Care. OttawaPhull, P.S., Green, C.J., Jacyna, M.R., A radical view of the stomach: The role of oxygen-derived free radicals and antioxidants in gastroduodenal disease (1995) Eur. J. Gastroenterol. Hepatol., 7, pp. 265-274Ren, X., Xue, Y., Liu, J., Zhang, K., Zheng, J., Luo, G., Guo, C., Shen, J., A novel cyclodextrin-derived tellurium compound with glutathione peroxidase (2002) Chembiochem., 3, pp. 356-363Rettori, R., Durán, N., Production, extraction and purification of violacein: An antibiotic pigment produced by Chromobacterium violaceum (1998) World J. Microbiol. Biotechnol., 14, pp. 685-688Robert, A., Nezamis, J.E., Lancaster, C., Hanchar, A.J., Cytoprotection by prostaglandins in rats. Prevention of gastric necroses produced by alcohol, HCl, NaOH, hypotonic and thermal injury (1979) Gastroenterology, 77, pp. 433-443Robert, A., Lancaster, C., Davis, J.P., Field, J.P., Nezamis, J.E., Distinction between antiulcer effect and cytoprotection (1984) Scand. J. Gastroenterol., 101, pp. 69-73Scheiman, J., Agents used in the prevention and treatment of nonsteroidal anti-inflammatory drug-associated symptoms and ulcers (1998) Am. J. Med., 105, pp. 32S-38SShii, D., Inaguma, K., Ito, M., Suzuki, Y., Role of oxygen radicals in healing process of acetic acid-induced ulcers in rats with limited food-intake-time and effects of oxygen radical scavengers on the healing (1992) Exp. Ulcer, 19, pp. 105-109Souza Brito, A.R.M., How to study the pharmacology of medicinal plants in underdeveloped countries (1996) J. Ethnopharmacol., 54, pp. 131-138Souza Brito, A.R.M., Rodriguez, J.A., Hiruma-Lima, C.A., Haun, M., Nunes, D.S., Anti-ulcerogenic activity of dehydrocrotonin, a diterpene isolated from Croton cajucara Benth (1998) Planta Med., 64, pp. 126-129Stella, V.J., Rajewski, R.A., Cyclodextrins: Their future in drug formulation and delivery (1997) Pharm. Res., 14, pp. 556-567Stella, V.J., Rao, V.M., Zannou, E.A., Zia, V.V., Mechanisms of drug release from cyclodextrin complexes (1999) Adv. Drug Deliv. Rev., 36, pp. 3-16Szejtli, J., Medicinal applications of cyclodextrins (1994) Med. Res. Rev., 14, pp. 353-386Szejtli, J., Bolla-Pusztai, E., Tardy-Lengyel, M., Szabo, P., Preparation, properties and biological activity of β-cyclodextrin inclusion complex of menadione (1983) Pharmazie, 38, pp. 189-193Szelenyi, I., Thiemer, K., Distention ulcer as a model for testing of drugs for ulcerogenic side effects (1978) Arch. Toxicol., 41, pp. 99-105Terano, A., Hiraishi, H., Ota, S., Shiga, J., Sugimoto, T., Role of superoxide and hydroxyl radicals in rat gastric mucosal injury induced by ethanol (1989) Gastroenterol. Jpn., 24, pp. 488-493Thompson, D.O., Cyclodextrins enabling excipients: Their present and future use in pharmaceuticals (1997) Crit. Rev. Ther. Drug Carrier Syst., 14, pp. 1-104Toma, W., Gracioso, J.S., Andrade, F.D.P., Hiruma-Lima, C.A., Vilegas, W., Souza Brito, A.R.M., Antiulcerogenic activity of four extracts obtained from the bark wood of Quassia amara L. (Simaroubaceae) (2002) Biol. Pharm. Bull., 25, pp. 1151-1155Uekama, K., Hirayama, F., Irie, T., Cyclodextrin drug carrier systems (1998) Chem. Rev., 98, pp. 2045-2076Vane, J.R., Botting, R.M., New insights into the mode of action of anti-inflammatory drugs (1995) Inflamm. Res., 44, pp. 1-10Vora, J., Boroujerdi, M., Enhanced aqueous solubility of phenolic antioxidants using modified beta-cyclodextrin (1995) Drug Develop. Ind. Pharm., 21, pp. 495-502Wallace, J.L., Nonsteroidal anti-inflammatory drugs and the gastrointestinal tract. Mechanisms of protection and healing: Current knowledge and future directions (2001) Am. J. Med., 110, pp. 19S-23SWallace, J.L., Granger, D.N., The cellular and molecular basis of gastric mucosal defense (1996) FASEB J., 10, pp. 731-740Wallace, J.L., Ma, L., Inflammatory mediators in gastrointestinal defense and injury (2001) Exp. Biol. Med., 226, pp. 1003-1115Yoshikawa, T., Naito, Y., Kishi, A., Tomii, Y., Kaneko, T., Iinuma, S., Ichikawa, H., Kondo, M., Role of active oxygen lipid peroxidation and antioxidants in the pathogenesis of gastric mucosal injury induced by indomethacin in rats (1993) Gut, 34, pp. 732-737Zimmermann, A.E., Katona, B.G., Lansoprazole: A comprehensive review (1997) Pharmacotherapy, 17, pp. 308-32

