Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.049
Filter
1.
PLoS One ; 19(5): e0299588, 2024.
Article in English | MEDLINE | ID: mdl-38718091

ABSTRACT

Corynebacterium glutamicum is a non-pathogenic species of the Corynebacteriaceae family. It has been broadly used in industrial biotechnology for the production of valuable products. Though it is widely accepted at the industrial level, knowledge about the genomic diversity of the strains is limited. Here, we investigated the comparative genomic features of the strains and pan-genomic characteristics. We also observed phylogenetic relationships among the strains based on average nucleotide identity (ANI). We found diversity between strains at the genomic and pan-genomic levels. Less than one-third of the C. glutamicum pan-genome consists of core genes and soft-core genes. Whereas, a large number of strain-specific genes covered about half of the total pan-genome. Besides, C. glutamicum pan-genome is open and expanding, which indicates the possible addition of new gene families to the pan-genome. We also investigated the distribution of biosynthetic gene clusters (BGCs) among the strains. We discovered slight variations of BGCs at the strain level. Several BGCs with the potential to express novel bioactive secondary metabolites have been identified. Therefore, by utilizing the characteristic advantages of C. glutamicum, different strains can be potential applicants for natural drug discovery.


Subject(s)
Corynebacterium glutamicum , Genetic Variation , Genome, Bacterial , Phylogeny , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Multigene Family , Genomics/methods
2.
Curr Microbiol ; 81(6): 167, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727744

ABSTRACT

Diabetes mellitus represents a persistent metabolic condition marked by heightened levels of blood glucose, presenting a considerable worldwide health concern, and finding targeted treatment for it is a crucial priority for global health. Gram-positive aerobic bacteria, predominantly inhabiting water and soil, are known carriers of various enzyme-encoding genetic material, which includes the malic enzyme gene that plays a role in insulin secretion. Corynebacterium glutamicum bacteria (ATCC 21799) were acquired from the Pasteur Institute and confirmed using microbiological and molecular tests, including DNA extraction. After identification, gene purification and cloning of the maeB gene were performed using the TA Cloning method. Additionally, the enhancement of enzyme expression was assessed using the expression vector pET-28a, and validation of simulation results was monitored through a real-time PCR analysis. Based on previous studies, the malic enzyme plays a pivotal role in maintaining glucose homeostasis, and increased expression of this enzyme has been associated with enhanced insulin sensitivity. However, the production of malic enzyme has encountered numerous challenges and difficulties. This study successfully isolated the malic enzyme genes via Corynebacterium glutamicum and introduced them into Escherichia coli for high-yield production. According to the results, the optimum temperature for the activity of enzymes has been identified as 39 °C.


Subject(s)
Cloning, Molecular , Corynebacterium glutamicum , Escherichia coli , Malate Dehydrogenase , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Escherichia coli/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/enzymology , Diabetes Mellitus/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression , Temperature , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Microb Cell Fact ; 23(1): 147, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783320

ABSTRACT

Aminopyrrolnitrin (APRN), a natural halogenated phenylpyrrole derivative (HPD), has strong antifungal and antiparasitic activities. Additionally, it showed 2.8-fold increased photostability compared to pyrrolnitrin, a commercially available HPD with antimicrobial activity. For microbial production of APRN, we first engineered anthranilate phosphoribosyltransferase encoded by trpD from Corynebacterium glutamicum, resulting in a TrpDA162D mutation that exhibits feedback-resistant against L-tryptophan and higher substrate affinity compared to wild-type TrpD. Plasmid-borne expression of trpDA162D in C. glutamicum TP851 strain with two copies of trpDA162D in the genome led to the production of 3.1 g/L L-tryptophan in flask culture. Subsequent step for L-tryptophan chlorination into 7-chloro-L-tryptophan was achieved by introducing diverse sources of genes encoding tryptophan 7-halogenase (PrnA or RebH) and flavin reductase (Fre, PrnF, or RebF). The combined expression of prnA from Serratia grimesii or Serratia plymuthica with flavin reductase gene from Escherichia coli, Pseudomonas fluorescens, or Lechevalieria aerocolonigenes yielded higher production of 7-chloro-L-tryptophan in comparison to other sets of two-component systems. In the next step, production of putative monodechloroaminopyrrolnitrin (MDAP) from 7-chloro-L-tryptophan was achieved through the expression of prnB encoding MDAP synthase from S. plymuthica or P. fluorescens. Finally, an artificial APRN biosynthetic pathway was constructed by simultaneously expressing genes coding for tryptophan 7-halogenase, flavin reductase, MDAP synthase, and MDAP halogenase (PrnC) from different microbial sources within the L-tryptophan-producing TP851 strain. As prnC from S. grimesii or S. plymuthica was introduced into the host strain, which carried plasmids expressing prnA from S. plymuthica, fre from E. coli, and prnB from S. plymuthica, APN3639 and APN3638 accumulated 29.5 mg/L and 28.1 mg/L of APRN in the culture broth. This study represents the first report on the fermentative APRN production by metabolically engineered C. glutamicum.


