1
|
Huang Y, Gao Y, Huang Y, Wang X, Xu M, Xu G, Zhang X, Li H, Shi J, Xu Z, Zhang X. Enhanced l-serine synthesis in Corynebacterium glutamicum by exporter engineering and Bayesian optimization of the medium composition. Synth Syst Biotechnol 2025; 10:835-845. [PMID: 40291977 PMCID: PMC12033900 DOI: 10.1016/j.synbio.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
l-serine is a versatile, high value-added amino acid, widely used in food, medicine and cosmetics. However, the low titer of l-serine has limited its industrial production. In this study, a cell factory without plasmid for efficient production of l-serine was constructed based on transport engineering. Firstly, the effects of l-serine exporter SerE overexpression and deletion on the cell growth and l-serine titer were investigated in Corynebacterium glutamicum (C. glutamicum) A36, overexpression of s erE using a plasmid led to a 15.1% increase in l-serine titer but also caused a 15.1% decrease in cell growth. Subsequently, to increase the export capacity of SerE, we conducted semi-rational design and bioinformatics analysis, combined with alanine mutation and site-specific saturation mutation. The mutant E277K was obtained and exhibited a 53.2% higher export capacity compared to wild-type SerE, resulting in l-serine titer increased by 39.6%. Structural analysis and molecular dynamics simulations were performed to elucidate the mechanism. The results showed that the mutation shortened the hydrogen bond distance between the exporter and l-serine, enhanced complex stability, and reduced the binding energy. Finally, Bayesian optimization was employed to further improve l-serine titer of the mutant strain C-E277K. Under the optimized conditions, 47.77 g/L l-serine was achieved in a 5-L bioreactor, representing the highest reported titer for C. glutamicum to date. This study provides a basis for the transformation of l-serine export pathway and offers a new strategy for increasing l-serine titer.
Collapse
Affiliation(s)
- Yifan Huang
- Laboratory of Pharmaceutical Engineering, School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yujie Gao
- Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yamin Huang
- Laboratory of Pharmaceutical Engineering, School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xiaogang Wang
- Key Laboratory of Advanced Control for Light Industry Processes, Ministry of Education, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Meijuan Xu
- Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Guoqiang Xu
- Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xiaojuan Zhang
- Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Hui Li
- Laboratory of Pharmaceutical Engineering, School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jinsong Shi
- Laboratory of Pharmaceutical Engineering, School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhenghong Xu
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Xiaomei Zhang
- Laboratory of Pharmaceutical Engineering, School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| |
Collapse
|
2
|
Wiechert J, Badia Roigé B, Dohmen-Olma D, Hindra, Zhang X, Stella RG, Elliot MA, Frunzke J. CRISPR/dCas-mediated counter-silencing: reprogramming dCas proteins into antagonists of xenogeneic silencers. mBio 2025:e0038225. [PMID: 40434115 DOI: 10.1128/mbio.00382-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 05/29/2025] Open
Abstract
Lsr2-like nucleoid-associated proteins function as xenogeneic silencers (XSs) inhibiting expression of horizontally acquired, adenine-thymine-rich DNA in actinobacteria. Interference by transcription factors can lead to counter-silencing of XS target promoters, but relief of this repression typically requires promoter engineering. In this study, we developed a novel clustered regularly interspaced short palindromic repeats (CRISPR)/dCas-mediated counter-silencing (CRISPRcosi) approach by using nuclease-deficient dCas enzymes to counteract the Lsr2-like XS protein CgpS in Corynebacterium glutamicum or Lsr2 in Streptomyces venezuelae. Systematic in vivo reporter studies with dCas9 and dCas12a and various guide RNAs revealed effective counter-silencing of different CgpS target promoters in response to guide RNA/dCas DNA binding, independent of promoter sequence modifications. The most prominent CRISPRcosi effect was observed when targeting the CgpS nucleation site, an effect that was also seen in S. venezuelae when targeting a known Lsr2 nucleation site within the chloramphenicol biosynthesis gene cluster. Analyzing the system in C. glutamicum strains lacking the XS protein CgpS revealed varying strengths of counteracting CRISPR interference effects based on the target position and strand. Genome-wide transcriptome profiling in single-guide RNA/dCas9 co-expressing C. glutamicum wild-type strains revealed high counter-silencing specificity with minimal off-target effects. Thus, CRISPRcosi provides a promising strategy for the precise upregulation of XS target genes with significant potential for studying gene networks as well as for developing applications in biotechnology and synthetic biology. IMPORTANCE Lsr2-like nucleoid-associated proteins act as xenogeneic silencers (XSs), repressing the expression of horizontally acquired, adenine-thymine-rich DNA in actinobacteria. The targets of Lsr2-like proteins are very diverse, including prophage elements, virulence gene clusters, and biosynthetic gene clusters. Consequently, the targeted activation of XS target genes is of interest for fundamental research and biotechnological applications. Traditional methods for counter-silencing typically require promoter modifications. In this study, we developed a novel clustered regularly interspaced short palindromic repeats (CRISPR)/dCas-mediated counter-silencing (CRISPRcosi) approach, utilizing nuclease-deficient dCas enzymes to counteract repression by Lsr2-like proteins in Corynebacterium glutamicum and Streptomyces venezuelae. The strongest effect was observed when targeting the Lsr2 nucleation site. Genome-wide transcriptome profiling revealed high specificity with minimal off-target effects. Overall, CRISPRcosi emerges as a powerful tool for the precise induction of genes silenced by xenogeneic silencers, offering new opportunities for exploring gene networks and advancing biotechnological applications.
Collapse
Affiliation(s)
- Johanna Wiechert
- Institute of Bio- and Geoscience-IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, North Rhine-Westphalia, Germany
| | - Biel Badia Roigé
- Institute of Bio- and Geoscience-IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, North Rhine-Westphalia, Germany
| | - Doris Dohmen-Olma
- Institute of Bio- and Geoscience-IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, North Rhine-Westphalia, Germany
| | - Hindra
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Xiafei Zhang
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Roberto G Stella
- Institute of Bio- and Geoscience-IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, North Rhine-Westphalia, Germany
| | - Marie A Elliot
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Julia Frunzke
- Institute of Bio- and Geoscience-IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, North Rhine-Westphalia, Germany
| |
Collapse
|
3
|
Krüger A, Weber U, Frunzke J. Genome-Wide Analysis of DtxR and HrrA Regulons Reveals Novel Targets and a High Level of Interconnectivity Between Iron and Heme Regulatory Networks in Corynebacterium glutamicum. Mol Microbiol 2025. [PMID: 40376914 DOI: 10.1111/mmi.15376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Iron is vital for most organisms, serving as a cofactor in enzymes, regulatory proteins, and respiratory cytochromes. In Corynebacterium glutamicum, iron and heme homeostasis are tightly interconnected and controlled by the global regulators DtxR and HrrA. While DtxR senses intracellular Fe2+, HrrSA is activated by heme. This study provides the first genome-wide analysis of DtxR and HrrA binding dynamics under varying iron and heme conditions using chromatin affinity purification and sequencing (ChAP-Seq). We revealed 25 novel DtxR targets and 210 previously unrecognized HrrA targets. Among these, metH, encoding homocysteine methyltransferase, and xerC, encoding a tyrosine recombinase, were bound by DtxR exclusively under heme conditions, underscoring condition-dependent variation. Activation of metH by DtxR links iron metabolism to methionine synthesis, potentially relevant for the mitigation of oxidative stress. Beyond novel targets, 16 shared targets between DtxR and HrrA, some with overlapping operator sequences, highlight their interconnected regulons. Strikingly, we demonstrate the significance of weak ChAP-Seq peaks that are often disregarded in global approaches, but feature an impact of the regulator on differential gene expression. These findings emphasize the importance of genome-wide profiling under different conditions to uncover novel targets and shed light on the complexity and dynamic nature of bacterial regulatory networks.
Collapse
Affiliation(s)
- Aileen Krüger
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, Jülich, Germany
| | - Ulrike Weber
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, Jülich, Germany
| | - Julia Frunzke
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, Jülich, Germany
| |
Collapse
|
4
|
Lv H, Li X, Peng Q, Niu X, Meng C, Niu L, Zhang S, Li P, Jiao H, Wang Z, Zhou Z. SodC is responsible for oxidative stress resistance and pathogenicity of Corynebacterium pseudotuberculosis, and the sodC-deleted C. pseudotuberculosis vaccine provides immunity in mice. Vet Microbiol 2025; 304:110484. [PMID: 40120522 DOI: 10.1016/j.vetmic.2025.110484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Corynebacterium pseudotuberculosis causes chronic inflammatory infectious diseases in animals and humans. Resistance to adverse environments, including oxidative stress, is required for the survival and pathogenicity of C. pseudotuberculosis. Superoxide dismutase (SOD) is a key enzyme to resist oxidative stress. However, the role of SODs in C. pseudotuberculosis has not been reported. In this study, we addressed this question using C. pseudotuberculosis XH02, sodA deleted (XH02ΔsodA), and sodC deleted (XH02ΔsodC) strains. We found that sodA or sodC deletion reduced the pathogenicity of C. pseudotuberculosis in mice, decreased bacterial loads and histopathological lesions in the infected organs. In addition, the deletion of sodC in C. pseudotuberculosis significantly decreased IL-1β secretion, lactate dehydrogenase (LDH) release, and propidium iodide (PI) uptake of the infected J774A.1 macrophages. Furthermore, sodC deletion weakened the biofilm formation ability of C. pseudotuberculosis, reduced the survival of C. pseudotuberculosis within macrophages, and decreased the ability of C. pseudotuberculosis to resist oxidative stress. We observed that mutations at H94E, H96E, H111A, and H166E reduced the enzyme activity of SodC and reduced the resistance to oxidative stress. Finally, XH02ΔsodC immunization in mice increased specific IgG level and CD4+/CD8+ T cells ratio, and protected mice against C. pseudotuberculosis challenge. Thus, this study confirmed that SodC is an important virulence-related factor of C. pseudotuberculosis, and plays crucial roles in oxidative stress resistance. XH02ΔsodC can be used as a potential candidate attenuated vaccine to prevent and control C. pseudotuberculosis infection.
Collapse
Affiliation(s)
- Hong Lv
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Xincan Li
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Qiuyue Peng
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Xiaoxin Niu
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Chi Meng
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Luting Niu
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Sixin Zhang
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Pei Li
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Hanwei Jiao
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Zhiying Wang
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| | - Zuoyong Zhou
- College of Veterinary Medicine, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing 402460, China.
| |
Collapse
|
5
|
Sohn YJ, Hwang S, Lee H, Jeon S, Park JY, Kim J, Kim D, Jeong KJ, Lee SY, Joo JC, Park J, Park SJ. Metabolic Engineering of Corynebacterium glutamicum for High-Level Production of 1,5-Pentanediol, a C5 Diol Platform Chemical. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412670. [PMID: 39731342 PMCID: PMC11967857 DOI: 10.1002/advs.202412670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/06/2024] [Indexed: 12/29/2024]
Abstract
The biobased production of chemicals is essential for advancing a sustainable chemical industry. 1,5-Pentanediol (1,5-PDO), a five-carbon diol with considerable industrial relevance, has shown limited microbial production efficiency until now. This study presents the development and optimization of a microbial system to produce 1,5-PDO from glucose in Corynebacterium glutamicum via the l-lysine-derived pathway. Engineering began with creating a strain capable of producing 5-hydroxyvaleric acid (5-HV), a key precursor to 1,5-PDO, by incorporating enzymes from Pseudomonas putida (DavB, DavA, and DavT) and Escherichia coli (YahK). Two conversion pathways for further converting 5-HV to 1,5-PDO are evaluated, with the CoA-independent pathway-utilizing Mycobacterium marinum carboxylic acid reductase (CAR) and E. coli YqhD-proving greater efficiency. Further optimization continues with chromosomal integration of the 5-HV module, increasing 1,5-PDO production to 5.48 g L-1. An additional screening of 13 CARs identifies Mycobacterium avium K-10 (MAP1040) as the most effective, and its engineered M296E mutant further increases production to 23.5 g L-1. A deep-learning analysis reveals that Gluconobacter oxydans GOX1801 resolves the limitations of NADPH, allowing the final strain to produce 43.4 g L-1 1,5-PDO without 5-HV accumulation in fed-batch fermentation. This study demonstrates systematic approaches to optimizing microbial biosynthesis, positioning C. glutamicum as a promising platform for sustainable 1,5-PDO production.
