Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters











Publication year range
1.
BMC Plant Biol ; 24(1): 804, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183318

ABSTRACT

BACKGROUND: 2-oxoglutarate-dependent dioxygenase (2ODD) superfamily is the second largest enzyme family in the plant genome and plays diverse roles in secondary metabolic pathways. The medicinal plant Scutellaria baicalensis Georgi contains various flavonoids, which have the potential to treat coronavirus disease 2019 (COVID-19), such as baicalein and myricetin. Flavone synthase I (FNSI) and flavanone 3-hydroxylase (F3H) from the 2ODDs of DOXC subfamily have been reported to participate in flavonoids biosynthesis. It is certainly interesting to study the 2ODD members involved in the biosynthesis of flavonoids in S. baicalensis. RESULTS: We provided a genome-wide analysis of the 2ODDs of DOXC subfamily in S. baicalensis, a total of 88 2ODD genes were identified, 82 of which were grouped into 25 distinct clades based on phylogenetic analysis of At2ODDs. We then performed a functional analysis of Sb2ODDs involved in the biosynthesis of flavones and dihydroflavonols. Sb2ODD1 and Sb2ODD2 from DOXC38 clade exhibit the activity of FNSI (Flavone synthase I), which exclusively converts pinocembrin to chrysin. Sb2ODD1 has significantly higher transcription levels in the root. While Sb2ODD7 from DOXC28 clade exhibits high expression in flowers, it encodes a F3H (flavanone 3-hydroxylase). This enzyme is responsible for catalyzing the conversion of both naringenin and pinocembrin into dihydrokaempferol and pinobanksin, kinetic analysis showed that Sb2ODD7 exhibited high catalytic efficiency towards naringenin. CONCLUSIONS: Our experiment suggests that Sb2ODD1 may serve as a supplementary factor to SbFNSII-2 and play a role in flavone biosynthesis specifically in the roots of S. baicalensis. Sb2ODD7 is mainly responsible for dihydrokaempferol biosynthesis in flowers, which can be further directed into the metabolic pathways of flavonols and anthocyanins.


Subject(s)
Dioxygenases , Flavonoids , Scutellaria baicalensis , Flavonoids/biosynthesis , Flavonoids/metabolism , Scutellaria baicalensis/genetics , Scutellaria baicalensis/metabolism , Scutellaria baicalensis/enzymology , Dioxygenases/genetics , Dioxygenases/metabolism , Phylogeny , Plant Proteins/genetics , Plant Proteins/metabolism , Flavanones/metabolism , Flavanones/biosynthesis , Gene Expression Regulation, Plant , Genes, Plant
2.
Appl Microbiol Biotechnol ; 108(1): 435, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39126431

ABSTRACT

Naringenin is a plant polyphenol, widely explored due to its interesting biological activities, namely anticancer, antioxidant, and anti-inflammatory. Due to its potential applications and attempt to overcome the industrial demand, there has been an increased interest in its heterologous production. The microbial biosynthetic pathway to produce naringenin is composed of tyrosine ammonia-lyase (TAL), 4-coumarate-CoA ligase (4CL), chalcone synthase (CHS), and chalcone isomerase (CHI). Herein, we targeted the efficient de novo production of naringenin in Escherichia coli by performing a step-by-step validation and optimization of the pathway. For that purpose, we first started by expressing two TAL genes from different sources in three different E. coli strains. The highest p-coumaric acid production (2.54 g/L) was obtained in the tyrosine-overproducing M-PAR-121 strain carrying TAL from Flavobacterium johnsoniae (FjTAL). Afterwards, this platform strain was used to express different combinations of 4CL and CHS genes from different sources. The highest naringenin chalcone production (560.2 mg/L) was achieved by expressing FjTAL combined with 4CL from Arabidopsis thaliana (At4CL) and CHS from Cucurbita maxima (CmCHS). Finally, different CHIs were tested and validated, and 765.9 mg/L of naringenin was produced by expressing CHI from Medicago sativa (MsCHI) combined with the other previously chosen genes. To our knowledge, this titer corresponds to the highest de novo production of naringenin reported so far in E. coli. KEY POINTS: • Best enzyme and strain combination were selected for de novo naringenin production. • After genetic and operational optimizations, 765.9 mg/L of naringenin was produced. • This de novo production is the highest reported so far in E. coli.


