Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Polymers (Basel) ; 12(1)2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31861436

RESUMO

Here, we synthesize zwitterionic polymer brushes on polyester fabrics by atom transfer radical polymerization (ATRP) after a prefunctionalization step involving an aminolysis reaction with ethylenediamine. Aminolysis is an easy method to achieve homogeneous distributions of functional groups on polyester fibers (PET) fabrics. Varying the polymerization time and the prefunctionalization conditions of the reaction, it is possible to tune the amount of water retained over the surface and study its effect on protein adhesion. This study revealed that the polymerization time plays a major role in preventing protein adhesion on the PET surface.

2.
RSC Adv ; 9(17): 9783-9791, 2019 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-35520694

RESUMO

Antimicrobial finishes for textiles and other surfaces that act without the release of biocides to the environment (contact biocides) or by inhibiting microbial adhesion (antifouling action) are viewed as promising and environmentally friendly alternatives to current products. We have used polyvinylamine polymers that were functionalized with zwitterionic sulfobetaine side chains with different degrees of substitution (DS) for the finishing of poly(ethylene terephthalate) (PET) and cotton fabrics in a water-based pad-dry-cure process. After washing with different surfactants, a stable finish with total polymer add-ons of 0.2-0.5 wt% was achieved. The finished textiles efficiently inhibited the adhesion of proteins and bacteria to the surface even with a small DS as low as 20%. Textiles finished with polymers with a low DS also showed significant antibacterial activity, most notably against Staphylococcus aureus. Accordingly, textile finishes with either pure antiadhesive (DS > 50%) or combined antiadhesive and antibacterial properties (DS = 20-50%) are accessible using this approach.

4.
Chem Biol ; 18(7): 907-19, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21802011

RESUMO

Iron acquisition by siderophores is crucial for survival and virulence of many microorganisms. Here, we investigated the binding of the exogenous siderophore ferric enterobactin and the synthetic siderophore mimic ferric mecam by the triscatecholate binding protein FeuA from Bacillus subtilis at the atomic level. The structural complexes provide molecular insights into the capture mechanism of FeuA for exogenous and synthetic siderophores. The protein-ligand complexes show an exclusive acceptance of Λ-stereoconfigured substrates. Ligand-induced cross-bridging of the complexes was not observed, revealing a different thermodynamic behavior especially of the ferric mecam substrate, which was previously shown to dimerize with the enterobactin binding protein CeuE. The nearly identical overall domain movement of FeuA upon binding of ferric enterobactin or ferric mecam compared with endogenously derived ferric bacillibactin implies the importance of the conserved domain rearrangement for recognition by the transmembrane permease FeuBC, for which the conserved FeuA residues E90 and E221 were proved to be essential.


Assuntos
Bacillus subtilis/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Sideróforos/metabolismo , Bacillus subtilis/química , Sítios de Ligação , Cristalografia por Raios X , Enterobactina/metabolismo , Compostos Férricos/metabolismo , Modelos Moleculares , Ligação Proteica
6.
Acta Crystallogr Sect E Struct Rep Online ; 65(Pt 11): o2741, 2009 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-21578336

RESUMO

The absolute configuration has been determined for the title compound, C(14)H(20)N(2)OS. Inter-molecular N-H⋯O hydrogen bonds are observed in the crystal packing, forming infinitive one-dimensional chains with the base vector [100].

7.
Acta Crystallogr Sect E Struct Rep Online ; 65(Pt 11): o2742, 2009 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-21578337

RESUMO

The absolute configuration has been determined for the title compound, C(14)H(20)N(2)OS. There are two independent mol-ecules in the asymmetric unit. Inter-molecular N-H⋯O hydrogen bonds are observed in the crystal packing, forming infinite chains with the base vectors [100] and [010]. Each chain contains only one of the two independent mol-ecules.

8.
J Am Chem Soc ; 130(8): 2656-66, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18237171

RESUMO

Here, we report a new experimental approach utilizing an amide ligation reaction for the characterization of acyl carrier protein (ACP)-bound reaction intermediates, which are otherwise difficult to analyze by traditional biochemical methods. To explore fatty acid tailoring enzymes of the calcium-dependent antibiotic (CDA) biosynthetic pathway, this strategy enabled the transformation of modified fatty acids, covalently bound as thioesters to an ACP, into amide ligation products that can be directly analyzed and compared to synthetic standards by HPLC-MS. The driving force of the amide formation is the thermodynamic activation inherent to thioester-bound compounds. Using this novel method, we were able to characterize the ACP-mediated biosynthesis of the unique 2,3-epoxyhexanoyl moiety of CDA, revealing a new type of FAD-dependent oxidase HxcO with intrinsic enoyl-ACP epoxidase activity, as well as a second enoyl-ACP epoxidase, HcmO. In general, our approach should be widely applicable for the in vitro characterization of other biosynthetic systems acting on carrier proteins, such as integrated enzymes from NRPS and PKS assembly lines or tailoring enzymes of fatty and amino acid precursor synthesis.