Subject(s)
Corynebacterium glutamicum , Metabolic Engineering , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/genetics , Metabolic Engineering/methods , Pyrrolnitrin/biosynthesis , Pyrrolnitrin/metabolism , Fermentation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Tryptophan/biosynthesis , Tryptophan/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Oxidoreductases
4.
J Chem Inf Model ; 64(10): 4250-4262, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38701175

ABSTRACT

The microbial enzyme diaminopimelate epimerase (DapF), a vital enzyme in the lysine biosynthetic pathway, catalyzes the conversion of L, L-diaminopimelate (L, L-DAP) to D, L-diaminopimelate (D, L-DAP) using a catalytic cysteine dyad with one cysteine in thiol state and another in thiolate. Under oxidizing conditions, the catalytic cysteines of apo DapF form a disulfide bond that alters the structure and function of DapF. Given its potential as a target for antimicrobial resistance treatments, understanding DapF's functional dynamics is imperative. In the present work, we employ microsecond-scale all-atom molecular dynamics simulations of product-bound DapF and apo-DapF under oxidized and reduced conditions. We employ a polarized charge model for the ligand and the active site residues, which was necessary to preserve the electrostatic environment in the active site and retain the ligand in the active site. The product-bound DapF and apo-DapF in oxidized and reduced conditions exhibit a closed, semi-open, and open conformation, respectively, as identified using the internal coordinates of the dimeric enzyme and the principal component analysis. The conformational switch is guided by the dynamic catalytic (DC) loop, loop II, and loop III movements in the active site. The time scale of the close-to-open conformational transition is estimated to be 0.8 µs through Markov state modeling (MSM) and transition path theory (TPT). The present study explains the role of various active site residues and loops in ligand binding and protein dynamics in the DapF enzyme under different redox conditions. Such information will be helpful in future inhibitor design studies targeting the DapF enzyme.


Subject(s)
Corynebacterium glutamicum , Markov Chains , Molecular Dynamics Simulation , Protein Conformation , Corynebacterium glutamicum/enzymology , Ligands , Amino Acid Isomerases/metabolism , Amino Acid Isomerases/chemistry , Catalytic Domain , Oxidation-Reduction
5.
Bioresour Technol ; 402: 130774, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701983

ABSTRACT

Formate as an ideal mediator between the physicochemical and biological realms can be obtained from electrochemical reduction of CO2 and used to produce bio-chemicals. Yet, limitations arise when employing natural formate-utilizing microorganisms due to restricted product range and low biomass yield. This study presents a breakthrough: engineered Corynebacterium glutamicum strains (L2-L4) through modular engineering. L2 incorporates the formate-tetrahydrofolate cycle and reverse glycine cleavage pathway, L3 enhances NAD(P)H regeneration, and L4 reinforces metabolic flux. Metabolic modeling elucidates C1 assimilation, guiding strain optimization for co-fermentation of formate and glucose. Strain L4 achieves an OD600 of 0.5 and produces 0.6 g/L succinic acid. 13C-labeled formate confirms C1 assimilation, and further laboratory evolution yields 1.3 g/L succinic acid. This study showcases a successful model for biologically assimilating formate in C. glutamicum that could be applied in C1-based biotechnological production, ultimately forming a formate-based bioeconomy.


