Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.328
Filtrar
1.
ACS Synth Biol ; 13(7): 2227-2237, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38975718

RESUMO

The inevitable transition from petrochemical production processes to renewable alternatives has sparked the emergence of biofoundries in recent years. Manual engineering of microbes will not be sufficient to meet the ever-increasing demand for novel producer strains. Here we describe the AutoBioTech platform, a fully automated laboratory system with 14 devices to perform operations for strain construction without human interaction. Using modular workflows, this platform enables automated transformations of Escherichia coli with plasmids assembled via modular cloning. A CRISPR/Cas9 toolbox compatible with existing modular cloning frameworks allows automated and flexible genome editing of E. coli. In addition, novel workflows have been established for the fully automated transformation of the Gram-positive model organism Corynebacterium glutamicum by conjugation and electroporation, with the latter proving to be the more robust technique. Overall, the AutoBioTech platform excels at versatility due to the modularity of workflows and seamless transitions between modules. This will accelerate strain engineering of Gram-negative and Gram-positive bacteria.


Assuntos
Sistemas CRISPR-Cas , Corynebacterium glutamicum , Escherichia coli , Edição de Genes , Plasmídeos , Escherichia coli/genética , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Plasmídeos/genética , Edição de Genes/métodos , Sistemas CRISPR-Cas/genética , Eletroporação/métodos , Engenharia Genética/métodos
2.
Biofabrication ; 16(4)2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-38996414

RESUMO

Riboflavin overproduction byCorynebacterium glutamicumwas achieved by screening synthetic operons, enabling fine-tuned expression of the riboflavin biosynthetic genesribGCAH.The synthetic operons were designed by means of predicted translational initiation rates of each open reading frame, with the best-performing selection enabling riboflavin overproduction without negatively affecting cell growth. Overexpression of the fructose-1,6-bisphosphatase (fbp) and 5-phosphoribosyl 1-pyrophosphate aminotransferase (purF) encoding genes was then done to redirect the metabolic flux towards the riboflavin precursors. The resulting strain produced 8.3 g l-1of riboflavin in glucose-based fed-batch fermentations, which is the highest reported riboflavin titer withC. glutamicum. Further genetic engineering enabled both xylose and mannitol utilization byC. glutamicum, and we demonstrated riboflavin overproduction with the xylose-rich feedstocks rice husk hydrolysate and spent sulfite liquor, and the mannitol-rich feedstock brown seaweed hydrolysate. Remarkably, rice husk hydrolysate provided 30% higher riboflavin yields compared to glucose in the bioreactors.


Assuntos
Corynebacterium glutamicum , Engenharia Metabólica , Riboflavina , Riboflavina/biossíntese , Riboflavina/química , Riboflavina/metabolismo , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Xilose/metabolismo , Fermentação , Glucose/metabolismo , Óperon , Manitol/metabolismo , Manitol/química , Reatores Biológicos , Engenharia Genética
3.
World J Microbiol Biotechnol ; 40(9): 267, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39004689

RESUMO

As an efficient and safe industrial bacterium, Corynebacterium glutamicum has extensive application in amino acid production. However, it often faces oxidative stress induced by reactive oxygen species (ROS), leading to diminished production efficiency. To enhance the robustness of C. glutamicum, numerous studies have focused on elucidating its regulatory mechanisms under various stress conditions such as heat, acid, and sulfur stress. However, a comprehensive review of its defense mechanisms against oxidative stress is needed. This review offers an in-depth overview of the mechanisms C. glutamicum employs to manage oxidative stress. It covers both enzymatic and non-enzymatic systems, including antioxidant enzymes, regulatory protein families, sigma factors involved in transcription, and physiological redox reduction pathways. This review provides insights for advancing research on the antioxidant mechanisms of C. glutamicum and sheds light on its potential applications in industrial production.