Subject(s)
Acyltransferases , Ammonia-Lyases , Biosynthetic Pathways , Coenzyme A Ligases , Escherichia coli , Flavanones , Flavanones/biosynthesis , Flavanones/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Biosynthetic Pathways/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Ammonia-Lyases/genetics , Ammonia-Lyases/metabolism , Metabolic Engineering/methods , Coumaric Acids/metabolism , Intramolecular Lyases/genetics , Intramolecular Lyases/metabolism , Tyrosine/metabolism
3.
Molecules ; 29(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38792058

ABSTRACT

The 1092 bp F3H gene from Trapa bispinosa Roxb., which was named TbF3H, was cloned and it encodes 363 amino acids. Bioinformatic and phylogenetic tree analyses revealed the high homology of TbF3H with flavanone 3-hydroxylase from other plants. A functional analysis showed that TbF3H of Trapa bispinosa Roxb. encoded a functional flavanone 3-hydroxylase; it catalyzed the formation of dihydrokaempferol (DHK) from naringenin in S. cerevisiae. The promoter strengths were compared by fluorescence microscopy and flow cytometry detection of the fluorescence intensity of the reporter genes initiated by each constitutive promoter (FITC), and DHK production reached 216.7 mg/L by the promoter adjustment strategy and the optimization of fermentation conditions. The results presented in this study will contribute to elucidating DHK biosynthesis in Trapa bispinosa Roxb.


Subject(s)
Flavanones , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Flavanones/biosynthesis , Flavanones/metabolism , Phylogeny , Promoter Regions, Genetic , Cloning, Molecular/methods , Flavonoids/biosynthesis , Plant Proteins/genetics , Plant Proteins/metabolism , Fermentation
4.
ACS Synth Biol ; 13(5): 1454-1466, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38662928

ABSTRACT

Chalcone synthase (CHS) catalyzes the rate-limiting step of (2S)-naringenin (the essential flavonoid skeleton) biosynthesis. Improving the activity of the CHS by protein engineering enhances (2S)-naringenin production by microbial fermentation and can facilitate the production of valuable flavonoids. A (2S)-naringenin biosensor based on the TtgR operon was constructed in Escherichia coli and its detection range was expanded by promoter optimization to 0-300 mg/L, the widest range for (2S)-naringenin reported. The high-throughput screening scheme for CHS was established based on this biosensor. A mutant, SjCHS1S208N with a 2.34-fold increase in catalytic activity, was discovered by directed evolution and saturation mutagenesis. A pathway for de novo biosynthesis of (2S)-naringenin by SjCHS1S208N was constructed in Saccharomyces cerevisiae, combined with CHS precursor pathway optimization, increasing the (2S)-naringenin titer by 65.34% compared with the original strain. Fed-batch fermentation increased the titer of (2S)-naringenin to 2513 ± 105 mg/L, the highest reported so far. These findings will facilitate efficient flavonoid biosynthesis and further modification of the CHS in the future.


Subject(s)
Acyltransferases , Biosensing Techniques , Directed Molecular Evolution , Escherichia coli , Fermentation , Flavanones , Saccharomyces cerevisiae , Flavanones/biosynthesis , Flavanones/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Directed Molecular Evolution/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Biosensing Techniques/methods , Protein Engineering/methods , Promoter Regions, Genetic , Operon/genetics , Metabolic Engineering/methods
5.
Metab Eng ; 83: 1-11, 2024 May.
Article in English | MEDLINE | ID: mdl-38447910

ABSTRACT

Flavonoids are a diverse set of natural products with promising bioactivities including anti-inflammatory, anti-cancer, and neuroprotective properties. Previously, the oleaginous host Yarrowia lipolytica has been engineered to produce high titers of the base flavonoid naringenin. Here, we leverage this host along with a set of E. coli bioconversion strains to produce the flavone apigenin and its glycosylated derivative isovitexin, two potential nutraceutical and pharmaceutical candidates. Through downstream strain selection, co-culture optimization, media composition, and mutant isolation, we were able to produce168 mg/L of apigenin, representing a 46% conversion rate of 2-(R/S)-naringenin to apigenin. This apigenin platform was modularly extended to produce isovitexin by addition of a second bioconversion strain. Together, these results demonstrate the promise of microbial production and modular bioconversion to access diversified flavonoids.