Subject(s)
Biomass , Corynebacterium glutamicum , Formates , Metabolic Engineering , Succinic Acid , Corynebacterium glutamicum/metabolism , Formates/metabolism , Metabolic Engineering/methods , Succinic Acid/metabolism , Fermentation , Models, Biological , Glucose/metabolism
6.
Molecules ; 29(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38792114

ABSTRACT

Flavonoids and stilbenoids, crucial secondary metabolites abundant in plants and fungi, display diverse biological and pharmaceutical activities, including potent antioxidant, anti-inflammatory, and antimicrobial effects. However, conventional production methods, such as chemical synthesis and plant extraction, face challenges in sustainability and yield. Hence, there is a notable shift towards biological production using microorganisms like Escherichia coli and yeast. Yet, the drawbacks of using E. coli and yeast as hosts for these compounds persist. For instance, yeast's complex glycosylation profile can lead to intricate protein production scenarios, including hyperglycosylation issues. Consequently, Corynebacterium glutamicum emerges as a promising alternative, given its adaptability and recent advances in metabolic engineering. Although extensively used in biotechnological applications, the potential production of flavonoid and stilbenoid in engineered C. glutamicum remains largely untapped compared to E. coli. This review explores the potential of metabolic engineering in C. glutamicum for biosynthesis, highlighting its versatility as a cell factory and assessing optimization strategies for these pathways. Additionally, various metabolic engineering methods, including genomic editing and biosensors, and cofactor regeneration are evaluated, with a focus on C. glutamicum. Through comprehensive discussion, the review offers insights into future perspectives in production, aiding researchers and industry professionals in the field.


Subject(s)
Corynebacterium glutamicum , Flavonoids , Metabolic Engineering , Stilbenes , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/genetics , Metabolic Engineering/methods , Flavonoids/biosynthesis , Flavonoids/metabolism , Stilbenes/metabolism
7.
J Agric Food Chem ; 72(21): 12219-12228, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38747135

ABSTRACT

Phycocyanobilin, an algae-originated light-harvesting pigment known for its antioxidant properties, has gained attention as it plays important roles in the food and medication industries and has surged in demand owing to its low-yield extraction from natural resources. In this study, engineered Corynebacterium glutamicum was developed to achieve high PCB production, and three strategies were proposed: reinforcement of the heme biosynthesis pathway with the introduction of two PCB-related enzymes, strengthening of the pentose phosphate pathway to generate an efficient cycle of NADPH, and fed-batch fermentation to maximize PCB production. Each approach increased PCB synthesis, and the final engineered strain successfully produced 78.19 mg/L in a flask and 259.63 mg/L in a 5 L bioreactor, representing the highest bacterial production of PCB reported to date, to our knowledge. The strategies applied in this study will be useful for the synthesis of PCB derivatives and can be applied in the food and pharmaceutical industries.


Subject(s)
Corynebacterium glutamicum , Metabolic Engineering , Phycobilins , Phycocyanin , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/genetics , Phycocyanin/metabolism , Phycocyanin/genetics , Phycobilins/metabolism , Phycobilins/genetics , Fermentation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Pentose Phosphate Pathway/genetics , Bioreactors/microbiology
8.
Int J Biol Macromol ; 267(Pt 2): 131415, 2024 May.
Article in English | MEDLINE | ID: mdl-38582485

ABSTRACT

The complete enzyme catalytic cycle includes substrate binding, chemical reaction and product release, in which different dynamic conformations are adopted. Due to the complex relationship among enzyme activity, stability and dynamics, the directed evolution of enzymes for improved activity or stability commonly leads to a trade-off in stability or activity. It hence remains a challenge to engineer an enzyme to have both enhanced activity and stability. Here, we have attempted to reconstruct the dynamics correlation network involved with active center to improve both activity and stability of a 2,3-butanediol dehydrogenase (2,3-BDH) by introducing inter-chain disulfide bonds. A computational strategy was first applied to evaluate the effect of introducing inter-chain disulfide bond on activity and stability of three 2,3-BDHs, and the N258C mutation of 2,3-BDH from Corynebacterium glutamicum (CgBDH) was proved to be effective in improving both activity and stability. In the results, CgBDH-N258C showed a different unfolding curve from the wild type, with two melting temperatures (Tm) of 68.3 °C and 50.8 °C, 19.7 °C and 2 °C higher than 48.6 °C of the wild type. Its half-life was also improved by 14.8-fold compared to the wild type. Catalytic efficiency (kcat/Km) of the mutant was increased by 7.9-fold toward native substrate diacetyl and 8.8-fold toward non-native substrate 2,5-hexanedione compared to the wild type. Molecular dynamics simulations revealed that an interaction network formed by Cys258, Arg162, Ala144 and the catalytic residues was reconstructed in the mutant and the dynamics change caused by the disulfide bond could be propagated through the interactions network. This improved the enzyme stability and activity by decreasing the flexibility and locking more "reactive" pose, respectively. Further construction of mutations including A144G showing a 44-fold improvement in catalytic efficiency toward meso-2,3-BD confirmed the role of modifying dynamics correlation network in tunning enzyme activity and selectivity. This study provided important insights into the relationship among dynamics, enzyme catalysis and stability, and will be useful in the designing new enzymes with co-evolution of stability, activity and selectivity.


