1
|
Barreto JA, Lacôrte E Silva MVM, Marin DC, Brienzo M, Jacobus AP, Contiero J, Gross J. Engineering adaptive alleles for Escherichia coli growth on sucrose using the EasyGuide CRISPR system. J Biotechnol 2025; 403:126-139. [PMID: 40252733 DOI: 10.1016/j.jbiotec.2025.04.016] [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: 12/25/2024] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Adaptive Laboratory Evolution (ALE) is a powerful approach for mining genetic data to engineer industrial microorganisms. This evolution-informed design requires robust genetic tools to incorporate the discovered alleles into target strains. Here, we introduce the EasyGuide CRISPR, a five-plasmid platform that exploits E. coli's natural recombination system to assemble gRNA plasmids from overlapping PCR fragments. The production of gRNAs and donor DNA is further facilitated by using recombination cassettes generated through PCR with 40-60-mer oligos. With the new CRISPR toolkit, we constructed 22 gene edits in E. coli DH5α, most of which corresponded to alleles mapped in E. coli DH5α and E2348/69 ALE populations selected for sucrose propagation. For DH5α ALE, sucrose consumption was supported by the cscBKA operon expression from a high-copy plasmid. During ALE, plasmid integration into the chromosome, or its copy number reduction due to the pcnB deletion, conferred a 30-35 % fitness gain, as demonstrated by CRISPR-engineered strains. A ∼5 % advantage was also associated with a ∼40.4 kb deletion involving fli operons for flagella assembly. In E2348/69 ALE, inactivation of the hfl system suggested selection pressures for maintaining λ-prophage dormancy (lysogeny). We further enhanced our CRISPR toolkit using yeast for in vivo assembly of donors and expression cassettes, enabling the establishment of polyhydroxybutyrate synthesis from sucrose. Overall, our study highlights the importance of combining ALE with streamlined CRISPR-mediated allele editing to advance microbial production using cost-effective carbon sources.
Collapse
Affiliation(s)
- Joneclei Alves Barreto
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil
| | - Matheus Victor Maso Lacôrte E Silva
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Rio Claro, SP 13506-900, Brazil
| | - Danieli Canaver Marin
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil
| | - Michel Brienzo
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil
| | - Ana Paula Jacobus
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil
| | - Jonas Contiero
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Rio Claro, SP 13506-900, Brazil
| | - Jeferson Gross
- Sao Paulo State University (Unesp), Institute for Research in Bioenergy, Rio Claro, SP 13500-230, Brazil; PhD Program in Bioenegy, São Paulo State University (Unesp), Rio Claro 13500-230, Brazil.
| |
Collapse
|
2
|
Cheng T, Cao X, Wang Y, Zhao L. Development of CRISPR-Cas9-based genome editing tools for non-model microorganism Erwinia persicina. Synth Syst Biotechnol 2025; 10:555-563. [PMID: 40092160 PMCID: PMC11909718 DOI: 10.1016/j.synbio.2025.02.006] [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: 11/20/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Erwinia persicina is a bacterium that has been known to produce secondary metabolites, such as andrimid, pink pigment, and exopolysaccharides, and to infect more than twenty plant species. However, traditional gene manipulation methods have been hindered by the inefficient of suicide plasmid-mediated genome editing. In this study, we describe the successful application of the CRISPR-Cas9 system in E. persicina. Efficient genome editing was achieved by substituting the native gRNA promoter with J23119 in a single-plasmid system (pRed_Cas9_ΔpoxB) and optimizing the gRNA design. The use of double gRNAs led to the deletion of a 42 kb genomic fragment, and the incorporation of a sacB screening marker facilitated iterative knockouts. Additionally, a 22 kb plasmid containing a self-resistance gene was conjugally transferred into E. persicina, resulting in the insertion of a 6.4 kb fragment with 100 % efficiency. Furthermore, we demonstrated the expression of shinorine, an anti-UV compound, within the E. persicina chassis. This study establishes E. persicina as a promising chassis for synthetic biology and provides a model for gene-editing systems in non-model microorganisms.
Collapse
Affiliation(s)
- Tingfeng Cheng
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xinyan Cao
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, PR China
| | - Yuchen Wang
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China
- College of Biological Sciences, China Agricultural University, Beijing, 100091, PR China
| | - Lei Zhao
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, PR China
| |
Collapse
|
3
|
Ebrecht AC, Luelf UJ, Govender K, Opperman DJ, Urlacher VB, Smit MS. Use of Whole Cells and Cell-Free Extracts of Catalase-Deficient E. coli for Peroxygenase-Catalyzed Reactions. Biotechnol Bioeng 2025; 122:1376-1385. [PMID: 40007105 PMCID: PMC12067033 DOI: 10.1002/bit.28959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025]
Abstract
Unspecific peroxygenases (UPOs) and cytochrome P450 monooxygenases (CYPs) with peroxygenase activity are becoming the preferred biocatalysts for oxyfunctionalization reactions. While whole cells (WCs) or cell-free extracts (CFEs) of Escherichia coli are often preferred for cofactor-dependent monooxygenase reactions, hydrogen peroxide (H2O2) driven peroxygenase reactions are generally performed with purified enzymes, because the catalases produced by E. coli are expected to quickly degrade H2O2. We used the CRISPR/Cas system to delete the catalase encoding chromosomal genes, katG, and katE, from E. coli BL21-Gold(DE3) to obtain a catalase-deficient strain. A short UPO, DcaUPO, and two CYP peroxygenases, SscaCYP_E284A and CYP102A1_21B3, were used to compare the strains for peroxygenase expression and subsequent sulfoxidation, epoxidation, and benzylic hydroxylation activity. While 10 mM H2O2 was depleted within 10 min after addition to WCs and CFEs of the wild-type strain, at least 60% remained after 24 h in WCs and CFEs of the catalase-deficient strain. CYP peroxygenase reactions, with generally lower turnover frequencies, benefited the most from the use of the catalase-deficient strain. Comparison of purified peroxygenases in buffer versus CFEs of the catalase-deficient strain revealed that the peroxygenases in CFEs generally performed as well as the purified proteins. We also used WCs from catalase-deficient E. coli to screen three CYP peroxygenases, wild-type SscaCYP, SscaCYP_E284A, and SscaCYP_E284I for activity against 10 substrates comparing H2O2 consumption with substrate consumption and product formation. Finally, the enzyme-substrate pair with highest activity, SscaCYP_E284I, and trans-β-methylstyrene, were used in a preparative scale reaction with catalase-deficient WCs. Use of WCs or CFEs from catalase-deficient E. coli instead of purified enzymes can greatly benefit the high-throughput screening of enzyme or substrate libraries for peroxygenase activity, while they can also be used for preparative scale reactions.