Collapse
Affiliation(s)
- Yu Jung Sohn
- Department of Chemical Engineering and Materials ScienceGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoul03760Republic of Korea
| | - Se‐Yeun Hwang
- Department of Food Science and BiotechnologyEwha Womans UniversitySeoul03760Republic of Korea
| | - Haeyoung Lee
- Department of Chemical Engineering and Materials ScienceGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoul03760Republic of Korea
| | - Subeen Jeon
- Department of Chemical Engineering and Materials ScienceGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoul03760Republic of Korea
| | - Ji Young Park
- Department of Chemical Engineering and Materials ScienceGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoul03760Republic of Korea
| | - Jaehyung Kim
- School of Energy and Chemical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Donghyuk Kim
- School of Energy and Chemical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Ki Jun Jeong
- Department of Chemical and Biomolecular EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Sang Yup Lee
- Metabolic and Biomolecular Engineering National Research LaboratorySystems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryDepartment of Chemical and Biomolecular Engineering (BK21 four)Korea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- BioProcess Engineering Research CenterCenter for Synthetic BiologyKAIST Institute for the BioCenturyKAIST Institute for Artificial IntelligenceKAISTDaejeon34141Republic of Korea
| | - Jeong Chan Joo
- Department of Chemical EngineeringKyung Hee UniversityYongin17104Republic of Korea
| | - Jin‐Byung Park
- Department of Food Science and BiotechnologyEwha Womans UniversitySeoul03760Republic of Korea
| | - Si Jae Park
- Department of Chemical Engineering and Materials ScienceGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoul03760Republic of Korea
| |
Collapse
|
6
|
Meyer F, Schmitt I, Wendisch VF, Henke NA. Response surface-based media optimization for astaxanthin production in Corynebacterium glutamicum. Front Bioeng Biotechnol 2025; 13:1516522. [PMID: 40134774 PMCID: PMC11933003 DOI: 10.3389/fbioe.2025.1516522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Introduction Astaxanthin is a C40 carotenoid that is used in animal feeds or cosmetics. Due to its high antioxidant property it is used for, e.g., anti-aging formulations and due to its intense red color it is used, e.g., in animal feed. While about 95% of commercial astaxanthin is currently chemically synthesized from fossil sources, the interest in natural and sustainable astaxanthin is growing. Corynebacterium glutamicum, an attractive host used in large-scale processes, e.g., industrial amino acid production, has been engineered for astaxanthin production. Methods Here, a design of experiment (DoE) approach was applied to optimize the standard minimal medium for astaxanthin production. The concentrations of carbon, nitrogen and phosphorus sources, magnesium, calcium, the iron chelator protocatechuic acid, the vitamin biotin, and the trace metals were varied and astaxanthin production was evaluated. Results and discussion By increasing the concentration of iron and decreasing that of manganese especially, it was possible to increase astaxanthin titers from 7.9 mg L-1-39.6 mg L-1 in a micro cultivation system and from 62 mg L-1-176 mg L-1 in a fed-batch fermentation.
Collapse
|
7
|
Ryabchenko LE, Titov II, Leonova TE, Kalinina TI, Gerasimova TV, Sheremetieva ME, Kolchanov NA, Khlebodarova TM, Yanenko AS. Mutational Analysis Supports Three-Hairpin Model of Attenuator for Transcription Regulation of ilvBNC Operon in Corynebacterium glutamicum. Microorganisms 2025; 13:291. [PMID: 40005659 PMCID: PMC11857589 DOI: 10.3390/microorganisms13020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
The ilvBNC operon in Corynebacterium glutamicum encodes key enzymes for the biosynthesis of branched-chain amino acids (L-isoleucine, L-leucine, and L-valine). This operon has been studied for quite a long time, and it is assumed that three hairpin mRNA structures can be formed in its regulatory region; however, their functionality and role in the attenuation mechanism of the ilvBNC operon are not completely clear. In the present work, we performed a mutational analysis of mRNA secondary structures in the regulatory region of the C. glutamicum ilvBNC operon, which allowed us to propose a model of the regulation of its transcription involving three mRNA hairpins that essentially act as a transcription terminator, an antiterminator, and an antiantiterminator. In this work, we proved the existence of a transcription terminator in this operon and experimentally confirmed the effectiveness of its influence on the expression of the ilvBNC operon, AHAS enzyme activity, and valine production. We demonstrated the unique functional features of this attenuator, which, due to the overlapping of the terminator and antiterminator hairpins, is capable of rapid low-energy transitions between them without the complete disruption of the hairpin structures.
Collapse
Affiliation(s)
- Ludmila E. Ryabchenko
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| | - Igor I. Titov
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (I.I.T.); (T.M.K.)
- Kurchatov Genomic Center, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia
| | - Tatyana E. Leonova
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| | - Tatyana I. Kalinina
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| | - Tatyana V. Gerasimova
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| | - Marina E. Sheremetieva
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| | - Nikolay A. Kolchanov
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (I.I.T.); (T.M.K.)
- Kurchatov Genomic Center, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia
| | - Tamara M. Khlebodarova
- Department of Systems Biology, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (I.I.T.); (T.M.K.)
- Kurchatov Genomic Center, Institute of Cytology and Genetics SB RAS, Akademika Lavrentyev Ave., 10, 630090 Novosibirsk, Russia
| | - Alexander S. Yanenko
- National Research Center “Kurchatov Institute”, Kurchatov Genomic Center, Akademika Kurchatova pl. 1, 123182 Moscow, Russia (A.S.Y.)
| |
Collapse
|
8
|
Saxena H, Patel R, Kelly J, Wakarchuk W. Differential substrate preferences IN ACTINOBACTERIAL protein O-MANNOSYLTRANSFERASES and alteration of protein-O-MANNOSYLATION by choice of secretion pathway. Glycobiology 2025; 35:cwae095. [PMID: 39673494 PMCID: PMC11727336 DOI: 10.1093/glycob/cwae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 12/16/2024] Open
Abstract
Protein-O-mannosylation (POM) is a form of O-glycosylation that is ubiquitous and has been studied extensively throughout in fungi and animals. The key glycosyltransferase, protein O-mannosyltransferase (PMT), a member of family GT-39, is also found in over 3,800 bacterial genomes but has only been minimally examined from prokaryotes. In prokaryotes POM has only been investigated in terms of pathogenicity (in Mycobacterium tuberculosis) even though there are far more non-pathogenic bacteria that appear to carry out POM. To date, there is no consensus on what benefit POM imparts to the non-pathogenic bacteria that can perform it. Through the generation of a POM deficient mutant of Corynebacterium glutamicum - a widely utilized and known protein O-mannosylating actinobacteria - this work shows that even closely related actinobacterial GT-39 s (the enzymes responsible for the initiation of POM) can have different substrate specificities for targets of POM. Moreover, presented here is evidence that POM does not only occur in a SEC-dependent manner; POM also occurs with TAT and non-SEC secreted substrates in a specific and likely tightly regulated manner. Together these results highlight the need for further biochemical characterization of POM in these and other bacterial species to help elucidate the true nature of its biological functions.
Collapse
Affiliation(s)
- Hirak Saxena
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3
| | - Rucha Patel
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3
| | - John Kelly
- Human Health Therapeutics, National Research Council of Canada, 100 Sussex Dr, Ottawa, ON K1N 1J1
| | - Warren Wakarchuk
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3
| |
Collapse
|
9
|
Blumenstein J, Dostálová H, Rucká L, Štěpánek V, Busche T, Kalinowski J, Pátek M, Barvík I. Promoter recognition specificity of Corynebacterium glutamicum stress response sigma factors σ D and σ H deciphered using computer modeling and point mutagenesis. J Comput Aided Mol Des 2024; 39:1. [PMID: 39585436 PMCID: PMC11588781 DOI: 10.1007/s10822-024-00577-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024]
Abstract
This study aimed to reveal interactions of the stress response sigma subunits (factors) σD and σH of RNA polymerase and promoters in Gram-positive bacterium Corynebacterium glutamicum by combining wet-lab obtained data and in silico modeling. Computer modeling-guided point mutagenesis of C. glutamicum σH subunit led to the creation of a panel of σH variants. Their ability to initiate transcription from naturally occurring hybrid σD/σH-dependent promoter Pcg0441 and two control canonical promoters (σD-dependent PrsdA and σH-dependent PuvrD3) was measured and interpreted using molecular dynamics simulations of homology models of all complexes. The results led us to design the artificial hybrid promoter PD35H10 combining the -10 element of the PuvrD3 promoter and the -35 element of the PrsdA promoter. This artificial hybrid promoter PD35-rsdAH10-uvrD3 showed almost optimal properties needed for the bio-orthogonal transcription (not interfering with the native biological processes).
Collapse
Affiliation(s)
- J Blumenstein
- Institute of Microbiology, CAS, v.v.i, Prague, Czech Republic
| | - H Dostálová
- Institute of Microbiology, CAS, v.v.i, Prague, Czech Republic
| | - L Rucká
- Institute of Microbiology, CAS, v.v.i, Prague, Czech Republic
| | - V Štěpánek
- Institute of Microbiology, CAS, v.v.i, Prague, Czech Republic
| | - T Busche
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - J Kalinowski
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - M Pátek
- Institute of Microbiology, CAS, v.v.i, Prague, Czech Republic
| | - I Barvík
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic.
| |
Collapse
|
10
|
Krüger A, Göddecke J, Osthege M, Navratil L, Weber U, Oldiges M, Frunzke J. Biosensor-based growth-coupling as an evolutionary strategy to improve heme export in Corynebacterium glutamicum. Microb Cell Fact 2024; 23:276. [PMID: 39402655 PMCID: PMC11472508 DOI: 10.1186/s12934-024-02556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
The iron-containing porphyrin heme is of high interest for the food industry for the production of artificial meat as well as for medical applications. Recently, the biotechnological platform strain Corynebacterium glutamicum has emerged as a promising host for animal-free heme production. Beyond engineering of complex heme biosynthetic pathways, improving heme export offers significant yet untapped potential for enhancing production strains. In this study, a growth-coupled biosensor was designed to impose a selection pressure on the increased expression of the hrtBA operon encoding an ABC-type heme exporter in C. glutamicum. For this purpose, the promoter region of the growth-regulating genes pfkA (phosphofructokinase) and aceE (pyruvate dehydrogenase) was replaced with that of PhrtB, creating biosensor strains with a selection pressure for hrtBA activation. Resulting sensor strains were used for plate-based selections and for a repetitive batch f(luorescent)ALE using a fully automated laboratory platform. Genome sequencing of isolated clones featuring increased hrtBA expression revealed three distinct mutational hotspots: (i) chrS, (ii) chrA, and (iii) cydD. Mutations in the genes of the ChrSA two-component system, which regulates hrtBA in response to heme levels, were identified as a promising target to enhance export activity. Furthermore, causal mutations within cydD, encoding an ABC-transporter essential for cytochrome bd oxidase assembly, were confirmed by the construction of a deletion mutant. Reversely engineered strains showed strongly increased hrtBA expression as well as increased cellular heme levels. These results further support the proposed role of CydDC as a heme transporter in bacteria. Mutations identified in this study therefore underline the potential of biosensor-based growth coupling and provide promising engineering targets to improve microbial heme production.
Collapse
Affiliation(s)
- Aileen Krüger
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Janik Göddecke
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Michael Osthege
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Luis Navratil
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Ulrike Weber
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Marco Oldiges
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany
| | - Julia Frunzke
- Institute for Bio- and Geosciences 1, Forschungszentrum Jülich GmbH, IBG-1, 52425, Jülich, Germany.
| |
Collapse
|
11
|
Rosch T, Tenhaef J, Stoltmann T, Redeker T, Kösters D, Hollmann N, Krumbach K, Wiechert W, Bott M, Matamouros S, Marienhagen J, Noack S. AutoBioTech─A Versatile Biofoundry for Automated Strain Engineering. ACS Synth Biol 2024; 13:2227-2237. [PMID: 38975718 PMCID: PMC11264319 DOI: 10.1021/acssynbio.4c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
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.
Collapse
Affiliation(s)
- Tobias
Michael Rosch
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Julia Tenhaef
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Tim Stoltmann
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Till Redeker
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Dominic Kösters
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
- Institute
of Biotechnology, RWTH Aachen University, Worringer Weg 3, D-52074 Aachen, Germany
| | - Niels Hollmann
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
- Institute
of Biotechnology, RWTH Aachen University, Worringer Weg 3, D-52074 Aachen, Germany
| | - Karin Krumbach
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Wolfgang Wiechert
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Michael Bott
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
- The
Bioeconomy Science Center (BioSC), Forschungszentrum
Jülich, D-52425 Jülich, Germany
| | - Susana Matamouros
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Jan Marienhagen
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
- Institute
of Biotechnology, RWTH Aachen University, Worringer Weg 3, D-52074 Aachen, Germany
| | - Stephan Noack
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, D-52425 Jülich, Germany
| |
Collapse
|
12
|
Junker N, Sariyar Akbulut B, Wendisch VF. Utilization of orange peel waste for sustainable amino acid production by Corynebacterium glutamicum. Front Bioeng Biotechnol 2024; 12:1419444. [PMID: 39050686 PMCID: PMC11266056 DOI: 10.3389/fbioe.2024.1419444] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Oranges are the most processed fruit in the world-it is therefore apparent that the industrial production of orange juice generates large quantities of orange peel as a by-product. Unfortunately, the management of the orange peel waste leads to economic and environmental problems. Meanwhile, the use of sustainable raw materials for the production of bulk chemicals, such as amino acids, is becoming increasingly attractive. To address both issues, this study focused on the use of orange peel waste as a raw material for media preparation for the production of amino acids by engineered Corynebacterium glutamicum. C. glutamicum grew on pure orange peel hydrolysate (OPH) and growth was enhanced by the addition of a nitrogen source and a pH buffer. Inhibitory effects by the combination of high concentrations of OPH, (NH4)2SO4, and MOPS buffer in the wild-type strain (WT), were overcome in the tyrosine-producing engineered C. glutamicum strain AROM3. Genetic modifications that we identified to allow for improved growth rates under these conditions included the deletions of the vanillin dehydrogenase gene vdh, the ʟ-lactate dehydrogenase gene ldhA and the 19 genes comprising cluster cg2663-cg2686. A growth inhibiting compound present in high concentrations in the OPH is 5-(hydroxymethyl)furfural (HMF). We identified vdh as being primarily responsible for the oxidation of HMF to its acid 5-hydroxymethyl-2-furancarboxylic acid (HMFCA), as the formation of HMFCA was reduced by 97% upon deletion of vdh in C. glutamicum WT. In addition, we showed that growth limitations could be overcome by adjusting the media preparation, using a combination of cheap ammonia water and KOH for pH neutralization after acidic hydrolysis. Overall, we developed a sustainable medium based on orange peel waste for the cultivation of C. glutamicum and demonstrated the successful production of the exemplary amino acids ʟ-arginine, ʟ-lysine, ʟ-serine, ʟ-valine and ʟ-tyrosine.