Subject(s)
Apigenin , Escherichia coli , Flavanones , Metabolic Engineering , Yarrowia , Apigenin/metabolism , Apigenin/biosynthesis , Flavanones/biosynthesis , Flavanones/metabolism , Yarrowia/metabolism , Yarrowia/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Glucosides/biosynthesis , Glucosides/metabolism
6.
Microb Cell Fact ; 22(1): 167, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37644530

ABSTRACT

BACKGROUND: Naringenin is an industrially relevant compound due to its multiple pharmaceutical properties as well as its central role in flavonoid biosynthesis. RESULTS: On our way to develop Streptomyces albidoflavus J1074 as a microbial cell factory for naringenin production, we have significantly increased the yields of this flavanone by combining various metabolic engineering strategies, fermentation strategies and genome editing approaches in a stepwise manner. Specifically, we have screened different cultivation media to identify the optimal production conditions and have investigated how the additive feeding of naringenin precursors influences the production. Furthermore, we have employed genome editing strategies to remove biosynthetic gene clusters (BGCs) associated with pathways that might compete with naringenin biosynthesis for malonyl-CoA precursors. Moreover, we have expressed MatBC, coding for a malonate transporter and an enzyme responsible for the conversion of malonate into malonyl-CoA, respectively, and have duplicated the naringenin BGC, further contributing to the production improvement. By combining all of these strategies, we were able to achieve a remarkable 375-fold increase (from 0.06 mg/L to 22.47 mg/L) in naringenin titers. CONCLUSION: This work demonstrates the influence that fermentation conditions have over the final yield of a bioactive compound of interest and highlights various bottlenecks that affect production. Once such bottlenecks are identified, different strategies can be applied to overcome them, although the efficiencies of such strategies may vary and are difficult to predict.


Subject(s)
Flavanones , Industrial Microbiology , Streptomyces , Metabolic Engineering , Streptomyces/growth & development , Streptomyces/metabolism , Flavanones/biosynthesis , Cerulenin/pharmacology , Phenylalanine/pharmacology , Tyrosine/pharmacology
7.
Sheng Wu Gong Cheng Xue Bao ; 38(4): 1565-1575, 2022 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-35470627

ABSTRACT

8-prenylnaringenin (8-PN) is a potent estrogen with high medicinal values. It also serves as an important precursor for many prenylated flavonoids. Microbial synthesis of 8-PN is mainly hindered by the low catalytic activity of prenyltransferases (PTS) and insufficient supply of precursors. In this work, a SfN8DT-1 from Sophora flavescens was used to improve the efficiency of (2S)-naringenin prenylation. The predicted structure of SfN8DT-1 showed that its main body is comprised of 9 α-helices and 8 loops, along with a long side chain formed by nearly 120 amino acids. SfN8DT-1 mutants with different side-chain truncated were tested in Saccharomyces cerevisiae. A mutant expressing the truncated enzyme at K62 site, designated as SfND8T-1-t62, produced the highest 8-PN titer. Molecular docking of SfN8DT-1-t62 with (2S)-naringenin and dimethylallyl diphosphate (DMAPP) showed that K185 was a potentially crucial residue. Alanine scanning within a range of 0.5 nm around these two substrates showed that the mutant K185A may decrease its affinity to substrates, which also indicated K185 was a potentially critical residue. Besides, the mutant K185W enhanced the affinity to ligands implied by the simulated saturation mutation, while the saturated mutation of K185 showed a great decrease in 8-PN production, indicating K185 is vital for the activity of SfN8DT-1. Subsequently, overexpressing the key genes of Mevalonate (MVA) pathway further improved the titer of 8-PN to 31.31 mg/L, which indicated that DMAPP supply is also a limiting factor for 8-PN synthesis. Finally, 44.92 mg/L of 8-PN was produced in a 5 L bioreactor after 120 h, which is the highest 8-PN titer reported to date.