Collapse
Affiliation(s)
- Ana C. Ebrecht
- Department of Microbiology and BiochemistryUniversity of the Free StateBloemfonteinSouth Africa
| | - U. Joost Luelf
- Institute of BiochemistryHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Kamini Govender
- Department of Microbiology and BiochemistryUniversity of the Free StateBloemfonteinSouth Africa
| | - Diederik J. Opperman
- Department of Microbiology and BiochemistryUniversity of the Free StateBloemfonteinSouth Africa
| | - Vlada B. Urlacher
- Institute of BiochemistryHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Martha S. Smit
- Department of Microbiology and BiochemistryUniversity of the Free StateBloemfonteinSouth Africa
| |
Collapse
|
4
|
Yang S, Guo CH, Tong WY, Liu XY, Li JC, Kang M. Identification and characterization of anaerobically activated promoters in Escherichia coli. J Biotechnol 2025; 402:30-38. [PMID: 40049517 DOI: 10.1016/j.jbiotec.2025.03.002] [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: 12/28/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Anaerobically activated promoters in Escherichia coli play crucial roles in transcriptional regulation during cellular responses to decreased oxygen concentrations and serve as essential tools for implementing dynamic regulation in metabolic engineering. These promoters exhibit transcriptional activity only under low-oxygen or anaerobic conditions. To discover novel anaerobically activated promoters, this study selected 11 native promoters from E. coli databases and characterized their activities using flow cytometry. Subsequently, we optimized the key elements of these promoters and re-evaluated their activities to investigate the impact of functional elements on promoter performance. Furthermore, we verified the regulatory mechanisms of these promoters by knocking out host regulatory genes. Finally, we characterized the promoters' responsiveness to aerobic-anaerobic transitions by rapidly switching cultivation environments during host growth. This study identified several novel anaerobically activated promoters and comprehensively characterized their performance and features from multiple aspects. The identified promoters provide new tools for oxygen-limited or anaerobic production in metabolic engineering, while the findings from promoter element optimization offer valuable references for the design of anaerobically activated promoters.
Collapse
Affiliation(s)
- Sen Yang
- College of Life Science, Hebei University, Baoding, Hebei 071002, China
| | - Chao-Hao Guo
- College of Life Science, Hebei University, Baoding, Hebei 071002, China
| | - Wen-Yue Tong
- College of Life Science, Hebei University, Baoding, Hebei 071002, China
| | - Xiao-Yun Liu
- College of Life Science, Hebei University, Baoding, Hebei 071002, China
| | - Jing-Chen Li
- College of Life Science, Hebei University, Baoding, Hebei 071002, China.
| | - Ming Kang
- College of Life Science, Hebei University, Baoding, Hebei 071002, China; Innovation Center for Bioengineering and Biotechnology, Hebei University, Baoding, Hebei 071002, China.
| |
Collapse
|
5
|
Xiong B, Yang T, Zhang Z, Li X, Yu H, Wang L, You Z, Peng W, Jin L, Song H. Metabolic reprogramming and machine learning-guided cofactor engineering to boost nicotinamide mononucleotide production in Escherichia coli. BIORESOURCE TECHNOLOGY 2025; 426:132350. [PMID: 40054751 DOI: 10.1016/j.biortech.2025.132350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Nicotinamide mononucleotide (NMN) is a bioactive compound in NAD(P)+ metabolism, which exhibits diverse pharmaceutical interests. However, enhancing NMN biosynthesis faces the challange of competing with cell growth and disturbing intracellular redox homeostasis. Herein, we boosted NMN production in Escherichia coli by reprogramming central carbon metabolism with a machine learning (ML)-guided cofactor engineering strategy. Engnieering NMN biosynthesis-related pathway directed carbon flux toward NMN with the NADPH level increased by 73 %, which, although enhanced NMN titer (2.45 g/L), impaired cell growth. A quorum sensing (QS)-controlled cofactor engineering system was thus contructed and optimized by ML models to address redox imbalance, which led to 3.04 g/L NMN with improved cell growth. The final strain S344 produced 20.13 g/L NMN in fed-batch fermentation. This study showed that perturbation on cofactor level is a crucial limiting factor for NMN biosynthesis, and proposed a novel ML-guided strategy to manipulate intracellular redox state for efficient NMN production.
Collapse
Affiliation(s)
- Bo Xiong
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Tianrui Yang
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Zixiong Zhang
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiang Li
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Huan Yu
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Lian Wang
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Zixuan You
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Wenbin Peng
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Luyu Jin
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Song
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| |
Collapse
|
6
|
Zhang Z, Liu P, Zhang B, Shen J, Wu J, Huang S, Chu X. De novo Biosynthesis of Caffeic Acid and Chlorogenic Acid in Escherichia coli via Enzyme Engineering and Pathway Engineering. ACS Synth Biol 2025; 14:1581-1593. [PMID: 40232288 DOI: 10.1021/acssynbio.4c00850] [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] [Indexed: 04/16/2025]
Abstract
Caffeic acid (CA) and chlorogenic acid (CGA) have diverse health benefits, including hemostatic, antioxidant, and antiinflammatory, highlighting their potential for medical applications. However, the absence of high-performance production strains increases production costs, limiting their wider application. In this study, we engineered Escherichia coli for the de novo production of CA and CGA. To improve production, a highly efficient mutant tyrosine ammonia-lyase from Rhodotorula taiwanensis (RtTALT415M/Y458F) was identified using genome mining and protein engineering. By engineering the tyrosine biosynthetic pathway through the deletion of pheA and tyrR, along with the overexpression of aroGfbr and tyrAfbr, we developed an engineered E. coli strain, CA11, which produced 6.36 g/L of CA with a yield of 0.06 g/g glucose and a productivity of 0.18 g/L/h. This represents the highest titer reported for microbial synthesis of CA using glucose as the sole carbon source in E. coli. Based on strain CA11, we further developed strain CGA13, with optimized replicons, promoters, and ribosome-binding sites, which produced 1.53 g/L of CGA in fed-batch fermentation, highlighting its potential for industrial-scale production.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| | - Pengfu Liu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| | - Bin Zhang
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jian Shen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| | - Jiequn Wu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| | - Shusheng Huang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| | - Xiaohe Chu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- ZhejiangYangtze River Delta Bio-Pharmaceutical Industry Technology Research Park, Deqing 313200, P. R. China
| |
Collapse
|
7
|
Wang Z, Cui J, Liu Q, Li J, Guo C, Liu Y, Jiang C, Xue H, Li J, Ke Y, Qi H. Self-authenticating genomic materials in Escherichia coli via advanced genome signatures. Commun Biol 2025; 8:762. [PMID: 40379889 PMCID: PMC12084327 DOI: 10.1038/s42003-025-08171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
The authenticity and integrity of synthetic genomic materials containing valuable intellectual property are essential for advancing scientific knowledge and enhancing biosafety. Nevertheless, existing DNA tags and watermarks have limited efficacy due to low mutation tolerance and inadequate digital encoding capacity. Here, we present "Genome Signature", a biochemically stable and tamper-resistant DNA labeling system that enables the creation of self-authenticating genomes. Central to this system is a Golomb-ruler-derived Genome-Comb, which efficiently maps extensive nucleotide sequences onto limited codons within endogenous genes, significantly improving error correction and data encoding across millions of nucleotides. Using our labeling system, we successfully recorded a 4.5-million-nucleotide genome in living E. coli. The Genome Signature effectively encodes data within codon orders, and autonomously identifies and corrects mutations in our computing test, ensuring genome integrity and authenticity. Furthermore, it allows precise tracking of coded sequences across different cells, potentially advancing the development of reliable genomic materials in synthetic biology.
