• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • 1
  • Tagged with
  • 4
  • 4
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Engineering carboxymethylproline synthases towards the biosynthetic productions of carbapenem antibiotics

Gómez Castellanos, José Rubén January 2013 (has links)
Mechanistic and biocatalytic studies of two carboxymethylproline synthases (CMPSs), CarB and ThnE, members of the crotonase superfamily of enzymes, both in isolation and in conjunction with the activity of the crotonyl-CoA carboxylase/reductase (Ccr) the malonyl-CoA synthetase (MatB) and the methylmalonyl-CoA epimerase (MCE) are presented. Protein engineering studies on carboxymethylproline synthases aimed at enabling stereoselective C–C bond formation leading to N-heterocycles via control of trisubstituted enolate intermediates were carried out. Active site substitutions, including at the oxyanion binding site, enabled the production of substituted N-heterocycles in high diastereomeric excesses via stereocontrolled enolate formation and reaction. The biocatalytic promiscuity of malonyl-CoA ligase and the stereoselectivity of crotonyl–CoA carboxylase/reductase were successfully coupled to the selective tri- substituted enolate forming capacity of engineered carboxymethylproline synthases for the preparation of functionalized 5- and 6-membered N-heterocycles substituted with a variety of alkyl side chains at the C-5/C-6 positions at high diastereomeric excess. The effect of methylmalonyl-CoA epimerase on the diastereoselectivity of the carboxymethylproline synthase-catalysed enolated alkylation was also demonstrated. The results illustrate the utility of the crotonase superfamily of enzymes for stereoselective biocatalysis and demonstrate the power of coupled enzyme systems to enhance diastereoselectivity and to expand the range of accepted substrates.
2

Caractérisation de voies de biosynthèse d’antibiotiques de la famille des pyrrolamides / Characterization of pyrrolamide antibiotics biosynthetic pathways

Vingadassalon, Audrey 17 May 2013 (has links)
Les pyrrolamides constituent une famille de produits naturels dotés de diverses activités biologiques et synthétisés par des actinobactéries. La congocidine et la distamycine, les molécules les plus connues de cette famille, sont capables de se lier à l'ADN de façon non covalente selon une certaine spécificité de séquence (succession de 4 paires de base A/T). Récemment, les gènes et la voie de biosynthèse de la congocidine ont été identifiés et caractérisés chez S. ambofaciens. Ceci a révélé un mécanisme original impliquant notamment de nouvelles enzymes et de nouvelles voies pour la biosynthèse des trois précurseurs nécessaires à l’assemblage de la congocidine. Nous avons entrepris d’étudier la régulation de la biosynthèse de la congocidine chez S. ambofaciens et d’isoler et de caractériser les groupes de gènes de biosynthèse de deux autres pyrrolamides, la distamycine et les pyrronamycines (produites respectivement par S distallicus et un streptomyces non caractérisé). L'objectif de cette étude est, dans un premier temps, d’améliorer notre compréhension des mécanismes impliqués lors de la biosynthèse de ces molécules (comme le mécanisme d’incorporation des pyrroles) et, par la suite, de manipuler les gènes identifiés pour synthétiser de nouvelles molécules pyrrolamides hybrides. / Pyrrolamides constitute a family of natural products with various biological activities, synthesized by actinobacteria. Congocidine (also called netropsin) and distamycin are the best characterized pyrrolamides, largely studied due to their ability to bind into the minor groove of the DNA double helix in a sequence specific manner (succession of four A/T bases). Recently, the congocidine biosynthetic pathway has been characterized in Streptomyces ambofaciens. We showed that an iterative Non Ribosomal Peptide Synthetase with an unusual architecture assembles congocidine, using precursors with undocumented biosynthetic pathways. With the aim of developing a combinatorial biosynthesis approach for the development of new pyrrolamides, we undertook the study of the regulation of congocidine biosynthesis in S. ambofaciens and the isolation of the distamycin and pyrronamycins biosynthetic gene clusters. Characterization of these clusters will result in a more detailed understanding of pyrrolamide biosynthesis (e.g. mechanism of pyrrole polymerization), and provide new tools (enzymes) and building blocks (precursors) necessary for combinatorial biosynthesis.
3

ISOLATION AND ELUCIDATION OF THE CHRYSOMYCIN BIOSYNTHETIC GENE CLUSTER AND ALTERING THE GLYCOSYLATION PATTERNS OF TETRACENOMYCINS AND MITHRAMYCIN-PATHWAY MOLECULES

