21 research outputs found

    Physico-chemical Characterization Of The Inclusion Complex Between A 2-propen-1-amine Derivative And β-cyclodextrin

    Get PDF
    Inclusion complexes and physical mixtures were prepared with isomeric mixture of E/Z (50:50) of 3-(4′-bromo-[1,1′-biphenyl]-4-yl)-3-(4- bromophenyl)-N,N-dimethyl-2-propen-1-amine (BBAP) and β-cyclodextrin (β-CD) in different proportions. In this study, theoretical calculations using Molecular Mechanics MM+ force field were applied to predict the structures of the inclusion complexes formed by interaction of BBAP and b-cyclodextrin. Circular dichroism, differential thermal analysis (DTA), X-ray diffraction and 13C CP/MAS NMR methods were used to characterize the inclusion complexes and provide information about the stoichiometry of the inclusion complexes. The combined spectroscopy techniques indicate the formation of a complex of BBAP/β-CD in the molar proportion of 1:1 and 1:2 by co-evaporation and no complexation was detected in the physical mixture of the compounds. © 2006 Sociedad Chilena de Química β-.503115127Bibby, D.C., Davies, N.M., Tucker, I.G., (2000) Int. J. Pharm., 197, p. 1Connors, K.A., (1995) J. Pharm. Sci., 84, p. 843Connors, K.A., Paulson, A., Toledo-Velasquez, D.J., (1988) J. Org. Chem., 53, p. 2123Dollo, G., Le Corre, P., Chollet, M., Chevanne, F., Bertault, M., Burgot, J.-L., Le Verge, R., (1999) J. Pharm. Sci., 88, p. 889Mucci, A., Schenetti, L., Vandelli, M.A., Ruozi, B., Forni, F., (1999) J. Chem. Research, (S), p. 414Keipert, S., Fedder, J., Bohm, A., Hanke, B., (1996) Int. J. Pharm., 142, p. 153Nigam, S., Durocher, G., (1999) J. Photochem. Photobiol. A: Chem., 103, p. 143De Souza, A.O., Sato, D.N., Aily, D.C.G., Duran, N., (1998) J. Antimicrob. Chemother., 42, p. 407Pereira, D.G., De Castro, S.L., Durán, N., (1998) Acta Tropica, 69, p. 205De Souza, A.O., Santos Júnior, R.R., Ferreira-Júlio, J.F., Rodrigues, J.A., Melo, P.S., Haun, M., Sato, D.N., Durán, N., (2001) Eur. J. Med. Chem., 36, p. 843De Souza, A.O., Hemerly, F.P., Busollo, A.C., Melo, P.S., Machado, G.M.C., Miranda, C.C., Santa-Rita, R.M., Durán, N., (2002) J. Antimicrob. Chemother., 50, p. 629De Conti, R., Gimenez, S.M.N., Haun, M., Pilli, R.A., De Castro, S.L., Durán, N., (1996) N. Eur. J. Med. Chem., 31, p. 915De Azevedo, M.B.M., Alderete, J.B., Lino, A.C.S., Loh, W., Faljoni-Alario, A., Durán, N., (2000) J. Incl. Phenom. Macrocyclic. Chem., 37, p. 67Zhou, D., Wu, Y., Xu, Q., Yang, L., Bai, C., (2000) Z. Tan. J. Incl. Phenom. Macrocyclic. Chem., 37, p. 273Ammar, H.O., Ghorab, M., El-Nahhas, S.A., Emara, I.H., Makram, T.S., (1999) Pharmazie, 54, p. 142Muñoz De La Pena, A., Ndou, T.T., Zung, J.B., Warner, I.M., (1991) J. Phys. Chem., 95, p. 3330Smith, V.K., Ndou, T.T., Warner, I.M., (1994) J. Phys. Chem., 98, p. 8627Blanco, M., Coello, J., Iturriaga, H., Maspoch, S., Pérez-Maseda, C., (2000) Anal. Chim. Acta, 407, p. 233Ohashi, M., Kasatani, K., Shinohara, H., Sato, H., (1990) J. Am. Chem. Soc., 112, p. 5824Harata, K., Uedaria, H., (1975) Bull. Chem. Soc. Jpn., 48, p. 375Schellman, J.A., (1968) Acc. Chem. Res., 1, p. 144Bettinetti, G.P., Gonzzaniga, A., Mura, P., Giordano, F., Setti, M., (1992) Drug Dev. Ind. Pharm., 18, p. 39Mura, P.P., Faucci, M.T., Parrini, P.L., Furlanetto, S., Pinzauti, S., (1999) Int. J. Pharm., 179, p. 117Cunha-Silva, L., Teixeira-Dias, J.J.C., (2002) J. Phys. Chem., 106, p. 3323Lai, S., Locci, E., Piras, A., Porcedda, S., Lai, A., Marangiu, B., (2003) Carbohydr. Res., 338, p. 222

    Chromobacterium Violaceum And Its Important Metabolites--review.

    No full text
    C. violaceum appeared as important bacterium in different applications and mainly these aspects are related to the production of violacein. This review discusses the last reports on biosynthetic pathways, production, genetic aspects, biological activities, pathological effects, antipathogenic screening through quorum sensing, environmental effects and the products of C. violaceum with industrial interest. An important discussion is on biological applications in medicine and as industrial products such as textile and in cosmetics.55535-4