Collapse
Affiliation(s)
- Zhaoguan Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Jingsong Cui
- School of Cyber Science and Engineering, Wuhan University, Wuhan, China.
| | - Qian Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Jiawei Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
- School of Cyber Science and Engineering, Wuhan University, Wuhan, China
| | - Chi Guo
- Artificial Intelligence Institute, Wuhan University, Wuhan, China
- Yangtze River Delta Technology Innovation Center of Wuhan University, Jiangyin, China
| | - Yiyang Liu
- School of Cyber Science and Engineering, Wuhan University, Wuhan, China
| | - Changyue Jiang
- School of Cyber Science and Engineering, Wuhan University, Wuhan, China
| | - Hui Xue
- School of Cyber Science and Engineering, Wuhan University, Wuhan, China
| | - Jiaojiao Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China.
| |
Collapse
|
8
|
Liu Q, Wang Z, Cui J, Li J, Jiang C, Tan G, Qi H. Bacterial Cells Engineered with Synthetic Genetic Materials for Blind Testing of Random Mutagenesis. ACS Synth Biol 2025. [PMID: 40354669 DOI: 10.1021/acssynbio.5c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Synthetic genetic materials, particularly those in genetically modified organisms (GMOs) deployed into complex environments, necessitate robust postmarket surveillance for continuous monitoring of both the materials and their applications throughout their lifecycle. Here, we introduce novel-coded genomic material for a blind mutation test that evaluates mutagenesis in synthetic genomic sequences without requiring direct sequence comparison. This test utilizes a Genome-Digest, which is embedded within essential genes, establishing mathematical correlation between the nucleotide sequence and codon order. This novel design allows for independent assessment of mutations by decoding the nucleotide sequence, thereby eliminating the need for reference sequences or extensive bioinformatic analysis. Furthermore, the test has the capability to analyze mixed genomic materials from a single sample and can be extended to the pooled testing of multiple samples as well. Building on this framework, we propose the 'Genome-ShockWatch' methodology. In proof-of-concept trials, it successfully detected mutations that exceeded a predefined threshold in long-read sequencing data from a yogurt sample containing Genome-Digest encoded Nissle 1917 E. coli cells and naturally occurring probiotic bacteria. Consequently, the Genome-Digest system provides a robust foundation for the routine surveillance and management of GMOs and related synthetic products, ensuring their safety and efficacy in diverse environmental contexts.
Collapse
Affiliation(s)
- Qian Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300354, China
| | - Zhaoguan Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300354, China
| | - Jingsong Cui
- School of Cyber Science and Engineering, Wuhan University, Wuhan 430072, China
| | - Jiawei Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300354, China
| | - Changyue Jiang
- School of Cyber Science and Engineering, Wuhan University, Wuhan 430072, China
| | - Gaoxu Tan
- School of Cyber Science and Engineering, Wuhan University, Wuhan 430072, China
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300354, China
| |
Collapse
|
9
|
Sun M, Gao J, Tang H, Wang H, Zhou L, Song C, Tian Y, Li Q. D-CAPS: an efficient CRISPR-Cas9-based phage defense system for E. coli. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40289704 DOI: 10.3724/abbs.2024208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Escherichia coli is widely used in industrial chemical synthesis but faces significant challenges due to bacteriophage contamination, which reduces product quality and yield. Therefore, developing an efficient antiphage system is essential. In this study, we develop a CRISPR-Cas9-based antiphage system (CAPS) targeting essential genes of the T7 phage (gene 5 and gene 19) with single gRNAs transformed into MG1655 strains expressing Cas9. While CAPS provides limited resistance, with plating efficiencies ranging from 10 -5 to 10 -1, further optimization is needed. To enhance efficacy, we design a double-site-targeting CRISPR-Cas9-based antiphage system (D-CAPS). D-CAPS demonstrates complete resistance, with no plaques observed even at a high multiplicity of infection (MOI of 2), and growth curve analysis reveals that antiphage E. coli strains grow normally, similar to the wild-type strain, even at a high multiplicity of infection. Furthermore, D-CAPS is effective against BL21(DE3) strains, showing strong resistance and demonstrating its versatility across different E . coli strains. Protein expression analysis via green fluorescent protein confirms that E. coli carrying D-CAPS could maintain normal protein expression levels even in the presence of phages, comparable to wild-type strains. Overall, D-CAPS offers a robust and versatile approach to enhancing E. coli resistance to phages, providing a practical solution for protecting industrial E. coli strains and improving fermentation processes.
Collapse
Affiliation(s)
- Mingjun Sun
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065 China
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jie Gao
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Hongjie Tang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Hengyi Wang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Liyan Zhou
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Chuan Song
- Luzhou Laojiao Co., Ltd., Luzhou 646000, China
- National Engineering Research Center of Solid-State Brewing, Luzhou 646000, China
| | - Yongqiang Tian
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065 China
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Qi Li
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| |
Collapse
|
10
|
Kang X, Wang Y, Liang Q, Luo W. Enhancing Ergothioneine Production by Combined Protein and Metabolic Engineering Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9234-9245. [PMID: 40181711 DOI: 10.1021/acs.jafc.5c01267] [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: 04/05/2025]
Abstract
Ergothioneine (ERG), a sulfur-containing histidine derivative recognized for its high stability, is of significant value across multiple sectors, including food, cosmetics, and medicine. In comparison to chemical synthesis, the establishment of microbial cell factories for ERG production represents a more efficient, environmentally friendly, and sustainable strategy. In this study, we achieved de novo synthesis of ERG in Escherichia coli by introducing genes from Trichoderma reesei. Protein engineering was subsequently employed to enable the soluble expression of the key genes Tr1 and Tr2, which resulted in a 198.1% increase in ERG production. Furthermore, strain modifications, including the knockout of competing pathways and optimization of key gene copies, were used to enhance ERG production. Following strategic combinations and medium optimization, strain E25 produced 430.9 mg/L ERG in an Erlenmeyer flask and 2331.58 mg/L via fed-batch fermentation in a 5 L bioreactor. This study not only establishes a solid foundation for the efficient and sustainable scale-up production of ERG and its derivatives but also provides valuable insights and references for its industrial production.