Nybo, Stephen Eric 01 January 2011 (has links)
Natural products occupy a central role as the majority of currently used antibiotic and anticancer agents. Among these are type-II polyketide synthase (PKS)-derived molecules, or polyketides, which are produced by many representatives of the genus Streptomyces. Some type-II polyketides, such as the tetracyclines and the anthracycline doxorubicin, are currently employed as therapeutics. However, several polyketide molecules exhibit promising biological activity, but due to toxic side effects or solubility concerns, remain undeveloped as drugs. Gilvocarcin V (GV) (topoisomerase II inhibitor) has a novel mechanism of action: [2+2] cycloaddition to thymine residues by the 8-vinyl side chain and cross-linking of histone H. Mithramycin blocks transcription of proto-oncogenes c-myc and c-src by forming an Mg2+-coordinated homodimer in the GC-rich minor groove of DNA. The purpose of this research was to investigate the biosynthesis of several type II polyketide compounds (e.g. chrysomycin, elloramycin, and mithramycin) with the goal of improving the bioactivities of these drugs through combinatorial biosynthesis. Alteration of the glycosylation pattern of these molecules is one promising way to improve or alter the bioactivities of these molecules. To this end, an understanding of the glycosyltransferases and post-polyketide tailoring enzymatic steps involved in these biosynthetic pathways must be established. Four specific aims were established to meet these goals. In specific aim 1, the biosynthetic locus of chrysomycin A was successfully cloned and elucidated, which afforded novel biosynthetic tools. Chrysomycin monooxygenases were found to catalyze identical roles to their gilvocarcin counterparts. Cloning of deoxysugar constructs (plasmids) which could direct biosynthesis of ketosugars, NDP-D-virenose, and NDP-D-fucofuranose in foreign pathways was undertaken in specific aim 2. Finally, these “sugar” plasmids were introduced into producer organisms of elloramycin and mithramycin pathways in specific aims 3 and 4 to interrogate the endogenous glycosyltransferases in order to alter their glycosylation patterns. These experiments resulted in the successful generation of a newly glycosylated tetracenomycin, as well as premithramycin, and mithramycin analogues. In specific aim 4, a new mithramycin analogue with an altered sugar pattern rationally designed and improved structural features was generated and structurally elucidated.
4

COMBINATORIAL BIOSYNTHETIC DERIVATIZATION OF THE ANTITUMORAL AGENT GILVOCARCIN V

Shepherd, Micah Douglas 01 January 2011 (has links)
Gilvocarcin V (GV), the principal product of Streptomyces griseoflavus Gö 3592 and other Streptomyces spp., is the most prominent member of a distinct class of antitumor antibiotics that share a polyketide derived coumarin-based aromatic core. GV and other members of this class including polycarcin V from Streptomyces polyformus, often referred to as gilvocarcin-like aryl C-glycosides, are particularly interesting because of their potent bactericidal, virucidal and antitumor activities at low concentrations while maintaining low in vivo toxicity. Although the precise molecular mechanism of GV bioactivity is unknown, gilvocarcin V has been shown to undergo a photoactivated [2+2] cycloaddition of its vinyl side chain with thymine residues of DNA in near-UV or visible blue light. In addition, GV was shown to selectively crosslink histone H3 with DNA, thereby effectively disrupting normal cellular processes such as transcription. Furthermore, GVs ability to inhibit topoisomerase II has also been attributed as a mechanism of action for gilvocarcin V activity. The excellent antitumor activity, as well as an unprecedented structural architecture, has made GV an ideal candidate for biosynthetic studies toward the development of novel analogues with improved pharmacological properties. Previous biosynthetic research has identified several candidate genes responsible for key steps during the biosynthesis of gilvocarcin V including an oxygenase cascade leading to C-C bond cleavage, methylations, lactone formation, C-glycosylation and vinyl side chain formation. In this study, we further examined two critical biosynthetic transformations essential for the bioactivity of gilvocarcin V, namely starter unit incorporation and C-glycosylation, through the following specific aims: 1) creation of functional chimeric C-glycosyltransferases through domain swapping of gilvocarcin-like glycosyltransferases and identification and evaluation of the donor substrate flexibility of PlcGT, the polycarcin V pathway specific C-glycosyltransferase; 2) creation of a library of O-methylated-L-rhamnose analogues of polycarcin V for structure activity relationship studies; 3) identification of the role of GilP and GilQ in starter unit specificity during gilvocarcin V biosynthesis; and 4) creation of a plasmid based approach in which selective gilvocarcin biosynthetic genes were utilized to produce important gilvocarcin intermediates for further in vivo and in vitro experimentation.

Page generated in 0.1042 seconds