    Chromobacterium Violaceum And Its Important Metabolites - Review

    No full text
    C. violaceum appeared as important bacterium in different applications and mainly these aspects are related to the production of violacein. This review discusses the last reports on biosynthetic pathways, production, genetic aspects, biological activities, pathological effects, antipathogenic screening through quorum sensing, environmental effects and the products of C. violaceum with industrial interest. An important discussion is on biological applications in medicine and as industrial products such as textile and in cosmetics. © 2010 Institute of Microbiology, v.v.i, Academy of Sciences of the Czech Republic.556535547Ahmetagic, A., Pemberton, J.M., Stable high level expression of the violacein indolocarbazole anti-tumour gene cluster and the Streptomyces lividans amyA gene in E. coli K12 (2010) Plasmid, 63, pp. 79-85Ajithdoss, D.K., Porter, B.F., Calise, D.V., Libal, M.C., Edwards, J.F., Septicemia in a neonatal calf associated with Chromobacterium violaceum (2009) Veter.Pathol., 46, pp. 71-74Al-Hussaini, R., Mahasneh, A.M., Microbial growth and quorum sensing antagonist activities of herbal plants extracts (2009) Molecules, 14, pp. 3425-3435Alves, O.L., Gimenez, I.F., de Azevedo, M.M.M., Durán, N., Melo, P.S., Phamacological use of cyclodextrine-Au-thiol-derivative/hydrophobic compound nanoparticles as antitumoral, antibacterial, antiviral and/or antiparasites, its obtention process and formulation (2005) Brazil. Pat., , PIBr 0502657-1Anah, M.U., Udo, J.J., Ochigbo, S.O., Abia-Bassey, L.N., Neonatal septicemia in calabar, Nigeria (2008) Tropical Doctor, 38, pp. 126-128Andrighetti-Frohner, C.R., Kratz, J.M., Antonio, R.V., Creczynski-Pasa, T.B., Barardi, C.R.M., Simoes, C.M.O., In vitro testing for genotoxicity of violacein assessed by Comet and Micronucleus assays (2006) Mutat.Res., 603, pp. 97-103Antônio, R.V., Creczynski-Pasa, T.B., Genetic analysis of violacein biosynthesis by Chromobacterium violaceum (2004) Genet.Mol.Res., 3, pp. 85-91Antonisamy, P., Kannan, P., Ignacimuthu, S., Anti-diarrheal 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) Japan Pat, , JP 10139612Asamizu, S., Kato, Y., Igarashia, Y., Onaka, H., VioE, a prodeoxyviolacein synthase involved in violacein biosynthesis, is responsible for intramolecular indole rearrangement (2007) Tetrahedron Lett., 48, pp. 2923-2926Baek, 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, , Republ. Korean Pat. KR 20070 88150 ABaker, S., Campbell, J.I., Stabler, R., Nguyen, H.V.M., To, D.S., Nguyen, D.V., Farrar, J., Fatal wound infection caused by Chromobacterium violaceum in Ho Chi Minh City, Vietnam (2008) J.Clin.Microbiol., 46, pp. 3853-3855Baldi, M., Morales, J.A., Hernandez, G., Jimenez, M., Alfaro, A., Arquero-Calvo, E., Chromobacterium Violaceum infection in a free-ranging howler monkey in Costa Rica (2010) J.Wildlife Dis., 46, pp. 306-310Balibar, C.J., Walsh, C.T., In vitro biosynthesis of violacein from L-tryptophan by the enzymes VioA-E from Chromobacterium violaceum (2006) Biochemistry, 45, pp. 15444-15457Barreto, E.S., Torres, A.R., Barreto, M.R., Vasconcelos, A.T.R., Astolfi-Filho, S., Hungria, M., Diversity in antifungal activity of strains of Chromobacterium violaceum from the Brazilian Amazon (2008) J.Ind.Microbiol.Biotechnol., 35, pp. 783-790Becker, M.H., Brucker, R.M., Schwantes, C.R., Harris, R.N., Minbiole, K.P.C., The bacterially produced metabolite violacein is associated with survival of amphibians infected with a lethal fungus (2009) Appl.Environ.Microbiol., 75, pp. 6635-6638Bodini, S.F., Manfredini, S., Epp, M., Valentini, S., Santori, F., Quorum sensing inhibition activity of garlic extract and p-coumaric acid (2009) Lett.Appl.Microbiol., 49, pp. 551-555Bosch, F.J., Badenhorst, L., le Roux, J.A., Louw, V.J., Successful treatment of Chromobacterium violacelum sepsis in South Africa (2008) J.Med.Microbiol., 57, pp. 1293-1295Bosgelmez-Tinaz, G., Ulusoy, S., Ugur, A., Ceylan, O., Inhibition of quorum sensing-regulated behaviors by Scorzonera sandrasica (2007) Curr.Microbiol., 55, pp. 114-118Bowers, A.A., Greshock, T.J., West, N., Schreiber, S.L., Wiest, O., Williams, R.M., Bradner, J.E., Synthesis and conformation-activity relationships of the peptide isosters of FK228 and largazole (2009) J.Am.Chem.Soc., 131, pp. 2901-2905Brandl, H., Lehmann, S., Faramarzi, M.A., Martinelli, D., Biomobilization of silver, gold, and platinum from solid waste materials