Collapse
Affiliation(s)
- Xiyue Kang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yi Wang
- Department of Biological and Agricultural Engineering, University of California, Davis, 1 Shields Ave, Davis, California 95616, United States
| | - Quanfeng Liang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Wei Luo
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| |
Collapse
|
11
|
Chang Z, Guo X, Li X, Wang Y, Zang Z, Pei S, Lu W, Li Y, Huang JD, Xiao Y, Liu C. Bacterial immunotherapy leveraging IL-10R hysteresis for both phagocytosis evasion and tumor immunity revitalization. Cell 2025; 188:1842-1857.e20. [PMID: 40037354 DOI: 10.1016/j.cell.2025.02.002] [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: 04/26/2024] [Revised: 12/07/2024] [Accepted: 02/05/2025] [Indexed: 03/06/2025]
Abstract
Bacterial immunotherapy holds promising cancer-fighting potential. However, unlocking its power requires a mechanistic understanding of how bacteria both evade antimicrobial immune defenses and stimulate anti-tumor immune responses within the tumor microenvironment (TME). Here, by harnessing an engineered Salmonella enterica strain with this dual proficiency, we unveil an underlying singular mechanism. Specifically, the hysteretic nonlinearity of interleukin-10 receptor (IL-10R) expression drives tumor-infiltrated immune cells into a tumor-specific IL-10Rhi state. Bacteria leverage this to enhance tumor-associated macrophages producing IL-10, evade phagocytosis by tumor-associated neutrophils, and coincidently expand and stimulate the preexisting exhausted tumor-resident CD8+ T cells. This effective combination eliminates tumors, prevents recurrence, and inhibits metastasis across multiple tumor types. Analysis of human samples suggests that the IL-10Rhi state might be a ubiquitous trait across human tumor types. Our study unveils the unsolved mechanism behind bacterial immunotherapy's dual challenge in solid tumors and provides a framework for intratumoral immunomodulation.
Collapse
Affiliation(s)
- Zhiguang Chang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xuan Guo
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xuefei Li
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongsheng Zang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Lu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yang Li
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jian-Dong Huang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China; Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chenli Liu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
12
|
Guo Q, Shen Q, Hao Q, Jiang XL, Zou LP, Xue YP, Zheng YG. Resolving the Trade-Off Between Toxicity and Efficiency of CRISPR-Cas9 System for Genome Editing Within Escherichia coli. Biotechnol J 2025; 20:e70010. [PMID: 40165637 DOI: 10.1002/biot.70010] [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: 12/12/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
Efficient gene editing of Escherichia coli BL21 (DE3) holds significant practical value as a host for heterologous protein expression. Recently reported CRISPR-Cas9 editing systems for this strain exhibit a trade-off between efficiency and toxicity. In this study, we addressed this trade-off by employing the strategy to transiently induce Cas9 expression in the high-copy plasmid during the editing stage. Furthermore, we demonstrated that eliminating the sgRNA-expressing plasmid using a temperature-sensitive replicon, combined with SacB for removing the Cas9-expressing plasmid, exhibited higher efficiency compared to previously reported strategies for editing system removal. We assigned this optimized CRISPR-Cas9 genome editing system as the pEBcas9/pEBsgRNA system, which has successfully achieved efficient five rounds of genome editing and simultaneous editing of multiple loci in E. coli BL21 (DE3). Using this system, we identified several loci suitable for multi-copy integrated expression of exogenous genes. Overall, the pEBcas9/pEBsgRNA system may facilitate the application of E. coli in both industrial and academic fields.
Collapse
Affiliation(s)
- Qian Guo
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Qi Shen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Qi Hao
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Xian-Long Jiang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Lu-Ping Zou
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Ya-Ping Xue
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| | - Yu-Guo Zheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
13
|
Xia Z, Lao C, Wu J, Jin Y, Chen X, Li H, Fan X, Yuan L, Sun L. Optimization of l-Fucose Biosynthesis in Escherichia coli through Pathway Engineering and Mixed Carbon Source Strategy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6102-6112. [PMID: 40029204 DOI: 10.1021/acs.jafc.4c12544] [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: 03/05/2025]
Abstract
This study presents an engineered strain of Escherichia coli specifically designed to enhance the production of l-fucose while minimizing residues of 2'-fucosyllactose. The optimization strategies employed include the selection of key enzymes, optimization of gene copy numbers, and fermentation using mixed carbon sources. The metabolic flux was directed toward l-fucose synthesis by integrating preferred 1,2-fucosyltransferase and α-l-fucosidase into the genome. Furthermore, the gene copy numbers were optimized to enhance enzyme expression, thereby increasing l-fucose production. Additionally, the supply of guanosine 5'-triphosphate was improved, and cofactors were regenerated to better regulate metabolism. Modifications to transporter proteins effectively reduced the accumulation of 2'-fucosyllactose. The implementation of a glucose/glycerol co-fermentation strategy enhanced carbon flux distribution and strain efficiency. The optimized strain achieved a yield of 91.90 g/L of l-fucose in a 5 L bioreactor, representing an 80.01% increase over previous yields, with a productivity of 1.18 g L-1 h-1. This yield is the highest reported for l-fucose, demonstrating its potential for industrial production.
Collapse
Affiliation(s)
- Zihan Xia
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Caiwen Lao
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Jinyong Wu
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Yiwen Jin
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Xiangsong Chen
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - He Li
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Xijie Fan
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Lixia Yuan
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Lijie Sun
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| |
Collapse
|
14
|
Ai Y, Huang Y, Zhao H, Su B, Lin J. Engineered phenylalanine hydroxylase coupled with an effective cofactor synthesis and regeneration system for high-yield production of 5-hydroxytryptophan. BIORESOUR BIOPROCESS 2025; 12:15. [PMID: 40047997 PMCID: PMC11885754 DOI: 10.1186/s40643-025-00846-z] [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: 01/06/2025] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
5-Hydroxytryptophan (5-HTP) is widely used as a natural remedy for sleep disorders. In terms of biosafety, bio-derived 5-HTP is preferred over chemically synthesized 5-HTP. However, the low titer of 5-HTP in the reported microbiological methods (< 10 g/L) limits the industrialization of 5-HTP biosynthesis. In the present study, a Trp-accumulating E. coli strain TRP1 was constructed by blocking the degradation path (ΔtnaA), branching paths (ΔpheA, ΔtyrA) and repression system (ΔtrpR, ΔtrpL). Next, the hydroxylation module employing a phenylalanine hydroxylase mutant XcPAHW179F (XC2) coupled with an MH4 regenerating system (CvPCD-EcFolM system) was screened to convert L-Trp into 5-HTP. Protein engineering was performed on hydroxylase XC2 based on the molecular dynamics simulation of the enzyme-substrate complex, and the strain TRP1-XC4 harboring the triple-mutant XcPAHL98I/A129K/W179F (XC4) was able to produce 319.4 mg/L 5-HTP. Genome editing was carried out focused on accelerating product efflux (strengthening YddG) and increasing MH4 supply (strengthening FolM, FolE and FolX), resulting in a strain TRP5-XC4 to produce 13.9 g/L 5-HTP in 5 L fed-batch fermentation with a space-time yield of 0.29 g/L/h, which is the highest production and productivity record for 5-HTP biosynthesis. This study successfully provided an engineered strain and an efficient green method for the industrial synthesis of 5-HTP.