Assuntos
Antioxidantes , Proteínas de Bactérias , Corynebacterium glutamicum , Regulação Bacteriana da Expressão Gênica , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio , Fator sigma , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Antioxidantes/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Espécies Reativas de Oxigênio/metabolismo , Fator sigma/metabolismo , Fator sigma/genética
4.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1711-1727, 2024 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-38914487

RESUMO

Amino acids as the building blocks of proteins are widely applied in food, medicine, feed, and chemical industries. Amino acid production by microbial cell factories from renewable resources is praised for the environmental friendliness, mild reaction conditions, and high product purity, which helps to achieve the goal of carbon neutrality. Researchers have employed the methods of metabolic engineering and synthetic biology to engineer Escherichia coli and Corynebacterium glutamicum and optimized the culture conditions to construct the microbial cell factories with high performance for producing branched chain amino acids, amino acids of the aspartic acid and glutamic acid families, and aromatic amino acids. We review the engineering process of microbial cell factories for high production of amino acids, in the hope of providing a reference for the creation of high-performance microbial cell factories.


Assuntos
Aminoácidos , Corynebacterium glutamicum , Escherichia coli , Engenharia Metabólica , Engenharia Metabólica/métodos , Aminoácidos/biossíntese , Aminoácidos/metabolismo , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Biologia Sintética , Microbiologia Industrial
5.
PLoS Genet ; 20(6): e1011127, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38829907

RESUMO

The cell envelope fortifies bacterial cells against antibiotics and other insults. Species in the Mycobacteriales order have a complex envelope that includes an outer layer of mycolic acids called the mycomembrane (MM) and a cell wall composed of peptidoglycan and arabinogalactan. This envelope architecture is unique among bacteria and contributes significantly to the virulence of pathogenic Mycobacteriales like Mycobacterium tuberculosis. Characterization of pathways that govern envelope biogenesis in these organisms is therefore critical in understanding their biology and for identifying new antibiotic targets. To better understand MM biogenesis, we developed a cell sorting-based screen for mutants defective in the surface exposure of a porin normally embedded in the MM of the model organism Corynebacterium glutamicum. The results revealed a requirement for the conserved σD envelope stress response in porin export and identified MarP as the site-1 protease, respectively, that activate the response by cleaving the membrane-embedded anti-sigma factor. A reporter system revealed that the σD pathway responds to defects in mycolic acid and arabinogalactan biosynthesis, suggesting that the stress response has the unusual property of being induced by activating signals that arise from defects in the assembly of two distinct envelope layers. Our results thus provide new insights into how C. glutamicum and related bacteria monitor envelope integrity and suggest a potential role for members of the σD regulon in protein export to the MM.


Assuntos
Membrana Celular , Parede Celular , Corynebacterium glutamicum , Ácidos Micólicos , Fator sigma , Parede Celular/metabolismo , Parede Celular/genética , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Ácidos Micólicos/metabolismo , Fator sigma/metabolismo , Fator sigma/genética , Membrana Celular/metabolismo , Estresse Fisiológico , Porinas/metabolismo , Porinas/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Galactanos/metabolismo , Regulação Bacteriana da Expressão Gênica , Peptidoglicano/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-38944415

RESUMO

Corynebacterium glutamicum ATCC 13032 is a promising microbial chassis for industrial production of valuable compounds, including aromatic amino acids derived from the shikimate pathway. In this work, we developed two whole-cell, transcription factor based fluorescent biosensors to track cis,cis-muconic acid (ccMA) and chorismate in C. glutamicum. Chorismate is a key intermediate in the shikimate pathway from which value-added chemicals can be produced, and a shunt from the shikimate pathway can divert carbon to ccMA, a high value chemical. We transferred a ccMA-inducible transcription factor, CatM, from Acinetobacter baylyi ADP1 into C. glutamicum and screened a promoter library to isolate variants with high sensitivity and dynamic range to ccMA by providing benzoate, which is converted to ccMA intracellularly. The biosensor also detected exogenously supplied ccMA, suggesting the presence of a putative ccMA transporter in C. glutamicum, though the external ccMA concentration threshold to elicit a response was 100-fold higher than the concentration of benzoate required to do so through intracellular ccMA production. We then developed a chorismate biosensor, in which a chorismate inducible promoter regulated by natively expressed QsuR was optimized to exhibit a dose-dependent response to exogenously supplemented quinate (a chorismate precursor). A chorismate-pyruvate lyase encoding gene, ubiC, was introduced into C. glutamicum to lower the intracellular chorismate pool, which resulted in loss of dose dependence to quinate. Further, a knockout strain that blocked the conversion of quinate to chorismate also resulted in absence of dose dependence to quinate, validating that the chorismate biosensor is specific to intracellular chorismate pool. The ccMA and chorismate biosensors were dually inserted into C. glutamicum to simultaneously detect intracellularly produced chorismate and ccMA. Biosensors, such as those developed in this study, can be applied in C. glutamicum for multiplex sensing to expedite pathway design and optimization through metabolic engineering in this promising chassis organism. ONE-SENTENCE SUMMARY: High-throughput screening of promoter libraries in Corynebacterium glutamicum to establish transcription factor based biosensors for key metabolic intermediates in shikimate and ß-ketoadipate pathways.