Subject(s)
Dimethylallyltranstransferase , Flavanones/biosynthesis , Sophora , Dimethylallyltranstransferase/genetics , Dimethylallyltranstransferase/metabolism , Flavonoids/metabolism , Molecular Docking Simulation , Prenylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sophora/genetics , Sophora/metabolism
8.
ACS Synth Biol ; 10(8): 2030-2039, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34251173

ABSTRACT

Abundant gene clusters of natural products are observed in the endophytic fungus Phomopsis liquidambaris; however, most of them are silent. Herein, a plug-and-play DNA assembly tool has been applied for flavonoid synthesis in P. liquidambaris. A shuttle plasmid was constructed based on S. cerevisiae, E. coli, and P. liquidambaris with screening markers URA, Amp, and hygR, respectively. Each fragment or cassette was successively assembled by overlap extension PCR with at least 40-50 bp homologous arms in S. cerevisiae for generating a new vector. Seven native promoters were screened by the DNA assembly based on the fluorescence intensity of the mCherry reporter gene in P. liquidambaris, and two of them were new promoters. The key enzyme chalcone synthase was the limiting step of the pathway. The naringenin and kaempferol pathways were refactored and activated with the titers of naringenin and kaempferol of 121.53 mg/L and 75.38 mg/L in P. liquidambaris using fed-batch fermentation, respectively. This study will be efficient and helpful for the biosynthesis of secondary metabolites.


Subject(s)
Ascomycota , Endophytes , Flavanones/biosynthesis , Kaempferols/biosynthesis , Ascomycota/genetics , Ascomycota/metabolism , Endophytes/genetics , Endophytes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Flavanones/genetics , Kaempferols/genetics , Plasmids/genetics , Plasmids/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
9.
Angew Chem Int Ed Engl ; 60(31): 16874-16879, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34129275

ABSTRACT

Chalcone isomerase (CHI) is a key enzyme in the biosynthesis of flavonoids in plants. The first bacterial CHI (CHIera ) was identified from Eubacterium ramulus, but its distribution, evolutionary source, substrate scope, and stereoselectivity are still unclear. Here, we describe the identification of 66 novel bacterial CHIs from Genbank using a novel Sequence-Structure-Function-Evolution (SSFE) strategy. These novel bacterial CHIs show diversity in substrate specificity towards various hydroxylated and methoxylated chalcones. The mutagenesis of CHIera according to the substrate binding models of these novel bacterial CHIs resulted in several variants with greatly improved activity towards these chalcones. Furthermore, the preparative scale conversion catalyzed by bacterial CHIs has been performed for five chalcones and revealed (S)-selectivity with up to 96 % ee, which provides an alternative biocatalytic route for the synthesis of (S)-flavanones in high yields.


Subject(s)
Eubacterium/enzymology , Flavanones/biosynthesis , Intramolecular Lyases/metabolism , Flavanones/chemistry , Intramolecular Lyases/chemistry , Molecular Structure , Substrate Specificity
10.
ACS Synth Biol ; 10(5): 1143-1154, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33835777

ABSTRACT

Dynamic control of microbial metabolism is an effective strategy to improve chemical production in fermentations. While dynamic control is most often implemented using chemical inducers, optogenetics offers an attractive alternative due to the high tunability and reversibility afforded by light. However, a major concern of applying optogenetics in metabolic engineering is the risk of insufficient light penetration at high cell densities, especially in large bioreactors. Here, we present a new series of optogenetic circuits we call OptoAMP, which amplify the transcriptional response to blue light by as much as 23-fold compared to the basal circuit (OptoEXP). These circuits show as much as a 41-fold induction between dark and light conditions, efficient activation at light duty cycles as low as ∼1%, and strong homogeneous light-induction in bioreactors of at least 5 L, with limited illumination at cell densities above 40 OD600. We demonstrate the ability of OptoAMP circuits to control engineered metabolic pathways in novel three-phase fermentations using different light schedules to control enzyme expression and improve production of lactic acid, isobutanol, and naringenin. These circuits expand the applicability of optogenetics to metabolic engineering.