Collapse
Affiliation(s)
- Yulin Ai
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Yusong Huang
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Hongru Zhao
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Bingmei Su
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
- Institute of Enzyme Catalysis and Synthetic Biotechnology, Fuzhou University, Fuzhou, 350108, China.
| | - Juan Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
- Institute of Enzyme Catalysis and Synthetic Biotechnology, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
15
|
Lu S, Lao C, Wang J, Yuan L, Yao J, Li H, Fan X, Zhang Q, Wu J, Chen X. Multistrategy Optimization for High-Yield 3-Fucosyllactose Production in Escherichia coli BL21 Star (DE3). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5385-5394. [PMID: 39967241 DOI: 10.1021/acs.jafc.4c11761] [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: 02/20/2025]
Abstract
3-Fucosyllactose (3-FL), an essential component of human milk oligosaccharides, is crucial for infant health. However, the extraction of 3-FL from milk presents a significant challenge. In this study, E. coli BL21 Star (DE3) was engineered for 3-FL biosynthesis, and the gene combinations and metabolic pathways were optimized to obtain a high-yield 3-FL production strain. First, the 3-FL production module was integrated into E. coli. Second, an efficient alpha-1,3-fucosyltransferase was screened. Then, different integration sites were used to optimize the precursor synthesis gene cluster and the transferase genes and to screen for transporter proteins and glycerol utilization genes that could enhance 3-FL production. The 3FL05-1 strain yielded a 3-FL titer of 11.26 g/L in shake flasks. Further amplification in a 5 L bioreactor resulted in a 3-FL titer of 60.24 g/L, which represents the highest reported titer to date.
Collapse
Affiliation(s)
- Shujie Lu
- University of Science and Technology of China, Hefei 230026, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Caiwen Lao
- Hefei CAS Health Bio-Industrial Technology Institute Co., Ltd., Hefei 230031, China
| | - Jin Wang
- University of Science and Technology of China, Hefei 230026, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lixia Yuan
- University of Science and Technology of China, Hefei 230026, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Jianming Yao
- University of Science and Technology of China, Hefei 230026, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - He Li
- Hefei CAS Health Bio-Industrial Technology Institute Co., Ltd., Hefei 230031, China
| | - Xijie Fan
- Hefei CAS Health Bio-Industrial Technology Institute Co., Ltd., Hefei 230031, China
| | - Qihong Zhang
- Hefei CAS Health Bio-Industrial Technology Institute Co., Ltd., Hefei 230031, China
| | - Jinyong Wu
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiangsong Chen
- University of Science and Technology of China, Hefei 230026, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
16
|
Pan TL, Cha JL, Wang H, Zhang JS, Xiao JL, Shen J, Zhou M, Li Y, Ma JZ, Zhao KY, Zhang YK, Xiao P, Gao H. The CRISPR/Cas9-Mediated Knockout of VgrG2 in Wild Pathogenic E. coli to Alleviate the Effects on Cell Damage and Autophagy. Vet Sci 2025; 12:249. [PMID: 40266908 PMCID: PMC11945575 DOI: 10.3390/vetsci12030249] [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: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 04/25/2025] Open
Abstract
CRISPR/Cas9, as a well-established gene editing technology, has been applied in numerous model organisms, but its application in wild-type E. coli remains limited. Pathogenic wild-type E. coli, a major cause of foodborne illnesses and intestinal inflammation in humans and animals, poses a significant global public health threat. The valine-glycine repeat protein G (VgrG) is a key virulence factor that enhances E. coli pathogenicity. In this study, PCR was used to identify 50 strains carrying the virulence gene VgrG2 out of 83 wild pathogenic E. coli strains, with only one strain sensitive to kanamycin and spectinomycin. A homologous repair template for VgrG2 was constructed using overlap PCR. A dual-plasmid CRISPR/Cas9 system, combining pTarget (spectinomycin resistance) and pCas (kanamycin resistance) with Red homologous recombination, was then used to induce genomic cleavage and knock out VgrG2. PCR and sequencing confirmed the deletion of a 1708 bp fragment of the VgrG2 gene in wild-type E. coli. IPEC-J2 cells were infected with E. coli-WT and E. coli ∆VgrG2, and treated with the mTOR inhibitor rapamycin to study the effects of VgrG2 on the mTOR signaling pathway. The qPCR results showed that VgrG2 activated the mTOR pathway, suppressed mTOR and p62 mRNA levels, and upregulated the autophagy-related genes and LC3-II protein expression. In conclusion, we utilized CRISPR/Cas9 technology to achieve large-fragment deletions in wild-type E. coli, revealing that VgrG2 activates the mTOR signaling pathway and upregulates autophagy markers. These findings offer new insights into E. coli genome editing and clarifies the pathogenic mechanisms through which VgrG2 induces cellular damage.
Collapse
Affiliation(s)
- Tian-Ling Pan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Long Cha
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Hao Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (H.W.); (M.Z.)
| | - Jing-Song Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Long Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jue Shen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Meng Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (H.W.); (M.Z.)
| | - Yue Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Zhi Ma
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Kai-Yuan Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Yong-Kang Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| |
Collapse
|
17
|
Zhou X, Yang S, Sun B, Dong F, Yin M, Jiang Y, Huang Z, Yang S. Implementation of RAGATH RNA-associated DNA Endonucleases as Genome Editing Tool in Escherichia coli. Biotechnol J 2025; 20:e70005. [PMID: 40111000 DOI: 10.1002/biot.70005] [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: 09/29/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
The preferred method for Escherichia coli genome editing relies on Cas9 from Streptococcus pyogenes (SpCas9) and λ-Red recombinase. Although SpCas9 is currently the most active RNA-guided DNA endonuclease, a significant number of escapers are often observed, making it inefficient across different sites, particularly when inserting large fragments. In this study, we identified two RAGATH RNA-associated DNA endonucleases (RADs) derived from IS607 transposons. Both of them exhibited high cleavage activity in E. coli. When combined with λ-Red recombinase, they achieved editing efficiencies approaching 100%. Even at target sites where SpCas9 exhibited low editing efficiency, RADs maintained efficiencies ranging from 57% to 94%. Moreover, RADs exhibited higher efficiencies in inserting large fragments in certain cases compared to SpCas9. Taken together, these RAD-based genome editing tools provide viable alternatives to SpCas9, particularly for challenging targets and/or large fragment insertions.
