Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Intern Med ; 269(6): 604-13, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21401739

RESUMO

Coeliac disease is a widespread, lifelong disorder for which dietary control represents the only accepted form of therapy. There is an unmet need for nondietary therapies to treat this condition. Most ongoing and emerging drug-discovery programmes are based on the understanding that coeliac disease is caused by an inappropriate T-cell-mediated immune response to dietary gluten proteins. Recent genome-wide association studies lend further support to this pathogenic model. The central role of human leucocyte antigen genes has been validated, and a number of new risk loci have been identified, most of which are related to the biology of T cells and antigen-presenting cells. Here, we review the status of potential nondietary therapies under consideration for coeliac disease. We conclude that future development of novel therapies will be aided considerably by the identification of new, preferably noninvasive, surrogate markers for coeliac disease activity.


Assuntos
Doença Celíaca/tratamento farmacológico , Fármacos Gastrointestinais/uso terapêutico , Dessensibilização Imunológica/métodos , Descoberta de Drogas/métodos , Inibidores Enzimáticos/uso terapêutico , Proteínas de Ligação ao GTP/antagonistas & inibidores , Glutens/imunologia , Glutens/farmacocinética , Humanos , Inativação Metabólica , Terapia de Alvo Molecular/métodos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/antagonistas & inibidores
2.
Oncogene ; 26(18): 2563-73, 2007 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-17099729

RESUMO

Transglutaminase 2 (TG2, a.k.a. tissue transglutaminase) belongs to a family of transglutaminase enzymes that stabilize proteins by affecting covalent crosslinking via formation of amide bonds. Cell surface TG2 is directly involved as an adhesive receptor in cell-extracellular matrix (ECM) interactions. Here, we show that TG2 activity is elevated in glioblastomas compared with non-neoplastic brain. Immunofluorescent studies showed increased staining of fibronectin colocalized with TG2 in the ECM in glioblastomas. In addition, small clusters of invading human glioblastoma cells present in non-neoplastic brain parenchyma secrete high levels of TG2 and fibronectin that distinguish them from normal brain stroma. Downregulation of TG2 in U87MG glioblastoma cells with RNAi demonstrated decreased assembly of fibronectin in the ECM. Treatment with KCC009 blocked the remodeling of fibronectin in the ECM in glioblastomas in both in vitro and in vivo studies. KCC009 treatment in mice harboring orthotopic glioblastomas (DBT-FG) sensitized the tumors to N,N'-bis(2-chloroethyl)-N-nitrosourea chemotherapy, as measured by reduced bioluminescence, increased apoptosis and prolonged survival. The ability of KCC009 to interfere with the permissive remodeling of fibronectin in the ECM in glioblastomas suggests a novel target to enhance sensitivity to chemotherapy directed not only at the tumor mass, but also invading glioblastoma cells.


Assuntos
Inibidores Enzimáticos/farmacologia , Matriz Extracelular/efeitos dos fármacos , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Isoxazóis/farmacologia , Transglutaminases/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Carmustina/farmacologia , Matriz Extracelular/metabolismo , Feminino , Imunofluorescência , Proteínas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Interferente Pequeno/farmacologia , Taxa de Sobrevida , Transglutaminases/metabolismo , Células Tumorais Cultivadas
3.
Cell Mol Life Sci ; 64(3): 345-55, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17160352

RESUMO

This review describes the structure and function of prolyl endopeptidase (PEP) enzymes and how they are being evaluated as drug targets and therapeutic agents. The most well studied PEP family has a two-domain structure whose unique seven-blade beta-propeller domain works with the catalytic domain to hydrolyze the peptide bond on the carboxyl side of internal proline residues of an oligopeptide substrate. Structural and functional studies on this protease family have elucidated the mechanism for peptide entry between the two domains. Other structurally unrelated PEPs have been identified, but have not been studied in detail. Human PEP has been evaluated as a pharmacological target for neurological diseases due to its high brain concentration and ability to cleave neuropeptides in vitro. Recently, microbial PEPs have been studied as potential therapeutics for celiac sprue, an inflammatory disease of the small intestine triggered by proline-rich gluten.