Assuntos
Técnicas Biossensoriais , Ácido Corísmico , Corynebacterium glutamicum , Ácido Sórbico , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Técnicas Biossensoriais/métodos , Ácido Sórbico/metabolismo , Ácido Sórbico/análogos & derivados , Ácido Corísmico/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regiões Promotoras Genéticas , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Acinetobacter/metabolismo , Acinetobacter/genética
7.
Metab Eng ; 84: 117-127, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38901555

RESUMO

Effective utilization of glucose, xylose, and acetate, common carbon sources in lignocellulose hydrolysate, can boost biomanufacturing economics. However, carbon leaks into biomass biosynthesis pathways instead of the intended target product remain to be optimized. This study aimed to enhance α-carotene production by optimizing glucose, xylose, and acetate utilization in a high-efficiency Corynebacterium glutamicum cell factory. Heterologous xylose pathway expression in C. glutamicum resulted in strain m4, exhibiting a two-fold increase in α-carotene production from xylose compared to glucose. Xylose utilization was found to boost the biosynthesis of pyruvate and acetyl-CoA, essential precursors for carotenoid biosynthesis. Additionally, metabolic engineering including pck, pyc, ppc, and aceE deletion, completely disrupted the metabolic connection between glycolysis and the TCA cycle, further enhancing α-carotene production. This strategic intervention directed glucose and xylose primarily towards target chemical production, while acetate supplied essential metabolites for cell growth recovery. The engineered strain C. glutamicum m8 achieved 30 mg/g α-carotene, 67% higher than strain m4. In fed-batch fermentation, strain m8 produced 1802 mg/L of α-carotene, marking the highest titer reported to date in microbial fermentation. Moreover, it exhibited excellent performance in authentic lignocellulosic hydrolysate, producing 216 mg/L α-carotene, 1.45 times higher than the initial strain (m4). These labor-division strategies significantly contribute to the development of clean processes for producing various valuable chemicals from lignocellulosic resources.


Assuntos
Corynebacterium glutamicum , Engenharia Metabólica , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Glucose/metabolismo , Xilose/metabolismo , Carotenoides/metabolismo , Carbono/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/biossíntese
8.
Molecules ; 29(12)2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38930958

RESUMO

The phosphoenol pyruvate-oxaloacetate-pyruvate-derived amino acids (POP-AAs) comprise native intermediates in cellular metabolism, within which the phosphoenol pyruvate-oxaloacetate-pyruvate (POP) node is the switch point among the major metabolic pathways existing in most living organisms. POP-AAs have widespread applications in the nutrition, food, and pharmaceutical industries. These amino acids have been predominantly produced in Escherichia coli and Corynebacterium glutamicum through microbial fermentation. With the rapid increase in market requirements, along with the global food shortage situation, the industrial production capacity of these two bacteria has encountered two bottlenecks: low product conversion efficiency and high cost of raw materials. Aiming to push forward the update and upgrade of engineered strains with higher yield and productivity, this paper presents a comprehensive summarization of the fundamental strategy of metabolic engineering techniques around phosphoenol pyruvate-oxaloacetate-pyruvate node for POP-AA production, including L-tryptophan, L-tyrosine, L-phenylalanine, L-valine, L-lysine, L-threonine, and L-isoleucine. Novel heterologous routes and regulation methods regarding the carbon flux redistribution in the POP node and the formation of amino acids should be taken into consideration to improve POP-AA production to approach maximum theoretical values. Furthermore, an outlook for future strategies of low-cost feedstock and energy utilization for developing amino acid overproducers is proposed.