Collapse
Affiliation(s)
- Xiaojie Zhou
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Siqi Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Bingbing Sun
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feng Dong
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingyu Yin
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yu Jiang
- Huzhou Center of Industrial Biotechnology, Shanghai Institutes for Biological Sciences, Huzhou, China
| | - Zhiwei Huang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Sheng Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
18
|
Zhang Z, Hu B, Zhou J, Li J, Chen J, Du G, Zhao X. Construction of a Plasmid-Free Escherichia coli Strain with Enhanced Heme Supply to Produce Active Hemoglobins. Metabolites 2025; 15:151. [PMID: 40137115 PMCID: PMC11943725 DOI: 10.3390/metabo15030151] [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: 02/04/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Heme is an important cofactor and plays crucial roles in the correct folding of hemoproteins. The synthesis of heme can be enhanced by the plasmid-based expression of heme biosynthetic genes. However, plasmid-based expression is genetically unstable and requires the utilization of antibiotics to maintain high copy numbers of plasmids. METHODS The rate-limiting steps in heme biosynthesis were first analyzed based on previous studies and the accumulation of heme intermediates was achieved by adding heme precursor (5-aminolevulinic acid, ALA). Next, the intracellular accumulation of porphyrin was increased by deleting the porphyrin transporter TolC. Finally, the heme synthetic genes were modified by integrating the hemA and hemL genes into the cheW and yciQ locus, assembling the rate-limiting enzymes HemC and HemD with RIAD-RIDD tags, replacing the promoters of hemE/hemH genes with the constitutive promoter PJ23100, and deleting the heme degradation gene yfeX. RESULTS An enhanced heme supply HEME2 strain was obtained with a heme titer of 0.14 mg/L, which was 4.60-fold higher than that of the C41(DE3) strain. The HEME2 strain was applied to produce human hemoglobin and leghemoglobin. The titer and peroxidase activity of human hemoglobin were 1.29-fold and 42.4% higher in the HEME2-hHb strain than the values in the control strain C41-hHb. In addition, the peroxidase activity and heme content of leghemoglobin were increased by 39.2% and 53.4% in the HEME2-sHb strain compared to the values in the control strain C41-sHb. CONCLUSIONS A plasmid-free Escherichia coli C41(DE3) strain capable of efficient and stable heme supply was constructed and can be used for the production of high-active hemoglobins.
Collapse
Affiliation(s)
- Zihan Zhang
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Baodong Hu
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Jingwen Zhou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Jianghua Li
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Guocheng Du
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Xinrui Zhao
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (Z.Z.); (B.H.); (J.Z.); (J.L.); (J.C.)
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| |
Collapse
|
19
|
Li N, Yan S, Xia H, Fang Y, Niu K, Li G, Xu Z, Sun Y, Xu H, Xu X. Metabolic Engineering of Escherichia coli BL21(DE3) for 2'-Fucosyllactose Synthesis in a Higher Productivity. ACS Synth Biol 2025; 14:441-452. [PMID: 39815725 DOI: 10.1021/acssynbio.4c00598] [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] [Indexed: 01/18/2025]
Abstract
2'-Fucosyllactose (2'-FL) is the most abundant human milk oligosaccharides (HMOs). 2'-FL exhibits great benefits for infant health, such as preventing infantile diarrhea and promoting the growth of intestinal probiotics. The microbial cell factory technique has shown promise for the massive production of 2'-FL. Here, we aimed to construct a recombinant E. coli BL21(DE3) strain for the hyperproduction of 2'-FL. Initially, multicopy genomic integration and expression of the lactose permease gene lacY reduced the formation of byproducts. Furthermore, a more efficient Shine-Dalgarno sequence was used to replace the wild-type sequence in the manC-manB and gmd-wcaG gene clusters, which significantly increased the 2'-FL titer. Based on these results, we overexpressed the sugar efflux transporter SetA and knocked out the pgi gene. This further improved 2'-FL synthesis when glycerol was used as the sole carbon source. Finally, a new α-1,2-fucosyltransferase was identified in Neisseria sp., which exhibited a higher capacity for 2'-FL production. Fed-batch fermentation produced 141.27 g/L 2'-FL in 45 h with a productivity of 3.14 g/L × h. This productivity rate achieved the highest recorded 2'-FL levels, indicating the potential of engineered E. coli BL21 (DE3) strains for use in the industrial production of 2'-FL.
Collapse
Affiliation(s)
- Na Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Saifeng Yan
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Hongzhi Xia
- Nantong Licheng Biological Engineering Co., Ltd, Shanghai 200000, China
| | - Yin Fang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Kun Niu
- Nantong Licheng Biological Engineering Co., Ltd, Shanghai 200000, China
| | - Guyue Li
- Nantong Licheng Biological Engineering Co., Ltd, Shanghai 200000, China
| | - Zheng Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Yang Sun
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Hong Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Xiaoqi Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
20
|
Tang H, Gao J, Wang H, Sun M, Zhang S, Song C, Li Q. Characterization of the genome editing with miniature DNA nucleases TnpB and IscB in Escherichia coli strains. Commun Biol 2025; 8:261. [PMID: 39972101 PMCID: PMC11840021 DOI: 10.1038/s42003-025-07521-1] [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: 08/12/2024] [Accepted: 01/11/2025] [Indexed: 02/21/2025] Open
Abstract
DNA endonucleases TnpB and IscB are emerging candidates for combating drug-resistant bacteria, particularly Escherichia coli, due to their specificity in targeting DNA and smaller size. However, the genome-editing of TnpB/IscB in E. coli remains unclear. This study characterized the genome editing of TnpB/IscB in different E. coli strains. First, the toxicity and cleavage results indicated TnpB was effective only in MG1655, whereas IscB and enIscB demonstrated functionality in ATCC9637/BL21(DE3). Subsequently, a genome-editing tool was established in MG1655 by using TnpB (as a thermophilic programmable endonuclease), achieving up to 100% editing efficiency, while IscB/enIscB achieved editing in ATCC9637/BL21(DE3). Additionally, the editing plasmids were successfully cured. Finally, the mechanism underlying the escape of E. coli during TnpB/IscB editing was elucidated. Overall, this study successfully applied TnpB/IscB/enIscB to genome editing in E. coli, which will expand the genetic manipulation toolbox in E. coli and facilitate the development of the antimicrobial drugs.
Collapse
Affiliation(s)
- Hongjie Tang
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Jie Gao
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Hengyi Wang
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Mingjun Sun
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Suyi Zhang
- Luzhou Laojiao Co. Ltd, Luzhou, 646000, China
- National Engineering Research Center of Solid-State Brewing, Luzhou, 646000, China
| | - Chuan Song
- Luzhou Laojiao Co. Ltd, Luzhou, 646000, China
- National Engineering Research Center of Solid-State Brewing, Luzhou, 646000, China
| | - Qi Li
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China.
| |
Collapse
|
21
|
Chu W, Tian R, Guo Y, Wu Y, Rehm FBH, Liu L, Li J, Du G, Chen J, Liu Y. An evolved, orthogonal ssDNA generator for targeted hypermutation of multiple genomic loci. Nucleic Acids Res 2025; 53:gkaf051. [PMID: 39878218 PMCID: PMC11775612 DOI: 10.1093/nar/gkaf051] [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: 12/08/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025] Open
Abstract
Achieving targeted hypermutation of specific genomic sequences without affecting other regions remains a key challenge in continuous evolution. To address this, we evolved a T7 RNA polymerase (RNAP) mutant that synthesizes single-stranded DNA (ssDNA) instead of RNA in vivo, while still exclusively recognizing the T7 promoter. By increasing the error rate of the T7 RNAP mutant, it generates mutated ssDNA that recombines with homologous sequences in the genome, leading to targeted genomic hypermutation. This approach, termed T7 RNAP mutant-assisted continuous evolution (T7ACE), functions effectively in both typical prokaryotic and eukaryotic microorganisms (Escherichia coli and Saccharomyces cerevisiae), achieving targeted hypermutations at rates 2800- and 1200-fold higher than the genomic mutation rates, respectively. Using T7ACE, we successfully evolved an eight-fold increase in tigecycline resistance within 7 days and doubled the efficiency of a xylose utilization pathway in 10 days, demonstrating the efficiency and broad applicability of this single-component tool for continuous evolution.