Collapse
Affiliation(s)
- Nora Junker
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | | | - Volker F. Wendisch
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| |
Collapse
|
13
|
Oliveira-Filho ER, Rodionov DA, Hanson AD. Comparative Genomic and Genetic Evidence on a Role for the OarX Protein in Thiamin Salvage. ACS OMEGA 2024; 9:28888-28894. [PMID: 38973919 PMCID: PMC11223231 DOI: 10.1021/acsomega.4c03514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
Salvage pathways for thiamin and its thiazole and pyrimidine moieties are poorly characterized compared to synthesis pathways. A candidate salvage gene is oarX, which encodes a short-chain dehydrogenase/reductase. In diverse bacteria, oarX clusters on the chromosome with genes of thiamin synthesis, salvage, or transport and is preceded by a thiamin pyrophosphate riboswitch. Thiamin and its moieties can undergo oxidations that convert a side-chain hydroxymethyl group to a carboxyl group, or the thiazole ring to a thiazolone, causing a loss of biological activity. To test if OarX participates in salvage of the carboxyl or thiazolone products, we used a genetic approach in Corynebacterium glutamicum ATCC 14067, which is auxotrophic for thiamin's pyrimidine moiety. This strain could not utilize the pyrimidine carboxyl derivative. This excluded a role in salvaging this product and narrowed the function search to metabolism of the carboxyl or thiazolone derivatives of thiamin or its thiazole moiety. However, a ΔthiG (thiazole auxotroph) strain was not rescued by any of these derivatives. Nor did deleting oarX affect rescue by the physiological pyrimidine and thiazole precursors of thiamin. These findings reinforce the genomic evidence that OarX has a function in thiamin metabolism and rule out five logical possibilities for what this function is.
Collapse
Affiliation(s)
- Edmar R. Oliveira-Filho
- Horticultural
Sciences Department, University of Florida, Gainesville, Florida 32611, United States
| | - Dmitry A. Rodionov
- Infectious
and Inflammatory Diseases Center, Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Andrew D. Hanson
- Horticultural
Sciences Department, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
14
|
Hart EM, Lyerly E, Bernhardt TG. The conserved σD envelope stress response monitors multiple aspects of envelope integrity in corynebacteria. PLoS Genet 2024; 20:e1011127. [PMID: 38829907 PMCID: PMC11175481 DOI: 10.1371/journal.pgen.1011127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/13/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Elizabeth M. Hart
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Evan Lyerly
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thomas G. Bernhardt
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
15
|
Kim EYS, Maltempi de Souza E, Müller-Santos M. Optimisation of DNA electroporation protocols for different plant-associated bacteria. J Microbiol Methods 2024; 220:106912. [PMID: 38452904 DOI: 10.1016/j.mimet.2024.106912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Electroporation is a vital process that facilitates the use of modern recombineering and other high-throughput techniques in a wide array of microorganisms, including non-model bacteria like plant growth-promoting bacteria (PGPB). These microorganisms play a significant role in plant health by colonizing plants and promoting growth through nutrient exchange and hormonal regulation. In this study, we introduce a sequential Design of Experiments (DOE) approach to obtain highly competent cells swiftly and reliably for electroporation. Our method focuses on optimizing the three stages of the electroporation procedure-preparing competent cells, applying the electric pulse field, and recovering transformed cells-separately. We utilized a split-plot fractional design with five factors and a covariate to optimize the first step, response surface methodology (RSM) for the second step, and Plackett-Burman design for two categorical factors and one continuous factor for the final step. Following the experimental sequence with three bacterial models, we achieved efficiencies 10 to 100 times higher, reaching orders of 105 to 106 CFU/μg of circular plasmid DNA. These results highlight the significant potential for enhancing electroporation protocols for non-model bacteria.
Collapse
Affiliation(s)
- Edson Yu Sin Kim
- Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Emanuel Maltempi de Souza
- Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Marcelo Müller-Santos
- Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil.
| |
Collapse
|
16
|
Göttl VL, Meyer F, Schmitt I, Persicke M, Peters-Wendisch P, Wendisch VF, Henke NA. Enhancing astaxanthin biosynthesis and pathway expansion towards glycosylated C40 carotenoids by Corynebacterium glutamicum. Sci Rep 2024; 14:8081. [PMID: 38582923 PMCID: PMC10998873 DOI: 10.1038/s41598-024-58700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/31/2024] [Indexed: 04/08/2024] Open
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 β-carotene production with strain BETA6 (18 mg g-1 CDW). Further advancements in astaxanthin production were made by fine-tuning the expression of the β-carotene hydroxylase gene crtZ and β-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-β-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.
Collapse
Affiliation(s)
- Vanessa L Göttl
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
| | - Florian Meyer
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
| | - Ina Schmitt
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
| | - Marcus Persicke
- CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
- Omics Core Facility - Proteom-Metabolom Unit (In Development), Bielefeld University, 33615, Bielefeld, Germany
| | - Petra Peters-Wendisch
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
| | - Volker F Wendisch
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany
| | - Nadja A Henke
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615, Bielefeld, Germany.
- CZS Junior Research Group, Microsystems in Bioprocess Engineering, Institute of Process Engineering in Life Sciences, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany.
| |
Collapse
|
17
|
Siebert D, Glawischnig E, Wirth MT, Vannahme M, Salazar-Quirós Á, Weiske A, Saydam E, Möggenried D, Wendisch VF, Blombach B. A genome-reduced Corynebacterium glutamicum derivative discloses a hidden pathway relevant for 1,2-propanediol production. Microb Cell Fact 2024; 23:62. [PMID: 38402147 PMCID: PMC10893638 DOI: 10.1186/s12934-024-02337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/16/2024] [Indexed: 02/26/2024] Open
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.
Collapse
Affiliation(s)
- Daniel Siebert
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany
- Chair of Genetics of Prokaryotes, Faculty of Biology & CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Erich Glawischnig
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany
| | - Marie-Theres Wirth
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Mieke Vannahme
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Álvaro Salazar-Quirós
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Annette Weiske
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Ezgi Saydam
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Dominik Möggenried
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Volker F Wendisch
- Chair of Genetics of Prokaryotes, Faculty of Biology & CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Bastian Blombach
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany.
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany.
| |
Collapse
|
18
|
Perera PGT, Linklater DP, Vilagosh Z, Nguyen THP, Hanssen E, Rubanov S, Wanjara S, Aadum B, Alfred R, Dekiwadia C, Juodkazis S, Croft R, Ivanova EP. Genetic Transformation of Plasmid DNA into Escherichia coli Using High Frequency Electromagnetic Energy. NANO LETTERS 2024; 24:1145-1152. [PMID: 38194429 DOI: 10.1021/acs.nanolett.3c03464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
We present a novel technique of genetic transformation of bacterial cells mediated by high frequency electromagnetic energy (HF EME). Plasmid DNA, pGLO (5.4 kb), was successfully transformed into Escherichia coli JM109 cells after exposure to 18 GHz irradiation at a power density between 5.6 and 30 kW m-2 for 180 s at temperatures ranging from 30 to 40 °C. Transformed bacteria were identified by the expression of green fluorescent protein (GFP) using confocal scanning microscopy (CLSM) and flow cytometry (FC). Approximately 90.7% of HF EME treated viable E. coli cells exhibited uptake of the pGLO plasmid. The interaction of plasmid DNA with bacteria leading to transformation was confirmed by using cryogenic transmission electron microscopy (cryo-TEM). HF EME-induced plasmid DNA transformation was shown to be unique, highly efficient, and cost-effective. HF EME-induced genetic transformation is performed under physiologically friendly conditions in contrast to existing techniques that generate higher temperatures, leading to altered cellular integrity. This technique allows safe delivery of genetic material into bacterial cells, thus providing excellent prospects for applications in microbiome therapeutics and synthetic biology.
Collapse
Affiliation(s)
- Palalle G Tharushi Perera
- STEM College, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Denver P Linklater
- STEM College, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
- Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Zoltan Vilagosh
- STEM College, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - The Hong Phong Nguyen
- STEM College, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Eric Hanssen
- Ian Holmes Imaging Centre, Bio21 institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Sergey Rubanov
- Ian Holmes Imaging Centre, Bio21 institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Steve Wanjara
- WaveCyte Biotechnologies, 9900 13th Ave N, Plymouth, Minnesota 55441, United States
| | - Bari Aadum
- WaveCyte Biotechnologies, 9900 13th Ave N, Plymouth, Minnesota 55441, United States
| | - Rebecca Alfred
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Chaitali Dekiwadia
- RMIT Microscopy and Microanalysis Facility, College of Science, Engineering and Health, RMIT University, P.O. Box 2476, Melbourne, VIC 3001, Australia
| | - Saulius Juodkazis
- Centre for Quantum and Optical Sciences, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Rodney Croft
- School of Psychology, Illawara Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Elena P Ivanova
- STEM College, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
19
|
Tripathi S, Voogdt CGP, Bassler SO, Anderson M, Huang PH, Sakenova N, Capraz T, Jain S, Koumoutsi A, Bravo AM, Trotter V, Zimmerman M, Sonnenburg JL, Buie C, Typas A, Deutschbauer AM, Shiver AL, Huang KC. Randomly barcoded transposon mutant libraries for gut commensals I: Strategies for efficient library construction. Cell Rep 2024; 43:113517. [PMID: 38142397 DOI: 10.1016/j.celrep.2023.113517] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023] Open
Abstract
Randomly barcoded transposon mutant libraries are powerful tools for studying gene function and organization, assessing gene essentiality and pathways, discovering potential therapeutic targets, and understanding the physiology of gut bacteria and their interactions with the host. However, construction of high-quality libraries with uniform representation can be challenging. In this review, we survey various strategies for barcoded library construction, including transposition systems, methods of transposon delivery, optimal library size, and transconjugant selection schemes. We discuss the advantages and limitations of each approach, as well as factors to consider when selecting a strategy. In addition, we highlight experimental and computational advances in arraying condensed libraries from mutant pools. We focus on examples of successful library construction in gut bacteria and their application to gene function studies and drug discovery. Given the need for understanding gene function and organization in gut bacteria, we provide a comprehensive guide for researchers to construct randomly barcoded transposon mutant libraries.
Collapse
Affiliation(s)
- Surya Tripathi
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Carlos Geert Pieter Voogdt
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; Structural and Computational Biology Unit, EMBL Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Stefan Oliver Bassler
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Grabengasse 1, 69117 Heidelberg, Germany
| | - Mary Anderson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nazgul Sakenova
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Tümay Capraz
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sunit Jain
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Alexandra Koumoutsi
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Afonso Martins Bravo
- Department of Fundamental Microbiology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Valentine Trotter
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Michael Zimmerman
- Structural and Computational Biology Unit, EMBL Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Justin L Sonnenburg
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cullen Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Athanasios Typas
- Genome Biology Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; Structural and Computational Biology Unit, EMBL Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Adam M Deutschbauer
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Anthony L Shiver
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Kerwyn Casey Huang
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Kataoka N, Matsushita K, Yakushi T. Development of a 2-hydroxyglutarate production system by Corynebacterium glutamicum. Appl Microbiol Biotechnol 2023; 107:5987-5997. [PMID: 37555949 DOI: 10.1007/s00253-023-12716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023]
Abstract
2-Oxoglutarate (2-OG) is a tricarboxylate cycle intermediate that can be biologically converted into several industrially important compounds. However, studies on the fermentative production of compounds synthesized from 2-OG, but not via glutamate (defined as 2-OG derivatives), have been limited. Herein, a system that can efficiently produce 2-hydroxyglutarate (2-HG), a 2-OG derivative biosynthesized by the hgdH-encoded NADH-dependent 2-HG dehydrogenase of Acidaminococcus fermentans, was developed as a model using Corynebacterium glutamicum. First, the D3 strain, which lacked the two NADH-consuming enzymes, lactate dehydrogenase and malate dehydrogenase, as well as isocitrate lyase, was constructed as a starting strain. Next, the growth conditions that induced the accumulation of 2-OG were investigated, and it was found that the biotin- and nitrogen-limited (B/N-limited) aerobic growth conditions were suitable for this purpose. Finally, the hgdH gene of A. fermentans became overexpressed in the D3 strain by inserting it into the intergenic regions with the strong constitutive promoter of the tuf gene of C. glutamicum; the engineered strain was cultured under the B/N-limited aerobic growth conditions. The engineered strain produced 80.1 mM 2-HG with a yield of 0.390 mol/mol glucose, which are the highest titer and yield reported thus far, to the best of our knowledge. Furthermore, reverse genetics showed that the produced 2-HG was partially exported via the YggB protein (NCgl1221 protein, a mechanosensitive channel) known as an exporter for glutamate under the conditions used herein. KEY POINTS: • An efficient 2-HG production system was developed with Corynebacterium glutamicum. • Biotin- and nitrogen-limited aerobic growth conditions induced 2-OG production. • Produced 2-HG was partially excreted via the glutamate exporter, YggB.
Collapse
Affiliation(s)
- Naoya Kataoka
- Department of Biological Chemistry, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan.