Subject(s)
Alcohol Oxidoreductases , Corynebacterium glutamicum , Disulfides , Enzyme Stability , Molecular Dynamics Simulation , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Disulfides/chemistry , Corynebacterium glutamicum/enzymology , Corynebacterium glutamicum/genetics , Mutation , Catalytic Domain , Kinetics , Protein Conformation , Protein Engineering/methods
9.
J Microbiol Biotechnol ; 34(5): 1154-1163, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38563097

ABSTRACT

Glucosylation is a well-known approach to improve the solubility, pharmacological, and biological properties of flavonoids, making flavonoid glucosides a target for large-scale biosynthesis. However, the low yield of products coupled with the requirement of expensive UDP-sugars limits the application of enzymatic systems for large-scale. C. glutamicum is a Gram-positive and generally regarded as safe (GRAS) bacteria frequently employed for the large-scale production of amino acids and bio-fuels. Due to the versatility of its cell factory system and its non-endotoxin producing properties, it has become an attractive system for the industrial-scale biosynthesis of alternate products. Here, we explored the cell factory of C. glutamicum for efficient glucosylation of flavonoids using apigenin as a model flavonoid, with the heterologous expression of a promiscuous glycosyltransferase, YdhE from Bacillus licheniformis and the endogenous overexpression of C. glutamicum genes galU1 encoding UDP-glucose pyrophosphorylase and pgm encoding phosphoglucomutase involved in the synthesis of UDP-glucose to create a C. glutamicum cell factory system capable of efficiently glucosylation apigenin with a high yield of glucosides production. Consequently, the production of various apigenin glucosides was controlled under different temperatures yielding almost 4.2 mM of APG1(apigenin-4'-O-ß-glucoside) at 25°C, and 0.6 mM of APG2 (apigenin-7-O-ß-glucoside), 1.7 mM of APG3 (apigenin-4',7-O-ß-diglucoside) and 2.1 mM of APG4 (apigenin-4',5-O-ß-diglucoside) after 40 h of incubation with the supplementation of 5 mM of apigenin and 37°C. The cost-effective developed system could be used to modify a wide range of plant secondary metabolites with increased pharmacokinetic activities on a large scale without the use of expensive UDP-sugars.


Subject(s)
Apigenin , Corynebacterium glutamicum , Glucosides , Metabolic Engineering , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/genetics , Apigenin/metabolism , Metabolic Engineering/methods , Glucosides/metabolism , Glucosides/biosynthesis , Glycosylation , Bacillus licheniformis/metabolism , Bacillus licheniformis/genetics , Bacillus licheniformis/enzymology , Uridine Diphosphate Glucose/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , UTP-Glucose-1-Phosphate Uridylyltransferase/metabolism , UTP-Glucose-1-Phosphate Uridylyltransferase/genetics , Glycosyltransferases/metabolism , Glycosyltransferases/genetics
10.
J Am Chem Soc ; 146(17): 12138-12154, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38635392