    Heterozygosis For Cyp21a2 Mutation Considered As 21-hydroxylase Deficiency In Neonatal Screening

    No full text
    Steroid 21-hydroxylase deficiency (21-OHD) accounts for more than 90% of congenital adrenal hyperplasia. CAH newborn screening, in general, is based on 17-hydroxyprogesterone dosage (17-OHP), however it is complicated by the fact that healthy preterm infants have high levels of 17-OHP resulting in false positive cases. We report on molecular features of a boy born pre-term (GA = 30 weeks; weight = 1,390 g) with elevated levels of 17-OHP (91.2 nmol/L, normal < 40) upon neonatal screening who was treated as having CAH up to the age of 8 months. He was brought to us for molecular diagnosis. Medication was gradually suspended and serum 17-OHP dosages mantained normal. The p.V281 L mutation was found in compound heterozygous status with a group of nucleotide alterations located at the 3′ end intron 4 and 5′ end exon 5 corresponding to the splice site acceptor region. Molecular studies continued in order to exclude the possibility of a nonclassical 21-OHD form. The group of three nucleotide changes was demonstrated to be a normal variant since they failed to interfere with the normal splicing process upon minigene studies.52813881392White, P.C., Speiser, P.W., Congenital adrenal hyperplasia due to 21-hydroxylase deficiency (2000) Endocr Rev, 21, pp. 245-291Riepe, F.G., Sippell, W.G., Recent advances in diagnosis, treatment, and outcome of congenital adrenal hyperplasia due to 21-hydroxylase deficiency (2007) Rev Endocr Metab Disord, 8 (4), pp. 349-363New, M., Lorenzen, F., Lerner, A.J., Kohn, B., Oberfield, S.E., Pollack, M.S., Genotyping steroid 21-hydroxylase deficiency: Hormonal reference data (1983) J Clin Endocrinol Metab, 57, pp. 320-326Azziz, R., Hincapie, L.A., Knochenhauer, E.S., Dewailly, D., Fox, L., Boots, L.R., Screening for 21-hydroxylase-deficient nonclassic adrenal hyperplasia among hyperandrogenic women (1999) Fertil Steril, 72, pp. 915-925Bachega, T.A., Brenlha, E.M., Billerbeck, A.E., Marcondes, J.A., Madureira, G., Arnhold, I.J., Mendonca, B.B., Variable ACTH-stimulated 17-hydroxyprogesterone values in 21-hydroxylase deficiency carriers are not related to the different CYP21 gene mutations (2002) J Clin Endocrinol Metab, 87 (2), pp. 786-790Pang, S., Hotchkiss, J., Drash, A.L., Levine, L.S., New, M., Microfilter paper method for 17-hydroxyprogesterone radioimmunoassay: Its application for rapid screening for congenital adrenal hyperplasia (1977) J Clin Endocrinol Metab, 45, pp. 1003-1008Valentino, R., Tommaselli, A.P., Rossi, R., Lombardi, G., Varrone, S., A pilot study for neonatal screening of congenital adrenal hyperplasia due to 21-hydroxylase and 11-hydroxylase deficiency in Campania region (1990) J Endocrinol lnvest, 13, pp. 221-225Janzen, N., Peter, M., Sander, S., Steuerwald, U., Terhardt, M., Holtkamp, U., Sander, J., Newborn screening for congenital adrenal hyperplasia: Additional steroid profile using liquid chromatography-tandem mass spectrometry (2007) J Clin Endocrinol Metab, 92 (7), pp. 2581-2589Pang, S., Murphey, W., Levine, L.S., Lorenzen, F., Levy, D., Lerner, A.J., Rondanini, G.F., New, M., A pilot newborn screening for congenital adrenal hyperplasia in Alaska (1982) J Clin Endocrinol Metab, 55, pp. 413-420Nordenström, A., Wedell, A., Hagenfeldt, L., Marcus, C., Larsson, A., Neonatal screening for congenital adrenal hyperplasia: 17-hydroxyprogesterone levels and CYP21 genotypes in preterm infants (2001) Pediatrics, 108 (4), pp. E68Cardoso, C.B., Fonseca, A.A., Oliveira Mde, F., Pereira, B.B., Guimarães, M.M., Congenital adrenal hyperplasia newborn screening: Rio de Janeiro experience. Arq Bras Endocrinol (2005) Metabol, 49 (1), pp. 112-119Silveira, EL, D.S., Bachega, T.A., van der, Linden Nader, I., Gross, J.L., Elnecave, R.H., The actual incidence of congenital adrenal hyperplasia in Brazil may not be as high as inferred-an estimate based on a public neonatal screening program in the state of Goiás (2008) J Pediatr Endocrinol Metab, 21 (5), pp. 455-460Sambrook, J., Fritsch, E.F., Maniatis, T.E., (1989) Molecular Cloning, a Laboratory Manual, , New York: Cold Spring HarborAraujo, M., Sanches, M.R., Suzuki, L.A., Guerra-Jr, G., Farah, S.B., De Mello, M.P., Molecular analysis