by HCN-forming microorganisms (2008) Hydrometallurgy, 94, pp. 14-17Brown, A.G., Discovery and development of new β-lactam antibiotics (1987) Pure Appl.Chem., 59, pp. 475-484Brucker, R.M., Harris, R.N., Schwantes, C.R., Gallaher, T.N., Flaherty, D.C., Lam, B.A., Minbiole, K.P.C., Amphibian chemical defense: antifungal metabolites of the microsymbiont Janthinobacterium lividum on the salamander Plethodon cinereus (2008) J.Chem.Ecol., 34, pp. 1422-1429Byrd, L.C., Marcucci, G., Parthun, M.R., Xiao, J.J., Klisovic, R.B., Moran, M., Lin, T.S., Grever, M.R., A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia (2005) Blood, 105, pp. 959-967Cao, 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. 1-4Carminatti, C.A., Oliveira, I.L., Recouvreux, D.O.S., Antonio, R.V., Porto, L.M., Anthranilate synthase subunit organization in Chromobacterium violaceum (2008) Genet.Mol.Res., 7, pp. 830-838Carter, E., Cain, K., Rutland, B., Chromobacterium violaceum cellulitis and sepsis following cutaneous marine trauma (2008) Cutis, 81, pp. 269-272Chen, C.N., Chen, C.J., Liao, C.T., Lee, C.Y., A probable aculeacin A acylase from the Ralstonia solanacearum GMI1000 is N-acylhomoserine-lactone acylase with quorum-quenching activity (2009) BMC Microbiol., 9, p. 89Cooper, R., Unger, S., Novel potentiators of β-lactam antibiotics. Structures of SQ 28504 and SQ 28546 (1986) J.Org.Chem., 51, pp. 3942-3946Costa, F.T.M., Justo, G.Z., Duran, N., Nogueira, P.A., Lopes, S.C.P., Uso da violaceina na forma livre ouencapsulada em sistemas polimericos como antimalárico (2005) Brazil. Pat., , PIBr 056399-0de Azevedo, M.B.M., Alderete, J.B., Rodriguez, J.A., de 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.Macrocyclic Chem., 37, pp. 93-101de Azevedo, M.B.M., Melo, P.S., Almeida, A.B.A., Souza-Brito, A.R.M., Haun, M., Durán, N., Antiulcerogenic activity of violacein/β-cyclodextrin inclusion complexes and violacein (2000) Proc.Intern.Symp.Control.Rel.Bioact.Mater., 27, pp. 508-509de Carvalho, D.D., Costa, F.T.M., Durán, 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. 77-81Deines, P., Matz, C., Jurgens, K., Toxicity of violacein-producing bacteria fed to bacterivorous freshwater plankton (2009) Limnol.Oceanogr., 54, pp. 1343-1352Dessaux, Y., Elmerich, C., Faure, D., Violacein: a molecule of biological interest originating from the soil-borne bacterium Chromobacterium violaceum (2004) Rev.Med.Internat., 25, pp. 659-662Dobretsov, S., Dahms, H.U., Huang Yili, H., Wahl, M., Qian, P.Y., The effect of quorum-sensing blockers on the formation of marine microbial communities and larval attachment (2007) FEMS Microbiol.Ecol., 60, pp. 177-188Dufosse, L., Microbial production of food grade pigments (2006) Food Technol.Biotechnol., 44, pp. 313-321Durán, N., Violacein: an antibiotic discovery (1990) Ciencia Hoje, 11, pp. 58-80Durán, 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) Brazil. Pat., , PIBr 0404306-5Durán, N., de Souza, A.O., Process of violacein application as antimycobacterial compound (2001) Brazil. Pat., , PIBr 0101346-7Durán, 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) Brazil. Pat., , PIBr 9702918Durán, N., Menck, C.F.M., Chromobacterium violaceum: a review of farmacological and indutrial perspectives (2001) Crit.Rev.Microbiol., 27, pp. 201-222Durán, N., Rettori, D., Menck, C.F.M., Who is Chromobacterium violaceum? (2001) Biotecnol.Ciên.Desenvol., 20, pp. 38-43Durán, 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 β-cyclodextrin (2003) Can.J.Physiol.Pharmacol., 81, pp. 387-396Durán, N., Justo, G.Z., Melo, P.S., Martins Jr., D., Cordi, L., Violacein: properties and biological activities (2007) Biotechnol.Appl.Biochem., 48, pp. 127-133Durán, N., Marcato, P.D., Alves, O.L., da Silva, J.P.S., de Souza, G.I.H., Rodrigues, F.A., Espósito, E., Ecosystem protection by effluent bioremediation: silver nanoparticles impregnation in a textile fabrics process (2010) J.Nanopart.Res., 12, pp. 285-292Fairbrother, L., Shapter, J., Brugger, J., Southam, G., Pring, A., Reith, F., Effect of the cyanide-producing bacterium Chromobacterium violaceum on ultraflat Au surfaces (2009) Chem.Geol., 265, pp. 313-320Ferreira, C.V., Bos, C.L., Versreeg, H.H., Justo, G.Z., Durán, N., Peppelenbosch, M.P., Molecular mechanism of violacein-mediated human promyelocytic leukemia cell-specific cytotoxicity (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-β-cyclodextrin-Au/violacein nanoparticles (2005) J.Biomed.Nanotechnol., 1, pp. 352-358Gootz, T.D., Discovery