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan.
| | - Kazunobu Matsushita
- Department of Biological Chemistry, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| | - Toshiharu Yakushi
- Department of Biological Chemistry, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
21
|
Sundermeyer L, Folkerts JG, Lückel B, Mack C, Baumgart M, Bott M. Cellular localization of the hybrid pyruvate/2-oxoglutarate dehydrogenase complex in the actinobacterium Corynebacterium glutamicum. Microbiol Spectr 2023; 11:e0266823. [PMID: 37754766 PMCID: PMC10581250 DOI: 10.1128/spectrum.02668-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 09/28/2023] Open
Abstract
For many bacterial proteins, specific localizations within the cell have been demonstrated, but enzymes involved in central metabolism are usually considered to be homogenously distributed within the cytoplasm. Here, we provide an example for a spatially defined localization of a unique enzyme complex found in actinobacteria, the hybrid pyruvate/2-oxoglutarate dehydrogenase complex (PDH-ODH). In non-actinobacterial cells, PDH and ODH form separate multienzyme complexes of megadalton size composed of three different subunits, E1, E2, and E3. The actinobacterial PDH-ODH complex is composed of four subunits, AceE (E1p), AceF (E2p), Lpd (E3), and OdhA (E1oE2o). Using fluorescence microscopy, we observed that in Corynebacterium glutamicum, all four subunits are co-localized in distinct spots at the cell poles, and in larger cells, additional spots are present at mid-cell. These results further confirm the existence of the hybrid complex. The unphosporylated OdhI protein, which binds to OdhA and inhibits ODH activity, was co-localized with OdhA at the poles, whereas phosphorylated OdhI, which does not bind OdhA, was distributed in the entire cytoplasm. Isocitrate dehydrogenase and glutamate dehydrogenase, both metabolically linked to ODH, were evenly distributed in the cytoplasm. Based on the available structural data for individual PDH-ODH subunits, a novel supramolecular architecture of the hybrid complex differing from classical PDH and ODH complexes has to be postulated. Our results suggest that localization at the poles or at mid-cell is most likely caused by nucleoid exclusion and results in a spatially organized metabolism in actinobacteria, with consequences yet to be studied. IMPORTANCE Enzymes involved in the central metabolism of bacteria are usually considered to be distributed within the entire cytoplasm. Here, we provide an example for a spatially defined localization of a unique enzyme complex of actinobacteria, the hybrid pyruvate dehydrogenase/2-oxoglutarate dehydrogenase (PDH-ODH) complex composed of four different subunits. Using fusions with mVenus or mCherry and fluorescence microscopy, we show that all four subunits are co-localized in distinct spots at the cell poles, and in larger cells, additional spots were observed at mid-cell. These results clearly support the presence of the hybrid PDH-ODH complex and suggest a similar localization in other actinobacteria. The observation of a defined spatial localization of an enzyme complex catalyzing two key reactions of central metabolism poses questions regarding possible consequences for the availability of substrates and products within the cell and other bacterial enzyme complexes showing similar behavior.
Collapse
Affiliation(s)
- Lea Sundermeyer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Jan-Gerrit Folkerts
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Benita Lückel
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Christina Mack
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Meike Baumgart
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Michael Bott
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
- Bioeconomy Science Center (BioSC), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
22
|
Werner F, Schwardmann LS, Siebert D, Rückert-Reed C, Kalinowski J, Wirth MT, Hofer K, Takors R, Wendisch VF, Blombach B. Metabolic engineering of Corynebacterium glutamicum for fatty alcohol production from glucose and wheat straw hydrolysate. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2023; 16:116. [PMID: 37464396 DOI: 10.1186/s13068-023-02367-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Fatty acid-derived products such as fatty alcohols (FAL) find growing application in cosmetic products, lubricants, or biofuels. So far, FAL are primarily produced petrochemically or through chemical conversion of bio-based feedstock. Besides the well-known negative environmental impact of using fossil resources, utilization of bio-based first-generation feedstock such as palm oil is known to contribute to the loss of habitat and biodiversity. Thus, the microbial production of industrially relevant chemicals such as FAL from second-generation feedstock is desirable. RESULTS To engineer Corynebacterium glutamicum for FAL production, we deregulated fatty acid biosynthesis by deleting the transcriptional regulator gene fasR, overexpressing a fatty acyl-CoA reductase (FAR) gene of Marinobacter hydrocarbonoclasticus VT8 and attenuating the native thioesterase expression by exchange of the ATG to a weaker TTG start codon. C. glutamicum ∆fasR cg2692TTG (pEKEx2-maqu2220) produced in shaking flasks 0.54 ± 0.02 gFAL L-1 from 20 g glucose L-1 with a product yield of 0.054 ± 0.001 Cmol Cmol-1. To enable xylose utilization, we integrated xylA encoding the xylose isomerase from Xanthomonas campestris and xylB encoding the native xylulose kinase into the locus of actA. This approach enabled growth on xylose. However, adaptive laboratory evolution (ALE) was required to improve the growth rate threefold to 0.11 ± 0.00 h-1. The genome of the evolved strain C. glutamicum gX was re-sequenced, and the evolved genetic module was introduced into C. glutamicum ∆fasR cg2692TTG (pEKEx2-maqu2220) which allowed efficient growth and FAL production on wheat straw hydrolysate. FAL biosynthesis was further optimized by overexpression of the pntAB genes encoding the membrane-bound transhydrogenase of E. coli. The best-performing strain C. glutamicum ∆fasR cg2692TTG CgLP12::(Ptac-pntAB-TrrnB) gX (pEKEx2-maqu2220) produced 2.45 ± 0.09 gFAL L-1 with a product yield of 0.054 ± 0.005 Cmol Cmol-1 and a volumetric productivity of 0.109 ± 0.005 gFAL L-1 h-1 in a pulsed fed-batch cultivation using wheat straw hydrolysate. CONCLUSION The combination of targeted metabolic engineering and ALE enabled efficient FAL production in C. glutamicum from wheat straw hydrolysate for the first time. Therefore, this study provides useful metabolic engineering principles to tailor this bacterium for other products from this second-generation feedstock.
Collapse
Affiliation(s)
- Felix Werner
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
| | - Lynn S Schwardmann
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Daniel Siebert
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany
| | | | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Marie-Theres Wirth
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
| | - Katharina Hofer
- Institute of Biochemical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Ralf Takors
- Institute of Biochemical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Volker F Wendisch
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Bastian Blombach
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany.
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany.
| |
Collapse
|
23
|
Ramp P, Mack C, Wirtz A, Bott M. Alternative routes for production of the drug candidate d-chiro-inositol with Corynebacterium glutamicum using endogenous or promiscuous plant enzymes. Metab Eng 2023; 78:1-10. [PMID: 37146873 DOI: 10.1016/j.ymben.2023.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/05/2023] [Accepted: 04/23/2023] [Indexed: 05/07/2023]
Abstract
d-chiro-Inositol (DCI) is a promising drug candidate for treating insulin resistance and associated diseases such as type 2 diabetes or polycystic ovary syndrome. In this study, we developed two production processes for DCI using Corynebacterium glutamicum as host. In the first process, myo-inositol (MI) is oxidized to 2-keto-myo-inositol (2KMI) by the inositol dehydrogenase (IDH) IolG and then isomerized to 1-keto-d-chiro-inositol (1KDCI) by the isomerases Cg0212 or Cg2312, both of which were identified in this work. 1KDCI is then reduced to DCI by IolG. Overproduction of IolG and Cg0212 in a chassis strain unable to degrade inositols allowed the production of 1.1 g/L DCI from 10 g/L MI. As both reactions involved are reversible, only a partial conversion of MI to DCI can be achieved. To enable higher conversion ratios, a novel route towards DCI was established by utilizing the promiscuous activity of two plant-derived enzymes, the NAD+-dependent d-ononitol dehydrogenase MtOEPa and the NADPH-dependent d-pinitol dehydrogenase MtOEPb from Medicago truncatula (barrelclover). Heterologous production of these enzymes in the chassis strain led to the production of 1.6 g/L DCI from 10 g/L MI. For replacing the substrate MI by glucose, the two plant genes were co-expressed with the endogenous myo-inositol-1-phosphate synthase gene ino1 either as a synthetic operon or using a novel, bicistronic T7-based expression vector. With the single operon construct, 0.75 g/L DCI was formed from 20 g/L glucose, whereas with the bicistronic construct 1.2 g/L DCI was obtained, disclosing C. glutamicum as an attractive host for of d-chiro-inositol production.
Collapse
Affiliation(s)
- Paul Ramp
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Christina Mack
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Astrid Wirtz
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Michael Bott
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
24
|
Hartmann FSF, Weiß T, Kastberg LLB, Workman CT, Seibold GM. Precise and versatile microplate reader-based analyses of biosensor signals from arrayed microbial colonies. Front Microbiol 2023; 14:1187228. [PMID: 37389345 PMCID: PMC10303141 DOI: 10.3389/fmicb.2023.1187228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Genetically encoded fluorescent biosensors have emerged as a powerful tool to support phenotypic screenings of microbes. Optical analyses of fluorescent sensor signals from colonies grown on solid media can be challenging as imaging devices need to be equipped with appropriate filters matching the properties of fluorescent biosensors. Toward versatile fluorescence analyses of different types of biosensor signals derived from arrayed colonies, we investigate here the use of monochromator equipped microplate readers as an alternative to imaging approaches. Indeed, for analyses of the LacI-controlled expression of the reporter mCherry in Corynebacterium glutamicum, or promoter activity using GFP as reporter in Saccharomyces cerevisiae, an improved sensitivity and dynamic range was observed for a microplate reader-based analyses compared to their analyses via imaging. The microplate reader allowed us to capture signals of ratiometric fluorescent reporter proteins (FRPs) with a high sensitivity and thereby to further improve the analysis of internal pH via the pH-sensitive FRP mCherryEA in Escherichia coli colonies. Applicability of this novel technique was further demonstrated by assessing redox states in C. glutamicum colonies using the FRP Mrx1-roGFP2. By the use of a microplate reader, oxidative redox shifts were measured in a mutant strain lacking the non-enzymatic antioxidant mycothiol (MSH), indicating its major role for maintaining a reduced redox state also in colonies on agar plates. Taken together, analyses of biosensor signals from microbial colonies using a microplate reader allows comprehensive phenotypic screenings and thus facilitates further development of new strains for metabolic engineering and systems biology.
Collapse
|
25
|
Schwardmann LS, Wu T, Dransfeld AK, Lindner SN, Wendisch VF. Formamide-based production of amines by metabolically engineering Corynebacterium glutamicum. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12592-3. [PMID: 37246985 DOI: 10.1007/s00253-023-12592-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/30/2023]
Abstract
Formamide is rarely used as nitrogen source by microorganisms. Therefore, formamide and formamidase have been used as protection system to allow for growth under non-sterile conditions and for non-sterile production of acetoin, a product lacking nitrogen. Here, we equipped Corynebacterium glutamicum, a renowned workhorse for industrial amino acid production for 60 years, with formamidase from Helicobacter pylori 26695, enabling growth with formamide as sole nitrogen source. Thereupon, the formamide/formamidase system was exploited for efficient formamide-based production of the nitrogenous compounds L-glutamate, L-lysine, N-methylphenylalanine, and dipicolinic acid by transfer of the formamide/formamidase system to established producer strains. Stable isotope labeling verified the incorporation of nitrogen from formamide into biomass and the representative product L-lysine. Moreover, we showed ammonium leakage during formamidase-based access of formamide to be exploitable to support growth of formamidase-deficient C. glutamicum in co-cultivation and demonstrated that efficient utilization of formamide as sole nitrogen source benefitted from overexpression of formate dehydrogenase. KEY POINTS: • C. glutamicum was engineered to access formamide. • Formamide-based production of nitrogenous compounds was established. • Nitrogen cross-feeding supported growth of a formamidase-negative strain.
Collapse
Affiliation(s)
- Lynn S Schwardmann
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - Tong Wu
- Department of Biochemistry, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Aron K Dransfeld
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - Steffen N Lindner
- Department of Biochemistry, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Volker F Wendisch
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany.
| |
Collapse
|
26
|
Göttl VL, Pucker B, Wendisch VF, Henke NA. Screening of Structurally Distinct Lycopene β-Cyclases for Production of the Cyclic C40 Carotenoids β-Carotene and Astaxanthin by Corynebacterium glutamicum. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7765-7776. [PMID: 37162369 DOI: 10.1021/acs.jafc.3c01492] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Lycopene β-cyclase (EC 5.5.1.19) is one of the key enzymes in the biosynthesis of β-carotene and derived carotenoids. It catalyzes isomerase reactions to form β-carotene from lycopene by β-cyclization of both of its ψ-ends. Lycopene β-cyclases are widespread in nature. We systematically analyzed the phylogeny of lycopene β-cyclases from all kingdoms of life and predicted their transmembrane structures. To this end, a collection of previously characterized lycopene β-cyclase polypeptide sequences served as bait sequences to identify their closest homologues in a range of bacteria, archaea, fungi, algae, and plant species. Furthermore, a DeepTMHMM scan was applied to search for the presence of transmembrane domains. A phylogenetic tree suggests at least five distinct clades, and the DeepTMHMM scan revealed that lycopene β-cyclases are a group of structurally different proteins: membrane-bound and cytosolic enzymes. Representative lycopene β-cyclases were screened in the lycopene-overproducing Corynebacterium glutamicum strain for β-carotene and astaxanthin production. This systematic screening facilitates the identification of new enzymes for carotenoid production. Higher astaxanthin production and less reduction of total carotenoids were achieved with the cytosolic lycopene β-cyclase CrtL from Synechococcus elongatus and the membrane-bound heterodimeric lycopene β-cyclase CrtYcd from Brevibacterium linens.