of CYP21 and C4 genes in Brazilian families with the classical form of steroid 21-hydroxylase deficiency (1996) Braz J Med Biol Res, 29, pp. 1-13Wilson, R.C., Wei, J.Q., Cheng, K.C., Mercado, A.B., New, M., Rapid deoxyribonucleic acid analysis by allele-specific polymerase chain reaction for detection of mutations in the steroid 21-hydroxylase gene (1995) J Clin Endocrinol Metab, 80 (5), pp. 1635-1640Lau, I.F., Soardi, F.C., Lemos-Marini, S.H., Guerra-Jr, G., Baptista, M.T., De Mello, M.P., H28+C insertion in the CYP21 gene: A novel frameshift mutation in Brazilian patient with the classical form of 21-hydroxylase deficiency (2001) J Clin Endocrinol Metab, 86, pp. 5877-5880Higashi, Y., Yoshioka, H., Yamane, M., Gotoh, O., Fujii-Kuriyama, Y., Complete nucleotide sequence of two steroid 21-hydroxylase genes tandemly arranged in human chromosome: A pseudogene and a genuine gene (1986) Proc Natl Acad Sci USA, 83, pp. 2841-2845Wilson, R.C., Nimkarn, S., Dumic, M., Obeid, J., Azar, M.R., Najmabadi, H., Ethnic-specific distribution of mutations in 716 patients with congenital adrenal hyperplasia owing to 21-hydroxylase deficiency (2007) Mol Genet Metab, 90 (4), pp. 414-421Tusie-Luna, M.T., Traktman, P., White, P.C., Determination of functional effects of mutations in the steroid 21-hydroxylase gene (CYP21) using recombinant vaccinia virus (1990) J Biol Chem, 265 (34), pp. 20916-20922Higashi, Y., Hiromasa, T., Tanae, A., Miki, T., Nakura, J., Kondo, T., Effects of individual mutations in the P-450(C21) pseudogene on the P-450(C21) activity and their distribution in the patient genomes of congenital steroid 21-hydroxylase deficiency (1991) J Biochem, 109 (4), pp. 638-644Bachega, T.A., Billerbeck, A.E., Madureira, G., Marcondes, J.A., Longui, C.A., Leite, M.V., Molecular genotyping in Brazilian patients with the classical and nonclassical forms of 21-hydroxylase deficiency (1998) J Clin Endocrinol Metab, 83 (12), pp. 4416-441

    Novel Mutations In Cyp11b1 Gene Leading To 11β-hydroxylase Deficiency In Brazilian Patients

    No full text
    Background: Deficiency of 11β-hydroxylase results in the impairment of the last step of cortisol synthesis. In females, the phenotype of this disorder includes different degrees of genital ambiguity and arterial hypertension. Mutations in the CYP11B1 gene are responsible for this disease. Objective: The objective of the study was to screen the CYP11B1 gene for mutations in two unrelated Brazilian females with congenital adrenal hyperplasia due to 11β-hydroxylase deficiency. Design: The coding and intron-exon junction regions of CYP11B1 were totally sequenced. A putative splice mutation was further investigated by minigene transcription. Results: We report two novel CYP11B1 mutations in these Brazilian patients. An Arabian Lebanese descendent female was found to be homozygous for a cytosine insertion at the beginning of exon 8, changing the 404 arginine to proline. It alters the open reading frame, creating a putative truncated protein at 421 residue, which eliminates the domain necessary for the association of heme prosthetic group. A severely virilized female was homozygous for the g.2791G>A transition in the last position of exon 4. This nucleotide is also part of 5′ intron 4 donor splice site consensus sequence. Minigene experiments demonstrated that g.2791G>A activated an alternative splice site within exon 4, leading to a 45-bp deletion in the transcript. The putative translation of such modified mRNA indicates a truncated protein at residue 280. Conclusions: We describe two novel mutations, g.4671-4672insC and g.2791G>A, that drastically affects normal protein structure. These mutations abolish normal enzyme activity, leading to a severe phenotype of congenital adrenal hyperplasia due to 11β-hydroxylase deficiency. Copyright © 2009 by The Endocrine Society.94934813485White, P.C., Curnow, K.M., Pascoe, L., Disorders of steroid 11β- hydroxylase isoenzymes (1994) Endocr Rev, 15, pp. 421-438White, P.C., Speiser, P.W., Congenital adrenal hyperplasia due to 21-hydroxylase deficiency (2000) Endocrine Reviews, 21 (3), pp. 245-291. , DOI 10.1210/er.21.3.245Mornet, E., Dupont, J., Vitek, A., White, P.C., Characterization of two genes encoding human steroid 11β-hydroxylase (P-450(11β)) (1989) Journal of Biological Chemistry, 264 (35), pp. 20961-20967Spoudeas, H.A., Slater, J.D., Rumsby, G., Honour, J.W., Brook, C.G., Deoxycorticosterone, 11β-hydroxylase and the adrenal cortex (1993) Clin Endocrinol, 39, pp. 245-251. , OxfHague, W., Honour, J., Malignant hypertension in congenital adrenal hyperplasia due to 11β-hydroxylase deficiency (1983) Clin Endocrinol, 18, pp. 505-510. , OxfKrawczak, M., Cooper, D.N., The human gene mutation database (1997) Trends Genet, 13, pp. 121-122Chabre, O., Portrat-Doyen, S., Vivier, J., Morel, Y., Defaye, G., Two novel mutations in splice donor sites of CYP11B1 in congenital adrenal hyperplasia due to 11β-hydroxylase deficiency (2000) Endocrine Res, 26, pp. 797-801Curnow, K.M., Slutsker, L., Vitek, J., Cole, T., Speiser, P.W., New, M.I., White, P.C., Pascoe, L., Mutations in the CYP11B1 gene causing congenital adrenal hyperplasia and hypertension cluster in exons 6, 7, and 8 (1993) Proc Natl Acad Sci USA, 90, pp. 4552-4556Skinner, C.A., Rumsby, G., Honour, J.W., Single strand conformation polymorphism (SSCP) analysis for the detection of mutations in the CYP11B1 gene (1996) Journal of Clinical Endocrinology and Metabolism, 81 (6), pp. 2389-2393. , DOI 10.1210/jc.81.6.2389De Carvalho, C.E., Castro, M., Moreira, A.C., De Mello, M.P., CYP11B1 mutation and polymorphisms in congenital adrenal hyperplasia due to 11β-hydroxylase deficiency (1999) J Endocr Genet, 1, pp. 79-86Moreira, A.C., Elias, L.L.K., Pituitary-adrenal responses to corticotropin- releasing hormone in different degrees of adrenal 21-hydroxylase deficiency (1992) J Clin Endocrinol Metab, 74, pp. 198-203Mermejo, L.M., Elias, L.L.K., Marui, S., Moreira, A.C., Mendonca, B.B., De Castro, M., Refining hormonal diagnosis of type II 3β-hydroxysteroid dehydrogenase deficiency in patients with premature pubarche and hirsutism based on HSD3B2 genotyping (2005) Journal of Clinical Endocrinology and Metabolism, 90 (3), pp. 1287-1293. , DOI 10.1210/jc.2004-1552De-Araujo, M., Sanches, M.R., Suzuki, L.A., Guerra Jr., G., Farah, S.B., De-Mello, M.P., Molecular analysis of CYP21 and C4 genes in Brazilian families with the classical form of steroid 21-hydroxylase deficiency (1996) Brazilian Journal of Medical and Biological Research, 29 (1), pp. 1-13Soardi, F.C., Lemos-Marini, S.H.V., Coeli, F.B., Maturana, V.G., Silva, M.D., Bernardi, R.D., Justo, G.Z., De Mello, M.P., Heterozygosis for CYP21A2 mutation considered as 21-hydroxylase deficiency in neonatal screening (2008) Arq Bras Endocrinol Metab, 52, pp. 1388-1392White, P.C., Slutsker, L., Haplotype analysis of CYP11B2 (1995) Endocr Res, 21, pp. 437-442Ravichandran, K.G., Boddupalli, S.S., Hasemann, C.A., Peterson, J.A., Deisenhofer, J., Crystal structure of hemoprotein domain of P450BM-3, a prototype for microsomal P450's (1993) Science, 261 (5122), pp. 731-736Roumen, L., Sanders, M.P.A., Pieterse, K., Hilbers, P.A.J., Plate, R., Custers, E., De Gooyer, M., Hermans, J.J.R., Construction of 3D models of the CYP11B family as a tool to predict ligand binding characteristics (2007) Journal of Computer-Aided Molecular Design, 21 (8), pp. 455-471. , DOI 10.1007/s10822-007-9128-9Mount, S.M., A catalogue of splice junction sequences (1982) Nucleic Acids Res, 10, pp. 459-472Buratti, E., Chivers, M., Královicová, J., Romano, M., Baralle, M., Krainer, A.R., Vorechovsky, I., Aberrant 5′ splice sites in human disease genes: Mutation pattern, nucleotide structure and comparison of computational tools that predict their utilization (2007) Nucleic Acids Res, 35, pp. 4250-426

    Growth Inhibitory Activity Of A Novel Lectin From Cliona Varians Against K562 Human Erythroleukemia Cells

    No full text