Assuntos
Proteínas de Transporte/química , Ésteres/química , Ácidos Graxos/química , Lipoproteínas/química , Oxirredutases/química , Compostos de Sulfidrila/química , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Clonagem Molecular , Conformação Molecular , Oxirredutases/genética , Oxirredutases/isolamento & purificação
9.
J Am Chem Soc ; 128(47): 15084-5, 2006 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-17117848

RESUMO

Moenomycin A is the only known natural antibiotic that inhibits bacterial cell wall synthesis by binding to the transglycosylases that catalyze formation of the carbohydrate chains of peptidoglycan. We report here the total synthesis of moenomycin A using the sulfoxide glycosylation method. A newly discovered byproduct of sulfoxide reactions was isolated that resulted in substantial loss of the glycosyl acceptor. A general method to suppress this byproduct was introduced, which enabled the glycosylations to proceed efficiently. The inverse addition protocol for sulfoxide glycosylations also proved essential in constructing some of the glycosidic linkages. The synthetic route is flexible and will allow for derivatives to be constructed to further analyze moenomycin A's mechanism of action.


Assuntos
Antibacterianos/síntese química , Bambermicinas/síntese química , Sequência de Carboidratos , Glicosilação , Dados de Sequência Molecular
10.
J Am Chem Soc ; 127(30): 10747-52, 2005 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-16045364

RESUMO

Glycosyltransferases involved in the biosynthesis of bacterial secondary metabolites may be useful for the generation of sugar-modified analogues of bioactive natural products. Some glycosyltransferases have relaxed substrate specificity, and it has been assumed that promiscuity is a feature of the class. As part of a program to explore the synthetic utility of these enzymes, we have analyzed the substrate selectivity of glycosyltransferases that attach similar 2-deoxy-L-sugars to glycopeptide aglycons of the vancomycin-type, using purified enzymes and chemically synthesized TDP beta-2-deoxy-L-sugar analogues. We show that while some of these glycopeptide glycosyltransferases are promiscuous, others tolerate only minor modifications in the substrates they will handle. For example, the glycosyltransferases GtfC and GtfD, which transfer 4-epi-L-vancosamine and L-vancosamine to C-2 of the glucose unit of vancomycin pseudoaglycon and chloroorienticin B, respectively, show moderately relaxed donor substrate specificities for the glycosylation of their natural aglycons. In contrast, GtfA, a transferase attaching 4-epi-L-vancosamine to a benzylic position, only utilizes donors that are closely related to its natural TDP sugar substrate. Our data also show that the spectrum of donors utilized by a given enzyme can depend on whether the natural acceptor or an analogue is used, and that GtfD is the most versatile enzyme for the synthesis of vancomycin analogues.


Assuntos
Antibacterianos/síntese química , Glicosiltransferases/química , Vancomicina/análogos & derivados , Antibacterianos/biossíntese , Glicosilação , Glicosiltransferases/isolamento & purificação , Glicosiltransferases/metabolismo , Hexosaminas/química , Hexosaminas/metabolismo , Isoenzimas , Cinética , Especificidade por Substrato , Vancomicina/síntese química , Vancomicina/química , Vancomicina/metabolismo
11.
Chem Biol ; 12(5): 527-34, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15911373

RESUMO

During biosynthesis of the anthracycline antitumor agents daunomycin, adriamycin, and aclacinomycin, the polyketide-derived tetracyclic aglycone is enzymatically glycosylated at the C7-OH by dedicated glycosyltransferases (Gtfs) that transfer L-2,3,6-trideoxy-3-aminohexoses. In aclacinomycins, the first deoxyhexose is predicted to be transferred via AknS action, then subjected to further elongation to a trisaccharide by the subsequent Gtf, AknK. We report here that purified AknS has very low activity in the absence of the adjacently encoded AknT; however, at a 3:1 ratio, AknT stimulates AknS k(cat) by 40-fold up to 0.22 min(-1) for transfer of L-2-deoxyfucose (2-dF) to the aglycone aklavinone. It is likely that several other Gtfs that glycosylate polyketide aglycones also act as two-component catalytic systems. Incubations of purified AknS/AknT/AknK with two aglycones and two dTDP-2-deoxyhexoses produced previously uncharacterized anthracycline disaccharides.