Subject(s)
Butanols/metabolism , Flavanones/biosynthesis , Lactic Acid/biosynthesis , Light , Metabolic Engineering/methods , Metabolic Networks and Pathways/radiation effects , Optogenetics/methods , Saccharomyces cerevisiae/metabolism , Signal Transduction/radiation effects , Bioreactors , DNA-Binding Proteins/genetics , Enzyme Activation/radiation effects , Fermentation/radiation effects , Gene Expression/radiation effects , Gene Expression Regulation/radiation effects , Metabolic Networks and Pathways/genetics , Microorganisms, Genetically-Modified , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription, Genetic/radiation effects
11.
ACS Synth Biol ; 10(5): 1087-1094, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33880917

ABSTRACT

Baicalein and scutellarein are bioactive flavonoids isolated from the traditional Chinese medicine Scutellaria baicalensis Georgi; however, there is a lack of effective strategies for producing baicalein and scutellarein. In this study, we developed a sequential self-assembly enzyme reactor involving two enzymes in the baicalein pathway with a pair of protein-peptide interactions in E. coli. These domains enabled us to optimize the stoichiometry of two baicalein biosynthetic enzymes recruited to be an enzymes complex. This strategy reduces the accumulation of intermediates and removes the pathway bottleneck. With this strategy, we successfully promoted the titer of baicalein by 6.6-fold (from 21.6 to 143.5 mg/L) and that of scutellarein by 1.4-fold (from 84.3 to 120.4 mg/L) in a flask fermentation, respectively. Furthermore, we first achieved the de novo biosynthesis of baicalein directly from glucose, and the strain was capable of producing 214.1 mg/L baicalein by fed-batch fermentation. This work provides novel insights for future optimization and large-scale fermentation of baicalein and scutellarein.


Subject(s)
Apigenin/biosynthesis , Bioreactors , Drugs, Chinese Herbal/metabolism , Escherichia coli/metabolism , Flavanones/biosynthesis , Metabolic Engineering/methods , Plant Extracts/biosynthesis , Batch Cell Culture Techniques/methods , Escherichia coli/genetics , Fermentation , Glucose/metabolism , Malonyl Coenzyme A/metabolism , Microorganisms, Genetically-Modified , PDZ Domains , Phenylalanine Ammonia-Lyase/chemistry , Phenylalanine Ammonia-Lyase/metabolism , Scutellaria baicalensis , Sirolimus/metabolism
12.
ACS Synth Biol ; 10(5): 1166-1175, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33877810

ABSTRACT

The (2S)-naringenin is an important natural flavonoid with several bioactive effects on human health. It is also a key precursor in the biosynthesis of other high value compounds. The production of (2S)-naringenin is significantly influenced by the acetyl-CoA available in the cytosol. In this study, we increased the acetyl-CoA supply via the ß-oxidation of fatty acids in the peroxisomes of Saccharomyces cerevisiae. Several lipases from different sources and PEX11, FOX1, FOX2, and FOX3, the key genes of the fatty acid ß-oxidation pathway, were overexpressed during the production of (2S)-naringenin in yeast. The level of acetyl-CoA was 0.205 nmol higher than that in the original strain and the production of (2S)-naringenin increased to 286.62 mg/g dry cell weight when PEX11 was overexpressed in S. cerevisiae strain L07. Remarkable (2S)-naringenin production (1129.44 mg/L) was achieved with fed-batch fermentation, with the highest titer reported in any microorganism. Our results demonstrated the use of fatty acid ß-oxidation to increase the level of cytoplasmic acetyl-CoA and the production of its derivatives.


Subject(s)
Fatty Acids/metabolism , Flavanones/biosynthesis , Metabolic Engineering/methods , Saccharomyces cerevisiae/metabolism , Acetyl Coenzyme A/metabolism , Cytosol/metabolism , Fermentation , Malonyl Coenzyme A/metabolism , Membrane Proteins/metabolism , Microorganisms, Genetically-Modified , Oxidation-Reduction , Peroxins/metabolism , Peroxisomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
13.
ACS Synth Biol ; 9(12): 3288-3297, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33226782

ABSTRACT

The compound (2S)-eriodictyol is an important flavonoid that can be derived from (2S)-naringenin through flavonoid 3'-hydroxylase (F3'H) catalyzation. F3'H is a cytochrome P450 enzyme that requires a cytochrome P450 reductase (CPR) to function. However, P450s have limited applications in industrial scale biosynthesis, owing to their low activity. Here, an efficient SmF3'H and a matched SmCPR were identified from Silybum marianum. To improve the efficiency of SmF3'H, we established a high-throughput detection method for (2S)-eriodictyol, in which the promoter combination of SmF3'H and SmCPR were optimized in Saccharomyces cerevisiae. The results revealed that SmF3'H/SmCPR should be expressed by using promoters with similar and strong expression levels. Furthermore, directed evolution was applied to further improve the efficiency of SmF3'H/SmCPR. With the optimized promoter and mutated combinations SmF3'HD285N/SmCPRI453V, the (2S)-eriodictyol titer was improved to 3.3 g/L, the highest titer in currently available reports. These results indicated that S. cerevisiae is an ideal platform for functional expression of flavonoid related P450 enzymes.