Assuntos
Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Animais , Doença Celíaca/enzimologia , Doença Celíaca/terapia , Humanos , Concentração de Íons de Hidrogênio , Prolil Oligopeptidases , Especificidade por Substrato , Termodinâmica , Distribuição Tecidual
4.
J Antibiot (Tokyo) ; 59(8): 464-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17080682

RESUMO

For the heterologous production of ansamycin polyketides such as rifamycin and geldanamycin in Escherichia coli, a number of unusual but important tools must be engineered into the bacterium. Here we demonstrate efficient production of the starter unit 3-amino-5-hydroxybenzoic acid (AHBA) and the methoxymalonyl extender unit in E. coli. Previous work has demonstrated the production of the ansamycin starter unit AHBA in E. coli in low yield. It was shown that the low yield is primarily due to acetylation of AHBA into N-acetyl-AHBA. Three methods for minimizing this side reaction were evaluated. First, a putative N-arylamine-acetyltransferase (NAT) was deleted from the E. coli chromosome, although this did not alter N-acetyl-AHBA production. Next, E. coli grown in media devoid of glucose yielded a nearly equal mixture of AHBA and N-acetyl-AHBA. Lastly, the NAT inhibitor glycyrrhizic acid was shown to further inhibit the acetylation reaction. The entire set of genes for synthesizing the methoxymalonyl extender unit was transferred from the geldanamycin producer Streptomyces hygroscopicus into E. coli. The pathway specific ACP isolated from the resulting recombinant strain was found to predominantly occur as methyoxymalonyl-ACP. Together, these findings set the stage for engineered biosynthesis of ansamycin polyketides in E. coli.


Assuntos
Antibacterianos/biossíntese , Escherichia coli/metabolismo , Rifabutina/metabolismo , Aminobenzoatos/metabolismo , Antibacterianos/química , Escherichia coli/genética , Genes Bacterianos , Engenharia Genética , Hidroxibenzoatos , Lactamas Macrocíclicas , Plasmídeos/genética , Rifabutina/análogos & derivados , Rifabutina/química
5.
Proc Natl Acad Sci U S A ; 98(26): 14808-13, 2001 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11752428

RESUMO

As the first structural elucidation of a modular polyketide synthase (PKS) domain, the crystal structure of the macrocycle-forming thioesterase (TE) domain from the 6-deoxyerythronolide B synthase (DEBS) was solved by a combination of multiple isomorphous replacement and multiwavelength anomalous dispersion and refined to an R factor of 24.1% to 2.8-A resolution. Its overall tertiary architecture belongs to the alpha/beta-hydrolase family, with two unusual features unprecedented in this family: a hydrophobic leucine-rich dimer interface and a substrate channel that passes through the entire protein. The active site triad, comprised of Asp-169, His-259, and Ser-142, is located in the middle of the substrate channel, suggesting the passage of the substrate through the protein. Modeling indicates that the active site can accommodate and orient the 6-deoxyerythronolide B precursor uniquely, while at the same time shielding the active site from external water and catalyzing cyclization by macrolactone formation. The geometry and organization of functional groups explain the observed substrate specificity of this TE and offer strategies for engineering macrocycle biosynthesis. Docking of a homology model of the upstream acyl carrier protein (ACP6) against the TE suggests that the 2-fold axis of the TE dimer may also be the axis of symmetry that determines the arrangement of domains in the entire DEBS. Sequence conservation suggests that all TEs from modular polyketide synthases have a similar fold, dimer 2-fold axis, and substrate channel geometry.


Assuntos
Esterases/química , Proteínas de Membrana/química , Complexos Multienzimáticos/química , Sequência de Aminoácidos , Dimerização , Leucina/química , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos
6.
Biochemistry ; 40(49): 14855-61, 2001 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-11732905

RESUMO

Biosynthesis of the carbon chain backbone of the R1128 substances is believed to involve the activity of a ketosynthase/chain length factor (ZhuB/ZhuA), an additional ketosynthase (ZhuH), an acyl transferase (ZhuC), and two acyl carrier proteins (ACPs; ZhuG and ZhuN). A subset of these proteins initiate chain synthesis via decarboxylative condensation between an acetyl-, propionyl-, isobutyryl-, or butyryl-CoA derived primer unit and a malonyl-CoA derived extender unit to yield an acetoacetyl-, beta-ketopentanoyl-, 3-oxo-4-methylpentanoyl-, or beta-ketohexanoyl-ACP product, respectively. To investigate the precise roles of ZhuH, ZhuC, ZhuG, and ZhuN, each protein was expressed in Escherichia coli and purified to homogeneity. Although earlier reports had proposed that ZhuC and its homologues played a role in primer unit selection, direct in vitro analysis of ZhuC showed that it was in fact a malonyl-CoA:ACP malonyltransferase (MAT). The enzyme could catalyze malonyl transfer but not acetyl- or propionyl-transfer onto R1128 ACPs or onto ACPs from other biosynthetic pathways, suggesting that ZhuC has broad substrate specificity with respect to the holo-ACP substrate but is specific for malonyl-CoA. Thus, ZhuC supplies extender units to both the initiating and elongating ketosynthases from this pathway. To interrogate the primer unit specificity of ZhuH, the kinetics of beta-ketoacyl-ACP formation in the presence of various acyl-CoAs and malonyl-ZhuG were measured. Propionyl-CoA and isobutyryl-CoA were the two most preferred substrates of ZhuH, although acetyl-CoA and butyryl-CoA could also be accepted and elongated. This specificity is not only consistent with earlier reports demonstrating that R1128B and R1128C are the major products of the R1128 pathway in vivo, but is also in good agreement with the properties of the ZhuH substrate binding pocket, as deduced from a recently solved crystal structure of the enzyme. Finally, to investigate the molecular logic for the occurrence of not one but two ACP genes within the R1128 gene cluster, the inhibition of ZhuH-catalyzed formation of beta-ketopentanoyl-ACP was quantified in the presence of apo-ZhuG or apo-ZhuN. Both apo-proteins were comparable inhibitors of the ZhuH catalyzed reaction, suggesting that the corresponding apo-proteins can be used interchangeably during chain initiation. Together, these results provide direct biochemical insights into the mechanism of chain initiation of an unusual bacterial aromatic PKS.


Assuntos
Complexos Multienzimáticos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/metabolismo , Coenzima A/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Estrutura Molecular , Complexos Multienzimáticos/genética , Família Multigênica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Transferases/genética , Transferases/isolamento & purificação , Transferases/metabolismo
7.
Biochemistry ; 40(41): 12407-11, 2001 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-11591161

RESUMO

Fatty acids and polyketides are synthesized by mechanistically and evolutionarily related multienzyme systems. Their carbon chain backbones are synthesized via repeated decarboxylative condensations of alpha-carboxylated building blocks onto a growing acyl chain. These alpha-carboxylated building blocks are transferred from the corresponding coenzyme A thioesters onto the phosphopantetheine arm of an acyl carrier protein (ACP) by acyl transferases, which operate by a ping-pong mechanism involving an acyl-O-serine intermediate. In the course of our studies on the malonyl-CoA:ACP transacylase (MAT) from Streptomyces coelicolor, we observed that an active-site Ser (97) --> Ala mutant retains activity as well as the ability to be covalently labeled by (14)C malonyl-CoA. Here we demonstrate that an alternative, catalytically competent nucleophile exists in the active site of this enzyme. Next to the active-site serine is a histidine residue that is conserved in some, but not all acyl transferases. The H96A mutant is also active and can be labeled, but an H96A/S97A double mutant is inactive and cannot be labeled. The ability of H96 to form a malonyl-imidazole adduct was confirmed by proteolysis, followed by radio-HPLC and mass spectrometric analysis of the S97A mutant enzyme. Kinetic analysis revealed that the k(cat) of the S97A mutant was within 10-fold that of the wild-type enzyme, whereas the K(M)s of the two enzymes were comparable. Sequence comparison with the E. coli MAT (whose X-ray structure is known) led to the identification of H201 as the putative base in the serine-histidine catalytic dyad of the S. coelicolor enzyme. The absence of MAT activity in the H201A mutant and the detection of weak activity in the H201Q mutant was consistent with this proposal. The implications of this unexpected finding are discussed.


Assuntos
Aciltransferases/química , Streptomyces/enzimologia , Proteína de Transporte de Acila S-Maloniltransferase , Aciltransferases/genética , Aciltransferases/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Domínio Catalítico/genética , Ácidos Graxos/biossíntese , Histidina/química , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Serina/química , Streptomyces/genética
8.
Biotechnol Prog ; 17(4): 612-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11485419

RESUMO

Polyketides, a large family of bioactive natural products, are synthesized from building blocks derived from alpha-carboxylated Coenzyme A thioesters such as malonyl-CoA and (2S)-methylmalonyl-CoA. The productivity of polyketide fermentation processes in natural and heterologous hosts is frequently limited by the availability of these precursors in vivo. We describe a metabolic engineering strategy to enhance both the yield and volumetric productivity of polyketide biosynthesis. The genes matB and matC from Rhizobium trifolii encode a malonyl-CoA synthetase and a putative dicarboxylate transport protein, respectively. These proteins can directly convert exogenous malonate and methylmalonate into their corresponding CoA thioesters with an ATP requirement of 2 mol per mol of acyl-CoA produced. Heterologous expression of matBC in a recombinant strain of Streptomyces coelicolor that produces the macrolactone 6-deoxyerythronolide B results in a 300% enhancement of macrolactone titers. The unusual efficiency of the bioconversion is illustrated by the fact that approximately one-third of the methylmalonate units added to the fermentation medium are converted into macrolactones. The direct conversion of inexpensive feedstocks such as malonate and methylmalonate into polyketides represents the most carbon- and energy-efficient route to these high value natural products and has implications for cost-effective fermentation of numerous commercial and development-stage small molecules.


Assuntos
Proteínas de Bactérias , Eritromicina/análogos & derivados , Eritromicina/biossíntese , Engenharia Genética/métodos , Streptomyces/genética , Streptomyces/metabolismo , Acil Coenzima A/biossíntese , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Ácido Metilmalônico/farmacologia , Rhizobium/genética , Streptomyces/efeitos dos fármacos
9.
J Am Chem Soc ; 123(27): 6465-74, 2001 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-11439032

RESUMO

6-Deoxyerythronolide B synthase (DEBS) is the modular polyketide synthase (PKS) that catalyzes the biosynthesis of 6-deoxyerythronolide B (6-dEB), the aglycon precursor of the antibiotic erythromycin. The biosynthesis of 6-dEB exemplifies the extraordinary substrate- and stereo-selectivity of this family of multifunctional enzymes. Paradoxically, DEBS has been shown to be an attractive scaffold for combinatorial biosynthesis, indicating that its constituent modules are also very tolerant of unnatural substrates. By interrogating individual modules of DEBS with a panel of diketides activated as N-acetylcysteamine (NAC) thioesters, it was recently shown that individual modules have a marked ability to discriminate among certain diastereomeric diketides. However, since free NAC thioesters were used as substrates in these studies, the modules were primed by a diffusive process, which precluded involvement of the covalent, substrate-channeling mechanism by which enzyme-bound intermediates are directly transferred from one module to the next in a multimodular PKS. Recent evidence pointing to a pivotal role for protein-protein interactions in the substrate-channeling mechanism has prompted us to develop novel assays to reassess the steady-state kinetic parameters of individual DEBS modules when primed in a more "natural" channeling mode by the same panel of diketide substrates used earlier. Here we describe these assays and use them to quantify the kinetic benefit of linker-mediated substrate channeling in a modular PKS. This benefit can be substantial, especially for intrinsically poor substrates. Examples are presented where the k(cat) of a module for a given diketide substrate increases >100-fold when the substrate is presented to the module in a channeling mode as opposed to a diffusive mode. However, the substrate specificity profiles for individual modules are conserved regardless of the mode of presentation. By highlighting how substrate channeling can allow PKS modules to effectively accept and process intrinsically poor substrates, these studies provide a rational basis for examining the enormous untapped potential for combinatorial biosynthesis via module rearrangement.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Proteína de Transporte de Acila/síntese química , Proteína de Transporte de Acila/metabolismo , Domínio Catalítico , Transporte de Elétrons , Cinética , Peptídeos/metabolismo , Ligação Proteica , Engenharia de Proteínas/métodos , Especificidade por Substrato
10.
Proc Natl Acad Sci U S A ; 98(15): 8548-53, 2001 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-11447274

RESUMO

Streptomyces lavendulae produces complestatin, a cyclic peptide natural product that antagonizes pharmacologically relevant protein-protein interactions including formation of the C4b,2b complex in the complement cascade and gp120-CD4 binding in the HIV life cycle. Complestatin, a member of the vancomycin group of natural products, consists of an alpha-ketoacyl hexapeptide backbone modified by oxidative phenolic couplings and halogenations. The entire complestatin biosynthetic and regulatory gene cluster spanning ca. 50 kb was cloned and sequenced. It consisted of 16 ORFs, encoding proteins homologous to nonribosomal peptide synthetases, cytochrome P450-related oxidases, ferredoxins, nonheme halogenases, four enzymes involved in 4-hydroxyphenylglycine (Hpg) biosynthesis, transcriptional regulators, and ABC transporters. The nonribosomal peptide synthetase consisted of a priming module, six extending modules, and a terminal thioesterase; their arrangement and domain content was entirely consistent with functions required for the biosynthesis of a heptapeptide or alpha-ketoacyl hexapeptide backbone. Two oxidase genes were proposed to be responsible for the construction of the unique aryl-ether-aryl-aryl linkage on the linear heptapeptide intermediate. Hpg, 3,5-dichloro-Hpg, and 3,5-dichloro-hydroxybenzoylformate are unusual building blocks that repesent five of the seven requisite monomers in the complestatin peptide. Heterologous expression and biochemical analysis of 4-hydroxyphenylglycine transaminon confirmed its role as an aminotransferase responsible for formation of all three precursors. The close similarity but functional divergence between complestatin and chloroeremomycin biosynthetic genes also presents a unique opportunity for the construction of hybrid vancomycin-type antibiotics.


Assuntos
Genes Bacterianos , Família Multigênica , Oligopeptídeos/genética , Peptídeos Cíclicos , Vancomicina/análogos & derivados , Sequência de Bases , Clorofenóis/química , Clonagem Molecular , DNA Bacteriano , Dados de Sequência Molecular , Estrutura Molecular , Oligopeptídeos/biossíntese , Oligopeptídeos/química , Análise de Sequência de DNA , Streptomyces/genética
11.
J Am Chem Soc ; 123(11): 2495-502, 2001 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-11456917

RESUMO

Streptomyces coelicolor CH999/pJRJ2 harbors a plasmid encoding DEBS(KS1 degrees ), a mutant form of 6-deoxyerythronolide B synthase that is blocked in the formation of 6-deoxyerythronolide B (1, 6-dEB) due to a mutation in the active site of the ketosynthase (KS1) domain that normally catalyzes the first polyketide chain elongation step of 6-dEB biosynthesis. Administration of (2E,4S,5R)-2,4-dimethyl-5-hydroxy-2-heptenoic acid, N-acetylcysteamine thioester (6) an unsaturated triketide analogue of the natural triketide chain elongation intermediate to cultures of S. coelicolor CH999/pJRJ2 results in formation of a 16-membered macrolactone, which is isolated in the hemiketal form 33. The formation of the octaketide 33 indicates that the triketide substrate has been processed by DEBS module 2 as if it were a diketide analogue. The substrate specificity of this novel reaction has been explored by the incubation of three additional analogues of the unsaturated triketide 6, compounds 18, 31, and 32, with S. coelicolor CH999/pJRJ2, resulting in the formation of the corresponding macrolactones 34, 35, and 36. By contrast, the unsaturated triketide 10, lacking a methyl group at C-2, did not give rise to any detectable macrolactone product when incubated with S. coelicolor CH999/pJRJ2.


Assuntos
Antibacterianos/biossíntese , Complexos Multienzimáticos/fisiologia , Macrolídeos , Conformação Molecular , Streptomyces/metabolismo
12.
Bioorg Med Chem Lett ; 11(12): 1477-9, 2001 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-11412964

RESUMO

Incubation of chirally deuterated NADPH with 6-deoxyerythronolide B synthase (DEBS) modules 5 and module 6 and analysis of the derived triketide lactones established that the two ketoreductase domains, KR5 and KR6, are both specific for the 4-pro-S hydride of the nicotinamide cofactor.