Assuntos
Antraciclinas/metabolismo , Proteínas de Bactérias/metabolismo , Glicosídeos/metabolismo , Glicosiltransferases/metabolismo , Aclarubicina/química , Aclarubicina/metabolismo , Antraciclinas/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Dissacarídeos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Vetores Genéticos , Glicosídeos/química , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/isolamento & purificação , Naftacenos/química , Naftacenos/metabolismo , Açúcares de Nucleosídeo Difosfato/metabolismo , Ligação Proteica , Streptomyces/genética , Streptomyces/metabolismo
12.
Biochemistry ; 44(12): 4949-56, 2005 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15779922

RESUMO

Simocyclinone D(8) consists of an anguicycline C-glycoside tethered by a tetraene diester linker to an aminocoumarin. Unlike the antibiotics novobiocin, clorobiocin, and coumermycin A(1), the phenolic hydroxyl group of the aminocoumarin in simocyclinone is not glycosylated with a decorated noviosyl moiety that is the pharmacophore for targeting bacterial DNA gyrase. We have expressed the Streptomyces antibioticus simocyclinone ligase SimL, purified it from Escherichia coli, and established its ATP-dependent amide bond forming activity with a variety of polyenoic acids including retinoic acid and fumagillin. We have then used the last three enzymes from the novobiocin pathway, NovM, NovP, and NovN, to convert a SimL product to a novel novobiocin analogue, in which the 3-prenyl-4-hydroxybenzoate of novobiocin is replaced with a tetraenoate moiety, to evaluate antibacterial activity.


Assuntos
Amida Sintases/química , Cumarínicos/química , Cumarínicos/metabolismo , Glicosídeos/biossíntese , Glicosídeos/química , Novobiocina/análogos & derivados , Novobiocina/metabolismo , Streptomyces antibioticus/enzimologia , Amida Sintases/biossíntese , Amida Sintases/genética , Amida Sintases/isolamento & purificação , Aminocumarinas , Antibacterianos/farmacologia , Ácidos Carboxílicos/química , Ácidos Carboxílicos/metabolismo , Regulação Bacteriana da Expressão Gênica , Novobiocina/biossíntese , Novobiocina/química , Streptomyces antibioticus/genética , Especificidade por Substrato
13.
J Am Chem Soc ; 127(10): 3250-1, 2005 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-15755121

RESUMO

The glycopeptide antibiotics prevent maturation of the bacterial cell wall by binding to the terminal d-alanyl-d-alanine moiety of peptidoglycan precursors, thereby inhibiting the enzymes involved in the final stages of peptidoglycan synthesis. However, there are significant differences in the biological activity of particular glycopeptide derivatives that are not related to their affinity for d-Ala-d-Ala. We compare the ability of vancomycin and a set of clinically relevant glycopeptides to inhibit Staphylococcus aureus PBP2 (penicillin binding protein), the major transglycosylase in a clinically relevant pathogen, S. aureus. We report experiments suggesting that activity differences between glycopeptides against this organism reflect a combination of substrate binding and secondary interactions with key enzymes involved in peptidoglycan synthesis.


Assuntos
Antibacterianos/farmacologia , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Staphylococcus aureus/enzimologia , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Vancomicina/farmacologia , Antibacterianos/química , Antibacterianos/metabolismo , Dipeptídeos/química , Dipeptídeos/metabolismo , Cinética , Testes de Sensibilidade Microbiana , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Especificidade por Substrato , Uridina Difosfato Ácido N-Acetilmurâmico/química , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo , Vancomicina/química , Vancomicina/metabolismo
14.
Chem Biol ; 12(1): 131-40, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15664522

RESUMO

The teicoplanin acyltransferase (Atf) responsible for N-acylation of the glucosamine moiety to create the teicoplanin lipoglycopeptide scaffold has recently been identified. Here we use that enzyme (tAtf) and the cognate acyltransferase from the related A-40,926 biosynthetic cluster (aAtf) to evaluate specificity for glycopeptide scaffolds and for the acyl-CoA donor. In addition to acylation of 2-aminoglucosyl glycopeptide scaffolds with k(cat) values of 400-2000 min(-1), both Atfs transfer acyl groups to regioisomeric 6-aminoglucosyl scaffolds and to glucosyl scaffolds at rates of 0.2-0.5 min(-1) to create variant lipoglycopeptides. Using the teicoplanin glycosyltransferase tGtfA, tAtf, and GtfD, a glycosyltransferase from the vancomycin producer, it is possible to assemble a novel lipoglycopeptide with GlcNAc at beta-OH-Tyr(6) and an N(6)-acyl-glucosaminyl-vancosamine at Phegly(4). This study illustrates the utility of chemo- and regioselective acyltransferases and glycosyltransferases to create novel lipoglycopeptides.