Subject(s)
Flavanones/biosynthesis , Flavanones/metabolism , Saccharomyces cerevisiae/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Directed Molecular Evolution , Flavanones/analysis , Silybum marianum/genetics , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae/chemistry
14.
ACS Synth Biol ; 9(11): 3042-3051, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33107298

ABSTRACT

Pinocembrin derived flavones are the major bioactive compounds presented in the Lamiaceae plants that have long been of interest due to their great pharmaceutical and economical significance. Modifications on the central skeleton of the flavone moiety have a huge impact on their biological activities. However, the enzymes responsible for structure modification of most flavones are either inefficient or remain unidentified. By integrating omics analysis of Scutellaria barbata and synthetic biology tools in yeast chassis, we characterized a novel gene encoding flavone 7-O-methyltransferase (F7OMT) and discovered a new flavone 8-hydroxylase (F8H) with increased activity. We also identified a series of flavone 6-hydroxylases (F6Hs) and flavone 8-O-methyltransferases (F8OMTs) in this study. Subsequently, we constructed the biosynthetic pathway for chrysin production by assembling catalytic elements from different species and improved the titer to 10.06 mg/L. Using the established chrysin production platform, we achieved the de novo biosynthesis of baicalein, baicalin, norwogonin, wogonin, isowogonin, and moslosooflavone in yeast. Our results indicated that the combination of omics and synthetic biology can greatly speed up the efficiency of gene mining in plants and the engineered yeasts established an alternative way for the production of pinocembrin derivatives.


Subject(s)
Flavanones/biosynthesis , Flavanones/metabolism , Saccharomyces cerevisiae/metabolism , Biosynthetic Pathways/physiology , Flavones/biosynthesis , Flavones/metabolism , Flavonoids/biosynthesis , Flavonoids/metabolism , Lamiaceae/metabolism , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/metabolism
15.
BMC Microbiol ; 20(1): 291, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32957914

ABSTRACT

BACKGROUND: The dried roots and rhizomes of medicinal licorices are widely used worldwide as a traditional medicinal herb, which are mainly attributed to a variety of bioactive compounds that can be extracted from licorice root. Endophytes and plants form a symbiotic relationship, which is an important source of host secondary metabolites. RESULTS: In this study, we used high-throughput sequencing technology and high-performance liquid chromatography to explore the composition and structure of the endophytic bacterial community and the content of bioactive compounds (glycyrrhizic acid, liquiritin and total flavonoids) in different species of medicinal licorices (Glycyrrhiza uralensis, Glycyrrhiza glabra, and Glycyrrhiza inflata) and in different planting years (1-3 years). Our results showed that the contents of the bioactive compounds in the roots of medicinal licorices were not affected by the species, but were significantly affected by the main effect growing year (1-3) (P < 0.05), and with a trend of stable increase in the contents observed with each growing year. In 27 samples, a total of 1,979,531 effective sequences were obtained after quality control, and 2432 effective operational taxonomic units (OTUs) were obtained at 97% identity. The phylum Proteobacteria, Actinobacteria, Bacteroidetes and Firmicutes, and the genera unified-Rhizobiaceae, Pseudomonas, Novosphingobium, and Pantoea were significantly dominant in the 27 samples. Distance-based redundancy analysis (db-RDA) showed that the content of total flavonoids explained the differences in composition and distribution of endophytic bacterial communities in roots of cultivated medicinal liquorices to the greatest extent. Total soil salt was the most important factor that significantly affected the endophytic bacterial community in soil factors, followed by ammonium nitrogen and nitrate nitrogen. Among the leaf nutrition factors, leaf water content had the most significant effect on the endophytic bacterial community, followed by total phosphorus and total potassium. CONCLUSIONS: This study not only provides information on the composition and distribution of endophytic bacteria in the roots of medicinal licorices, but also reveals the influence of abiotic factors on the community of endophytic bacteria and bioactive compounds, which provides a reference for improving the quality of licorice.