    Purpose: In this study, the antitumoral potential of a novel lectin (CvL) purified from the marine sponge Cliona varians was studied in different cancer cell lines. Methods: CvL cytotoxicity was evaluated in mammalian tumor cells and in normal human peripheral blood lymphocytes by the MTT assay using the same range of concentrations (1-150 μg ml-1). The mechanisms involved in K562 cell death were investigated by confocal fluorescence microscopy, flow cytometry and immunoblot. Results: CvL inhibited the growth of human leukemia cells, with IC50 values of 70 and 100 μg ml-1 for K562 and JURKAT cells, respectively, but it was ineffective on blood lymphocytes and solid tumor cell lines. K562 cell death occurred 72 h after exposure to the lectin and with signs of apoptosis, as analyzed by DAPI and annexin V/PI staining. Investigation of the possible mediators of this process showed that cell death occurred via a caspase-independent pathway. Confocal fluorescence microscopy indicated a pivotal role for the lysosomal protease cathepsin B in mediating cell death. Accordingly, pre-incubation of K562 cells with the cathepsin inhibitor l-trans-epoxysuccinyl-l-leucylamido-(4-guanidino)butane (E-64) abolished CvL cytotoxic effect. Furthermore, we found upregulation of tumor necrosis factor receptor 1 (TNFR1) and down-modulation of p65 subunit of nuclear factor kappa B (NFκB) expression in CvL-treated cells. These effects were accompanied by increased levels of p21 and reduced expression of pRb, suggesting that CvL can induce cell cycle arrest. Conclusions: Collectively, these findings indicate an antileukemic effect for CvL and suggest that cathepsin B acts as a death mediator in CvL-induced cytotoxicity possibly in an uncharacterized connection with the membrane death receptor pathway. © 2008 Springer-Verlag.63610231033Broker, L.E., Kruyt, F.A., Giaccone, G., Cell death independent of caspases: A review (2005) Clin Cancer Res, 11, pp. 3155-3162Fehrenbacher, N., Jaattela, M., Lysosomes as targets for cancer therapy (2005) Cancer Res, 65, pp. 2993-2995Guicciardi, M.E., Leist, M., Gores, G.J., Lysosomes in cell death (2004) Oncogene, 23, pp. 2881-2890Sharon, N., Carbohydrates as recognition determinants in phagocytosis and in lectin-mediated killing of target cells (1984) Biol Cell, 51 (2), pp. 239-245Abdullaev, F.I., De Mejia, E.G., Antitumor effect of plant lectins (1997) Nat Toxins, 5, pp. 157-163Atta, A.M., Barralnetto, M., Peixinho, S., Sousaatta, M.L.B., Isolation and functional-characterization of a mitogenic lectin from the marine sponge Cinachyrella alloclada (1989) Braz J Med Biol Res, 22, pp. 379-385Hajto, T., Hostanska, K., Berki, T., Palinkas, L., Boldizsar, F., Nemeth, P., Oncopharmacological perspectives of a plant lectin (Viscum album agglutinin-I): Overview of recent results from in vitro experiments and in vivo animal models, and their possible relevance for clinical applications (2005) Evid Based Complement Alternat Med, 2, pp. 59-67Wang, H., Ng, T.B., Ooi, V.E., Liu, W.K., Effects of lectins with different carbohydrate-binding specificities on hepatoma, choriocarcinoma, melanoma and osteosarcoma cell lines (2000) Int J Biochem Cell Biol, 32, pp. 365-372Arenas, M.I., Romo, E., De Gaspar, I., De Bethencourt, F.R., Sanchez-Chapado, M., Fraile, B., Paniagua, R., A lectin histochemistry comparative study in human normal prostate, benign prostatic hyperplasia, and prostatic carcinoma (1999) Glycoconj J, 16, pp. 375-382Gorelik, E., Galili, U., Raz, A., On the role of cell surface carbohydrates and their binding proteins (lectins) in tumor metastasis (2001) Cancer Metastasis Rev, 20, pp. 245-277Opric, M.M., Poznanovic, S., Kljajic, Z., Sladic, D., Pupic, G., Perunovic, B., Gasic, M.J., Labelling of breast carcinoma, thyroid carcinoma and melanoma with manno- and galacto-specific lectins from marine invertebrates (1996) Eur J Histochem, 40, pp. 211-218Bantel, H., Engels, I.H., Voelter, W., Schulze-Osthoff, K., Wesselborg, S., Mistletoe lectin activates caspase-8/FLICE independently of death receptor signaling and enhances anticancer drug-induced apoptosis (1999) Cancer Res, 59, pp. 2083-2090Miyoshi, N., Koyama, Y., Katsuno, Y., Hayakawa, S., Mita, T., Ohta, T., Kaji, K., Isemura, M., Apoptosis induction associated with cell cycle dysregulation by rice bran agglutinin (2001) J Biochem (Tokyo), 130, pp. 799-805Ohba, H., Moriwaki, S., Bakalova, R., Yasuda, S., Yamasaki, N., Plant-derived abrin-a induces apoptosis in cultured leukemic cell lines by different mechanisms (2004) Toxicol Appl Pharmacol, 195, pp. 182-193Park, R., Kim, M.S., So, H.S., Jung, B.H., Moon, S.R., Chung, S.Y., Ko, C.B., Chung, H.T., Activation of c-Jun N-terminal kinase 1 (JNK1) in mistletoe lectin II-induced apoptosis of human myeloleukemic U937 cells (2000) Biochem Pharmacol, 60, pp. 