Collapse
Affiliation(s)
- Vanessa L Göttl
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615 Bielefeld, Germany
| | - Boas Pucker
- Institute of Plant Biology & BRICS, TU Braunschweig, 38106 Braunschweig, Germany
| | - Volker F Wendisch
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615 Bielefeld, Germany
| | - Nadja A Henke
- Genetics of Prokaryotes, Faculty of Biology and CeBiTec, Bielefeld University, 33615 Bielefeld, Germany
| |
Collapse
|
27
|
Schmitt I, Meyer F, Krahn I, Henke NA, Peters-Wendisch P, Wendisch VF. From Aquaculture to Aquaculture: Production of the Fish Feed Additive Astaxanthin by Corynebacterium glutamicum Using Aquaculture Sidestream. Molecules 2023; 28:molecules28041996. [PMID: 36838984 PMCID: PMC9958746 DOI: 10.3390/molecules28041996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/31/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Circular economy holds great potential to minimize the use of finite resources, and reduce waste formation by the creation of closed-loop systems. This also pertains to the utilization of sidestreams in large-scale biotechnological processes. A flexible feedstock concept has been established for the industrially relevant Corynebacterium glutamicum, which naturally synthesizes the yellow C50 carotenoid decaprenoxanthin. In this study, we aimed to use a preprocessed aquaculture sidestream for production of carotenoids, including the fish feed ingredient astaxanthin by C. glutamicum. The addition of a preprocessed aquaculture sidestream to the culture medium did not inhibit growth, obviated the need for addition of several components of the mineral salt's medium, and notably enhanced production of astaxanthin by an engineered C. glutamicum producer strain. Improved astaxanthin production was scaled to 2 L bioreactor fermentations. This strategy to improve astaxanthin production was shown to be transferable to production of several native and non-native carotenoids. Thus, this study provides a proof-of-principle for improving carotenoid production by C. glutamicum upon supplementation of a preprocessed aquaculture sidestream. Moreover, in the case of astaxanthin production it may be a potential component of a circular economy in aquaculture.
Collapse
|
28
|
Pashou E, Reich SJ, Reiter A, Weixler D, Eikmanns BJ, Oldiges M, Riedel CU, Goldbeck O. Identification and Characterization of Corynaridin, a Novel Linaridin from Corynebacterium lactis. Microbiol Spectr 2023; 11:e0175622. [PMID: 36541778 PMCID: PMC9927463 DOI: 10.1128/spectrum.01756-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Genome analysis of Corynebacterium lactis revealed a bacteriocin gene cluster encoding a putative bacteriocin of the linaridin family of ribosomally synthesized and posttranslationally modified peptides (RiPPs). The locus harbors typical linaridin modification enzymes but lacks genes for a decarboxylase and methyltransferase, which is unusual for type B linaridins. Supernatants of Corynebacterium lactis RW3-42 showed antimicrobial activity against Corynebacterium glutamicum. Deletion of the precursor gene crdA clearly linked the antimicrobial activity of the producer strain to the identified gene cluster. Following purification, we observed potent activity of the peptide against Actinobacteria, mainly other members of the genus Corynebacterium, including the pathogenic species Corynebacterium striatum and Corynebacterium amycolatum. Also, low activity against some Firmicutes was observed, but there was no activity against Gram-negative species. The peptide is resilient towards heat but sensitive to proteolytic degradation by trypsin and proteinase K. Analysis by mass spectrometry indicates that corynaridin is processed by cleaving off the leader sequence at a conserved motif and posttranslationally modified by dehydration of all threonine and serin residues, resulting in a monoisotopic mass of 3,961.19 Da. Notably, time-kill kinetics and experiments using live biosensors to monitor membrane integrity suggest bactericidal activity that does not involve formation of pores in the cytoplasmic membrane. As Corynebacterium species are ubiquitous in nature and include important commensals and pathogens of mammalian organisms, secretion of bacteriocins by species of this genus could be a hitherto neglected trait with high relevance for intra- and interspecies competition and infection. IMPORTANCE Bacteriocins are antimicrobial peptides produced by bacteria to fend off competitors in ecological niches and are considered to be important factors influencing the composition of microbial communities. However, bacteriocin production by bacteria of the genus Corynebacterium has been a hitherto neglected trait, although its species are ubiquitous in nature and make up large parts of the microbiome of humans and animals. In this study, we describe and characterize a novel linaridin family bacteriocin from Corynebacterium lactis and show its narrow-spectrum activity, mainly against other actinobacteria. Moreover, we were able to extend the limited knowledge on linaridin bioactivity in general and for the first time describe the bactericidal activity of such a bacteriocin. Interestingly, the peptide, which was named corynaridin, appears bactericidal, but without formation of pores in the bacterial membrane.
Collapse
Affiliation(s)
- Efthimia Pashou
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Sebastian J. Reich
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Alexander Reiter
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Dominik Weixler
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | | | - Marco Oldiges
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Christian U. Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Oliver Goldbeck
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| |
Collapse
|
29
|
Reich SJ, Goldbeck O, Lkhaasuren T, Weixler D, Weiß T, Eikmanns BJ. C-di-AMP Is a Second Messenger in Corynebacterium glutamicum That Regulates Expression of a Cell Wall-Related Peptidase via a Riboswitch. Microorganisms 2023; 11:296. [PMID: 36838266 PMCID: PMC9960051 DOI: 10.3390/microorganisms11020296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Cyclic di-adenosine monophosphate (c-di-AMP) is a bacterial second messenger discovered in Bacillus subtilis and involved in potassium homeostasis, cell wall maintenance and/or DNA stress response. As the role of c-di-AMP has been mostly studied in Firmicutes, we sought to increase the understanding of its role in Actinobacteria, namely in Corynebacterium glutamicum. This organism is a well-known industrial production host and a model organism for pathogens, such as C. diphtheriae or Mycobacterium tuberculosis. Here, we identify and analyze the minimal set of two C. glutamicum enzymes, the diadenylate cyclase DisA and the phosphodiesterase PdeA, responsible for c-di-AMP metabolism. DisA synthesizes c-di-AMP from two molecules of ATP, whereas PdeA degrades c-di-AMP, as well as the linear degradation intermediate phosphoadenylyl-(3'→5')-adenosine (pApA) to two molecules of AMP. Here, we show that a ydaO/kimA-type c-di-AMP-dependent riboswitch controls the expression of the strictly regulated cell wall peptidase gene nlpC in C. glutamicum. In contrast to previously described members of the ydaO/kimA-type riboswitches, our results suggest that the C. glutamicum nlpC riboswitch likely affects the translation instead of the transcription of its downstream gene. Although strongly regulated by different mechanisms, we show that the absence of nlpC, the first known regulatory target of c-di-AMP in C. glutamicum, is not detrimental for this organism under the tested conditions.
Collapse
Affiliation(s)
- Sebastian J. Reich
- Institute of Microbiology and Biotechnology, Ulm University, 89081 Ulm, Germany
| | - Oliver Goldbeck
- Institute of Microbiology and Biotechnology, Ulm University, 89081 Ulm, Germany
- Institute of Biochemistry, Department of Chemistry, University of Cologne, 50674 Cologne, Germany
| | | | - Dominik Weixler
- Institute of Microbiology and Biotechnology, Ulm University, 89081 Ulm, Germany
| | - Tamara Weiß
- Institute of Microbiology and Biotechnology, Ulm University, 89081 Ulm, Germany
| | | |
Collapse
|
30
|
Helleckes LM, Müller C, Griesbach T, Waffenschmidt V, Moch M, Osthege M, Wiechert W, Oldiges M. Explore or exploit? A model-based screening strategy for PETase secretion by Corynebacterium glutamicum. Biotechnol Bioeng 2023; 120:139-153. [PMID: 36225165 DOI: 10.1002/bit.28261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
Extracellular production of target proteins simplifies downstream processing due to obsolete cell disruption. However, optimal combinations of a heterologous protein, suitable signal peptide, and secretion host can currently not be predicted, resulting in large strain libraries that need to be tested. On the experimental side, this challenge can be tackled by miniaturization, parallelization, and automation, which provide high-throughput screening data. These data need to be condensed into a candidate ranking for decision-making to focus bioprocess development on the most promising candidates. We screened for Bacillus subtilis signal peptides mediating Sec secretion of two polyethylene terephthalate degrading enzymes (PETases), leaf-branch compost cutinase (LCC) and polyester hydrolase mutants, by Corynebacterium glutamicum. We developed a fully automated screening process and constructed an accompanying Bayesian statistical modeling framework, which we applied in screenings for highest activity in 4-nitrophenyl palmitate degradation. In contrast to classical evaluation methods, batch effects and biological errors are taken into account and their uncertainty is quantified. Within only two rounds of screening, the most suitable signal peptide was identified for each PETase. Results from LCC secretion in microliter-scale cultivation were shown to be scalable to laboratory-scale bioreactors. This work demonstrates an experiment-modeling loop that can accelerate early-stage screening in a way that experimental capacities are focused to the most promising strain candidates. Combined with high-throughput cloning, this paves the way for using large strain libraries of several hundreds of strains in a Design-Build-Test-Learn approach.
Collapse
Affiliation(s)
- Laura M Helleckes
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Carolin Müller
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Tim Griesbach
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Vera Waffenschmidt
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Matthias Moch
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Michael Osthege
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wiechert
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.,Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Marco Oldiges
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
31
|
An NADPH-auxotrophic Corynebacterium glutamicum recombinant strain and used it to construct L-leucine high-yielding strain. INTERNATIONAL MICROBIOLOGY : THE OFFICIAL JOURNAL OF THE SPANISH SOCIETY FOR MICROBIOLOGY 2023; 26:11-24. [PMID: 35925494 DOI: 10.1007/s10123-022-00270-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 01/06/2023]
Abstract
The NADPH-regeneration enzymes in Corynebacterium glutamicum were inactivated to construct an NADPH-auxotrophic C. glutamicum strain by gene knockout and gene replacement. The resultant NADPH-auxotrophic C. glutamicum XL-1 ΔZMICg::ISm (i.e., strain Leu-1) grew well in the basic medium only with gluconate as carbon source. Replacement of the native glyceraldehyde 3-phosphate dehydrogenase (NAD-GapDHCg) by NADP-GapDHCa from Clostridium acetobutylicum is an effective strategy for producing L-leucine in NADPH-prototrophic strain XL-1 and NADPH-auxotrophic strain Leu-1, whereas the L-leucine yield did not differ significantly between these strains (14.1 ± 1.8 g/L vs 16.2 ± 1.1 g/L). Enhancing the carbon flux in biosynthetic pathway by recombinant expression plasmid pEC-ABNCE promoted L-leucine production, but the shortage NADPH supply limited the L-leucine yield. The mutated promoters of zwf and icdCg were introduced into C. glutamicum with NADP-GapDHCa and pEC-ABNCE increased L-leucine yield (54.3 ± 2.9 g/L) and improved cell growth (OD562 = 83.4 ± 7.5) in fed-batch fermentation because the resultant strain C. glutamicum XL-1 ΔMICg::ISm GCg::GCa Pzwf-D1 Picd-D2/pEC-ABNCE (i.e., strain Leu-9) exhibited the proper intracellular NADPH and NADH level. This is the first report of constructing an L-leucine high-yielding strain that reasonably supplies NADPH by optimizing the biosynthetic pathway of NADPH from an NADPH-auxotrophic strain.
Collapse
|
32
|
Sundermeyer L, Bosco G, Gujar S, Brocker M, Baumgart M, Willbold D, Weiergräber OH, Bellinzoni M, Bott M. Characteristics of the GlnH and GlnX Signal Transduction Proteins Controlling PknG-Mediated Phosphorylation of OdhI and 2-Oxoglutarate Dehydrogenase Activity in Corynebacterium glutamicum. Microbiol Spectr 2022; 10:e0267722. [PMID: 36445153 PMCID: PMC9769921 DOI: 10.1128/spectrum.02677-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/05/2022] [Indexed: 12/03/2022] Open
Abstract
In Corynebacterium glutamicum the protein kinase PknG phosphorylates OdhI and thereby abolishes the inhibition of 2-oxoglutarate dehydrogenase activity by unphosphorylated OdhI. Our previous studies suggested that PknG activity is controlled by the periplasmic binding protein GlnH and the transmembrane protein GlnX, because ΔglnH and ΔglnX mutants showed a growth defect on glutamine similar to that of a ΔpknG mutant. We have now confirmed the involvement of GlnH and GlnX in the control of OdhI phosphorylation by analyzing the OdhI phosphorylation status and glutamate secretion in ΔglnH and ΔglnX mutants and by characterizing ΔglnX suppressor mutants. We provide evidence for GlnH being a lipoprotein and show by isothermal titration calorimetry that it binds l-aspartate and l-glutamate with moderate to low affinity, but not l-glutamine, l-asparagine, or 2-oxoglutarate. Based on a structural comparison with GlnH of Mycobacterium tuberculosis, two residues critical for the binding affinity were identified and verified. The predicted GlnX topology with four transmembrane segments and two periplasmic domains was confirmed by PhoA and LacZ fusions. A structural model of GlnX suggested that, with the exception of a poorly ordered N-terminal region, the entire protein is composed of α-helices and small loops or linkers, and it revealed similarities to other bacterial transmembrane receptors. Our results suggest that the GlnH-GlnX-PknG-OdhI-OdhA signal transduction cascade serves to adapt the flux of 2-oxoglutarate between ammonium assimilation via glutamate dehydrogenase and energy generation via the tricarboxylic acid (TCA) cycle to the availability of the amino group donors l-glutamate and l-aspartate in the environment. IMPORTANCE Actinobacteria comprise a large number of species playing important roles in biotechnology and medicine, such as Corynebacterium glutamicum, the major industrial amino acid producer, and Mycobacterium tuberculosis, the pathogen causing tuberculosis. Many actinobacteria use a signal transduction process in which the phosphorylation status of OdhI (corynebacteria) or GarA (mycobacteria) regulates the carbon flux at the 2-oxoglutarate node. Inhibition of 2-oxoglutarate dehydrogenase by unphosphorylated OdhI shifts the flux of 2-oxoglutarate from the TCA cycle toward glutamate formation and, thus, ammonium assimilation. Phosphorylation of OdhI/GarA is catalyzed by the protein kinase PknG, whose activity was proposed to be controlled by the periplasmic binding protein GlnH and the transmembrane protein GlnX. In this study, we combined genetic, biochemical, and structural modeling approaches to characterize GlnH and GlnX of C. glutamicum and confirm their roles in the GlnH-GlnX-PknG-OdhI-OdhA signal transduction cascade. These findings are relevant also to other Actinobacteria employing a similar control process.