and development of new antimicrobial agents (1990) Clin.Microbiol.Rev., 3, pp. 13-31Gregory, A., Ribollet, D., Pailla, K., Bourgade, M., Alma, S., Chromobacterium violaceum septicemia: first case report in the French West Indies (2007) Méd.Malad.Infect., 37, pp. 613-615Greshock, T.J., Johns, D.M., Noguchi, Y., Williams, R.M., Improved total synthesis of the potent HDAC inhibitor FK228 (FR-901228) (2008) Org.Lett., 10, pp. 613-616Guevara, A., Salomon, M., Oliveros, M., Guevara, E., Guevara, M., Medina, Z., Sepsis caused by pigmented and no pigmented Chromobacterium violaceum (2007) Rev.Chilena Infect., 24, pp. 402-406Gupta, R., Shah, P., Swiatlo, E., Differential gene expression in Streptococcus pneumoniae in response to various iron sources (2009) Microb.Pathogen., 47, pp. 101-109Hakvag, S., Fjaervik, E., Klinkenberg, G., Borgos, S.E.F., Josefsen, K.D., Ellingsen, T.E., Zotchev, S.B., Violacein-producing Collimonas sp. from the sea surface microlayer of costal waters in Trondelag, Norway (2009) Marine Drugs, 7, pp. 576-588Harris, R.N., Brucker, R.M., Walke, J.B., Becker, M.H., Schwantes, C.R., Flaherty, D.C., Lam, B.A., Minbiole, K.P.C., Skin microbes on frogs prevent morbidity and mortality caused by a lethal skin fungus (2009) ISME J., 3, pp. 818-824Haun, M., Pereira, M.F., Hoffman, M.F., 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, p. 21Hirano, S., Asamizu, S., Onaka, H., Shiro, Y., Nagano, S., Crystal structure of VioE, a key player in the construction of the molecular skeleton of violacein (2008) J.Biol.Chem., 283, pp. 6459-6466Kato, 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) Japan Pat, , JP 1115 2687 AKhan, M.S.A., Zahin, M., Hasan, S., Husain, F.M., Ahmad, I., Inhibition of quorum sensing regulated bacterial functions by plant essential oils with special reference to clove oil (2009) Lett.Appl.Microbiol., 49, pp. 354-360Kita, Y., Nishikawa, H., Ike, M., Takemoto, T., Enhancement of Au dissolution by microorganisms using an accelerating cathode reaction (2009) Metallurg.Mat.Trans.B, 40 B, pp. 2009-2044Kodach, L.L., Bos, C.L., Durán, N., Peppelenbosh, M.P., Ferreira, C.V., Hardwick, J.C.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. 8307-8313Kubo, H., Kotani, H., Yamamoto, Y., Hazato, T., Involvement of sperm proteases in the binding of sperm to the vitelline envelope in Xenopus laevis (2008) Zoolog.Sci., 25, pp. 80-87Lai, M.T., Yang, C.C., Lin, T.Y., Tsai, F.J., Chen, W.C., Depsipeptide (FK228) inhibits growth of human prostate cancer cells (2008) Urol. Oncol.Sem.Orig.Invest., 26, pp. 182-189Leon, L.L., Miranda, C.C., de Souza, A.O., Durán, N., Antileishmanial activity of the violacein extracted from Chromobacterium violaceum (2001) J.Antimicrob.Chemother., 48, pp. 449-450Lim, I.W.M., Stride, P.J., Horvath, R.L., Hamilton-Craig, C.R., Chau, P.P., Chromobacterium violaceum endocarditis and hepatic abscesses treated successfully with meropenem and ciprofloxacin (2009) Med.J.Austral., 190, pp. 386-387Lima-Bittencourt, C.I., Astolfi-Filho, S., Chartone-Souza, E., Santos, F.R., Nascimento, A.M.A., Analysis of Chromobacterium sp. natural isolates from different Brazilian ecosystems (2007) BMC Microbiol., 7, p. 58Lopes, S.C.P., 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, China (2009) Biochem.Eng.J., 43, pp. 135-141Manjunath, M., Fatal septicemia due to Chromobacterium violaceum (2007) West Indian Med.J., 56, pp. 380-381Martins, D., Frungillo, L., Anazzetti, M.C., Melo, P.S., Durán, N., Antitumoral activity of L-ascorbic acid-poly-d,l-(lactide-coglycolide) nanoparticles containing violacein (2010) Internat.J.Nanomed., 5, pp. 77-85Masuoka, Y., Shindoh, N., Inamura, N., Histone deacetylase inhibitors from microorganisms: the Astellas experience (2008) Natur. Comp. Drugs, 2, pp. 335-359. , in F. Petersen, R. AmstutzMatz, C., Webb, J.S., Schupp, P.J., Phang, S.Y., Penesyan, A., Egan, S., Steinberg, P., Kjelleberg, S., Marine biofilm bacteria evade eukaryotic predation by targeted chemical defense (2008) PLoS One, 3, pp. e2744Meiring, U., Lanzendoerfer, G., Riedel, H., Kallmayer, V., Viala, S., Mocigemba, N., Schaefer, J., Cosmetic preparations containing violacein as dye (2007) Ger. Pat., , DE 102 005 051 869 A1Melo, P.S., Justo, G.Z., de Azevedo, M.B.M., Durán, N., Haun, M., Violacein and its β-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 violacein-loaded 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) Brazil. Pat., , 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. 