ABSTRACT

Protein lipidation dynamically controls protein localization and function within cellular membranes. A unique form of protein O-fatty acylation in Corynebacterium, termed protein O-mycoloylation, involves the attachment of mycolic acids─unusually large and hydrophobic fatty acids─to serine residues of proteins in these organisms' outer mycomembrane. However, as with other forms of protein lipidation, the scope and functional consequences of protein O-mycoloylation are challenging to investigate due to the inherent difficulties of enriching and analyzing lipidated peptides. To facilitate the analysis of protein lipidation and enable the comprehensive profiling and site mapping of protein O-mycoloylation, we developed a chemical proteomics strategy integrating metabolic labeling, click chemistry, cleavable linkers, and a novel liquid chromatography-tandem mass spectrometry (LC-MS/MS) method employing LC separation and complementary fragmentation methods tailored to the analysis of lipophilic, MS-labile O-acylated peptides. Using these tools in the model organism Corynebacterium glutamicum, we identified approximately 30 candidate O-mycoloylated proteins, including porins, mycoloyltransferases, secreted hydrolases, and other proteins with cell envelope-related functions─consistent with a role for O-mycoloylation in targeting proteins to the mycomembrane. Site mapping revealed that many of the proteins contained multiple spatially proximal modification sites, which occurred predominantly at serine residues surrounded by conformationally flexible peptide motifs. Overall, this study (i) discloses the putative protein O-mycoloylome for the first time, (ii) yields new insights into the undercharacterized proteome of the mycomembrane, which is a hallmark of important pathogens (e.g., Corynebacterium diphtheriae, Mycobacterium tuberculosis), and (iii) provides generally applicable chemical strategies for the proteomic analysis of protein lipidation.


Subject(s)
Bacterial Proteins , Corynebacterium glutamicum , Proteomics , Proteomics/methods , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/chemistry , Mycolic Acids/metabolism , Mycolic Acids/chemistry , Tandem Mass Spectrometry , Chromatography, Liquid , Acylation , Click Chemistry
11.
Sci Rep ; 14(1): 8081, 2024 04 06.
Article in English | MEDLINE | ID: mdl-38582923

ABSTRACT

Astaxanthin, a versatile C40 carotenoid prized for its applications in food, cosmetics, and health, is a bright red pigment with powerful antioxidant properties. To enhance astaxanthin production in Corynebacterium glutamicum, we employed rational pathway engineering strategies, focused on improving precursor availability and optimizing terminal oxy-functionalized C40 carotenoid biosynthesis. Our efforts resulted in an increased astaxanthin precursor supply with 1.5-fold higher ß-carotene production with strain BETA6 (18 mg g-1 CDW). Further advancements in astaxanthin production were made by fine-tuning the expression of the ß-carotene hydroxylase gene crtZ and ß-carotene ketolase gene crtW, yielding a nearly fivefold increase in astaxanthin (strain ASTA**), with astaxanthin constituting 72% of total carotenoids. ASTA** was successfully transferred to a 2 L fed-batch fermentation with an enhanced titer of 103 mg L-1 astaxanthin with a volumetric productivity of 1.5 mg L-1 h-1. Based on this strain a pathway expansion was achieved towards glycosylated C40 carotenoids under heterologous expression of the glycosyltransferase gene crtX. To the best of our knowledge, this is the first time astaxanthin-ß-D-diglucoside was produced with C. glutamicum achieving high titers of microbial C40 glucosides of 39 mg L-1. This study showcases the potential of pathway engineering to unlock novel C40 carotenoid variants for diverse industrial applications.


Subject(s)
Carotenoids , Corynebacterium glutamicum , Carotenoids/metabolism , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Xanthophylls/metabolism , beta Carotene/metabolism , Metabolic Engineering/methods
12.
J Agric Food Chem ; 72(15): 8674-8683, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38569079

ABSTRACT

The enhancement of intracellular glutamate synthesis in glutamate-independent poly-γ-glutamic acid (γ-PGA)-producing strains is an essential strategy for improving γ-PGA production. Bacillus tequilensis BL01ΔpgdSΔggtΔsucAΔgudB:P43-ppc-pyk-gdhA for the efficient synthesis of γ-PGA was constructed through expression of glutamate synthesis features of Corynebacterium glutamicum, which increased the titer of γ-PGA by 2.18-fold (3.24 ± 0.22 g/L) compared to that of B. tequilensis BL01ΔpgdSΔggtΔsucAΔgudB (1.02 ± 0.11 g/L). To further improve the titer of γ-PGA and decrease the production of byproducts, three enzymes (Ppc, Pyk, and AceE) were assembled to a complex using SpyTag/Catcher pairs. The results showed that the γ-PGA titer of the assembled strain was 31.31% higher than that of the unassembled strain. To further reduce the production cost, 25.73 ± 0.69 g/L γ-PGA with a productivity of 0.48 g/L/h was obtained from cheap molasses. This work provides new metabolic engineering strategies to improve the production of γ-PGA in B. tequilensis BL01. Furthermore, the engineered strain has great potential for the industrial production of γ-PGA from molasses.