Collapse
Affiliation(s)
- Lea Sundermeyer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Graziella Bosco
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Srushti Gujar
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
- IBI-7: Structural Biochemistry, Institute of Biological Information Processing, Forschungszentrum Jülich, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Melanie Brocker
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Meike Baumgart
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Dieter Willbold
- IBI-7: Structural Biochemistry, Institute of Biological Information Processing, Forschungszentrum Jülich, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Oliver H. Weiergräber
- IBI-7: Structural Biochemistry, Institute of Biological Information Processing, Forschungszentrum Jülich, Jülich, Germany
| | - Marco Bellinzoni
- Institut Pasteur, Université de Paris Cité, CNRS UMR3528, Unité de Microbiologie Structurale, Paris, France
| | - Michael Bott
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
- Bioeconomy Science Center (BioSC), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
33
|
Schmollack M, Werner F, Huber J, Kiefer D, Merkel M, Hausmann R, Siebert D, Blombach B. Metabolic engineering of Corynebacterium glutamicum for acetate-based itaconic acid production. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:139. [PMID: 36517879 PMCID: PMC9753420 DOI: 10.1186/s13068-022-02238-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Itaconic acid is a promising platform chemical for a bio-based polymer industry. Today, itaconic acid is biotechnologically produced with Aspergillus terreus at industrial scale from sugars. The production of fuels but also of chemicals from food substrates is a dilemma since future processes should rely on carbon sources which do not compete for food or feed. Therefore, the production of chemicals from alternative substrates such as acetate is desirable to develop novel value chains in the bioeconomy. RESULTS In this study, Corynebacterium glutamicum ATCC 13032 was engineered to efficiently produce itaconic acid from the non-food substrate acetate. Therefore, we rewired the central carbon and nitrogen metabolism by inactivating the transcriptional regulator RamB, reducing the activity of isocitrate dehydrogenase, deletion of the gdh gene encoding glutamate dehydrogenase and overexpression of cis-aconitate decarboxylase (CAD) from A. terreus optimized for expression in C. glutamicum. The final strain C. glutamicum ΔramB Δgdh IDHR453C (pEKEx2-malEcadopt) produced 3.43 ± 0.59 g itaconic acid L-1 with a product yield of 81 ± 9 mmol mol-1 during small-scale cultivations in nitrogen-limited minimal medium containing acetate as sole carbon and energy source. Lowering the cultivation temperature from 30 °C to 25 °C improved CAD activity and further increased the titer and product yield to 5.01 ± 0.67 g L-1 and 116 ± 15 mmol mol-1, respectively. The latter corresponds to 35% of the theoretical maximum and so far represents the highest product yield for acetate-based itaconic acid production. Further, the optimized strain C. glutamicum ΔramB Δgdh IDHR453C (pEKEx2-malEcadopt), produced 3.38 ± 0.28 g itaconic acid L-1 at 25 °C from an acetate-containing aqueous side-stream of fast pyrolysis. CONCLUSION As shown in this study, acetate represents a suitable non-food carbon source for itaconic acid production with C. glutamicum. Tailoring the central carbon and nitrogen metabolism enabled the efficient production of itaconic acid from acetate and therefore this study offers useful design principles to genetically engineer C. glutamicum for other products from acetate.
Collapse
Affiliation(s)
- Marc Schmollack
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
| | - Felix Werner
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
| | - Janine Huber
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
| | - Dirk Kiefer
- Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, University of Hohenheim, Stuttgart, Germany
| | - Manuel Merkel
- Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, University of Hohenheim, Stuttgart, Germany
| | - Rudolf Hausmann
- Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, University of Hohenheim, Stuttgart, Germany
| | - Daniel Siebert
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany
| | - Bastian Blombach
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Uferstraße 53, 94315, Straubing, Germany.
- SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany.
| |
Collapse
|
34
|
Ghiffary MR, Prabowo CPS, Adidjaja JJ, Lee SY, Kim HU. Systems metabolic engineering of Corynebacterium glutamicum for the efficient production of β-alanine. Metab Eng 2022; 74:121-129. [DOI: 10.1016/j.ymben.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
35
|
Physiological, Biochemical, and Structural Bioinformatic Analysis of the Multiple Inositol Dehydrogenases from Corynebacterium glutamicum. Microbiol Spectr 2022; 10:e0195022. [PMID: 36094194 PMCID: PMC9603128 DOI: 10.1128/spectrum.01950-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inositols (cyclohexanehexols) comprise nine isomeric cyclic sugar alcohols, several of which occur in all domains of life with various functions. Many bacteria can utilize inositols as carbon and energy sources via a specific pathway involving inositol dehydrogenases (IDHs) as the first step of catabolism. The microbial cell factory Corynebacterium glutamicum can grow with myo-inositol as a sole carbon source. Interestingly, this species encodes seven potential IDHs, raising the question of the reason for this multiplicity. We therefore investigated the seven IDHs to determine their function, activity, and selectivity toward the biologically most important isomers myo-, scyllo-, and d-chiro-inositol. We created an ΔIDH strain lacking all seven IDH genes, which could not grow on the three inositols. scyllo- and d-chiro-inositol were identified as novel growth substrates of C. glutamicum. Complementation experiments showed that only four of the seven IDHs (IolG, OxiB, OxiD, and OxiE) enabled growth of the ΔIDH strain on two of the three inositols. The kinetics of the four purified enzymes agreed with the complementation results. IolG and OxiD are NAD+-dependent IDHs accepting myo- and d-chiro-inositol but not scyllo-inositol. OxiB is an NAD+-dependent myo-IDH with a weak activity also for scyllo-inositol but not for d-chiro-inositol. OxiE on the other hand is an NAD+-dependent scyllo-IDH showing also good activity for myo-inositol and a very weak activity for d-chiro-inositol. Structural models, molecular docking experiments, and sequence alignments enabled the identification of the substrate binding sites of the active IDHs and of residues allowing predictions on the substrate specificity. IMPORTANCE myo-, scyllo-, and d-chiro-inositol are C6 cyclic sugar alcohols with various biological functions, which also serve as carbon sources for microbes. Inositol catabolism starts with an oxidation to keto-inositols catalyzed by inositol dehydrogenases (IDHs). The soil bacterium C. glutamicum encodes seven potential IDHs. Using a combination of microbiological, biochemical, and modeling approaches, we analyzed the function of these enzymes and identified four IDHs involved in the catabolism of inositols. They possess distinct substrate preferences for the three isomers, and modeling and sequence alignments allowed the identification of residues important for substrate specificity. Our results expand the knowledge of bacterial inositol metabolism and provide an important basis for the rational development of producer strains for these valuable inositols, which show pharmacological activities against, e.g., Alzheimer's disease, polycystic ovarian syndrome, or type II diabetes.
Collapse
|
36
|
Kranz A, Polen T, Kotulla C, Arndt A, Bosco G, Bussmann M, Chattopadhyay A, Cramer A, Davoudi CF, Degner U, Diesveld R, Freiherr von Boeselager R, Gärtner K, Gätgens C, Georgi T, Geraths C, Haas S, Heyer A, Hünnefeld M, Ishige T, Kabus A, Kallscheuer N, Kever L, Klaffl S, Kleine B, Kočan M, Koch-Koerfges A, Kraxner KJ, Krug A, Krüger A, Küberl A, Labib M, Lange C, Mack C, Maeda T, Mahr R, Majda S, Michel A, Morosov X, Müller O, Nanda AM, Nickel J, Pahlke J, Pfeifer E, Platzen L, Ramp P, Rittmann D, Schaffer S, Scheele S, Spelberg S, Schulte J, Schweitzer JE, Sindelar G, Sorger-Herrmann U, Spelberg M, Stansen C, Tharmasothirajan A, Ooyen JV, van Summeren-Wesenhagen P, Vogt M, Witthoff S, Zhu L, Eikmanns BJ, Oldiges M, Schaumann G, Baumgart M, Brocker M, Eggeling L, Freudl R, Frunzke J, Marienhagen J, Wendisch VF, Bott M. A manually curated compendium of expression profiles for the microbial cell factory Corynebacterium glutamicum. Sci Data 2022; 9:594. [PMID: 36182956 PMCID: PMC9526701 DOI: 10.1038/s41597-022-01706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/18/2022] [Indexed: 11/12/2022] Open
Abstract
Corynebacterium glutamicum is the major host for the industrial production of amino acids and has become one of the best studied model organisms in microbial biotechnology. Rational strain construction has led to an improvement of producer strains and to a variety of novel producer strains with a broad substrate and product spectrum. A key factor for the success of these approaches is detailed knowledge of transcriptional regulation in C. glutamicum. Here, we present a large compendium of 927 manually curated microarray-based transcriptional profiles for wild-type and engineered strains detecting genome-wide expression changes of the 3,047 annotated genes in response to various environmental conditions or in response to genetic modifications. The replicates within the 927 experiments were combined to 304 microarray sets ordered into six categories that were used for differential gene expression analysis. Hierarchical clustering confirmed that no outliers were present in the sets. The compendium provides a valuable resource for future fundamental and applied research with C. glutamicum and contributes to a systemic understanding of this microbial cell factory. Measurement(s) Gene Expression Analysis Technology Type(s) Two Color Microarray Factor Type(s) WT condition A vs. WT condition B • Plasmid-based gene overexpression in parental strain vs. parental strain with empty vector control • Deletion mutant vs. parental strain Sample Characteristic - Organism Corynebacterium glutamicum Sample Characteristic - Environment laboratory environment Sample Characteristic - Location Germany.
Collapse
Affiliation(s)
- Angela Kranz
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany.
- IBG-4: Bioinformatics, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany.
| | - Tino Polen
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Christian Kotulla
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Annette Arndt
- Institute of Microbiology and Biotechnology, University of Ulm, D-89069, Ulm, Germany
| | - Graziella Bosco
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Michael Bussmann
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Ava Chattopadhyay
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Annette Cramer
- Institute of Microbiology and Biotechnology, University of Ulm, D-89069, Ulm, Germany
| | - Cedric-Farhad Davoudi
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Ursula Degner
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Ramon Diesveld
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | | | - Kim Gärtner
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Cornelia Gätgens
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Tobias Georgi
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Christian Geraths
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Sabine Haas
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Antonia Heyer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Max Hünnefeld
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Takeru Ishige
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Armin Kabus
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Nicolai Kallscheuer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Larissa Kever
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Simon Klaffl
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Britta Kleine
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Martina Kočan
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Abigail Koch-Koerfges
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Kim J Kraxner
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Andreas Krug
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Aileen Krüger
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Andreas Küberl
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Mohamed Labib
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Christian Lange
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Christina Mack
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Tomoya Maeda
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Regina Mahr
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Stephan Majda
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Andrea Michel
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Xenia Morosov
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Olga Müller
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Arun M Nanda
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Jens Nickel
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Jennifer Pahlke
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Eugen Pfeifer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Laura Platzen
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Paul Ramp
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Doris Rittmann
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Steffen Schaffer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Sandra Scheele
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Stephanie Spelberg
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Julia Schulte
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Jens-Eric Schweitzer
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Georg Sindelar
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Ulrike Sorger-Herrmann
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Markus Spelberg
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Corinna Stansen
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Apilaasha Tharmasothirajan
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Jan van Ooyen
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | | | - Michael Vogt
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Sabrina Witthoff
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Lingfeng Zhu
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Bernhard J Eikmanns
- Institute of Microbiology and Biotechnology, University of Ulm, D-89069, Ulm, Germany
| | - Marco Oldiges
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Georg Schaumann
- SenseUp GmbH, c/o Campus Forschungszentrum, Wilhelm-Johnen-Strasse, D-52425, Jülich, Germany
| | - Meike Baumgart
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Melanie Brocker
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Lothar Eggeling
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Roland Freudl
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Julia Frunzke
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Jan Marienhagen
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Volker F Wendisch
- Genetics of Prokaryotes, Biology & CeBiTec, Bielefeld University, Universitaetsstr. 25, D-33615, Bielefeld, Germany
| | - Michael Bott
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, D-52425, Jülich, Germany.
| |
Collapse
|
37
|
Krüger A, Frunzke J. A pseudokinase version of the histidine kinase ChrS promotes high heme tolerance of Corynebacterium glutamicum. Front Microbiol 2022; 13:997448. [PMID: 36160252 PMCID: PMC9491836 DOI: 10.3389/fmicb.2022.997448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Heme is an essential cofactor for almost all living cells by acting as prosthetic group for various proteins or serving as alternative iron source. However, elevated levels are highly toxic for cells. Several corynebacterial species employ two paralogous, heme-responsive two-component systems (TCS), ChrSA and HrrSA, to cope with heme stress and to maintain intracellular heme homeostasis. Significant cross-talk at the level of phosphorylation between these systems was previously demonstrated. In this study, we have performed a laboratory evolution experiment to adapt Corynebacterium glutamicum to increasing heme levels. Isolated strains showed a highly increased tolerance to heme growing at concentrations of up to 100 μM. The strain featuring the highest heme tolerance harbored a frameshift mutation in the catalytical and ATPase-domain (CA-domain) of the chrS gene, converting it into a catalytically-inactive pseudokinase (ChrS_CA-fs). Reintroduction of the respective mutation in the parental C. glutamicum strain confirmed high heme tolerance and showed a drastic upregulation of hrtBA encoding a heme export system, conserved in Firmicutes and Actinobacteria. The strain encoding the ChrS pseudokinase variant showed significantly higher heme tolerance than a strain lacking chrS. Mutational analysis revealed that induction of hrtBA in the evolved strain is solely mediated via the cross-phosphorylation of the response regulator (RR) ChrA by the kinase HrrS and BACTH assays revealed the formation of heterodimers between HrrS and ChrS. Overall, our results emphasize an important role of the ChrS pseudokinase in high heme tolerance of the evolved C. glutamicum and demonstrate the promiscuity in heme-dependent signaling of the paralogous two-component systems facilitating fast adaptation to changing environmental conditions.