1685-1691Yoon, T.J., Yoo, Y.C., Kang, T.B., Shimazaki, K., Song, S.K., Lee, K.H., Kim, S.H., Kim, J.B., Lectins isolated from Korean mistletoe (Viscum album coloratum) induce apoptosis in tumor cells (1999) Cancer Lett, 136, pp. 33-40Inamori, K., Saito, T., Iwaki, D., Nagira, T., Iwanaga, S., Arisaka, F., Kawabata, S., A newly identified horseshoe crab lectin with specificity for blood group A antigen recognizes specific O-antigens of bacterial lipopolysaccharides (1999) J Biol Chem, 274, pp. 3272-3278Okino, N., Kawabata, S., Saito, T., Hirata, M., Takagi, T., Iwanaga, S., Purification, characterization, and cDNA cloning of a 27-kDa lectin (L10) from horseshoe crab hemocytes (1995) J Biol Chem, 270, pp. 31008-31015Pajic, I., Kljajic, Z., Dogovic, N., Sladic, D., Juranic, Z., Gasic, M.J., A novel lectin from the sponge Haliclona cratera: Isolation, characterization and biological activity (2002) Comp Biochem Physiol C Toxicol Pharmacol, 132, pp. 213-221Schroder, H.C., Kljajic, Z., Wegner, R., Reuter, P., Gasic, M., Uhlenbruck, G., Kurelac, B., Muller, W.E.G., The galactose-specific lectin from the sponge Chondrilla nucula displays anti-human immunodeficiency virus activity in vitro via stimulation of the (2'-5') oligoadenylate metabolism (1990) Antivir Chem Chemother, 1 (2), pp. 99-105Fukuda, Y., Sugahara, T., Ueno, M., Fukuta, Y., Ochi, Y., Akiyama, K., Miyazaki, T., Kato, K., The anti-tumor effect of Euchema serra agglutinin on colon cancer cells in vitro and in vivo (2006) Anticancer Drugs, 17, pp. 943-947Moura, R.M., Queiroz, A.F., Fook, J.M., Dias, A.S., Monteiro, N.K., Ribeiro, J.K., Moura, G.E., Sales, M.P., CvL, a lectin from the marine sponge Cliona varians: Isolation, characterization and its effects on pathogenic bacteria and Leishmania promastigotes (2006) Comp Biochem Physiol A Mol Integr Physiol, 145, pp. 517-523Debray, H., Decout, D., Strecker, G., Spik, G., Montreuil, J., Specificity of 12 Lectins towards oligosaccharides and glycopeptides related to N-glycosylproteins (1981) Eur J Biochem, 117, pp. 41-55Laemmli, U.K., Cleavage of structural proteins during the assembly of the head of the bacteriophage T4 (1970) Nature, 227, pp. 680-683Bromberg, N., Justo, G.Z., Haun, M., Duran, N., Ferreira, C.V., Violacein cytotoxicity on human blood lymphocytes and effect on phosphatases (2005) J Enzyme Inhib Med Chem, 20, pp. 449-454De Carvalho, D.D., Costa, F.T., Duran, N., Haun, M., Cytotoxic activity of violacein in human colon cancer cells (2006) Toxicol in Vitro, 20, pp. 1514-1521Guicciardi, M.E., Deussing, J., Miyoshi, H., Bronk, S.F., Svingen, P.A., Peters, C., Kaufmann, S.H., Gores, G.J., Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c (2000) J Clin Invest, 106, pp. 1127-1137Hartree, E.F., Determination of protein: A modification of the Lowry method that gives a linear photometric response (1972) Anal Biochem, 48, pp. 422-427Foghsgaard, L., Wissing, D., Mauch, D., Lademann, U., Bastholm, L., Boes, M., Elling, F., Jaattela, M., Cathepsin B acts as a dominant execution protease in tumor cell apoptosis induced by tumor necrosis factor (2001) J Cell Biol, 153, pp. 999-1010Mathiasen, I.S., Hansen, C.M., Foghsgaard, L., Jaattela, M., Sensitization to TNF-induced apoptosis by 1, 25-dihydroxy vitamin D(3) involves up-regulation of the TNF receptor 1 and cathepsin B (2001) Int J Cancer, 93, pp. 224-231Turk, B., Stoka, V., Protease signalling in cell death: Caspases versus cysteine cathepsins (2007) FEBS Lett, 581, pp. 2761-2767Nakanishi, C., Toi, M., Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs (2005) Nat Rev Cancer, 5, pp. 297-309Kirchner, D., Duyster, J., Ottmann, O., Schmid, R.M., Bergmann, L., Munzert, G., Mechanisms of Bcr-Abl-mediated NF-kappaB/Rel activation (2003) Exp Hematol, 31, pp. 504-511Reuther, J.Y., Reuther, G.W., Cortez, D., Pendergast, A.M., Baldwin Jr, A.S., A requirement for NF-kappaB activation in Bcr-Abl-mediated transformation (1998) Genes Dev, 12, pp. 968-981Karin, M., Lin, A., NF-kappaB at the crossroads of life and death (2002) Nat Immunol, 3, pp. 221-227Sanchez-Garcia, I., Grutz, G., Tumorigenic activity of the BCR-ABL oncogenes is mediated by BCL2 (1995) Proc Natl Acad Sci USA, 92, pp. 5287-5291Desrivieres, S., Volarevic, S., Mercep, L., Ferrari, S., Evidence for different mechanisms