Collapse
Affiliation(s)
- Weiran Chu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| | - Rongzhen Tian
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 1TN, United Kingdom
| | - Yaxin Guo
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| | - Yaokang Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| | - Fabian B H Rehm
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 1TN, United Kingdom
| | - Long Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| | - Jianghua Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Guocheng Du
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| | - Yanfeng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
22
|
Fu P, Wang Y, Liu Y, Han Z, Peng Z, Liu L, Han W. A mobile genetic element-derived primase-polymerase harbors multiple activities implicated in DNA replication and repair. Nucleic Acids Res 2025; 53:gkae1318. [PMID: 39797730 PMCID: PMC11724360 DOI: 10.1093/nar/gkae1318] [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: 06/24/2024] [Revised: 12/21/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Primase-polymerases (PrimPols) play divergent functions from DNA replication to DNA repair in all three life domains. In archaea and bacteria, numerous and diverse PPs are encoded by mobile genetic elements (MGEs) and act as the replicases for their MGEs. However, their varying activities and functions are not fully understood. In this study, we characterized a group of PrimPols that are genetically associated with prokaryotic argonaute proteins (pAgos). The pAgo-associated PrimPol (AgaPP) is likely derived from a MGE. AgaPP has polymerase and primase activities and physically interacts with a helicase encoded by its downstream gene, suggesting that they constitute a functional replication module. Further, AgaPP performs translesion DNA synthesis, terminal transfer and microhomology-mediated end joining (MMEJ), showing striking similarity to human DNA repair polymerase θ. AgaPP can promote the MMEJ repair of Cas9-induced double-stranded DNA breaks and increase cell survival post DNA damage in Escherichia coli. In addition, the MMEJ activity of AgaPP can be repurposed to assist DNA assembly in vitro. Together, the findings reveal dual role of AgaPP in both DNA replication and repair.
Collapse
Affiliation(s)
- Pan Fu
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Yuwei Wang
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Yanqiu Liu
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Zhenhao Han
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Zhangzhong Peng
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Linfeng Liu
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| | - Wenyuan Han
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Shizishan Road No.1, Hongshan District, 430070 Wuhan, China
| |
Collapse
|
23
|
Wang K, Song X, Cui B, Wang Y, Luo W. Metabolic Engineering of Escherichia coli for Efficient Production of Ectoine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:646-654. [PMID: 39723826 DOI: 10.1021/acs.jafc.4c07640] [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: 12/28/2024]
Abstract
Ectoine is a valuable compatible solute with extensive applications in bioengineering, cosmetics, medicine, and the food industry. While certain halophilic bacteria can naturally produce ectoine, as a model organism for biomanufacturing, Escherichia coli offers significant advantages to be engineered for potentially high-level ectoine production. However, complex metabolic flux distributions and byproduct formation present bottlenecks that limit ectoine production in E. coli. In this study, we aimed to enhance ectoine production in E. coli BL21(DE3) through systematic metabolic engineering strategies. We investigated the effects of the ectABC gene cluster sequence, plasmid copy number, and key gene copy number on ectoine synthesis. Using the original ectABC sequence with the high-copy-number plasmid pRSFDuet-1 resulted in the highest level of ectoine production. Knocking out genes encoding homoserine dehydrogenase and diaminopimelate decarboxylase reduced competing pathways, further increasing ectoine yield. Overexpression of aspartate semialdehyde dehydrogenase, aspartate kinase I (thrA*), aspartate aminotransferase, and aspartate ammonia-lyase (aspA) was performed, and optimal gene copy numbers were determined. When the copy numbers of thrA* and aspA were both three, ectoine synthesis improved, reaching 1.91 g/L. Enhancing the oxaloacetate pool by overexpressing phosphoenolpyruvate carboxylase (ppc) or introducing pyruvate carboxylase (pyc) from Corynebacterium glutamicum further increased ectoine production to 4.99 g/L. Balancing NADPH and ATP levels through cofactor engineering contributed to additional production improvements. Combining these strain engineering strategies, we ultimately constructed strain C24, which produced 35.33 g/L ectoine in a 5 L fermenter with a glucose conversion rate of 0.21 g/g. These results demonstrate that targeted metabolic engineering can significantly enhance ectoine production in E. coli, providing a foundation for industrial-scale production.
Collapse
Affiliation(s)
- Ke Wang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xitong Song
- Key Laboratory of Ecological Environment and Information Atlas, Fujian Provincial University (Putian University), Putian 351100, China
- Fujian Provincial Key Laboratory of Ecology-Toxicological Effects & Control for Emerging Contaminants, College of Environmental and Biological Engineering, Putian University, Putian 351100, China
| | - Boya Cui
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Yi Wang
- Department of Biological and Agricultural Engineering, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Wei Luo
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
24
|
Rahman KMT, Amaratunga R, Butzin XY, Singh A, Hossain T, Butzin NC. Rethinking dormancy: Antibiotic persisters are metabolically active, non-growing cells. Int J Antimicrob Agents 2025; 65:107386. [PMID: 39551274 DOI: 10.1016/j.ijantimicag.2024.107386] [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: 02/19/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVES Bacterial persisters are a subpopulation of multidrug-tolerant cells capable of surviving and resuming activity after exposure to bactericidal antibiotic concentrations, contributing to relapsing infections and the development of antibiotic resistance. In this study, we challenge the conventional view that persisters are metabolically dormant by providing compelling evidence that an isogenic population of Escherichia coli remains metabolically active in persistence. METHODS Using transcriptomic analysis, we examined E. coli persisters at multiple time points following exposure to bactericidal concentrations of ampicillin (Amp). Some genes were consistently upregulated in Amp treated persisters compared to the untreated controls, a change that can only occur in metabolically active cells capable of increasing RNA levels. RESULTS Some of the identified genes have been previously linked to persister cells, while others have not been associated with them before. If persister cells were metabolically dormant, gene expression changes over time would be minimal during Amp treatment. However, network analysis revealed major shifts in gene network activity at various time points of antibiotic exposure. CONCLUSIONS These findings reveal that persisters are metabolically active, non-dividing cells, thereby challenging the traditional view that they are dormant.