Assuntos
Aminoácidos , Engenharia Metabólica , Engenharia Metabólica/métodos , Aminoácidos/metabolismo , Ácido Oxaloacético/metabolismo , Escherichia coli/metabolismo , Escherichia coli/genética , Fosfoenolpiruvato/metabolismo , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Ácido Pirúvico/metabolismo , Redes e Vias Metabólicas , Fermentação
9.
J Agric Food Chem ; 72(23): 13186-13195, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38814711

RESUMO

Ketopantoate hydroxymethyltransferase (KPHMT) plays a pivotal role in d-pantothenic acid biosynthesis. Most KPHMTs are homodecamers with low thermal stability, posing challenges for protein engineering and limiting output enhancement. Previously, a high-enzyme activity KPHMT mutant (K25A/E189S) from Corynebacterium glutamicum was screened as mother strain (M0). Building upon this strain, our study focused on interface engineering modifications, employing a multifaceted approach including integrating folding-free energy calculation, B-factor analysis, and conserved site analysis. Preliminary screening led to the selection of five mutants in the interface─E106S, E98T, E98N, S247I, and S247D─showing improved thermal stability, culminating in the double-site mutant M8 (M0-E98N/S247D). M8 exhibited a T1/2 value of 288.79 min at 50 °C, showing a 3.29-fold increase compared to M0. Meanwhile, the Tm value of M8 was elevated from 53.2 to 59.6 °C. Investigations of structural and molecular dynamics simulations revealed alterations in surface electrostatic charge distribution and the formation of increased hydrogen bonds between subunits, contributing to enhanced thermal stability. This investigation corroborates the efficacy of interface engineering modifications in bolstering KPHMT stability while showing its potential for positively impacting industrial d-pantothenic acid synthesis.


Assuntos
Proteínas de Bactérias , Corynebacterium glutamicum , Estabilidade Enzimática , Engenharia de Proteínas , Corynebacterium glutamicum/enzimologia , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Simulação de Dinâmica Molecular , Cinética , Temperatura Alta
10.
Curr Microbiol ; 81(6): 167, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38727744

RESUMO

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.


Assuntos
Clonagem Molecular , Corynebacterium glutamicum , Escherichia coli , Malato Desidrogenase , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Clonagem Molecular/métodos , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/enzimologia , Diabetes Mellitus/genética , Escherichia coli/genética , Expressão Gênica , Malato Desidrogenase/genética , Malato Desidrogenase/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Temperatura
11.
Microb Cell Fact ; 23(1): 147, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783320

RESUMO

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.


Assuntos
Corynebacterium glutamicum , Engenharia Metabólica , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Engenharia Metabólica/métodos , Pirrolnitrina/biossíntese , Pirrolnitrina/metabolismo , Fermentação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Triptofano/biossíntese , Triptofano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Oxirredutases
12.
Molecules ; 29(10)2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38792114

RESUMO

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.


Assuntos
Corynebacterium glutamicum , Flavonoides , Engenharia Metabólica , Estilbenos , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Engenharia Metabólica/métodos , Flavonoides/biossíntese , Flavonoides/metabolismo , Estilbenos/metabolismo
13.
J Agric Food Chem ; 72(21): 12219-12228, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38747135

RESUMO

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.


Assuntos
Corynebacterium glutamicum , Engenharia Metabólica , Ficobilinas , Ficocianina , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Ficocianina/metabolismo , Ficocianina/genética , Ficobilinas/metabolismo , Ficobilinas/genética , Fermentação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Via de Pentose Fosfato/genética , Reatores Biológicos/microbiologia
14.
PLoS One ; 19(5): e0299588, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38718091

RESUMO

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.


Assuntos
Corynebacterium glutamicum , Variação Genética , Genoma Bacteriano , Filogenia , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Família Multigênica , Genômica/métodos
15.
J Microbiol Biotechnol ; 34(5): 1154-1163, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38563097

RESUMO

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.