of growth inhibition of T-cell lymphoma by phorbol esters and concanavalin A (1997) J Biol Chem, 272, pp. 2470-2476Kulkarni, G.V., McCulloch, C.A., Concanavalin A induced apoptosis in fibroblasts: The role of cell surface carbohydrates in lectin mediated cytotoxicity (1995) J Cell Physiol, 165, pp. 119-133Cragg, G.M., Newman, D.J., Yang, S.S., Natural product extracts of plant and marine origin having antileukemia potential. the NCI experience (2006) J Nat Prod, 69, pp. 488-498McGahon, A., Bissonnette, R., Schmitt, M., Cotter, K.M., Green, D.R., Cotter, T.G., BCR-ABL maintains resistance of chronic myelogenous leukemia cells to apoptotic cell death (1994) Blood, 83, pp. 1179-1187Vancompernolle, K., Van Herreweghe, F., Pynaert, G., Van De Craen, M., De Vos, K., Totty, N., Sterling, A., Grooten, J., Atractyloside-induced release of cathepsin B, a protease with caspase-processing activity (1998) FEBS Lett, 438, pp. 150-158Dai, Y., Rahmani, M., Corey, S.J., Dent, P., Grant, S., A Bcr/Abl-independent, Lyn-dependent form of imatinib mesylate (STI-571) resistance is associated with altered expression of Bcl-2 (2004) J Biol Chem, 279, pp. 34227-34239Cortez, D., Reuther, G., Pendergast, A.M., The Bcr-Abl tyrosine kinase activates mitogenic signaling pathways and stimulates G1-to-S phase transition in hematopoietic cells (1997) Oncogene, 15, pp. 2333-2342Joyce, D., Albanese, C., Steer, J., Fu, M., Bouzahzah, B., Pestell, R.G., NF-kappaB and cell-cycle regulation: The cyclin connection (2001) Cytokine Growth Factor Rev, 12, pp. 73-90Niculescu III, A.B., Chen, X., Smeets, M., Hengst, L., Prives, C., Reed, S.I., Effects of p21(Cip1/Waf1) at both the G1/S and the G2/M cell cycle transitions: PRb is a critical determinant in blocking DNA replication and in preventing endoreduplication (1998) Mol Cell Biol, 18, pp. 629-64

    Anti-inflammatory Properties Of A Heparin-like Glycosaminoglycan With Reduced Anti-coagulant Activity Isolated From A Marine Shrimp

    No full text
    The anti-inflammatory properties of a heparin-like compound from the shrimp Litopenaeus vannamei are related. Besides reducing significantly (p < 0.001) the influx of inflammatory cells to injury site in a model of acute inflammation, shrimp heparin-like compound was able to reduce the matrix metalloproteinase (MMPs) activity in the peritoneal lavage of inflamed animals. Moreover, this compound also reduced almost 90% the activity of MMP-9 secreted by human activated leukocytes. Negligible anti-coagulant activities in aPPT assay and a poor bleeding potential make this compound a better alternative than mammalian heparin as a possible anti-inflammatory drug. © 2008 Elsevier Ltd. All rights reserved.162195889595Fransson, L.A., (1985) Aspinall GO, 3, pp. 337-413. , Orlando F.L. (Ed), Academic PressYoung, E., (2007) Thromb. Res., , 10.1016/j.thromres.2006.10.026Weiler, J.M., Edens, R.E., Linhardt, R.J., Kapelanski, D.P., (1992) J. Immunol., 148, p. 3210Teixeira, M.M., Rossi, A.G., Hellewell, P.G., (1996) J. Leukocyte Biol., 59, p. 89Miller, M.D., Krangel, M.S., (1984) Crit. Rev. Immunol., 12, p. 17Brown, R.A., Lever, R., Jones, N.A., Page, C.P., (2003) Br. J. Pharmacol., 139, p. 845Teixeira, M.M., Hellewell, P.G., (1993) Br. J. Pharmacol., 110, p. 1496Riesenberg, K., Schlaeffer, F., Katz, A., Levy, R., (1995) Br. Heart J., 73, p. 14Nelson, R.M., Cecconi, O., Roberts, W.G., Aruffo, A., Linhardt, R.J., Bevilacqua, M.P., (1993) Blood, 82, p. 3253Xie, X., Thorlacius, H., Raud, J., Hedqvist, P., Lindbom, L., (1997) Br. J. Pharmacol., 122, p. 906Pasini, F.L., Pasqui, A.L., Ceccatelli, L., Capecchi, P.L., Orrico, A., Di Perri, T., (1984) Thromb. Res., 35, p. 527Kenagy, R.D., Nikkari, S.T., Welgus, H.G., Clowes, A.W., (1994) J. Clin. Invest., 93, p. 987Kumagai, K., Ohno, I., Okada, S., Ohkawara, T., Suzuki, K., Shinya, T., Nagase, H., Shirato, K., (1999) J. Immunol., 162, p. 4212Wang, J.G., Brown, J.R., Varki, A., Esko, D., (2002) Inflamm. Res., 51, p. 435Gao, Y., Li, N., Fei, R., Chen, Z., Zheng, S., Zeng, X., (2005) Mol. Cells, 19, p. 350Lindahl, U., Li, J.P., Kusche-Gullberg, M., Salmivirta, M., Alaranta, S., Veromaa, T., Emeis, J., Casu, B., (2005) J. Med. Chem., 48, p. 349Dietrich, C.P., Paiva, J.F., Castro, R.A.B., Chavante, S.F., Jeske, W., Fareed, J., Gorin, P.A.J., Nader, H.B., (1999) Biochim. Biophys. Acta, 