Subject(s)
Flavonoids/biosynthesis , Glycyrrhiza uralensis/microbiology , Glycyrrhiza/microbiology , Plant Roots/microbiology , Rhizome/microbiology , Actinobacteria/classification , Actinobacteria/genetics , Actinobacteria/isolation & purification , Ammonia/pharmacology , Bacteroidetes/classification , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , DNA Barcoding, Taxonomic , DNA, Bacterial/genetics , Endophytes/physiology , Firmicutes/classification , Firmicutes/genetics , Firmicutes/isolation & purification , Flavanones/biosynthesis , Flavanones/isolation & purification , Flavonoids/classification , Flavonoids/isolation & purification , Glucosides/biosynthesis , Glucosides/isolation & purification , Glycyrrhiza/drug effects , Glycyrrhiza/metabolism , Glycyrrhiza uralensis/drug effects , Glycyrrhiza uralensis/metabolism , Glycyrrhizic Acid/isolation & purification , Glycyrrhizic Acid/metabolism , Microbial Consortia/drug effects , Microbial Consortia/genetics , Nitrates/pharmacology , Phylogeny , Plant Roots/metabolism , Proteobacteria/classification , Proteobacteria/genetics , Proteobacteria/isolation & purification , Rhizobiaceae/classification , Rhizobiaceae/genetics , Rhizobiaceae/isolation & purification , Rhizome/metabolism , Seasons , Secondary Metabolism , Soil/chemistry , Soil Microbiology , Symbiosis
16.
J Microbiol Biotechnol ; 30(10): 1574-1582, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32830192

ABSTRACT

Flavonoids have diverse biological functions in human health. All flavonoids contain a common 2-phenyl chromone structure (C6-C3-C6) as a scaffold. Hence, in using such a scaffold, plenty of highvalue-added flavonoids can be synthesized by chemical or biological catalyzation approaches. (2S)-Naringenin is one of the most commonly used flavonoid scaffolds. However, biosynthesizing (2S)-naringenin has been restricted not only by low production but also by the expensive precursors and inducers that are used. Herein, we established an induction-free system to de novo biosynthesize (2S)-naringenin in Escherichia coli. The tyrosine synthesis pathway was enhanced by overexpressing feedback inhibition-resistant genes (aroGfbr and tyrAfbr) and knocking out a repressor gene (tyrR). After optimizing the fermentation medium and conditions, we found that glycerol, glucose, fatty acids, potassium acetate, temperature, and initial pH are important for producing (2S)-naringenin. Using the optimum fermentation medium and conditions, our best strain, Nar-17LM1, could produce 588 mg/l (2S)-naringenin from glucose in a 5-L bioreactor, the highest titer reported to date in E. coli.


Subject(s)
Anti-Ulcer Agents/metabolism , Biosynthetic Pathways , Escherichia coli/metabolism , Estrogen Antagonists/metabolism , Fermentation , Batch Cell Culture Techniques , Bioreactors , Culture Media , Escherichia coli/genetics , Flavanones/biosynthesis , Flavonoids/metabolism , Hydrogen-Ion Concentration , Metabolic Engineering , Temperature , Tyrosine/metabolism
17.
Food Res Int ; 136: 109291, 2020 10.
Article in English | MEDLINE | ID: mdl-32846510

ABSTRACT

Citrus peel wastes are an important renewable resource and rich in naringin, a flavonoid compound with multiple bioactivities. To cope with the low bioavailability of naringin, a new bienzyme whole-cell system was developed for bioconversion of naringin into two lipophilic derivatives. A series of naringin esters with different fatty acid chain length were successfully synthesized via cell-bound lipase catalyzed acylation, and another lipophilic product naringenin was simultaneously yielded via intracellular naringinase-catalyzed hydrolysis. The naringin esters obtained showed higher log P values and free radical-scavenging capacities against DPPH and ABTS than naringin itself. These esters also showed markedly enhanced permeability across the human intestinal Caco-2 cells. The whole-cell mediated conversion of naringin offers a two-fold advantage: naringin esters are produced as new high-valued derivatives with high lipophilicity and antioxidant activity; and the tasteless product naringenin was obtained simultaneously, which can reduce the bitterness of the total product and benefited its industrial applications.