1963-1969Morohoshi, T., Kato, M., Fukamachi, K., Kato, N., Ikeda, T., N-Acylhomoserine lactone regulates violacein production in Chromobacterium violaceum type strain ATCC 12472 (2008) FEMS Microbiol.Lett., 279, pp. 124-130Nomura, T., Natural dye violacein manufacture with Chromobacterium and Janthinobacterium (1994) Japan Pat, , JP 0625 3864 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. 992-999Pearson, R.N., Chromobacterium violaceum endocarditis and hepatic abscesses treated successfully with meropenem and ciprofloxacin (2009) Med.J.Austral., 191, p. 416Perpetuo, E.A., Marques, R.C.P., Mendes, M.A., de Lima, W.C., Menck, C.F.M., do Nascimento, C.A.O., Characterization of the phenol monooxygenase gene from Chromobacterium violaceum: potential use for phenol biodegradation (2009) Biotechnol.Bioproc. Eng., 14, pp. 694-701Pham, V.A., Ting, Y.P., Gold bioleaching of electronic waste by cyanogenic bacteria and its enhancement with bio-oxidation (2009) Adv.Mat. Res., 71-73, pp. 661-664Philip, 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. 39-43Praneenararat, T., Geske, G.D., Blackwell, H.E., Efficient synthesis and evaluation of quorum-sensing modulators using small molecule macroarrays (2009) Org.Lett., 11, pp. 4600-4603Rettori, D., Durán, N., Stationary tray bioreactor for the bacterial metabolite production (1997) Brazil. Pat., , PIBr 9702986-6Ryan, K.S., Balibar, C.J., Turo, K.E., Walsh, C.T., Drennan, C.L., The violacein biosynthetic enzyme VioE shares a fold with lipoprotein transporter proteins (2008) J.Biol.Chem., 283, pp. 6467-6475Sanchez, C., Braña, A.F., Mendez, C., Salas, J.A., Reevaluation of the violacein biosynthetic pathway and its relationship to indolocarbazole biosynthesis (2006) ChemBioChem., 7, pp. 1231-1240Sarovich, D.S., Pemberton, J.M., pPSX: a novel vector for the cloning and heterologous expression of antitumor antibiotic gene clusters (2007) Plasmid, 57, pp. 306-313Shinoda, K., Hasegawa, T., Sato, H., Shinozaki, M., Kuramoto, H., Takamiya, Y., Sato, T., Hoshino, T., Biosynthesis of violacein: a genuine intermediate, protoviolaceinic acid, produced by VioABDE, and insight into VioC function (2007) Chem. Commun., pp. 4140-4142Shirata, 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) Japan Kokai Tokkyo Koho JP 10113169 AShukla, S.R., Damle, A.J., Colorants from microorganisms (2007) Asian Dyer, 4, pp. 33-34Singh, B.N., Singh, B.R., Singh, R.L., Prakash, D., Dhakarey, R., Upadhyay, G., Singh, H.B., Oxidative DNA damage protective activity, antioxidant and anti-quorum sensing potentials of Moringa oleifera (2009) Food Chem.Toxicol., 47, pp. 1109-1116Slesak, G., Douangdala, P., Inthalad, S., Silisouk, J., Vongsouvath, M., Sengduangphachanh, A., Moore, C.E., Newton, P.N., Fatal Chromobacterium violaceum septicaemia in northern Laos, a modified oxidase test and post-mortem forensic family G6PD analysis (2009) Ann.Clin.Microbiol.Antimicrob., 8, p. 24Swem, L.R., Swem, D.L., O'Loughlin, C.T., Gatmaitan, R., Zhao, B., Ulrich, S.M., Bassler, B.L., A quorum-sensing antagonist targets both membrane-bound and cytoplasmic receptors and controls bacterial pathogenicity (2009) Mol.Cell, 35, pp. 143-153Tan, T.L., Montforts, F.P., Meyer, D., Microbiological method for the biosynthesis of natural blue-violet colorants violacein and desoxyviolacein (2002) PCT Internat. Appl. WO 20020 50299 A2Teasdale, M.E., Liu, J.Y., Wallace, J., Akhlaghi, F., Rowley, D.C., Secondary metabolites produced by the marine bacterium Halobacillus salinus that inhibit quorum sensing-controlled phenotypes in Gram-negative bacteria (2009) Appl.Environ.Microbiol., 75, pp. 567-572Uroz, S., Oger, P., Chhabra, S.R., Cámara, M., Williams, P., Dessaux, Y., N-Acylhomoserine lactones are degraded via an amidolytic activity in Comamonas sp. strain D1 (2007) Arch.Microbiol., 187, pp. 249-256Vasconcelos, A.T.R., (Brazilian National Genome Project Consortium109 authors): The complete genome of Chromobacterium violaceum reveals remarkable and exploitable bacterial adaptability (2003) Proc.Nat.Acad.Sci.USA, 100, pp. 11660-11665Vattem, D.A., Mihalik, K., Crixell, S.H., McLean, R.J.C., Dietary phytochemicals as quorum sensing inhibitors (2007) Fitoterapia, 78, pp. 302-310Vijayan, A.P., Anand, M.R., Remesh, P., Chromobacterium violaceum sepsis in an infant (2009) Indian Pedriat., 46, pp. 721-722Wang, Y., Ikawa, A., Okaue, S., Taniguchi, S., Osaka, I., Yoshimoto, A., Kishida, Y., Enomoto, K., Quorum sensing signaling molecules involved in the produc