1428, p. 273Medeiros, G.F., Mendes, A., Castro, R.A.B., Baú, E.C., Nader, H.B., Dietrich, C.P., (2000) Biochim. Biophys. Acta, 1475, p. 287Chavante, S.F., Santos, E.A., Oliveira, F.W., Guerrini, M., Torri, G., Casu, B., Dietrich, C.P., Nader, H.B., (2000) Int. J. Biol. Macromol., 27, p. 49Santos, E.A., Rocha, L.R.M., Pereira, N.M.L., Andrade, G.P.V., Nader, H.B., Dietrich, C.P., (2002) Histochem. J., 34, p. 553Casu, B., Grazioli, G., Razi, N., Guerrini, M., Naggi, A., Torri, G., Oreste, P., Lindahl, U., (1994) Carbohydr. Res., 263, p. 271Naggi, A., Torri, G., Casu, B., Oreste, P., Zoppetti, G., Li, J.P., Lindahl, U., (2001) Semin. Thromb. Hemost., 27, p. 437Nader, H.B., Chavante, S.F., Santos, E.A., Oliveira, T.W., Paiva, J.F., Jerônimo, S.M., Medeiros, G.F., Dietrich, C.P., (1999) Braz. J. Med. Biol. Res., 32, p. 529Nader, H.B., Lopes, C.C., Rocha, H.A.O., Santos, E.A., Dietrich, C.P., (2004) Curr. Pharm. Des., 10, p. 951Frenette, P.S., Mayadas, T.N., Rayburn, H., Hynes, R.O., Wagner, D.D., (1996) Cell, 84, p. 563Milligan, K.E., Marquez, B.L., Williamson, R.T., Gerwick, W.H., (2000) J. Nat. Prod., 63, p. 1440Nader, H.B., Porcionatto, M.A., Tersariol, I.L.S., Pinhal, M.A.S., Oliveira, F.W., Moraes, C.T., Dietrich, C.P., (1990) J. Biol. Chem., 265, p. 16807Dietrich, C.P., Paiva, J.F., Moraes, C.T., Takahashi, H.K., Porcionatto, M.A., Nader, H.B., (1985) Biochim. Biophys. Acta, 843, p. 1Dietrich, C.P., Nader, H.B., Paiva, J.F., Tersariol, I.L.S., Santos, E.A., Holme, K.R., Perlin, A.S., (1989) Int. J. Biol. Macromol., 11, p. 361Pejler, G., Danielsson, A., Björk, I., Lindahl, U., Nader, H.B., Dietrich, C.P., (1987) J. Biol. Chem., 268, p. 11413Casu, B., (2005) Chemistry and Biology of Heparin and Heparan sulfate, pp. 1-28. , Garg H.G., Linhardt R.J., and Hales C.A. (Eds), Elsevier LtdBouças, R.L., Sampaio, L.O., Andrade, G.P.V., Lopes, C.C., Nascimento, F.D., Tersariol, I.L.S., Rocha, H.A.O., Nader, H.B., (2006) Insights Carbohydr. Struct. Biol. Funct., p. 145Casu, B., Lindahl, U., (2001) Adv. Carbohydr. Chem. Biochem., 57, p. 159Nader, H.B., Pinhal, M.A.S., Baú, E.C., Castro, R.A.B., Medeiros, G.F., Chavante, S.F., Leite, E.L., Dietrich, C.P.Braz., (2001) J. Med. Biol. Res., 34, p. 699Bianchini, P., Osima, B., Parma, B., Nader, H.B., Dietrich, C.P., (1985) Thromb. Res., 40, p. 597Rocha, H.A., Moraes, F.A., Trindade, E.S., Franco, C.R., Torquato, R.J., Veiga, S.S., Valente, A.P., Dietrich, C.P., (2005) J. Biol. Chem., 280, p. 41278Pinhal, M.A.S., Santos, I.A., Silva, I.F., Dietrich, C.P., Nader, H.B., (1995) Thromb. Haemost., 74, p. 1169Borsig, L., Wang, L., Cavalcante, M.C.M., Cardilo-Reis, L., Ferreira, P.L., Mourão, P.A.S., Esko, J.D., Pavão, M.S.G., (2007) J. Biol. Chem., 282, p. 14984Wang, L., Brown, J.R., Varki, A., Esko, D., (2002) J. Clin. Invest., 110, p. 127Betsuyaku, T., Shipley, J.M., Liu, Z., Senior, R.M., (1999) Am. J. Respir. Cell. Mol. Biol., 20, p. 1303Schonbeck, U., Mach, F., Libby, P., (1998) J. Immunol, 161, p. 3340English, W.R., Puente, X.S., Freije, J.M., Knauper, V., Amour, A., Merryweather, A., Lopez-Otin, C., Murphy, G., (2000) J. Biol. Chem., 275, p. 14046Mohan, M.J., Seaton, T., Mitchell, J., Howe, A., Blackburn, K., Burkhart, W., Moyer, M., Milla, M.E., (2002) Biochemistry, 41, p. 9462Leppert, D., Waubant, E., Galardy, R., Bunnett, N.W., Hauser, S.L., (1995) J. Immunol., 154, p. 4379Gogly, B., Hornebeck, W., Groult, N., Godeau, G., Pellat, B., (1998) Biochem. Pharmacol., 56, p. 1447Kitamura, M., Marayama, N., Mitarai, T., Nagasawa, R., Yokoo, T., Sakai, O., (1994) Biochem. Biophys. Res. Commun., 203, p. 1333Lee, H.M., Ciancio, S.G., Tüter, G., Ryan, M.E., Komaroff, E., Golub, L.M., (2004) J. Periodontol., 75, p. 453Dietrich, C.P., Nader, H.B., Paiva, J.F., Tersariol, I.L.S., Santos, E.A., Holme, K.R., Perlin, A.S., (1989) Int. J. Biol. Macromol., 11, p. 361Dietrich, C.P., Dietrich, S.M.C., (1976) Anal. Biochem., 70, p. 645Trevelyan, W.E., Procter, D.P., Harrison, J.S., (1950) Nature, 166, p. 444Cruz, W.O., Dietrich, C.P., (1967) Proc. Soc. Exp. Biol. Med., 126, p. 420Bradford, M.M., (1976) Anal. Biochem., 72, p. 248Lowry, O.H., Rosbrough, N.J., Farr, A.L., Randall, R.J., (1951) J. Biol. Chem., 193, p. 26
    corecore