Subject(s)
Bacillus , Corynebacterium glutamicum , Polyglutamic Acid/analogs & derivatives , Glutamic Acid/metabolism , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism
13.
World J Microbiol Biotechnol ; 40(5): 154, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38568465

ABSTRACT

D-chiro-inositol (DCI) is a potential drug for the treatment of type II diabetes and polycystic ovary syndrome. In order to effectively synthesize DCI in Corynebacterium glutamicum, the genes related to inositol catabolism in clusters iol1 and iol2 were knocked out in C. glutamicum SN01 to generate the chassis strain DCI-1. DCI-1 did not grow in and catabolize myo-inositol (MI). Subsequently, different exogenous and endogenous inosose isomerases were expressed in DCI-1 and their conversion ability of DCI from MI were compared. After fermentation, the strain DCI-7 co-expressing inosose isomerase IolI2 and inositol dehydrogenase IolG was identified as the optimal strain. Its DCI titer reached 3.21 g/L in the presence of 20 g/L MI. On this basis, the pH, temperature and MI concentration during whole-cell conversion of DCI by strain DCI-7 were optimized. Finally, the optimal condition that achieved the highest DCI titer of 6.96 g/L were obtained at pH 8.0, 37 °C and addition of 40 g/L MI. To our knowledge, it is the highest DCI titer ever reported.


Subject(s)
Corynebacterium glutamicum , Diabetes Mellitus, Type 2 , Inositol/analogs & derivatives , Female , Humans , Corynebacterium glutamicum/genetics , Metabolic Engineering
14.
World J Microbiol Biotechnol ; 40(5): 159, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38607454

ABSTRACT

Gamma-aminobutyric acid (GABA) is a non-protein amino acid which is widely applied in agriculture and pharmaceutical additive industries. GABA is synthesized from glutamate through irreversible α-decarboxylation by glutamate decarboxylase. Recently, microbial synthesis has become an inevitable trend to produce GABA due to its sustainable characteristics. Therefore, reasonable microbial platform design and metabolic engineering strategies for improving production of GABA are arousing a considerable attraction. The strategies concentrate on microbial platform optimization, fermentation process optimization, rational metabolic engineering as key metabolic pathway modification, promoter optimization, site-directed mutagenesis, modular transporter engineering, and dynamic switch systems application. In this review, the microbial producers for GABA were summarized, including lactic acid bacteria, Corynebacterium glutamicum, and Escherichia coli, as well as the efficient strategies for optimizing them to improve the production of GABA.


Subject(s)
Corynebacterium glutamicum , gamma-Aminobutyric Acid , Agriculture , Corynebacterium glutamicum/genetics , Drug Industry , Engineering , Escherichia coli/genetics
15.
Sheng Wu Gong Cheng Xue Bao ; 40(3): 621-643, 2024 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-38545968

ABSTRACT

L-tryptophan is an essential amino acid that is widely used in food, medicine and feed sectors. L-tryptophan can be produced through fermentation, and the main producing strains are engineered Escherichia coli and Corynebacterium glutamicum, which are constructed by rational design methods based on metabolic engineering and synthetic biology. However, due to the long metabolic pathway, complex and unclear regulatory mechanism for L-tryptophan production in microbial cells, the production efficiency and robustness of L-tryptophan producing strains are still low. In this connection, irrational design methods such as laboratory adaptive evolution, are often applied to improve the performance of L-tryptophan producing strains. This review summarizes the recent progress on biosynthesis metabolism of L-tryptophan and its regulation, the construction and optimization of L-tryptophan producing strains, and fermentative production of L-tryptophan, and prospects future development perspective. This review may facilitate research and development for fermentative production of L-tryptophan.


Subject(s)
Corynebacterium glutamicum , Tryptophan , Fermentation , Metabolic Engineering , Metabolic Networks and Pathways , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
16.
J Agric Food Chem ; 72(14): 8039-8051, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38545740

ABSTRACT

d-Amino acids (d-AAs) have wide applications in industries such as pharmaceutical, food, and cosmetics due to their unique properties. Currently, the production of d-AAs has relied on chemical synthesis or enzyme catalysts, and it is challenging to produce d-AAs via direct fermentation from glucose. We observed that Corynebacterium glutamicum exhibits a remarkable tolerance to high concentrations of d-Ala, a crucial characteristic for establishing a successful fermentation process. By optimizing meso-diaminopilmelate dehydrogenases in different C. glutamicum strains and successively deleting l-Ala biosynthetic pathways, we developed an efficient d-Ala fermentation system. The d-Ala titer was enhanced through systems metabolic engineering, which involved strengthening glucose assimilation and pyruvate supply, reducing the formation of organic acid byproducts, and attenuating the TCA cycle. During fermentation in a 5-L bioreactor, a significant accumulation of l-Ala was observed in the broth, which was subsequently diminished by introducing an l-amino acid deaminase. Ultimately, the engineered strain DA-11 produced 85 g/L d-Ala with a yield of 0.30 g/g glucose, accompanied by an optical purity exceeding 99%. The fermentation platform has the potential to be extended for the synthesis of other d-AAs, as demonstrated by the production of d-Val and d-Glu.


Subject(s)
Amino Acids , Corynebacterium glutamicum , Amino Acids/metabolism , Fermentation , Alanine/metabolism , Corynebacterium glutamicum/metabolism , Metabolic Engineering , Glucose/metabolism
17.
Bioprocess Biosyst Eng ; 47(4): 549-556, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499686

ABSTRACT

Heme, found in hemoproteins, is a valuable source of iron, an essential mineral. The need for an alternative hemoprotein source has emerged due to the inherent risks of large-scale livestock farming and animal proteins. Corynebacterium glutamicum, regarded for Qualified Presumption of Safety or Generally Recognized as Safe, can biosynthesize hemoproteins. C. glutamicum single-cell protein (SCP) can be a valuable alternative hemoprotein for supplying heme iron without adversely affecting blood fat levels. We constructed the chemostat culture system to increase hemoprotein content in C. glutamicum SCP. Through adaptive evolution, hemoprotein levels could be naturally increased to address oxidative stress resulting from enhanced growth rate. In addition, we used several specific plasmids containing growth-accelerating genes and the hemA promoter to expedite the evolutionary process. Following chemostat culture for 15 days, the plasmid in selected descendants was cured. The evolved strains showed improved specific growth rates from 0.59 h-1 to 0.62 h-1, 20% enhanced resistance to oxidative stress, and increased heme concentration from 12.95 µg/g-DCW to 14.22-15.24 µg/g-DCW. Notably, the putative peptidyl-tRNA hydrolase-based evolved strain manifested the most significant increase (30%) of hemoproteins. This is the first report presenting the potential of a growth-acceleration-targeted evolution (GATE) strategy for developing non-GMO industrial strains with increased bio-product productivity.


Subject(s)
Corynebacterium glutamicum , Animals , Plasmids , Iron/metabolism , Heme/metabolism , Acceleration , Metabolic Engineering
18.
J Biosci Bioeng ; 137(5): 396-402, 2024 May.
Article in English | MEDLINE | ID: mdl-38433040

ABSTRACT

To improve the cell productivity of Corynebacterium glutamicum, its initial specific growth rate was improved by medium improvement using deep neural network (DNN)-assisted design with Bayesian optimization (BO) and a genetic algorithm (GA). To obtain training data for the DNN, experimental design with an orthogonal array was set up using a chemically defined basal medium (GC XII). Based on the cultivation results for the training data, specific growth rates were observed between 0.04 and 0.3/h. The resulting DNN model estimated the test data with high accuracy (R2test ≥ 0.98). According to the validation cultivation, specific growth rates in the optimal media components estimated by DNN-BO and DNN-GA increased from 0.242 to 0.355/h. Using the optimal media (UCB_3), the specific growth rate, along with other parameters, was evaluated in batch culture. The specific growth rate reached 0.371/h from 3 to 12 h, and the dry cell weight was 28.0 g/L at 22.5 h. From the cultivation, the cell yields against glucose, ammonium ion, phosphate ion, sulfate ion, potassium ion, and magnesium ion were calculated. The cell yield calculation was used to estimate the required amounts of each component, and magnesium was found to limit the cell growth. However, in the follow-up fed-batch cultivation, glucose and magnesium addition was required to achieve the high initial specific growth rate, while appropriate feeding of glucose and magnesium during cultivation resulted in maintaining the high specific growth rate, and obtaining a cell yield of 80 g/Lini.


Subject(s)
Corynebacterium glutamicum , Deep Learning , Corynebacterium glutamicum/genetics , Bayes Theorem , Magnesium , Glucose , Cell Count
19.
Biochemistry ; 63(7): 939-951, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38507812

ABSTRACT

MshA is a GT-B glycosyltransferase catalyzing the first step in the biosynthesis of mycothiol. While many GT-B enzymes undergo an open-to-closed transition, MshA is unique because its 97° rotation is beyond the usual range of 10-25°. Molecular dynamics (MD) simulations were carried out for MshA in both ligand bound and unbound states to investigate the effect of ligand binding on localized protein dynamics and its conformational free energy landscape. Simulations showed that both the unliganded "opened" and liganded "closed" forms of the enzyme sample a wide degree of dihedral angles and interdomain distances with relatively low overlapping populations. Calculation of the free energy surface using replica exchange MD for the apo "opened" and an artificial generated apo "closed" structure revealed overlaps in the geometries sampled, allowing calculation of a barrier of 2 kcal/mol for the open-to-closed transition in the absence of ligands. MD simulations of fully liganded MshA revealed a smaller sampling of the dihedral angles. The localized protein fluctuation changes suggest that UDP-GlcNAc binding activates the motions of loops in the 1-l-myo-inositol-1-phosphate (I1P)-binding site despite little change in the interactions with UDP-GlcNAc. Circular dichroism, intrinsic fluorescence spectroscopy, and mutagenesis studies were used to confirm the ligand-induced structural changes in MshA. The results support a proposed mechanism where UDP-GlcNAc binds with rigid interactions to the C-terminal domain of MshA and activates flexible loops in the N-terminal domain for binding and positioning of I1P. This model can be used for future structure-based drug development of inhibitors of the mycothiol biosynthetic pathway.


Subject(s)
Corynebacterium glutamicum , Cysteine , Glycopeptides , Glycosyltransferases , Inositol , Glycosyltransferases/metabolism , Ligands , Inositol Phosphates/metabolism , Uridine Diphosphate/metabolism , Protein Conformation , Molecular Dynamics Simulation
20.
Microb Cell Fact ; 23(1): 62, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402147

ABSTRACT

BACKGROUND: 1,2-propanediol (1,2-PDO) is widely used in the cosmetic, food, and drug industries with a worldwide consumption of over 1.5 million metric tons per year. Although efforts have been made to engineer microbial hosts such as Corynebacterium glutamicum to produce 1,2-PDO from renewable resources, the performance of such strains is still improvable to be competitive with existing petrochemical production routes. RESULTS: In this study, we enabled 1,2-PDO production in the genome-reduced strain C. glutamicum PC2 by introducing previously described modifications. The resulting strain showed reduced product formation but secreted 50 ± 1 mM D-lactate as byproduct. C. glutamicum PC2 lacks the D-lactate dehydrogenase which pointed to a yet unknown pathway relevant for 1,2-PDO production. Further analysis indicated that in C. glutamicum methylglyoxal, the precursor for 1,2-PDO synthesis, is detoxified with the antioxidant native mycothiol (MSH) by a glyoxalase-like system to lactoylmycothiol and converted to D-lactate which is rerouted into the central carbon metabolism at the level of pyruvate. Metabolomics of cell extracts of the empty vector-carrying wildtype, a 1,2-PDO producer and its derivative with inactive D-lactate dehydrogenase identified major mass peaks characteristic for lactoylmycothiol and its precursors MSH and glucosaminyl-myo-inositol, whereas the respective mass peaks were absent in a production strain with inactivated MSH synthesis. Deletion of mshA, encoding MSH synthase, in the 1,2-PDO producing strain C. glutamicum ΔhdpAΔldh(pEKEx3-mgsA-yqhD-gldA) improved the product yield by 56% to 0.53 ± 0.01 mM1,2-PDO mMglucose-1 which is the highest value for C. glutamicum reported so far. CONCLUSIONS: Genome reduced-strains are a useful basis to unravel metabolic constraints for strain engineering and disclosed in this study the pathway to detoxify methylglyoxal which represents a precursor for 1,2-PDO production. Subsequent inactivation of the competing pathway significantly improved the 1,2-PDO yield.


Subject(s)
Corynebacterium glutamicum , Propylene Glycol , Propylene Glycols , Propylene Glycol/metabolism , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Pyruvaldehyde/metabolism , Lactates/metabolism , Metabolic Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...