Assuntos
Apigenina , Corynebacterium glutamicum , Glucosídeos , Engenharia Metabólica , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/genética , Apigenina/metabolismo , Engenharia Metabólica/métodos , Glucosídeos/metabolismo , Glucosídeos/biossíntese , Glicosilação , Bacillus licheniformis/metabolismo , Bacillus licheniformis/genética , Bacillus licheniformis/enzimologia , Uridina Difosfato Glucose/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , UTP-Glucose-1-Fosfato Uridililtransferase/metabolismo , UTP-Glucose-1-Fosfato Uridililtransferase/genética , Glicosiltransferases/metabolismo , Glicosiltransferases/genética
16.
Curr Opin Microbiol ; 79: 102478, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38653035

RESUMO

Members of the order Mycobacteriales are distinguished by a characteristic diderm cell envelope, setting them apart from other Actinobacteria species. In addition to the conventional peptidoglycan cell wall, these organisms feature an extra polysaccharide polymer composed of arabinose and galactose, termed arabinogalactan. The nonreducing ends of arabinose are covalently linked to mycolic acids (MAs), forming the immobile inner leaflet of the highly hydrophobic MA membrane. The contiguous outer leaflet of the MA membrane comprises trehalose mycolates and various lipid species. Similar to all actinobacteria, Mycobacteriales exhibit apical growth, facilitated by a polar localized elongasome complex. A septal cell envelope synthesis machinery, the divisome, builds instead of the cell wall structures during cytokinesis. In recent years, a growing body of knowledge has emerged regarding the cell wall synthesizing complexes of Mycobacteriales., focusing particularly on three model species: Corynebacterium glutamicum, Mycobacterium smegmatis, and Mycobacterium tuberculosis.


Assuntos
Parede Celular , Galactanos , Ácidos Micólicos , Parede Celular/metabolismo , Ácidos Micólicos/metabolismo , Galactanos/metabolismo , Peptidoglicano/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/genética , Corynebacterium glutamicum/metabolismo , Corynebacterium glutamicum/crescimento & desenvolvimento , Corynebacterium glutamicum/genética , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/crescimento & desenvolvimento , Mycobacterium smegmatis/genética , Arabinose/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética
17.
ACS Synth Biol ; 13(7): 2081-2090, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38607270

RESUMO

Ectoine is a compatible solute that functions as a cell protector from various stresses, protecting cells and stabilizing biomolecules, and is widely used in medicine, cosmetics, and biotechnology. Microbial fermentation has been widely used for the large-scale production of ectoine, and a number of fermentation strategies have been developed to increase the ectoine yield, reduce production costs, and simplify the production process. Here, Corynebacterium glutamicum was engineered for ectoine production by heterologous expression of the ectoine biosynthesis operon ectBAC gene from Halomonas elongata, and a series of genetic modifications were implemented. This included introducing the de3 gene from Escherichia coli BL21 (DE3) to express the T7 promoter, eliminating the lysine transporter protein lysE to limit lysine production, and performing a targeted mutation lysCS301Y on aspartate kinase to alleviate feedback inhibition of lysine. The new engineered strain Ect10 obtained an ectoine titer of 115.87 g/L in an optimized fed-batch fermentation, representing the highest ectoine production level in C. glutamicum and achieving the efficient production of ectoine in a low-salt environment.


Assuntos
Diamino Aminoácidos , Corynebacterium glutamicum , Escherichia coli , Fermentação , Halomonas , Engenharia Metabólica , Diamino Aminoácidos/biossíntese , Diamino Aminoácidos/metabolismo , Diamino Aminoácidos/genética , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Engenharia Metabólica/métodos , Halomonas/genética , Halomonas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Lisina/metabolismo , Lisina/biossíntese , Regiões Promotoras Genéticas , Óperon/genética , Aspartato Quinase/genética , Aspartato Quinase/metabolismo , Sistemas de Transporte de Aminoácidos Básicos
18.
Sci Rep ; 14(1): 8081, 2024 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-38582923

RESUMO

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.


Assuntos
Carotenoides , Corynebacterium glutamicum , Carotenoides/metabolismo , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Xantofilas/metabolismo , beta Caroteno/metabolismo , Engenharia Metabólica/métodos
19.
J Agric Food Chem ; 72(15): 8674-8683, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38569079

RESUMO

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.


Assuntos
Bacillus , Corynebacterium glutamicum , Ácido Poliglutâmico/análogos & derivados , Ácido Glutâmico/metabolismo , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo
20.
World J Microbiol Biotechnol ; 40(5): 154, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38568465

RESUMO

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.


Assuntos
Corynebacterium glutamicum , Diabetes Mellitus Tipo 2 , Inositol/análogos & derivados , Feminino , Humanos , Corynebacterium glutamicum/genética , Engenharia Metabólica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...