Assuntos
Aciltransferases/química , Antibacterianos/química , Glicopeptídeos/síntese química , Óperon , Teicoplanina/química , Acil Coenzima A/química , Aciltransferases/genética , Aciltransferases/isolamento & purificação , Clonagem Molecular , Glicopeptídeos/química , Concentração de Íons de Hidrogênio , Cinética , Conformação Molecular , Sensibilidade e Especificidade , Estereoisomerismo , Teicoplanina/síntese química , Fatores de Tempo
15.
Biochemistry ; 43(47): 15022-36, 2004 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-15554710

RESUMO

Coumermycin A(1) is a member of the aminocoumarin family of antibiotics. Unlike its structural relatives, novobiocin and clorobiocin, coumermycin A(1) is a dimer built on a 3-methyl-2,4-dicarboxypyrrole scaffold and bears two decorated noviose sugar components which are the putative target binding motifs for DNA gyrase. Starting with this scaffold, we have utilized the ligase CouL for mono- and bisamide formation with aminocoumarins to provide substrates for the glycosyltransferase CouM. CouM was subsequently shown to catalyze mono- and bisnoviosylation of the resulting CouL products. CouP was shown to possess 4'-O-methyltransferase activity on products from tandem CouL, CouM assays. A fourth enzyme, NovN, the 3'-O-carbamoyltransferase from the novobiocin operon, was then able to carbamoylate either or both arms of the CouP product. The tandem action of CouL, CouM, CouP, and NovN thus generates a biscarbamoyl analogue of the pseudodimer coumermycin A(1). Starting from alternative dicarboxy scaffolds, these four enzymes can be utilized in tandem to create additional variants of dimeric aminocoumarin antibiotics.


Assuntos
Antibacterianos/biossíntese , Antibacterianos/química , Cumarínicos/química , Cumarínicos/metabolismo , Variação Genética , Ligases/metabolismo , Aminocumarinas , Antibacterianos/isolamento & purificação , Cromatografia Líquida , Cumarínicos/isolamento & purificação , Dimerização , Escherichia coli/genética , Glicosiltransferases/metabolismo , Ligases/química , Espectrometria de Massas , Estrutura Molecular , Streptomyces/genética , Especificidade por Substrato
16.
Org Lett ; 6(17): 2873-6, 2004 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15330636

RESUMO

Beta-2-Deoxy sugar nucleotides are substrates used by a variety of glycosyltransferases (Gtfs). We have developed a chemical route to synthesize beta-2-deoxy sugar phosphates that starts from alpha-glycosyl chlorides. Our approach reliably provides access to a range of NDP beta-2-deoxy sugars essential for studying glycosyltransferases involved in the synthesis of biologically active natural products.


Assuntos
Glicosiltransferases/metabolismo , Fosfatos Açúcares/síntese química , Modelos Moleculares , Estrutura Molecular , Estereoisomerismo
17.
Biochemistry ; 43(15): 4548-58, 2004 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15078101

RESUMO

The antitumor drug aclacinomycin A is a representative member of the anthracycline subgroup that contains a C(7)-O-trisaccharide chain composed of L-2-deoxysugars. The sugar portion of the molecule, which greatly affects its biological activity, is assembled by dedicated glycosyltransferases; however, these enzymes have not been well-studied. Here we report the heterologous expression and purification of one of these enzymes, AknK, as well as the preparation of dTDP-L-2-deoxysugar donors, dTDP-L-2-deoxyfucose and dTDP-L-daunosamine, and the monoglycosyl aglycone, rhodosaminyl aklavinone. Our experiments show that AknK catalyzes the addition of the second sugar to the chain, using dTDP-L-2-deoxyfucose and rhodosaminyl aklavinone, to create the L-2-deoxyfucosyl-L-rhodosaminyl aklavinone. AknK also accepts an alternate dTDP-L-sugar, dTDP-L-daunosamine, and other monoglycosylated anthracyclines, including daunomycin, adriamycin, and idarubicin, to build alternate disaccharides on variant anthracycline backbones. Remarkably, AknK also catalyzes a tandem addition of a second L-2-deoxyfucosyl moiety, albeit with reduced activity, to the natural disaccharide chain to produce L-deoxyfucosyl-L-deoxyfucosyl-L-rhodosaminyl aklavinone, a variant of the natural aclacinomycin A. These results demonstrate that AknK may be a useful enzyme for the chemoenzymatic synthesis of anthracycline variants.