Collapse
|
38
|
Zhang D, Shen J, Peng X, Gao S, Wang Z, Zhang H, Sun W, Niu H, Ying H, Zhu C, Chen Y, Liu D. Physiological changes and growth behavior of Corynebacterium glutamicum cells in biofilm. Front Microbiol 2022; 13:983545. [PMID: 36110303 PMCID: PMC9468548 DOI: 10.3389/fmicb.2022.983545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilm cells are well-known for their increased survival and metabolic capabilities and have been increasingly implemented in industrial and biotechnological processes. Corynebacterium glutamicum is one of the most widely used microorganisms in the fermentation industry. However, C. glutamicum biofilm has been rarely reported and little is known about its cellular basis. Here, the physiological changes and characteristics of C. glutamicum biofilm cells during long-term fermentation were studied for the first time. Results showed that the biofilm cells maintained stable metabolic activity and cell size was enlarged after repeated-batch of fermentation. Cell division was slowed, and chromosome content and cell proliferation efficiency were reduced during long-term fermentation. Compared to free cells, more biofilm cells were stained by the apoptosis indicator dyes Annexin V-FITC and propidium iodide (PI). Overall, these results suggested slow-growing, long-lived cells of C. glutamicum biofilm during fermentation, which could have important industrial implications. This study presents first insights into the physiological changes and growth behavior of C. glutamicum biofilm cell population, which would be valuable for understanding and developing biofilm-based processes.
Collapse
Affiliation(s)
- Di Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Jiawen Shen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Xiwei Peng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Shansong Gao
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Zhenyu Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Huifang Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Wenjun Sun
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Huanqing Niu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| | - Hanjie Ying
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| | - Chenjie Zhu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| | - Yong Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| | - Dong Liu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
- *Correspondence: Dong Liu,
| |
Collapse
|
39
|
Kataoka N, Matsutani M, Matsushita K, Yakushi T. Stepwise metabolic engineering of Corynebacterium glutamicum for the production of phenylalanine. J GEN APPL MICROBIOL 2022. [PMID: 35989300 DOI: 10.2323/jgam.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Corynebacterium glutamicum was metabolically engineered to produce phenylalanine, a valuable aromatic amino acid that can be used as a raw material in the food and pharmaceutical industries. First, a starting phenylalanine-producer was constructed by overexpressing tryptophan-sensitive 3-deoxy-D-arabino-heptulosonate-7-phosphate synthase and phenylalanine- and tyrosine-insensitive bifunctional enzyme chorismate mutase prephenate dehydratase from Escherichia coli, followed by the inactivation of enzymes responsible for the formation of dihydroxyacetone and the consumption of shikimate pathway-related compounds. Second, redirection of the carbon flow from tyrosine to phenylalanine was attempted by deleting of the tyrA gene encoding prephenate dehydrogenase, which catalyzes the committed step for tyrosine biosynthesis from prephenate. However, suppressor mutants were generated, and two mutants were isolated and examined for phenylalanine production and genome sequencing. The suppressor mutant harboring an amino acid exchange (L180R) on RNase J, which was experimentally proven to lead to a loss of function of the enzyme, showed significantly enhanced production of phenylalanine. Finally, modifications of phosphoenolpyruvate-pyruvate metabolism were investigated, revealing that the inactivation of either phosphoenolpyruvate carboxylase or pyruvate carboxylase, which are enzymes of the anaplerotic pathway, is an effective means for improving phenylalanine production. The resultant strain, harboring a phosphoenolpyruvate carboxylase deficiency, synthesized 50.7 mM phenylalanine from 444 mM glucose. These results not only provided new insights into the practical mutations in constructing a phenylalanine-producing C. glutamicum but also demonstrated the creation of a potential strain for the biosynthesis of phenylalanine-derived compounds represented by plant secondary metabolites.
Collapse
Affiliation(s)
- Naoya Kataoka
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University
| | | | - Kazunobu Matsushita
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University
| | - Toshiharu Yakushi
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University
| |
Collapse
|
40
|
Müller C, Bakkes PJ, Lenz P, Waffenschmidt V, Helleckes LM, Jaeger KE, Wiechert W, Knapp A, Freudl R, Oldiges M. Accelerated strain construction and characterization of C. glutamicum protein secretion by laboratory automation. Appl Microbiol Biotechnol 2022; 106:4481-4497. [PMID: 35759036 PMCID: PMC9259529 DOI: 10.1007/s00253-022-12017-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/02/2022]
Abstract
Secretion of bacterial proteins into the culture medium simplifies downstream processing by avoiding cell disruption for target protein purification. However, a suitable signal peptide for efficient secretion needs to be identified, and currently, there are no tools available to predict optimal combinations of signal peptides and target proteins. The selection of such a combination is influenced by several factors, including protein biosynthesis efficiency and cultivation conditions, which both can have a significant impact on secretion performance. As a result, a large number of combinations must be tested. Therefore, we have developed automated workflows allowing for targeted strain construction and secretion screening using two platforms. Key advantages of this experimental setup include lowered hands-on time and increased throughput. In this study, the automated workflows were established for the heterologous production of Fusarium solani f. sp. pisi cutinase in Corynebacterium glutamicum. The target protein was monitored in culture supernatants via enzymatic activity and split GFP assay. Varying spacer lengths between the Shine-Dalgarno sequence and the start codon of Bacillus subtilis signal peptides were tested. Consistent with previous work on the secretory cutinase production in B. subtilis, a ribosome binding site with extended spacer length to up to 12 nt, which likely slows down translation initiation, does not necessarily lead to poorer cutinase secretion by C. glutamicum. The best performing signal peptides for cutinase secretion with a standard spacer length were identified in a signal peptide screening. Additional insights into the secretion process were gained by monitoring secretion stress using the C. glutamicum K9 biosensor strain. KEY POINTS: • Automated workflows for strain construction and screening of protein secretion • Comparison of spacer, signal peptide, and host combinations for cutinase secretion • Signal peptide screening for secretion by C. glutamicum using the split GFP assay.
Collapse
Affiliation(s)
- Carolin Müller
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, 52062, Aachen, Germany
| | - Patrick J Bakkes
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Patrick Lenz
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Vera Waffenschmidt
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Laura M Helleckes
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Institute of Biotechnology, RWTH Aachen University, 52062, Aachen, Germany
| | - Karl-Erich Jaeger
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Wolfgang Wiechert
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, 52062, Aachen, Germany
| | - Andreas Knapp
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich, 52425, Jülich, Germany.,Castrol Germany GmbH, 41179, Mönchengladbach, Germany
| | - Roland Freudl
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Marco Oldiges
- Institute of Bio- and Geosciences IBG-1, Biotechnology, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany. .,Institute of Biotechnology, RWTH Aachen University, 52062, Aachen, Germany.
| |
Collapse
|
41
|
Merkel M, Kiefer D, Schmollack M, Blombach B, Lilge L, Henkel M, Hausmann R. Acetate-based production of itaconic acid with Corynebacterium glutamicum using an integrated pH-coupled feeding control. BIORESOURCE TECHNOLOGY 2022; 351:126994. [PMID: 35288270 DOI: 10.1016/j.biortech.2022.126994] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
To date, most bio-based products of industrial biotechnology stem from sugar-based carbon sources originating from food and feed competing resources. Exemplary for bioproducts converted from glucose, the potential C5 platform chemical itaconic acid is presently produced by the filamentous fungus Aspergillus terreus. Here, an engineered strain of the industrial platform organism Corynebacterium glutamicum ATCC 13032 was used for acetate-based production of itaconic acid to overcome current production difficulties. For this purpose, C. glutamicum ICDR453C (pEKEx2-malEcadopt) with a mutated icd variant for reduced isocitrate dehydrogenase activity was constructed harbouring pEKEx2-malEcadopt, that includes a cis-aconitate dehydrogenase gene originating from A. terreus. Overall, a peak volumetric productivity of 1.01 gL-1h-1 was achieved resulting in an itaconate titer of 29.2 g/L, by using an integrated pH-coupled acetate feeding control in a fed-batch process without base titration. The results support the high potential of acetate as alternative substrate for bioproduction.
Collapse
Affiliation(s)
- Manuel Merkel
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, Fruwirthstrasse 12, 70599 Stuttgart, Germany
| | - Dirk Kiefer
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, Fruwirthstrasse 12, 70599 Stuttgart, Germany
| | - Marc Schmollack
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany
| | - Bastian Blombach
- Microbial Biotechnology, Campus Straubing for Biotechnology and Sustainability, Technical University of Munich, Straubing, Germany; SynBiofoundry@TUM, Technical University of Munich, Straubing, Germany
| | - Lars Lilge
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, Fruwirthstrasse 12, 70599 Stuttgart, Germany
| | - Marius Henkel
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, Fruwirthstrasse 12, 70599 Stuttgart, Germany.
| | - Rudolf Hausmann
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Bioprocess Engineering, Fruwirthstrasse 12, 70599 Stuttgart, Germany
| |
Collapse
|
42
|
Mhatre A, Shinde S, Jha AK, Rodriguez A, Wardak Z, Jansen A, Gladden JM, George A, Davis RW, Varman AM. Corynebacterium glutamicum as an Efficient Omnivorous Microbial Host for the Bioconversion of Lignocellulosic Biomass. Front Bioeng Biotechnol 2022; 10:827386. [PMID: 35433642 PMCID: PMC9011048 DOI: 10.3389/fbioe.2022.827386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/03/2022] [Indexed: 01/07/2023] Open
Abstract
Corynebacterium glutamicum has been successfully employed for the industrial production of amino acids and other bioproducts, partially due to its native ability to utilize a wide range of carbon substrates. We demonstrated C. glutamicum as an efficient microbial host for utilizing diverse carbon substrates present in biomass hydrolysates, such as glucose, arabinose, and xylose, in addition to its natural ability to assimilate lignin-derived aromatics. As a case study to demonstrate its bioproduction capabilities, L-lactate was chosen as the primary fermentation end product along with acetate and succinate. C. glutamicum was found to grow well in different aromatics (benzoic acid, cinnamic acid, vanillic acid, and p-coumaric acid) up to a concentration of 40 mM. Besides, 13C-fingerprinting confirmed that carbon from aromatics enter the primary metabolism via TCA cycle confirming the presence of β-ketoadipate pathway in C. glutamicum. 13C-fingerprinting in the presence of both glucose and aromatics also revealed coumarate to be the most preferred aromatic by C. glutamicum contributing 74 and 59% of its carbon for the synthesis of glutamate and aspartate respectively. 13C-fingerprinting also confirmed the activity of ortho-cleavage pathway, anaplerotic pathway, and cataplerotic pathways. Finally, the engineered C. glutamicum strain grew well in biomass hydrolysate containing pentose and hexose sugars and produced L-lactate at a concentration of 47.9 g/L and a yield of 0.639 g/g from sugars with simultaneous utilization of aromatics. Succinate and acetate co-products were produced at concentrations of 8.9 g/L and 3.2 g/L, respectively. Our findings open the door to valorize all the major carbon components of biomass hydrolysate by using C. glutamicum as a microbial host for biomanufacturing.
Collapse
Affiliation(s)
- Apurv Mhatre
- Chemical Engineering Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, United States
| | - Somnath Shinde
- Department of Bioresource and Environmental Security, Sandia National Laboratories, Livermore, CA, United States
| | - Amit Kumar Jha
- Chemical Engineering Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, United States,Department of Bioresource and Environmental Security, Sandia National Laboratories, Livermore, CA, United States
| | - Alberto Rodriguez
- Department of Biomaterials and Biomanufacturing, Sandia National Laboratories, Livermore, CA, United States,Joint BioEnergy Institute, Emeryville, CA, United States
| | - Zohal Wardak
- Department of Bioresource and Environmental Security, Sandia National Laboratories, Livermore, CA, United States
| | - Abigail Jansen
- Chemical Engineering Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, United States
| | - John M. Gladden
- Department of Biomaterials and Biomanufacturing, Sandia National Laboratories, Livermore, CA, United States,Joint BioEnergy Institute, Emeryville, CA, United States
| | - Anthe George
- Department of Bioresource and Environmental Security, Sandia National Laboratories, Livermore, CA, United States,Department of Biomaterials and Biomanufacturing, Sandia National Laboratories, Livermore, CA, United States
| | - Ryan W. Davis
- Department of Bioresource and Environmental Security, Sandia National Laboratories, Livermore, CA, United States,*Correspondence: Ryan W. Davis, ; Arul M. Varman,
| | - Arul M. Varman
- Chemical Engineering Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, United States,*Correspondence: Ryan W. Davis, ; Arul M. Varman,
| |
Collapse
|
43
|
Liu B, Sun X, Liu Y, Yang M, Wang L, Li Y, Wang J. Increased NADPH Supply Enhances Glycolysis Metabolic Flux and L-methionine Production in Corynebacterium glutamicum. Foods 2022; 11:foods11071031. [PMID: 35407118 PMCID: PMC8998051 DOI: 10.3390/foods11071031] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/19/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Corynebacterium glutamicum is an important strain for the industrial production of amino acids, but the fermentation of L-methionine has not been realized. The purpose of this study is to clarify the effect of reducing power NADPH on L-methionine synthesis. Site-directed mutagenesis of zwf and gnd genes in pentose phosphate pathway relieved feedback inhibition, increased NADPH supply by 151.8%, and increased L-methionine production by 28.3%; Heterologous expression of gapC gene to introduce NADP+ dependent glyceraldehyde-3-phosphate dehydrogenase increased NADPH supply by 75.0% and L-methionine production by 48.7%; Heterologous expression of pntAB gene to introduce membrane-integral nicotinamide nucleotide transhydrogenase increased NADPH by 89.2% and L-methionine production by 35.9%. Finally, the engineering strain YM6 with a high NADPH supply was constructed, which increased the NADPH supply by 348.2% and the L-methionine production by 64.1%. The analysis of metabolic flux showed that YM6 significantly increased the glycolytic flux, including the metabolic flux of metabolites such as glycosyldehyde-3-phosphate, dihydroxyacetate phosphate, 3-phosphoglycate and pyruvate, and the significant increase of L-methionine flux also confirmed the increase of its synthesis. This study provides a research basis for the systematic metabolic engineering construction of L-methionine high-yield engineering strains.
Collapse
Affiliation(s)
- Bingnan Liu
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
| | - Xinyu Sun
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
| | - Yue Liu
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
| | - Mengmeng Yang
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
| | - Liang Wang
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
| | - Ying Li
- School of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
- Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Dalian 116600, China
- Correspondence: (Y.L.); (J.W.); Tel.: +86-411-863-24050 (Y.L.); +86-769-222-61545 (J.W.)
| | - Jihui Wang
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China; (B.L.); (X.S.); (Y.L.); (M.Y.); (L.W.)
- Engineering Research Center of Health Food Design & Nutrition Regulation, School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China
- Correspondence: (Y.L.); (J.W.); Tel.: +86-411-863-24050 (Y.L.); +86-769-222-61545 (J.W.)
| |
Collapse
|
44
|
Carbohydrate-binding module of cycloisomaltooligosaccharide glucanotransferase from Thermoanaerobacter thermocopriae improves its cyclodextran production. Enzyme Microb Technol 2022; 157:110023. [DOI: 10.1016/j.enzmictec.2022.110023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 11/23/2022]
|
45
|
Blumenstein J, Rädisch R, Štěpánek V, Grulich M, Dostálová H, Pátek M. Identification of Rhodococcus erythropolis Promoters Controlled by Alternative Sigma Factors Using In Vivo and In Vitro Systems and Heterologous RNA Polymerase. Curr Microbiol 2022; 79:55. [PMID: 34982253 DOI: 10.1007/s00284-021-02747-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/10/2021] [Indexed: 11/03/2022]
Abstract
Rhodococcus erythropolis CCM2595 is a bacterial strain, which has been studied for its capability to degrade phenol and other toxic aromatic compounds. Its cell wall contains mycolic acids, which are also an attribute of other bacteria of the Mycolata group, such as Corynebacterium and Mycobacterium species. We suppose that many genes upregulated by phenol stress in R. erythropolis are controlled by the alternative sigma factors of RNA polymerase, which are active in response to the cell envelope or oxidative stress. We developed in vitro and in vivo assays to examine the connection between the stress sigma factors and genes activated by various extreme conditions, e.g., heat, cell surface, and oxidative stress. These assays are based on the procedures of such tests carried out in the related species, Corynebacterium glutamicum. We showed that the R. erythropolis CCM2595 genes frmB1 and frmB2, which encode S-formylglutathione hydrolases (named corynomycolyl transferases in C. glutamicum), are controlled by SigD, just like the homologous genes cmt1 and cmt2 in C. glutamicum. The new protocol of the in vivo and in vitro assays will enable us to classify R. erythropolis promoters according to their connection to sigma factors and to assign the genes to the corresponding sigma regulons. The complex stress responses, such as that induced by phenol, could, thus, be analyzed with respect to the gene regulation by sigma factors.
Collapse
Affiliation(s)
- Jan Blumenstein
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic.,Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Robert Rädisch
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic.,Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Václav Štěpánek
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | - Michal Grulich
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | - Hana Dostálová
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | - Miroslav Pátek
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic.
| |
Collapse
|
46
|
Henke NA, Göttl VL, Schmitt I, Peters-Wendisch P, Wendisch VF. A synthetic biology approach to study carotenoid production in Corynebacterium glutamicum: Read-out by a genetically encoded biosensor combined with perturbing native gene expression by CRISPRi. Methods Enzymol 2022; 671:383-419. [DOI: 10.1016/bs.mie.2021.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
47
|
Li CL, Ruan HZ, Liu LM, Zhang WG, Xu JZ. Rational reformation of Corynebacterium glutamicum for producing L-lysine by one-step fermentation from raw corn starch. Appl Microbiol Biotechnol 2021; 106:145-160. [PMID: 34870736 DOI: 10.1007/s00253-021-11714-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 11/27/2022]
Abstract
This article focuses on engineering Corynebacterium glutamicum to produce L-lysine efficiently from starch using combined method of "classical breeding" and "genome breeding." Firstly, a thermo-tolerable L-lysine-producing C. glutamicum strain KT45-6 was obtained after multi-round of acclimatization at high temperature. Then, amylolytic enzymes were introduced into strain KT45-6, and the resultant strains could use starch for cell growth and L-lysine production except the strain with expression of isoamylase. In addition, co-expression of amylolytic enzymes showed a good performance in starch degradation, cell growth and L-lysine production, especially co-expression of α-amylase (AA) and glucoamylase (GA). Moreover, L-lysine yield was increased by introducing AA-GA fusion protein (i.e., strain KT45-6S-5), and finally reached to 23.9 ± 2.3 g/L in CgXIIIPM-medium. It is the first report of an engineered L-lysine-producing strain with maximum starch utilization that may be used as workhorse for producing amino acid using starch as the main feedstock. KEY POINTS: • Thermo-tolerable C. glutamicum was obtained by temperature-induced adaptive evolution. • The fusion order between AA and GA affects the utilization efficiency of starch. • C. glutamicum with starch utilization was constructed by optimizing amylases expression.
Collapse
Affiliation(s)
- Chang-Long Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China
| | - Hao-Zhe Ruan
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China
| | - Li-Ming Liu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China.,State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China
| | - Wei-Guo Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China
| | - Jian-Zhong Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, WuXi, 214122, People's Republic of China.
| |
Collapse
|
48
|
Wang JY, Rao ZM, Xu JZ, Zhang WG. Enhancing β-alanine production from glucose in genetically modified Corynebacterium glutamicum by metabolic pathway engineering. Appl Microbiol Biotechnol 2021; 105:9153-9166. [PMID: 34837493 DOI: 10.1007/s00253-021-11696-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
Abstract
To directly produce β-alanine from glucose by microbial fermentation, a recombinant Corynebacterium glutamicum strain with high efficiency of β-alanine production was constructed in this study. To do this, the biosynthetic pathway of β-alanine in an L-lysine-producing strain XQ-5 was modified by enhancing carbon flux in biosynthetic pathway and limiting carbon flux in competitive pathway. This study showed that replacement of L-aspartate kinase (AK) with wild-type AK and disruption of lactate dehydrogenase and alanine/valine aminotransferases increase β-alanine production because of decreasing the by-products accumulation. Moreover, L-aspartate-α-decarboxylase (ADC) from Bacillus subtilis was designed as the best enzyme for increasing β-alanine production, and its variant (BsADCE56S/I88M) showed the highest activity for catalyzing L-aspartate to generate β-alanine. To further increase β-alanine production, expression level of BsADCE56S/I88M was controlled by optimizing promoter and RBS, indicating that Pgro plus ThirRBS is the best combination for BsADCE56S/I88M expression and β-alanine production. The resultant strain XQ-5.5 produced 30.7 ± 2.3 g/L of β-alanine with a low accumulation of lactate (from 5.2 ± 0.14 to 0.2 ± 0.09 g/L) and L-alanine (from 7.6 ± 0.22 to 3.8 ± 0. 32 g/L) in shake-flask fermentation and produced 56.5 ± 3.2 g/L of β-alanine with a productivity of 0.79 g/(L·h) and the glucose conversion efficiency (α) of 39.5% in feed-batch fermentation. This is the first report of genetically modifying the biosynthetic pathway of β-alanine that improves the efficiency of β-alanine production in an L-lysine-producing strain, and these results give us a new insight for constructing the other valuable biochemical. KEY POINTS: • Optimization and overexpression of the key enzyme BsADC increased the accumulation of β-alanine. • The AK was replaced with wild-type AK to increase the conversion of aspartic acid to β-alanine. • A 56.5-g/L β-alanine production in fed-batch fermentation was achieved.
Collapse
Affiliation(s)
- Jin-Yu Wang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, Wuxi, 214122, People's Republic of China
| | - Zhi-Ming Rao
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, Wuxi, 214122, People's Republic of China.,National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800# Lihu Road, Wuxi, 214122, People's Republic of China
| | - Jian-Zhong Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, Wuxi, 214122, People's Republic of China.
| | - Wei-Guo Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800# Lihu Road, Wuxi, 214122, People's Republic of China.
| |
Collapse
|
49
|
MmpA, a Conserved Membrane Protein Required for Efficient Surface Transport of Trehalose Lipids in Corynebacterineae. Biomolecules 2021; 11:biom11121760. [PMID: 34944401 PMCID: PMC8698533 DOI: 10.3390/biom11121760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/23/2022] Open
Abstract
Cell walls of bacteria of the genera Mycobacterium and Corynebacterium contain high levels of (coryno)mycolic acids. These very long chain fatty acids are synthesized on the cytoplasmic leaflet of the inner membrane (IM) prior to conjugation to the disaccharide, trehalose, and transport to the periplasm. Recent studies on Corynebacterium glutamicum have shown that acetylation of trehalose monohydroxycorynomycolate (hTMCM) promotes its transport across the inner membrane. Acetylation is mediated by the membrane acetyltransferase, TmaT, and is dependent on the presence of a putative methyltransferase, MtrP. Here, we identify a third protein that is required for the acetylation and membrane transport of hTMCM. Deletion of the C. glutamicum gene NCgl2761 (Rv0226c in Mycobacterium tuberculosis) abolished synthesis of acetylated hTMCM (AcTMCM), resulting in an accumulation of hTMCM in the inner membrane and reduced synthesis of trehalose dihydroxycorynomycolate (h2TDCM), a major outer membrane glycolipid. Complementation with the NCgl2761 gene, designated here as mmpA, restored the hTMCM:h2TDCM ratio. Comprehensive lipidomic analysis of the ΔtmaT, ΔmtrP and ΔmmpA mutants revealed strikingly similar global changes in overall membrane lipid composition. Our findings suggest that the acetylation and membrane transport of hTMCM is regulated by multiple proteins: MmpA, MtrP and TmaT, and that defects in this process lead to global, potentially compensatory changes in the composition of inner and outer membranes.
Collapse
|
50
|
He P, Li S, Xu S, Fan H, Wang Y, Zhou W, Fu G, Han G, Wang YY, Zheng SJ. Monitoring Tritrophic Biocontrol Interactions Between Bacillus spp., Fusarium oxysporum f. sp. cubense, Tropical Race 4, and Banana Plants in vivo Based on Fluorescent Transformation System. Front Microbiol 2021; 12:754918. [PMID: 34721361 PMCID: PMC8550332 DOI: 10.3389/fmicb.2021.754918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/22/2021] [Indexed: 11/27/2022] Open
Abstract
Bacillus spp. is effective biocontrol agents for Fusarium wilt of banana (FWB), tropical race 4 (TR4). This study explores the colonization by Bacillus subtilis, Bacillus velezensis, and Bacillus amyloliquefaciens of host banana plants and elucidates the mechanism of antagonistic TR4 biocontrol. The authors selected one B. subtilis strain, three B. velezensis strains, and three B. amyloliquefaciens strains that are proven to significantly inhibit TR4 in vitro, optimized the genetic transformation conditions and explored their colonization process in banana plants. The results showed that we successfully constructed an optimized fluorescent electro-transformation system (OD600 of bacteria concentration=0.7, plasmid concentration=50ng/μl, plasmid volume=2μl, transformation voltage=1.8kV, and transformation capacitance=400Ω) of TR4-inhibitory Bacillus spp. strains. The red fluorescent protein (RFP)-labeled strains were shown to have high stability with a plasmid-retention frequency above 98%, where bacterial growth rates and TR4 inhibition are unaffected by fluorescent plasmid insertion. In vivo colonizing observation by Laser Scanning Confocal Microscopy (LSCM) and Scanning Electron Microscopy (SEM) showed that Bacillus spp. can colonize the internal cells of banana plantlets roots. Further, fluorescent observation by LSCM showed these RFP-labeled bacteria exhibit chemotaxis (chemotaxis ratio was 1.85±0.04) toward green fluorescent protein (GFP)-labeled TR4 hyphae in banana plants. We conclude that B. subtilis, B. velezensis, and B. amyloliquefaciens can successfully colonize banana plants and interact with TR4. Monitoring its dynamic interaction with TR4 and its biocontrol mechanism is under further study.
Collapse
Affiliation(s)
- Ping He
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Ministry of Education Key Laboratory of Agriculture Biodiversity for Plant Disease Management, College of Plant Protection, Yunnan Agricultural University, Kunming, China.,Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Shu Li
- Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Shengtao Xu
- Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Huacai Fan
- Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Yongfen Wang
- Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China.,Institute of Tropical and Subtropical Industry Crops, Yunnan Academy of Agricultural Sciences, Baoshan, China
| | - Wei Zhou
- Biotechnology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China
| | - Gang Fu
- Institute of Plant Protection, Guangxi Academy of Agricultural Sciences, Nanning, China
| | - Guangyu Han
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Ministry of Education Key Laboratory of Agriculture Biodiversity for Plant Disease Management, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Yun-Yue Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Ministry of Education Key Laboratory of Agriculture Biodiversity for Plant Disease Management, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Si-Jun Zheng
- Yunnan Key Laboratory of Green Prevention and Control of Agricultural Transboundary Pests, Agricultural Environment and Resources Institute, Yunnan Academy of Agricultural Sciences, Kunming, China.,Bioversity International, Kunming, China
| |
Collapse
|