Subject(s)
Antioxidants , Aspergillus oryzae/enzymology , Flavanones/metabolism , Lipids , Acylation , Biological Availability , Caco-2 Cells , Citrus/chemistry , Esters/metabolism , Fatty Acids/chemistry , Flavanones/biosynthesis , Flavanones/chemistry , Flavanones/pharmacokinetics , Fruit/chemistry , Humans , Intestinal Absorption , Lipase/metabolism , Multienzyme Complexes/metabolism , beta-Glucosidase/metabolism
18.
ACS Synth Biol ; 9(7): 1823-1832, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32525654

ABSTRACT

Combinatorial biosynthesis has great potential for designing synthetic circuits and amplifying the production of new active compounds. Studies on multienzyme cascades are extremely useful for improving our knowledge on enzymatic catalysis. In particular, the elucidation of enzyme substrate promiscuity can be potentially used for bioretrosynthetic approaches, leading to the design of alternative and more convenient routes to produce relevant molecules. In this perspective, plant-derived polyketides are extremely adaptable to those synthetic biological applications. Here, we present a combination of an in vitro CoA ligase activity assay coupled with a bacterial multigene expression system that leads to precursor-directed biosynthesis of 21 flavonoid derivatives. When the vast knowledge from protein databases is exploited, the herein presented procedure can be easily repeated with additional plant-derived polyketides. Lastly, we report an efficient in vivo expression system that can be further exploited to heterologously express pathways not necessarily related to plant polyketide synthases.


Subject(s)
Arabidopsis/genetics , Arabidopsis/metabolism , Flavanones/biosynthesis , Polyketide Synthases/metabolism , Polyketides/metabolism , Acyltransferases , Arabidopsis Proteins , Chalcones/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors , Malonyl Coenzyme A/metabolism , Plasmids/genetics , Substrate Specificity , Synthetic Biology/methods
19.
J Agric Food Chem ; 68(4): 1015-1021, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31690080

ABSTRACT

(2S)-Naringenin, a (2S)-flavanone, is widely used in the food, chemical, and pharmaceutical industries because of its diverse physiological activities. The production of (2S)-naringenin in microorganisms provides an ideal source that reduces the cost of the flavonoid. To achieve efficient production of (2S)-naringenin in Saccharomyces cerevisiae (S. cerevisiae), we constructed a biosynthetic pathway from p-coumaric acid, a cost-effective and more efficient precursor. The (2S)-naringenin synthesis pathway genes were integrated into the yeast genome to obtain a (2S)-naringenin production strain. After gene dosage experiments, the genes negatively regulating the shikimate pathway and inefficient chalcone synthase activity were verified as factors limiting (2S)-naringenin biosynthesis. With fed-batch process optimization of the engineered strain, the titer of (2S)-naringenin reached 648.63 mg/L from 2.5 g/L p-coumaric acid. Our results indicate that the constitutive production of (2S)-naringenin from p-coumaric acid in S. cerevisiae is highly promising.


Subject(s)
Coumaric Acids/metabolism , Flavanones/biosynthesis , Saccharomyces cerevisiae/metabolism , Biosynthetic Pathways , Saccharomyces cerevisiae/genetics
20.
J Ind Microbiol Biotechnol ; 46(12): 1707-1713, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31595455

ABSTRACT

Methylated flavonoids possess improved bioactivities compared to their unmethylated counterparts. In this study, for the efficient production of O-methylated flavonoids from simple methylated phenylpropanoic acids, a recombinant Escherichia coli strain expressing 4-coumarate:coenzyme A ligase (4CL) from Oryza sativa and chalcone synthase (CHS) from Hordeum vulgare was constructed; this strain produced significant amount of homoeriodictyol (~ 52 mg/L) as well as a few amount of hesperetin (0.4 mg/L), respectively, from ferulic acid and 4-methylcaffeic acid. This demonstrates, for the first time, that the scarce but valuable methylated flavanones can be successfully produced from methylated phenylpropanoic acids in a microbial host via an artificial biosynthetic pathway consisting of 4CL and CHS that can accept O-methylated precursors.


Subject(s)
Acids, Carbocyclic/metabolism , Escherichia coli/metabolism , Flavanones/biosynthesis , Acyltransferases/metabolism , Biosynthetic Pathways , Coenzyme A Ligases/metabolism , Coumaric Acids/metabolism , Escherichia coli/genetics
SELECTION OF CITATIONS
SEARCH DETAIL