Assuntos
Aclarubicina/biossíntese , Fucosiltransferases/isolamento & purificação , Streptomyces/enzimologia , Aclarubicina/análogos & derivados , Catálise , Clonagem Molecular , Fucose/análogos & derivados , Fucose/biossíntese , Fucose/química , Fucosiltransferases/biossíntese , Fucosiltransferases/genética , Glicosilação , Hexosaminas/biossíntese , Hexosaminas/química , Naftacenos/síntese química , Streptomyces/genética , Especificidade por Substrato , Nucleotídeos de Timina/síntese química , Trissacarídeos/química
18.
Proc Natl Acad Sci U S A ; 101(13): 4390-5, 2004 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-15070728

RESUMO

Chloroeremomycin, a vancomycin family glycopeptide antibiotic has three sugars, one D-glucose and two L-4-epi-vancosamines, attached to the crosslinked heptapeptide backbone by three glycosyltransferases, GtfA, -B, and -C. Prior efforts have revealed that GtfB and -C in tandem build an epivancosaminyl-1,2-glucosyldisaccharide chain on residue 4 of the aglycone; however, the characterization of GtfA and glycosylation sequence of chloroeremomycin have been lacking. Here, we report the expression and purification of GtfA, as well as synthesis of its sugar donor, 2'-deoxy-thymidine 5'-diphosphate (dTDP)-beta-L-4-epi-vancosamine. GtfA transfers 4-epi-vancosamine from the chemically synthesized dTDP-4-epi-vancosamine to the beta-OH-Tyr6 residue of the aglycone, preferentially after GtfB action, to generate chloroorienticin B. With the preferred kinetic order of GtfB, then GtfA, then GtfC established, we have succeeded in reconstitution of chloroeremomycin from the heptapeptide aglycone by the sequential actions of the three enzymes.


Assuntos
Actinomycetales/enzimologia , Transaminases/genética , Transaminases/metabolismo , Vancomicina/análogos & derivados , Vancomicina/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Clonagem Molecular , Escherichia coli/enzimologia , Escherichia coli/genética , Glicosilação , Cinética , Especificidade por Substrato , Transaminases/isolamento & purificação
20.
Biochemistry ; 42(14): 4179-89, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12680772

RESUMO

The aminocoumarin class of antibiotics, exemplified by novobiocin, is composed of tripartite l-noviosylaminocoumarin prenylbenzoate natural products. The decorated noviosyl sugar component interacts with the target bacterial enzyme DNA gyrase. We have subcloned the putative 40 kDa l-noviosyl transferase from Streptomyces spheroides into Escherichia coli, expressed it in soluble form, and purified it to homogeneity as a C-terminal His(8) fusion protein. The aglycone novobiocic acid, obtained from selective degradation of novobiocin, and TDP-l-noviose, obtained by an 11-step chemical synthesis from l-rhamnose, were shown to be robust substrates for NovM to produce the desmethyldescarbamoyl novobiocin intermediate with a k(cat) of >300 min(-1). NovM displays activity with variant coumarin aglycones, suggesting it may be a promiscuous catalyst for noviosylation of a range of planar scaffolds. Conversely, NovM shows no activity with and is inhibited by TDP-l-rhamnose (K(i) = 83.5 +/- 5.5 microM), the sugar donor that most closely structurally resembles the natural substrate TDP-l-noviose. The NovM reaction products generated during the course of this work will serve as substrates for subsequent analysis of the NovP and NovN tailoring enzymes that impart the noviose decorations required for DNA gyrase binding and antibiotic activity.


Assuntos
Antibacterianos/biossíntese , Novobiocina/biossíntese , Streptomyces/enzimologia , Transferases/metabolismo , Sequência de Bases , Primers do DNA , Inibidores Enzimáticos/farmacologia , Escherichia coli/genética , Ressonância Magnética Nuclear Biomolecular , Especificidade por Substrato , Transferases/antagonistas & inibidores , Transferases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA