Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 12(5): e0173121, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34488446

RESUMO

In natural environments, antibiotics are important means of interspecies competition. At subinhibitory concentrations, they act as cues or signals inducing antibiotic production; however, our knowledge of well-documented antibiotic-based sensing systems is limited. Here, for the soil actinobacterium Streptomyces lincolnensis, we describe a fundamentally new ribosome-mediated signaling cascade that accelerates the onset of lincomycin production in response to an external ribosome-targeting antibiotic to synchronize antibiotic production within the population. The entire cascade is encoded in the lincomycin biosynthetic gene cluster (BGC) and consists of three lincomycin resistance proteins in addition to the transcriptional regulator LmbU: a lincomycin transporter (LmrA), a 23S rRNA methyltransferase (LmrB), both of which confer high resistance, and an ATP-binding cassette family F (ABCF) ATPase, LmrC, which confers only moderate resistance but is essential for antibiotic-induced signal transduction. Specifically, antibiotic sensing occurs via ribosome-mediated attenuation, which activates LmrC production in response to lincosamide, streptogramin A, or pleuromutilin antibiotics. Then, ATPase activity of the ribosome-associated LmrC triggers the transcription of lmbU and consequently the expression of lincomycin BGC. Finally, the production of LmrC is downregulated by LmrA and LmrB, which reduces the amount of ribosome-bound antibiotic and thus fine-tunes the cascade. We propose that analogous ABCF-mediated signaling systems are relatively common because many ribosome-targeting antibiotic BGCs encode an ABCF protein accompanied by additional resistance protein(s) and transcriptional regulators. Moreover, we revealed that three of the eight coproduced ABCF proteins of S. lincolnensis are clindamycin responsive, suggesting that the ABCF-mediated antibiotic signaling may be a widely utilized tool for chemical communication. IMPORTANCE Resistance proteins are perceived as mechanisms protecting bacteria from the inhibitory effect of their produced antibiotics or antibiotics from competitors. Here, we report that antibiotic resistance proteins regulate lincomycin biosynthesis in response to subinhibitory concentrations of antibiotics. In particular, we show the dual character of the ABCF ATPase LmrC, which confers antibiotic resistance and simultaneously transduces a signal from ribosome-bound antibiotics to gene expression, where the 5' untranslated sequence upstream of its encoding gene functions as a primary antibiotic sensor. ABCF-mediated antibiotic signaling can in principle function not only in the induction of antibiotic biosynthesis but also in selective gene expression in response to any small molecules targeting the 50S ribosomal subunit, including clinically important antibiotics, to mediate intercellular antibiotic signaling and stress response induction. Moreover, the resistance-regulatory function of LmrC presented here for the first time unifies functionally inconsistent ABCF family members involving antibiotic resistance proteins and translational regulators.


Assuntos
Adenosina Trifosfatases/metabolismo , Antibacterianos/biossíntese , Antibacterianos/farmacologia , Lincomicina/biossíntese , Lincomicina/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Metiltransferases , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Família Multigênica , Ribossomos/metabolismo , Transdução de Sinais , Streptomyces/metabolismo , Fatores de Transcrição
2.
J Basic Microbiol ; 61(9): 772-781, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34313330

RESUMO

Lincomycin is an important antimicrobial agent which is widely used in clinical and animal husbandry. The biosynthetic pathway of lincomycin comes to light in the past 10 years, however, the regulatory mechanism is still unclear. In this study, a redox-sensing regulator Rex from Streptomyces lincolnensis (Rexlin ) was identified and characterized to affect cell growth and lincomycin biosynthesis. Disruption of rex resulted in an increase in cell growth, but a decrease in lincomycin production. The results of quantitative real-time polymerase chain reaction showed that Rexlin can promote transcription of the regulatory gene lmbU and the structural genes lmbA, lmbC, lmbJ, lmbV, and lmbW. However, electrophoretic mobility shift assay analysis demonstrated that Rexlin can not bind to the promoter regions of these genes above. Findings in this study broadened our horizons in the regulatory mechanism of lincomycin production and laid a foundation for strain improvement of antibiotic producers.


Assuntos
Antibacterianos/biossíntese , Vias Biossintéticas/genética , Regulação Bacteriana da Expressão Gênica , Lincomicina/biossíntese , Streptomyces/genética , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Família Multigênica , Oxirredução , Fatores de Transcrição/genética
3.
Proc Natl Acad Sci U S A ; 117(40): 24794-24801, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-32958639

RESUMO

The structure of lincomycin A consists of the unusual eight-carbon thiosugar core methyllincosamide (MTL) decorated with a pendent N-methylprolinyl moiety. Previous studies on MTL biosynthesis have suggested GDP-ᴅ-erythro-α-ᴅ-gluco-octose and GDP-ᴅ-α-ᴅ-lincosamide as key intermediates in the pathway. However, the enzyme-catalyzed reactions resulting in the conversion of GDP-ᴅ-erythro-α-ᴅ-gluco-octose to GDP-ᴅ-α-ᴅ-lincosamide have not yet been elucidated. Herein, a biosynthetic subpathway involving the activities of four enzymes-LmbM, LmbL, CcbZ, and CcbS (the LmbZ and LmbS equivalents in the closely related celesticetin pathway)-is reported. These enzymes catalyze the previously unknown biosynthetic steps including 6-epimerization, 6,8-dehydration, 4-epimerization, and 6-transamination that convert GDP-ᴅ-erythro-α-ᴅ-gluco-octose to GDP-ᴅ-α-ᴅ-lincosamide. Identification of these reactions completes the description of the entire lincomycin biosynthetic pathway. This work is significant since it not only resolves the missing link in octose core assembly of a thiosugar-containing natural product but also showcases the sophistication in catalytic logic of enzymes involved in carbohydrate transformations.


Assuntos
Lincomicina/biossíntese , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vias Biossintéticas , Lincomicina/química , Lincosamidas/química , Lincosamidas/metabolismo , Streptomyces/química , Streptomyces/enzimologia , Streptomyces/genética
4.
Microb Cell Fact ; 19(1): 30, 2020 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-32050973

RESUMO

BACKGROUND: Lincomycin, produced by Streptomyces lincolnensis, is a lincosamide antibiotic and widely used for the treatment of the infective diseases caused by Gram-positive bacteria. The mechanisms of lincomycin biosynthesis have been deeply explored in recent years. However, the regulatory effects of LmbU that is a transcriptional regulator in lincomycin biosynthetic (lmb) gene cluster have not been fully addressed. RESULTS: LmbU was used to search for homologous LmbU (LmbU-like) proteins in the genomes of actinobacteria, and the results showed that LmbU-like proteins are highly distributed regulators in the biosynthetic gene clusters (BGCs) of secondary metabolites or/and out of the BGCs in actinomycetes. The overexpression, inactivation and complementation of the lmbU gene indicated that LmbU positively controls lincomycin biosynthesis in S. lincolnensis. Comparative transcriptomic analysis further revealed that LmbU activates the 28 lmb genes at whole lmb cluster manner. Furthermore, LmbU represses the transcription of the non-lmb gene hpdA in the biosynthesis of L-tyrosine, the precursor of lincomycin. LmbU up-regulates nineteen non-lmb genes, which would be involved in multi-drug flux to self-resistance, nitrate and sugar transmembrane transport and utilization, and redox metabolisms. CONCLUSIONS: LmbU is a significant pleiotropic transcriptional regulator in lincomycin biosynthesis by entirely activating the lmb cluster and regulating the non-lmb genes in Streptomyces lincolnensis. Our results first revealed the pleiotropic regulatory function of LmbU, and shed new light on the transcriptional effects of LmbU-like family proteins on antibiotic biosynthesis in actinomycetes.


Assuntos
Regulação Bacteriana da Expressão Gênica/genética , Lincomicina/biossíntese , Streptomyces , Fatores de Transcrição/genética , Transcriptoma/genética , Proteínas de Bactérias/genética , Perfilação da Expressão Gênica/métodos , Pleiotropia Genética/genética , Família Multigênica/genética , Metabolismo Secundário/genética , Streptomyces/genética , Streptomyces/metabolismo
5.
J Am Chem Soc ; 142(7): 3440-3448, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-31944685

RESUMO

Antitumor pyrrolobenzodiazepines (PBDs), lincosamide antibiotics, quorum-sensing molecule hormaomycin, and antimicrobial griselimycin are structurally and functionally diverse groups of actinobacterial metabolites. The common feature of these compounds is the incorporation of l-tyrosine- or l-leucine-derived 4-alkyl-l-proline derivatives (APDs) in their structures. Here, we report that the last reaction in the biosynthetic pathway of APDs, catalyzed by F420H2-dependent Apd6 reductases, contributes to the structural diversity of APD precursors. Specifically, the heterologous overproduction of six Apd6 enzymes demonstrated that Apd6 from the biosynthesis of PBDs and hormaomycin can reduce only an endocyclic imine double bond, whereas Apd6 LmbY and partially GriH from the biosyntheses of lincomycin and griselimycin, respectively, also reduce the more inert exocyclic double bond of the same 4-substituted Δ1-pyrroline-2-carboxylic acid substrate, making LmbY and GriH unusual, if not unique, among reductases. Furthermore, the differences in the reaction specificity of the Apd6 reductases determine the formation of the fully saturated APD moiety of lincomycin versus the unsaturated APD moiety of PBDs, providing molecules with optimal shapes to bind their distinct biological targets. Moreover, the Apd6 reductases establish the first F420H2-dependent enzymes from the luciferase-like hydride transferase protein superfamily in the biosynthesis of bioactive molecules. Finally, our bioinformatics analysis demonstrates that Apd6 and their homologues, widely distributed within several bacterial phyla, play a role in the formation of novel yet unknown natural products with incorporated l-proline-like precursors and likely in the microbial central metabolism.


Assuntos
Benzodiazepinas/metabolismo , Lincomicina/biossíntese , Oxirredutases/metabolismo , Pirróis/metabolismo , Benzodiazepinas/química , Benzodiazepinas/farmacologia , Catálise , Depsipeptídeos/biossíntese , Depsipeptídeos/química , Depsipeptídeos/farmacologia , Lincomicina/química , Lincomicina/farmacologia , Modelos Moleculares , Oxirredutases/química , Peptídeos Cíclicos/biossíntese , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Prolina/análogos & derivados , Prolina/metabolismo , Pirróis/química , Pirróis/farmacologia , Riboflavina/análogos & derivados , Riboflavina/química , Riboflavina/metabolismo , Especificidade por Substrato , Tirosina/análogos & derivados , Tirosina/metabolismo
6.
Prep Biochem Biotechnol ; 50(6): 529-537, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31916478

RESUMO

As a lincosamide antibiotic, lincomycin is still important for treating diseases caused by Gram-positive bacteria. Manufacturing of lincomycin needs efforts to, e.g. maximize desirable species and minimizing unwanted fermentation byproducts. Analysis of the lincomycin biosynthetic gene cluster of Streptomyces lincolnensis, lmbB1, was shown to catalyze the conversion of L-dopa but not of L-tyrosine and then further generated the precursor of lincomycin A. Based on the principle of directed breeding, a strain termed as S. lincolnensis 24-2, was obtained in this work. By overexpressing the lmbB1 gene, this strain produces efficacious lincomycin A and suppresses melanin generation, whereas contains unwanted lincomycin B. The good fermentation performance of the mutant-lmbB1 (M-lmbB1) was also confirmed in a 15 L-scale bioreactor, which increased the lincomycin A production by 37.6% compared with control of 6435 u/mL and reduced the accumulation of melanin by 29.9% and lincomycin B by 73.4%. This work demonstrated that the amplification of lmbB1 gene mutation and metabolic engineering could promote lincomycin biosynthesis and might be helpful for reducing the production of other industrially unnecessary byproduct.


Assuntos
Antibacterianos/biossíntese , Proteínas de Bactérias/biossíntese , Fermentação/genética , Lincomicina/biossíntese , Engenharia Metabólica/métodos , Família Multigênica , Streptomyces/genética , Streptomyces/metabolismo , Reatores Biológicos , Escherichia coli/genética , Expressão Gênica , Levodopa/metabolismo , Lincosamidas/metabolismo , Melaninas/biossíntese , Microrganismos Geneticamente Modificados , Transcrição Gênica/genética
7.
Appl Microbiol Biotechnol ; 104(6): 2575-2587, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31993701

RESUMO

Leucine-responsive regulatory proteins (Lrps) are a family of transcription factors involved in diverse biological processes in bacteria. So far, molecular mechanism of Lrps for regulating antibiotics biosynthesis in actinomycetes remains largely unexplored. This study, for the first time in Streptomyces lincolnensis, identified an Lrp (named as SLCG_Lrp) associated with lincomycin production. SLCG_Lrp was validated to be a positive regulator for lincomycin biosynthesis by directly stimulating transcription of two structural genes (lmbA and lmbV), three resistance genes (lmrA, lmrB and lmrC), and a regulatory gene (lmbU) within the lincomycin biosynthetic gene (lin) cluster. SLCG_Lrp was transcriptionally self-inhibited and triggered the expression of its adjacent gene SLCG_3127 encoding a LysE superfamily protein. Further, the binding site of SLCG_Lrp in the intergenic region of SLCG_3127 and SLCG_Lrp was precisely identified. Inactivation of SLCG_3127 in S. lincolnensis resulted in yield improvement of lincomycin, which was caused by intracellular accumulation of proline and cysteine. Arginine and phenylalanine were identified as specific regulatory ligands, respectively, to reduce and promote DNA-binding affinity of SLCG_Lrp. We further found that SLCG_Lrp was directly repressed by SLCG_2919, the first identified transcription factor outside lin cluster for lincomycin production. Therefore, our findings revealed SLCG_Lrp-mediated transcriptional regulation of lincomycin biosynthesis. This study extends the understanding of molecular mechanisms underlying lincomycin biosynthetic regulation.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Proteína Reguladora de Resposta a Leucina/genética , Lincomicina/biossíntese , Streptomyces/genética , Transcrição Gênica , Vias Biossintéticas , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Família Multigênica , Fatores de Transcrição/genética
8.
Biochemistry ; 58(48): 4794-4798, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31710815

RESUMO

Extradiol dioxygenase chemistry is essential for catechol breakdown. The largest natural reservoir of catechols, or 1,2-dihydroxybenzenes, is the plant woody-tissue polymer lignin. Vicinal-oxygen-chelate (VOC) dioxygenases make up the largest group of characterized extradiol dioxygenases, and while most are found as part of catabolic pathways degrading a variety of natural and human-made aromatic rings, L-DOPA (l-3,4-dihydroxyphenylalanine) dioxygenase is a VOC enzyme that participates in the biosynthesis of a natural product. All VOC superfamily members shared conserved elements of catalysis, yet despite decades of investigation of VOC enzymes, the relationships between VOC domain architecture and enzymatic function remain complex and poorly understood. Herein, we present evidence that L-DOPA dioxygenase is the representative member of a new topological class of VOC extradiol dioxygenases. Guided by its evolutionary similarity to glyoxylase enzymes, we performed a careful investigation of the Streptomyces lincolnensis L-DOPA dioxygenase (LmbB1) active site through mutagenesis, kinetic, and pH studies. Our results demonstrate that the L-DOPA dioxygenase reaction depends upon an active-site tyrosine and histidine and is remarkably resilient to mutation, even at the iron-ligating residues. Evaluation of the cleavage reaction as a function of pH supports the role of a histidine in acid-base catalysis. The active-site architecture is functionally consistent with the existing knowledge of VOC extradiol dioxygenase catalysis.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Dioxigenases/química , Dioxigenases/metabolismo , Lincomicina/biossíntese , Família Multigênica , Streptomyces/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Domínio Catalítico , Dioxigenases/genética , Cinética , Levodopa/metabolismo , Alinhamento de Sequência , Streptomyces/química , Streptomyces/metabolismo
9.
Appl Environ Microbiol ; 85(1)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30341075

RESUMO

Lincomycin A (Lin-A) is a widely used antibacterial antibiotic fermented by Streptomyces lincolnensis However, the transcriptional regulatory mechanisms underlying lincomycin biosynthesis have seldom been investigated. Here, we first identified a TetR family transcriptional regulator (TFR), SLCG_2919, which negatively modulates lincomycin biosynthesis in S. lincolnensis LCGL. SLCG_2919 was found to specifically bind to promoter regions of the lincomycin biosynthetic gene cluster (lin cluster), including 25 structural genes, three resistance genes, and one regulatory gene, and to inhibit the transcription of these genes, demonstrating a directly regulatory role in lincomycin biosynthesis. Furthermore, we found that SLCG_2919 was not autoregulated, but directly repressed its adjacent gene, SLCG_2920, which encodes an ATP/GTP binding protein whose overexpression increased resistance against lincomycin and Lin-A yields in S. lincolnensis The precise SLCG_2919 binding site within the promoter region of SLCG_2920 was determined by a DNase I footprinting assay and by electrophoretic mobility shift assays (EMSAs) based on base substitution mutagenesis, with the internal 10-nucleotide (nt) AT-rich sequence (AAATTATTTA) shown to be essential for SLCG_2919 binding. Our findings indicate that SLCG_2919 is a negative regulator for controlling lincomycin biosynthesis in S. lincolnensis The present study improves our understanding of molecular regulation for lincomycin biosynthesis.IMPORTANCE TetR family transcriptional regulators (TFRs) are generally found to regulate diverse cellular processes in bacteria, especially antibiotic biosynthesis in Streptomyces species. However, knowledge of their function in lincomycin biosynthesis in S. lincolnensis remains unknown. The present study provides a new insight into the regulation of lincomycin biosynthesis through a TFR, SLCG_2919, that directly modulates lincomycin production and resistance. Intriguingly, SLCG_2919 and its adjoining gene, SLCG_2920, which encodes an ATP/GTP binding protein, were extensively distributed in diverse Streptomyces species. In addition, we revealed a new TFR binding motif, in which SLCG_2919 binds to the promoter region of SLCG_2920, dependent on the intervening AT-rich sequence rather than on the flanking inverted repeats found in the binding sites of other TFRs. These insights into transcriptional regulation of lincomycin biosynthesis by SLCG_2919 will be valuable in paving the way for genetic engineering of regulatory elements in Streptomyces species to improve antibiotic production.


Assuntos
Antibacterianos/biossíntese , Proteínas de Bactérias/genética , Lincomicina/biossíntese , Streptomyces/genética , Fatores de Transcrição/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Streptomyces/metabolismo , Fatores de Transcrição/metabolismo
10.
Appl Microbiol Biotechnol ; 102(9): 4101-4115, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29549449

RESUMO

Global regulator BldA, the only tRNA for a rare leucine codon UUA, is best known for its ability to affect morphological differentiation and secondary metabolism in the genus Streptomyces. In this study, we confirmed the regulatory function of the bldA gene (Genbank accession no. EU124663.1) in Streptomyces lincolnensis. Disruption of bldA hinders the sporulation and lincomycin production, that can recur when complemented with a functional bldA gene. Western blotting assays demonstrate that translation of the lmbB2 gene which encodes a L-tyrosine hydroxylase is absolutely dependent on BldA; however, mistranslation of the lmbU gene which encodes a cluster-situated regulator (CSR) is observed in a bldA mutant. Intriguingly, when the preferential cognate codon CTG was used, the expression level of LmbU was not the highest compared to the usage of rare codon TTA or CTA, indicating the rare codon in this position is significant for the regulation of lmbU expression. Moreover, replacement of TTA codons in both genes with another leucin codon in the bldA mutant did not restore lincomycin production. Thus, we believe that the bldA gene regulates lincomycin production via controlling the translation of not only lmbB2 and lmbU, but also the other TTA-containing genes. In conclusion, the present study demonstrated the importance of the bldA gene in morphological differentiation and lincomycin production in S. lincolnensis.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Lincomicina/biossíntese , RNA de Transferência de Leucina/metabolismo , Streptomyces/genética , Streptomyces/metabolismo , RNA Bacteriano/metabolismo , Streptomyces/citologia
11.
J Bacteriol ; 200(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29038257

RESUMO

Lincomycin A is a clinically important antimicrobial agent produced by Streptomyces lincolnensis In this study, a new regulator designated LmbU (GenBank accession no. ABX00623.1) was identified and characterized to regulate lincomycin biosynthesis in S. lincolnensis wild-type strain NRRL 2936. Both inactivation and overexpression of lmbU resulted in significant influences on lincomycin production. Transcriptional analysis and in vivo neomycin resistance (Neor) reporter assays demonstrated that LmbU activates expression of the lmbA, lmbC, lmbJ, and lmbW genes and represses expression of the lmbK and lmbU genes. Electrophoretic mobility shift assays (EMSAs) demonstrated that LmbU can bind to the regions upstream of the lmbA and lmbW genes through the consensus and palindromic sequence 5'-CGCCGGCG-3'. However, LmbU cannot bind to the regions upstream of the lmbC, lmbJ, lmbK, and lmbU genes as they lack this motif. These data indicate a complex transcriptional regulatory mechanism of LmbU. LmbU homologues are present in the biosynthetic gene clusters of secondary metabolites of many other actinomycetes. Furthermore, the LmbU homologue from Saccharopolyspora erythraea (GenBank accession no. WP_009944629.1) also binds to the regions upstream of lmbA and lmbW, which suggests widespread activity for this regulator. LmbU homologues have no significant structural similarities to other known cluster-situated regulators (CSRs), which indicates that they belong to a new family of regulatory proteins. In conclusion, the present report identifies LmbU as a novel transcriptional regulator and provides new insights into regulation of lincomycin biosynthesis in S. lincolnensisIMPORTANCE Although lincomycin biosynthesis has been extensively studied, its regulatory mechanism remains elusive. Here, a novel regulator, LmbU, which regulates transcription of its target genes in the lincomycin biosynthetic gene cluster (lmb gene cluster) and therefore promotes lincomycin biosynthesis, was identified in S. lincolnensis strain NRRL 2936. Importantly, we show that this new regulatory element is relatively widespread across diverse actinomycetes species. In addition, our findings provide a new strategy for improvement of yield of lincomycin through manipulation of LmbU, and this approach could also be evaluated in other secondary metabolite gene clusters containing this regulatory protein.


Assuntos
Regulação Bacteriana da Expressão Gênica , Lincomicina/biossíntese , Streptomyces/genética , Streptomyces/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/metabolismo , Família Multigênica , Saccharopolyspora/genética , Metabolismo Secundário , Fatores de Transcrição/genética
12.
Appl Microbiol Biotechnol ; 101(13): 5341-5352, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28455615

RESUMO

Nitrate is necessary for primary and secondary metabolism of actinomycetes and stimulates the production of a few antibiotics, such as lincomycin and rifamycin. However, the mechanism of this nitrate-stimulating effect was not fully understood. Two putative ABC-type nitrate transporters were identified in Streptomyces lincolnensis NRRL2936 and verified to be involved in lincomycin biosynthesis. With nitrate supplementation, the transcription of nitrogen assimilation genes, nitrate-specific ABC1 transporter genes, and lincomycin exporter gene lmrA was found to be enhanced and positively regulated by the global regulator GlnR, whose expression was also improved. Moreover, heterologous expression of ABC2 transporter genes in Streptomyces coelicolor M145 resulted in an increased actinorhodin production. Further incorporation of a nitrite-specific transporter gene nirC, as in nirC-ABC2 cassette, led to an even higher actinorhodin production. Similarly, the titers of salinomycin, ansamitocin, lincomycin, and geldanamycin were increased with the integration of this cassette to Streptomyces albus BK3-25, Actinosynnema pretiosum ATCC31280, S. lincolnensis LC-G, and Streptomyces hygroscopicus XM201, respectively. Our work expanded the nitrate-stimulating effect to many antibiotic producers by utilizing the nirC-ABC2 cassette for enhanced nitrate utilization, which could become a general tool for titer increase of antibiotics in actinomycetes.


Assuntos
Actinobacteria/genética , Antibacterianos/biossíntese , Lincomicina/biossíntese , Nitratos/metabolismo , Streptomyces/genética , Actinobacteria/metabolismo , Proteínas de Transporte de Ânions/genética , Antraquinonas/metabolismo , Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Nitrogênio/metabolismo , Piranos/metabolismo , Streptomyces/metabolismo , Streptomyces coelicolor/genética , Transativadores/genética , Transativadores/metabolismo
13.
Biochim Biophys Acta ; 1864(6): 724-737, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26963649

RESUMO

The metabolic pathways for the production of lincomycin, hormaomycin and the antitumor pyrrolo [1,4] benzodiazepines share a vinyl substituted pyrroline carboxylic acid (3-vinyl-2,3-pyrroline-5-carboxylic acid, VPCA) as a common intermediate. Biosynthesis of this vinyl substituted pyrroline carboxylic acid intermediate requires a short, three-enzyme pathway containing two metalloenzymes: a heme-dependent l-tyrosine hydroxylase and a non-heme Fe(2+) dependent l-DOPA dioxygenase. The l-tyrosine hydroxylase is an unprecedented type of peroxidase that specifically monohydroxylates tyrosine, while the l-DOPA extradiol cleaving enzyme is a single-domain vicinal-oxygen-chelate (VOC) dioxygenase. The dioxygenase product subsequently undergoes an, as yet uncharacterized, C-C bond cleavage reaction. This mini-pathway demonstrates the use of metal-dependent chemistry typically associated with natural product degradation in order to build a compact, functionalized building block for larger, bioactive molecules.


Assuntos
Benzodiazepinas/metabolismo , Depsipeptídeos/biossíntese , Enzimas/metabolismo , Lincomicina/biossíntese , Metaloproteínas/metabolismo , Sequência de Aminoácidos , Enzimas/química , Metaloproteínas/química , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
14.
J Appl Microbiol ; 119(4): 1064-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26248490

RESUMO

AIMS: To improve lincomycin A production and decrease the content of byproduct lincomycin B in an industrial lincomycin-producing strain. METHODS AND RESULTS: The in silico analysis indicated that LmbW could be involved in propylproline biosynthesis of lincomyin A. In this study, we constructed an lmbW deletion mutant and found that the mutant lost the ability to produce lincomycin A, but increased the accumulation of lincomycin B. The loss of lincomycin A production can be restored by complementing the mutant with the expression of lmbW gene. When lmbW and metK (encoding S-adenosylmethionine synthetase) was co-overexpressed, lincomycin A titre was 1744·6 mg l(-1) , a 35·83% improvement over the original strain. Meanwhile, the content of lincomycin B was reduced to 4·41%, a remarkable decrease of 34·76%, compared to that of the original strain. CONCLUSIONS: lmbW encodes a C-methyltransferase involved in the biosynthesis of lincomycin A but not lincomycin B. Co-overexpression of lmbW and metK improved lincomycin A production and decreased the content of lincomycin B. SIGNIFICANCE AND IMPACT OF THE STUDY: The engineered Streptomyces lincolnensis strain shows promising application in the fermentation production of lincomycin A, which may help cut production costs and simplify downstream separation processes.


Assuntos
Lincomicina/biossíntese , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fermentação , Lincosamidas/biossíntese , Metionina Adenosiltransferase/metabolismo , Dados de Sequência Molecular , Streptomyces/genética
15.
PLoS One ; 10(3): e0118850, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25741696

RESUMO

In the biosynthesis of lincosamide antibiotics lincomycin and celesticetin, the amino acid and amino sugar units are linked by an amide bond. The respective condensing enzyme lincosamide synthetase (LS) is expected to be an unusual system combining nonribosomal peptide synthetase (NRPS) components with so far unknown amino sugar related activities. The biosynthetic gene cluster of celesticetin was sequenced and compared to the lincomycin one revealing putative LS coding ORFs shared in both clusters. Based on a bioassay and production profiles of S. lincolnensis strains with individually deleted putative LS coding genes, the proteins LmbC, D, E, F and V were assigned to LS function. Moreover, the newly recognized N-terminal domain of LmbN (LmbN-CP) was also assigned to LS as a NRPS carrier protein (CP). Surprisingly, the homologous CP coding sequence in celesticetin cluster is part of ccbZ gene adjacent to ccbN, the counterpart of lmbN, suggesting the gene rearrangement, evident also from still active internal translation start in lmbN, and indicating the direction of lincosamide biosynthesis evolution. The in vitro test with LmbN-CP, LmbC and the newly identified S. lincolnensis phosphopantetheinyl transferase Slp, confirmed the cooperation of the previously characterized NRPS A-domain LmbC with a holo-LmbN-CP in activation of a 4-propyl-L-proline precursor of lincomycin. This result completed the functional characterization of LS subunits resembling NRPS initiation module. Two of the four remaining putative LS subunits, LmbE/CcbE and LmbV/CcbV, exhibit low but significant homology to enzymes from the metabolism of mycothiol, the NRPS-independent system processing the amino sugar and amino acid units. The functions of particular LS subunits as well as cooperation of both NRPS-based and NRPS-independent LS blocks are discussed. The described condensing enzyme represents a unique hybrid system with overall composition quite dissimilar to any other known enzyme system.


Assuntos
Cisteína/metabolismo , Glicopeptídeos/metabolismo , Inositol/metabolismo , Lincomicina/biossíntese , Lincosamidas/biossíntese , Peptídeo Sintases/metabolismo
16.
Nature ; 518(7537): 115-9, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25607359

RESUMO

Low-molecular-mass thiols in organisms are well known for their redox-relevant role in protection against various endogenous and exogenous stresses. In eukaryotes and Gram-negative bacteria, the primary thiol is glutathione (GSH), a cysteinyl-containing tripeptide. In contrast, mycothiol (MSH), a cysteinyl pseudo-disaccharide, is dominant in Gram-positive actinobacteria, including antibiotic-producing actinomycetes and pathogenic mycobacteria. MSH is equivalent to GSH, either as a cofactor or as a substrate, in numerous biochemical processes, most of which have not been characterized, largely due to the dearth of information concerning MSH-dependent proteins. Actinomycetes are able to produce another thiol, ergothioneine (EGT), a histidine betaine derivative that is widely assimilated by plants and animals for variable physiological activities. The involvement of EGT in enzymatic reactions, however, lacks any precedent. Here we report that the unprecedented coupling of two bacterial thiols, MSH and EGT, has a constructive role in the biosynthesis of lincomycin A, a sulfur-containing lincosamide (C8 sugar) antibiotic that has been widely used for half a century to treat Gram-positive bacterial infections. EGT acts as a carrier to template the molecular assembly, and MSH is the sulfur donor for lincomycin maturation after thiol exchange. These thiols function through two unusual S-glycosylations that program lincosamide transfer, activation and modification, providing the first paradigm for EGT-associated biochemical processes and for the poorly understood MSH-dependent biotransformations, a newly described model that is potentially common in the incorporation of sulfur, an element essential for life and ubiquitous in living systems.


Assuntos
Antibacterianos/biossíntese , Cisteína/metabolismo , Ergotioneína/metabolismo , Glicopeptídeos/metabolismo , Inositol/metabolismo , Lincomicina/biossíntese , Streptomyces/metabolismo , Produtos Biológicos/metabolismo , Vias Biossintéticas/genética , Biotransformação , Cisteína/química , Ergotioneína/química , Glicopeptídeos/química , Glicosilação , Inositol/química , Lincosamidas/metabolismo , Dados de Sequência Molecular , Streptomyces/genética
18.
J Am Chem Soc ; 136(3): 906-9, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24380627

RESUMO

Lincomycin A is a clinically useful antibiotic isolated from Streptomyces lincolnensis. It contains an unusual methylmercapto-substituted octose, methylthiolincosamide (MTL). While it has been demonstrated that the C8 backbone of MTL moiety is derived from D-fructose 6-phosphate and D-ribose 5-phosphate via a transaldol reaction catalyzed by LmbR, the subsequent enzymatic transformations leading to the MTL moiety remain elusive. Here, we report the identification of GDP-D-erythro-α-D-gluco-octose (GDP-D-α-D-octose) as a key intermediate in the MTL biosynthetic pathway. Our data show that the octose 1,8-bisphosphate intermediate is first converted to octose 1-phosphate by a phosphatase, LmbK. The subsequent conversion of the octose 1-phosphate to GDP-D-α-D-octose is catalyzed by the octose 1-phosphate guanylyltransferase, LmbO. These results provide significant insight into the lincomycin biosynthetic pathway, because the activated octose likely serves as the acceptor for the installation of the C1 sulfur appendage of MTL.


Assuntos
Açúcares de Guanosina Difosfato/metabolismo , Lincomicina/biossíntese , Monossacarídeos/metabolismo , Nucleotidiltransferases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Streptomyces/enzimologia
19.
PLoS One ; 8(12): e79974, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324587

RESUMO

The gene lmbB2 of the lincomycin biosynthetic gene cluster of Streptomyces lincolnensis ATCC 25466 was shown to code for an unusual tyrosine hydroxylating enzyme involved in the biosynthetic pathway of this clinically important antibiotic. LmbB2 was expressed in Escherichia coli, purified near to homogeneity and shown to convert tyrosine to 3,4-dihydroxyphenylalanine (DOPA). In contrast to the well-known tyrosine hydroxylases (EC 1.14.16.2) and tyrosinases (EC 1.14.18.1), LmbB2 was identified as a heme protein. Mass spectrometry and Soret band-excited Raman spectroscopy of LmbB2 showed that LmbB2 contains heme b as prosthetic group. The CO-reduced differential absorption spectra of LmbB2 showed that the coordination of Fe was different from that of cytochrome P450 enzymes. LmbB2 exhibits sequence similarity to Orf13 of the anthramycin biosynthetic gene cluster, which has recently been classified as a heme peroxidase. Tyrosine hydroxylating activity of LmbB2 yielding DOPA in the presence of (6R)-5,6,7,8-tetrahydro-L-biopterin (BH4) was also observed. Reaction mechanism of this unique heme peroxidases family is discussed. Also, tyrosine hydroxylation was confirmed as the first step of the amino acid branch of the lincomycin biosynthesis.


Assuntos
Antibacterianos/biossíntese , Proteínas de Bactérias/metabolismo , Hemeproteínas/metabolismo , Lincomicina/biossíntese , Streptomyces/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas de Bactérias/genética , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Di-Hidroxifenilalanina/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Expressão Gênica , Heme/química , Heme/metabolismo , Hemeproteínas/genética , Hidroxilação , Ferro/química , Ferro/metabolismo , Família Multigênica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptomyces/genética , Tirosina/metabolismo , Tirosina 3-Mono-Oxigenase/genética
20.
Chembiochem ; 14(17): 2259-62, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24166757

RESUMO

Chemical diversity: Two SAM-dependent N-methyltransferases-LmbJ from the biosynthesis of the antibiotic lincomycin and CcbJ from celesticetin biosynthesis-have been characterized and compared. Both tested enzymes form multimers and are able to utilize N-demethyllincomycin, the natural substrate of LmbJ, with comparable efficiency.


Assuntos
Antibacterianos/biossíntese , Biocatálise , Lincomicina/biossíntese , Lincosamidas/biossíntese , Metiltransferases/metabolismo , Antibacterianos/química , Lincomicina/química , Lincosamidas/química , Metiltransferases/química , Conformação Molecular , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...