Assuntos
Eritromicina/análogos & derivados , Eritromicina/biossíntese , Complexos Multienzimáticos/metabolismo , NADP/metabolismo , Bactérias/enzimologia , Bactérias/metabolismo , Domínio Catalítico , Deutério , Eritromicina/química , Cromatografia Gasosa-Espectrometria de Massas , Complexos Multienzimáticos/química , Oxirredução , Estrutura Terciária de Proteína , Especificidade por Substrato
14.
Org Lett ; 3(1): 57-9, 2001 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11429871

RESUMO

[figure: see text] We describe a semi-synthetic deglycosylated derivative of apoptolidin that retains considerable activity against the mitochondrial ATPase but has greatly reduced cellular cytotoxicity. We also demonstrate that a related antifungal natural product, cytovaricin, inhibits the same molecular target. Structural comparison of these macrolides provides insights into their conserved features that are presumably important for biological activity and identifies promising avenues at the interface of organic synthesis and biosynthesis for the generation of new selective cytotoxic agents.


Assuntos
Antibacterianos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Antifúngicos/farmacologia , Produtos Biológicos/farmacologia , Inibidores Enzimáticos/farmacologia , Macrolídeos , Oligomicinas/farmacologia , ATPases Translocadoras de Prótons/antagonistas & inibidores , Bactérias , Estrutura Molecular , ATPases Translocadoras de Prótons/efeitos dos fármacos , Relação Estrutura-Atividade , Leveduras/enzimologia
15.
Biochemistry ; 40(20): 6116-23, 2001 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-11352749

RESUMO

The rifamycin synthetase is primed with a 3-amino-5-hydroxybenzoate starter unit by a loading module that contains domains homologous to the adenylation and thiolation domains of nonribosomal peptide synthetases. Adenylation and thiolation activities of the loading module were reconstituted in vitro and shown to be independent of coenzyme A, countering literature proposals that the loading module is a coenzyme A ligase. Kinetic parameters for covalent arylation of the loading module were measured directly for the unnatural substrates benzoate and 3-hydroxybenzoate. This analysis was extended through competition experiments to determine the relative rates of incorporation of a series of substituted benzoates. Our results show that the loading module can accept a variety of substituted benzoates, although it exhibits a preference for the 3-, 5-, and 3,5-disubstituted benzoates that most closely resemble its biological substrate. The considerable substrate tolerance of the loading module of rifamycin synthetase suggests that the module has potential as a tool for generating substituted derivatives of natural products.


Assuntos
Benzoatos/metabolismo , Complexos Multienzimáticos/metabolismo , Peptídeo Sintases/metabolismo , Rifamicinas/biossíntese , Actinomycetales/enzimologia , Monofosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Aminobenzoatos/metabolismo , Coenzima A Ligases/metabolismo , Vetores Genéticos/síntese química , Vetores Genéticos/isolamento & purificação , Holoenzimas/metabolismo , Hidroxibenzoatos/metabolismo , Cinética , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/isolamento & purificação , Estrutura Terciária de Proteína , Especificidade por Substrato
16.
Biochemistry ; 40(8): 2317-25, 2001 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11327851

RESUMO

Although the role of protein-protein interactions in transducing signals within biological systems has been extensively explored, their relevance to the channeling of intermediates in metabolism is not widely appreciated. Polyketide synthases (PKSs) and nonribosomal peptide synthetases (NRPSs) are two related families of modular megasynthases that channel covalently bound intermediates from one active site to the next. Recent biochemical studies have highlighted the importance of protein-protein interactions in these chain transfer processes. The information available on this subject is reviewed, and its possible mechanistic implications are placed in context by comparisons with selected well-studied multicomponent protein systems.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Proteínas/química , Proteínas/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Animais , Humanos , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-fos/química , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/química , Proteínas Proto-Oncogênicas c-jun/metabolismo , Complexo Piruvato Desidrogenase/química , Complexo Piruvato Desidrogenase/metabolismo , Quinases da Família src/química , Quinases da Família src/metabolismo
17.
Biochemistry ; 40(8): 2326-31, 2001 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11327852