    Peroxidase-hydrogen Peroxide System Acting On Lignin(1)

    No full text
    Horseradish peroxidase (HRP)/H2O2 system, as a possible model of lignolytic enzyme, was evaluated. Aerobic peroxidation of lignin involved chemiluminescence, which arose mainly from recombination of OH radicals and carbonate ions (dissolved CO2). The peroxidase cycle followed by ultraviolet spectrophotometry and by circular dichroism pointed out the HRP-III (Fe III-O2{minus sign, dot below}) species, instead of the free-activated oxygen species, as the most reactive intermediate acting on lignin. HRP-I and HRP-II participate only in the oxidation of the phenolic moiety. The participation of HRP-III in lignin degradation open the way for more detailed studies of biological oxidation by enzyme-bound oxygen species. A general mechanism for lignin degradation by peroxidase was proposed. © 1988.342105115Kirk, (1983) Recent Advances in Lignin Biodegradation, pp. 1-11. , T. Higuchi, H.-M. Chang, T.K. Kirk, Japan: UNI Publ. Co. Ltd, Tokyo, Proc. Intern. Sem. US-Japan Cooperative Sci. ProgramKirk, (1984) Microbiological Degradation of Organic Compounds, Microbiology Series, 13, pp. 399-437. , D.T. Gibson, Marcel Dekker, Inc, New YorkKutsuki, Enoki, Gold, RIBOFLAVIN-PHOTOSENSITIZED OXIDATIVE DEGRADATION OF A VARIETY OF LIGNIN MODEL COMPOUNDS (1983) Photochemistry and Photobiology, 37, pp. 1-7Gold, Kutsuki, Morgan, OXIDATIVE DEGRADATION OF LIGNIN BY PHOTOCHEMICAL AND CHEMICAL RADICAL GENERATING SYSTEMS (1983) Photochemistry and Photobiology, 38, pp. 647-651Greene, Gould, (1984) Biochem. Biophys. Res. Commun., 118, pp. 437-443Faison, Kirk, (1984) Appl. Environ. Microbiol., 46, pp. 1140-1145Kirk, Mozuch, Tien, (1985) Biochem. J., 226, pp. 455-460Tien, Kirk, (1983) Science, 221, pp. 661-663Faison, Kirk, (1985) Appl. Environ. Microbiol., 49, pp. 299-304Tien, Kirk, Lignin-degrading enzyme from Phanerochaete chrysosporium: Purification, characterization, and catalytic properties of a unique H2O2-requiring oxygenase (1984) Proceedings of the National Academy of Sciences, 81, pp. 2280-2284Kuwahara, Glenn, Morgan, Gold, (1984) FEBS Lett., 169, pp. 247-250Gold, Kuwahara, Chiu, Glend, (1984) Arch. Biochem. Biophys., 234, pp. 353-362Leisola, Meussdoerffer, Waldner, Fiechter, (1985) J. Biotechnol., 2, pp. 379-382Leisola, Thanei-Wyss, Fiechter, (1985) J. Biotechnol., 2, pp. 97-107Kirk, Croan, Tien, Murtagh, Farrel, (1986) Enzyme Microb. Technol., 8, pp. 27-32Kirk, Tien, Johnsrud, Eriksson, (1986) Enzyme Microb. Technol., 8, pp. 75-80Kirk, Tien, Kersten, Mozuch, Kalyanaraman, (1986) Biochem. J., 236, pp. 279-287Durán, Rodriguez, Ferraz, Campos, Chrysonila sitophila (TFB-27441): A hyperlignolytic strain (1987) Biotechnology Letters, 9, pp. 357-360Renganathan, Miki, Gold, (1987) Biochemistry, 26, pp. 5127-5132Andersson, Renganathan, Loehr, Gold, (1987) Biochemistry, 26, pp. 2258-2263Leisola, Kozulic, Meussdoerffer, Fiechter, (1987) J. Biol. Chem., 262, pp. 419-424Asada, Miyabe, Kikawa, Kuwahara, (1987) J. Ferment. Technol., 65, pp. 483-487Renganathan, Miki, Gold, (1985) Arch. Biochem. Biophys., 241, pp. 304-314Glenn, Akileswaran, Gold, (1986) Arch. Biochem. Biophys., 251, pp. 688-696Shimada, Habe, Umezaba, Higuchi, Okamoto, (1984) Biochem. Biophys. Res. Commun., 122, pp. 1247-1252Schoemaker, Harvey, Palmer, (1985) FEBS Lett., 183, pp. 7-12Ander, Eriksson, (1976) Arch. Microbiol., 109, pp. 1-8Harstenstein, Neuhauser, Mulligan, (1977) Phytochemistry, 16, pp. 1855-1857Young, Steelink, (1973) Phytochemistry, 12, pp. 2851-2861Caldwell, Steelink, (1969) Biochim. Biophys. Acta, 184, pp. 420-431Strel'skii, Chupka, (1982) Khim. Prir. Soedin., 6, pp. 762-764Renganathan, Miki, Gold, (1986) Arch. Biochem. Biophys., 246, pp. 155-161Renganathan, Gold, (1986) Biochemistry, 25, pp. 1626-1631Harvey, Schoemaker, Bowen, Palmer, (1985) FEBS Lett., 183, pp. 13-16Kuila, Tien, Fee, Ondrias, (1985) Biochemistry, 24, pp. 3394-3397Durán, Mansilla, Leite, (1985) Atual. Fis. Quim. Org. (Brazil), 4, pp. 200-207Durán, Ferrer, Rodriguez, Mansilla, Baeza, Biomass photochemistry XIII: pre-irradiated lignin from Pinus Radiata D. Don and its degradation by ligninase and horse-radish peroxidase (1988) Journal of Photochemistry and Photobiology A: Chemistry, 41, pp. 267-273Durán, Ferrer, Rodriguez, (1988) Appl. Biochem. Biotechnol., , in pressFerrer, Durán, (1987) Biotechnol. Lett., 9, pp. 361-364Durán, Mansilla, Biomass Photochemistry. V. Modifications of Lignin by Photochemical Treatment and Its Chemiluminescence (1984) Journal of Macromolecular Science: Part A - Chemistry, 21 A, pp. 1467-1485Pepper, Baylis, Adler, THE ISOLATION AND PROPERTIES OF LIGNINS OBTAINED BY THE ACIDOLYSIS OF SPRUCE AND ASPEN WOODS IN DIOXANE–WATER MEDIUM (1959) Canadian Journal of Chemistry, 37, pp. 1241-1248Durán, (1982) Chemical and Biological Excited State Generation, pp. 345-369. , W. Adam, G. Cilento, Academic Press, New YorkAnderson, Patel, (1978) Photochem. Photobiol., 28, pp. 881-885Durán, Rojas, Nascimento, Freer, Baeza, (1984) Cellul. Chem. Technol., 18, pp. 411-419Dordick, Marletta, Klibanov, Peroxidases depolymerize lignin in organic media but not in water (1986) Proceedings of the National Academy of Sciences, 83, pp. 6255-6257Hammel, Kalyanaraman, Kirk, (1986) Proc. Natl. Acad. Sci. USA, 83, pp. 3708-3712Dunford, Stillman, (1976) Coordination Chem. Rev., 19, pp. 187-251Santimone, (1975) Can. J. Biochem., 53, pp. 649-657Bors, Saram, Lengfelder, Spotti, Michel, (1974) Curr. Topics Radial. Res. Quart., 9, pp. 247-309Yokota, Yamazaki, (1965) Biochem. Biophys. Res. Commun., 18, pp. 48-53Job, Dunford, (1977) Can. J. Biochem., 55, pp. 804-811Kirk, (1975) Biotechnol. Bioeng. Symp., 5, pp. 139-150Deng, (1975) Biochim. Biophys. Acta, 180, pp. 271-290Smith, Morrison, Milham, (1982) Biochemistry, 21, pp. 4414-4419Dunford, (1982) Advan. Inorg. Biochem., 4, pp. 41-68Andrawir, Johnson, Tien, (1988) J. Biol. Chem., 263, pp. 1195-1198Harvey, Schoemaker, Palmer, (1987) Plant Cell Environ., 10, pp. 709-710. , and references thereinRodriguez, Durán, (1988) Brazilian J. Med. Biol. Res., , and references therei