RESUMO

Polyketide synthases (PKSs) have represented fertile targets for rational manipulation via protein engineering ever since their modular architecture was first recognized. However, the mechanistic principles by which biosynthetic intermediates are sequentially channeled between modules remain poorly understood. Here we demonstrate the importance of complementarity in a remarkably simple, repetitive structural motif within these megasynthases that has been implicated to affect intermodular chain transfer [Gokhale, R. S., et al. (1999) Science 284, 482]. The C- and N-terminal ends of adjacent PKS polypeptides are capped by short peptides of 20-40 residues. Mismatched sequences abolish intermodular chain transfer without affecting the activity of individual modules, whereas matched sequences can facilitate the channeling of intermediates between ordinarily nonconsecutive modules. Thus, in addition to substrate-PKS interactions and domain-domain interactions, these short interpolypeptide sequences represent a third determinant of selective chain transfer that must be taken into consideration in the protein engineering of PKSs. Preliminary biophysical studies on synthetic peptide mimics of these linkers suggest that they may adopt coiled-coil conformations.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Proteínas/química , Proteínas/metabolismo , Sequência de Aminoácidos , Dicroísmo Circular , Cicloexanonas/antagonistas & inibidores , Dissacarídeos/antagonistas & inibidores , Dissacarídeos/biossíntese , Eritromicina/análogos & derivados , Eritromicina/química , Cinética , Dados de Sequência Molecular , Complexos Multienzimáticos/genética , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Engenharia de Proteínas , Estrutura Secundária de Proteína/genética , Estrutura Terciária de Proteína/genética , Proteínas/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Tioléster Hidrolases/química , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo
19.
Microbiol Mol Biol Rev ; 65(1): 106-18, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238987

RESUMO

Polyketide natural products show great promise as medicinal agents. Typically the products of microbial secondary biosynthesis, polyketides are synthesized by an evolutionarily related but architecturally diverse family of multifunctional enzymes called polyketide synthases. A principal limitation for fundamental biochemical studies of these modular megasynthases, as well as for their applications in biotechnology, is the challenge associated with manipulating the natural microorganism that produces a polyketide of interest. To ameliorate this limitation, over the past decade several genetically amenable microbes have been developed as heterologous hosts for polyketide biosynthesis. Here we review the state of the art as well as the difficulties associated with heterologous polyketide production. In particular, we focus on two model hosts, Streptomyces coelicolor and Escherichia coli. Future directions for this relatively new but growing technological opportunity are also discussed.


Assuntos
Engenharia Genética/métodos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Biotecnologia/métodos , Proteínas de Transporte/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fungos/genética , Fungos/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Plantas/genética , Plantas/metabolismo , Processamento de Proteína Pós-Traducional , Streptomyces/genética , Streptomyces/metabolismo , Especificidade por Substrato
20.
Science ; 291(5509): 1790-2, 2001 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-11230695

RESUMO

The macrocyclic core of the antibiotic erythromycin, 6-deoxyerythronolide B (6dEB), is a complex natural product synthesized by the soil bacterium Saccharopolyspora erythraea through the action of a multifunctional polyketide synthase (PKS). The engineering potential of modular PKSs is hampered by the limited capabilities for molecular biological manipulation of organisms (principally actinomycetes) in which complex polyketides have thus far been produced. To address this problem, a derivative of Escherichia coli has been genetically engineered. The resulting cellular catalyst converts exogenous propionate into 6dEB with a specific productivity that compares well with a high-producing mutant of S. erythraea that has been incrementally enhanced over decades for the industrial production of erythromycin.


Assuntos
Proteínas de Bactérias , Eritromicina/biossíntese , Escherichia coli/genética , Complexos Multienzimáticos/genética , Saccharopolyspora/genética , Acil Coenzima A/metabolismo , Clonagem Molecular , Eritromicina/análogos & derivados , Escherichia coli/enzimologia , Complexos Multienzimáticos/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/metabolismo , Saccharopolyspora/enzimologia , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Transformação Bacteriana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...