    Liposome effect on the cytochrome c-catalyzed peroxidation of carbonyl substrates to triplet species

    No full text
    Cytochrome c exhibits peroxidase activity on diphenylacetaldehyde (DPAA) and 3-methylacetoacetone (MAA), which is greatly affected by the presence and nature of charged liposome or micelle interfaces interacting with the enzyme. The ferricytochrome c reaction with DPAA is accelerated when the enzyme is attached to negatively charged interfaces. Whatever the medium, bulk solution or negatively charged dicetylphosphate (DCP), phosphatidyl coline/phosphatidylethanolamine/cardiolipin (PC/PE/CL) liposomes, this chemiluminescent reaction is accompanied by reduction of cytochrome c to its ferrous form. In turn, MAA is oxidized by cytochrome c exclusively when bound to DCP liposomes. Contrary to DPAA oxidation, the MAA reaction is followed by bleaching of cytochrome c, reflecting damage to the hemeprotein chromophore. The cytochrome-c-catalyzed oxidation of either DPAA or MAA leads to concomitant disappearance of the enzyme charge transfer absorption band at 695 nm. This suggests that the peroxidase activity of cytochrome c involves substrate-induced loss of the methionine ligand at the iron sixth coordination position, which is favored by interaction of cytochrome c with negatively charged interfaces. Accordingly, a decrease and blue shift of the charge transfer band could be observed in cytochrome-c-containing negatively charged DCP, PC/PE/CL liposomes or lysophosphatidylethanolamine micelles in the presence of DPAA or MAA.254176354655

    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

    Physico-chemical characterization of the inclusion complex between a 2-propen-1-amine derivative and beta-cyclodextrin

    No full text
    Inclusion complexes and physical mixtures were prepared with isomeric mixture of E/Z (50:50) of 3-(4'-bromo-[1,1'-biphenyl)-4-yl)-3-(4-bromophenyl)-N,N-dimethyl-2-propen-1-amine (BBAP) and beta-cyclodextrin (beta-CD) in different proportions. In this study, theoretical calculations using Molecular Mechanics MM+ force field were applied to predict the structures of the inclusion complexes formed by interaction of BBAP and beta-cyclodextrin. Circular dichroism, differential thermal analysis (DTA), X-ray diffraction and (13)C CP/MAS NMR methods were used to characterize the inclusion complexes and provide information about the stoichiometry of the inclusion complexes. The combined spectroscopy techniques indicate the formation of a complex of BBAP/beta-CD in the molar proportion of 1:1 and 1:2 by co-evaporation and no complexation was detected in the physical mixture of the compounds.50359159

    Violacein/β-cyclodextrin Inclusion Complex Formation Studied By Measurements Of Diffusion Coefficient And Circular Dichroism

    No full text
    The formation of inclusion compounds between violacein and β-cyclodextrin was studied by diffusion and circular dichroism measurements. The present work was undertaken to explore the feasibility of the β-cyclodextrin in reducing the toxicity and enhancing the antitumoral efficacy of violacein by forming an inclusion complex. The results of the two experiments are in good agreement, suggesting the formation of 1 : 1 and 1 : 2 complexes. The diffusion coefficient measurements enabled estimates of the sizes of the complexes involved. From the circular dichroism and computational calculations it was possible to view a preference for inclusion of the most polar part of the molecule to form a 1 : 2 inclusion complex. We expect that this work proves the potential of these techniques to determining complex stoichiometry.3701/04/156774Bortolus, P., Monti, S., (1996) Advances in Photochemistry, 21, p. 1. , D. C. Neckers, D. H. Volman and G. von Bün Wiley-Interscience, New YorkLipkowitz, K.B., (1998) Chem. Rev., 98, p. 1829Dekharsky, M.V., Inoue, Y., (1998) Chem. Rev., 98, p. 1875Saenger, W., (1980) Angew. Chem. Int. Ed. Engl., 19, p. 344Parker, D., Kataky, R., (1997) J. Chem. Soc., Chem. Commun., 2, p. 141Connors, K.A., (1997) Chem. Rev., 97, p. 1325Stella, V.J., Rajewski, R.A., (1997) Pharm. Res., 14, p. 556Koehler, G., Grabner, G., Klein, C.T.H., Marconi, G., Mayer, B., Monti, S., Rechthaler, K., Wolschann, P., (1996) J. Incl. Phenom. Mol. Recognit. Chem., 25, p. 103Beezer, A.E., Mitchell, J.C., (1992) Andrews: Pestic. Sci., 35, p. 375Loh, W., Beezer, A.E., Mitchell, J.C., (1994) Lagmuir, 10, p. 3431Lino, A.C.S., Loh, W., J. Incl. Phenom. Mol. Recognit. Chem., p. 1998. , submittedCohen, A.G.Y., (1997) J. Org. Chem., 62, p. 120Li, M.-X., Li, N.-Q., Gu, Z.-N., Zhou, N.-H., Sun, Y.-L., Wu, Y.-Q., (1996) Electrochim. Acta., 41, p. 2877Manzanares, M.I., Solis, V., De Rossi, R.H., (1996) J. Electroanal. Chem., 407, p. 141Uekama, K., Otagiri, M., Kanie, Y., Tanaka, S., Ikeda, K., (1975) Chem. Pharm. Bull., 23, p. 1421Nakahara, H., Tanaka, H., Kukuda, K., Matsumoto, M., Tagaki, W., (1990) Thin Solid Films, p. 284Guo, R., Chang, J., Lin, S., Chen, R., Hu, J., Zhang, H., (1996) Guangpuxue Yu Guangpu Fenxi., 16, p. 38(1994) Chem. Abstr., 125, p. 86074Hamai, S., (1997) J. Incl. Phenom. Mol. Recognit. Chem., 27, p. 57Mayer, B., Marconi, G., Klein, C., Köhler, G., Wolschann, P., (1997) J. Incl. Phenom. Mol. Recognit. Chem., 29, p. 79Marconi, G., Mayer, B., (1997) Pure Appl. Chem., 69, p. 779Wu, Y., Jin, J., (1966) Sichuan Daxue Xuebao, Ziran Kexueban., 33, p. 560(1997) Chem. Abstr., 126, p. 317579Shen, X., Belletête, M., Durocher, G., (1998) J. Phys. Chem. B, 102, p. 1877Harada, A., Lin, J., Kamachi, M., (1992) Nature, 356, p. 325Harada, A., Lin, J., Kamachi, M., (1994) Nature, 370, p. 126Durán, N., Faljoni-Alario, A., (1980) An. Acad. Brasil. Ciên., 52, p. 287Rettori, D., Durán, N., (1998) World J. Microbiol. Biotechnol., 14, p. 685Haun, M., Pereira, M.F., Hoffmann, M.E., Riveros, R., Joyas, A., Campos, V., Durán, N., (1992) Biol. Res., 25, p. 21Durán, N., Antônio, R.V., Haun, M., Pilli, R.A., (1994) World J. Microbiol. Biotechnol., 10, p. 686Durán, N., Erazo, S., Campos, V., (1983) An. Acad. Brasil. Ciênc., 55, p. 231(1984) Chem. Abstr., 100, pp. 48417bDurán, N., Melo, P.S., Haun, M., (1996) Proc. XXV Annual Meeting Brazilian Biochemical Society, p. 150. , (Escritorio & Editorial, São Paulo, S.P., Brazil), Caxambu, M.G., Brazil O-24Durán, N., Melo, P.S., Haun, M., Proc. VIII Brazilian National Meeting in Virology, p. 1996. , (Brazilian Soc. Virology Publ., UNESP, Jaboticabal, S. P., Brazil), São Lourenço, M.G., BrazilMelo, P.S., Haun, M., Durán, N., (1997) FASEB J., 11 (SUPPL.), pp. A1418Durán, N., Haun, M., Brazilian Patent PI 9702918 (1997)Singh, U.V., Udupa, N., (1997) Indian J. Physiol. Pharmacol., 41, p. 171Singh, U.V., Udupa, N., (1997) Pharm. Sci., 3, p. 573Loh, W., Tonegutti, C.A., Volpe, P.L.O., (1993) J. Chem. Soc. Faraday Trans., 89, p. 113Akizadeth, A., Nieto De Castro, C.A., Wakeham, W.A., (1980) Int. J. Thermophys., 1, p. 243Price, W.E., Trickett, R.A., Harris, K.R., (1989) J. Chem. Soc. Faraday Trans. 1, 85, p. 3281Eastel, A.E., Woolf, L.A., (1989) J. Chem. Soc. Faraday Trans. 1, 80, p. 1287Noulty, R.A., Leaist, D.G., (1987) J. Chem. Eng. Data, 32, p. 418Grabner, G., Monti, S., Marconi, G., Mayer, B., Klein, C., Köhler, G., (1996) J. Phys. Chem., 100, p. 2006

    Violacein/beta-cyclodextrin inclusion complex formation studied by measurements of diffusion coefficient and circular dichroism

    No full text
    The formation of inclusion compounds between violacein and beta-cyclodextrin was studied by diffusion and circular dichroism measurements. The present work was undertaken to explore the feasibility of the beta-cyclodextrin in reducing the toxicity and enhancing the antitumoral efficacy of violacein by forming an inclusion complex. The results of the two experiments are in good agreement, suggesting the formation of 1 : 1 and 1 : 2 complexes. The diffusion coefficient measurements enabled estimates of the sizes of the complexes involved. From the circular dichroism and computational calculations it was possible to view a preference for inclusion of the most polar part of the molecule to form a 1 : 2 inclusion complex. We expect that this work proves the potential of these techniques to determining complex stoichiometry.3741730677

    Liposome Effect On The Cytochrome C-catalyzed Peroxidation Of Carbonyl Substrates To Triplet Species.

    No full text
    Cytochrome c exhibits peroxidase activity on diphenylacetaldehyde (DPAA) and 3-methylacetoacetone (MAA), which is greatly affected by the presence and nature of charged liposome or micelle interfaces interacting with the enzyme. The ferricytochrome c reaction with DPAA is accelerated when the enzyme is attached to negatively charged interfaces. Whatever the medium, bulk solution or negatively charged dicetylphosphate (DCP), phosphatidylcholine/phosphatidylethanolamine/cardiolipin (PC/PE/CL) liposomes, this chemiluminescent reaction is accompanied by reduction of cytochrome c to its ferrous form. In turn, MAA is oxidized by cytochrome c exclusively when bound to DCP liposomes. Contrary to DPAA oxidation, the MAA reaction is followed by bleaching of cytochrome c, reflecting damage to the hemeprotein chromophore. The cytochrome-c-catalyzed oxidation of either DPAA or MAA leads to concomitant disappearance of the enzyme charge transfer absorption band at 695 nm. This suggests that the peroxidase activity of cytochrome c involves substrate-induced loss of the methionine ligand at the iron sixth coordination position, which is favored by interaction of cytochrome c with negatively charged interfaces. Accordingly, a decrease and blue shift of the charge transfer band could be observed in cytochrome-c-containing negatively charged DCP, PC/PE/CL liposomes or lysophosphatidylethanolamine micelles in the presence of DPAA or MAA.25546-5
    corecore