Collapse
Affiliation(s)
- K M Taufiqur Rahman
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Ruqayyah Amaratunga
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Xuan Yi Butzin
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, Delaware, USA
| | - Tahmina Hossain
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA.
| | - Nicholas C Butzin
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA; Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, South Dakota, USA.
| |
Collapse
|
25
|
Wei T, Lai W, Chen Q, Sun C. Engineered Phage Enables Efficient Control of Gene Expression upon Infection of the Host Cell. Int J Mol Sci 2024; 26:250. [PMID: 39796105 PMCID: PMC11720261 DOI: 10.3390/ijms26010250] [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: 12/08/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Recently, we developed a spatial phage-assisted continuous evolution (SPACE) system. This system utilizes chemotaxis coupled with the growth of motile bacteria during their spatial range expansion in soft agar to provide fresh host cells for iterative phage infection and selection pressure for preserving evolved genes of interest carried by phage mutants. Controllable mutagenesis activated only in a subpopulation of the migrating cells is essential in this system to efficiently generate mutated progeny phages from which desired individuals are selected during the directed evolution process. But, the widely adopted small molecule-dependent inducible system could hardly fulfill this purpose because it always affects all cells homogeneously. In this study, we developed a phage infection-induced gene expression system using modified Escherichia coli (E. coli) phage shock protein operon or sigma factors from Bacillus subtilis. Results showed that this system enabled efficient control of gene expression upon phage infection with dynamic output ranges from small to large using combinations of different engineered phages and corresponding promoters. This system was incorporated into SPACE to function as a phage infection-induced mutagenesis module and successfully facilitated the evolution of T7 RNA polymerase, which generated diverse mutants with altered promoter recognition specificity. We expect that phage infection-induced gene expression system could be further extended to more applications involving partial induction in a portion of a population and targeted induction in specific strains among a mixed bacterial community, which provides an important complement to small molecule-dependent inducible systems.
Collapse
Affiliation(s)
- Ting Wei
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wangsheng Lai
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| | - Qian Chen
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| | - Chenjian Sun
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| |
Collapse
|
26
|
Sandoval Hurtado CP, Kelly SP, Shende V, Perez M, Curtis BJ, Newmister SA, Ott K, Pereira F, Sherman DH. Engineering a Biosynthetic Pathway for the Production of (+)-Brevianamides A and B in Escherichia coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627567. [PMID: 39713314 PMCID: PMC11661150 DOI: 10.1101/2024.12.10.627567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The privileged fused-ring system comprising the bicyclo[2.2.2]diazaoctane (BDO) core is prevalent in diketopiperazine (DKP) natural products with potent and diverse biological activities, with some being explored as drug candidates. Typically, only low yields of these compounds can be extracted from native fungal producing strains and the available synthetic routes remain challenging due to their structural complexity. BDO-containing DKPs including (+)-brevianamides A and B are assembled via multi-component biosynthetic pathways incorporating non-ribosomal peptide synthetases, prenyltransferases, flavin monooxygenases, cytochrome P450s and semi-pinacolases. To simplify access to this class of alkaloids, we designed an engineered biosynthetic pathway in Escherichia coli , composed of six enzymes sourced from different kingdoms of life. The pathway includes a cyclodipeptide synthase (NascA), a cyclodipeptide oxidase (DmtD2/DmtE2), a prenyltransferase (NotF), a flavin-dependent monooxygenase (BvnB), and kinases (PhoN and IPK). Cultivated in glycerol supplemented with prenol, the engineered E. coli strain produces 5.3 mg/L of (-)-dehydrobrevianamide E ( 4 ), which undergoes a terminal, ex vivo lithium hydroxide catalyzed rearrangement reaction to yield (+)-brevianamides A and B with a 46% yield and a 92:8 diastereomeric ratio. Additionally, titers of 4 were increased eight-fold by enhancing NADPH pools in the engineered E. coli strain. Our study combines synthetic biology, biocatalysis and synthetic chemistry approaches to provide a five-step engineered biosynthetic pathway for producing complex indole alkaloids in E. coli . Abstract Figure
Collapse
|
27
|
Li X, Shi PW, Du F, Zhang ZX, Li ZJ, Wu N, Yang G, Ma W, Sun XM. Creating a System of Dual Regulation of Translation and Transcription to Enhance the Production of Recombinant Protein. Biotechnol J 2024; 19:e202400679. [PMID: 39676520 DOI: 10.1002/biot.202400679] [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: 11/07/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
When constructing cell factories, it is crucial to reallocate intracellular resources towards the synthesis of target compounds. However, imbalanced resource allocation can lead to a tradeoff between cell growth and production, reducing overall efficiency. Reliable gene expression regulation tools are needed to coordinate cell growth and production effectively. The orthogonal translation system, developed based on genetic code expansion (GCE), incorporates non-canonical amino acids (ncAAs) into proteins by assigning them to expanded codons, which enables the control of target protein expression at the translational level in an ncAA-dependent manner. However, the stringency of this regulatory tool remains inadequate. This study achieved strict translational-level control of the orthogonal translation system by addressing the abnormal leakage caused by the arabinose-inducible promoter. Further validation was conducted on the relationship between ncAA concentration and expression level, as well as the host's adaptability to the system. Subsequently, the system's applicability across multiple Escherichia coli hosts was verified by examining the roles of RF1 (peptide chain release factor 1) and endogenous TAG codons. By combining this strategy with inducible promoters, dual-level regulation of target gene expression at both transcriptional and translational levels was achieved and the dynamic range was further increased to over 20-fold. When using ncAA to control the expression of T7 RNA polymerase (T7 RNAP), the leakage expression was reduced by 82.7%, mitigating the low production efficiency caused by extensive leakage in the T7 system. As proof of concept, the strategy enhanced the production of alcohol dehydrogenase (ADH) by 9.82-fold, demonstrating its excellent capability in controlling gene expression in developing cell factories.
Collapse
Affiliation(s)
- Xin Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Peng-Wei Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Fei Du
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Zi-Xu Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Zi-Jia Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Na Wu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu, China
| | - Guang Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Wang Ma
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Xiao-Man Sun
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Zhu J, Liu W, Guo L, Tan X, Sun W, Zhang H, Zhang H, Tian W, Jiang T, Meng W, Liu Y, Kang Z, Gao C, Lü C, Xu P, Ma C. Acetate production from corn stover hydrolysate using recombinant Escherichia coli BL21 (DE3) with an EP-bifido pathway. Microb Cell Fact 2024; 23:300. [PMID: 39523316 PMCID: PMC11552437 DOI: 10.1186/s12934-024-02575-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Acetate is an important chemical feedstock widely applied in the food, chemical and textile industries. It is now mainly produced from petrochemical materials through chemical processes. Conversion of lignocellulose biomass to acetate by biotechnological pathways is both environmentally beneficial and cost-effective. However, acetate production from carbohydrate in lignocellulose hydrolysate via glycolytic pathways involving pyruvate decarboxylation often suffers from the carbon loss and results in low acetate yield. RESULTS Escherichia coli BL21 (DE3) was confirmed to have high tolerance to acetate in this work. Thus, it was selected from seven laboratory E. coli strains for acetate production from lignocellulose hydrolysate. The byproduct-producing genes frdA, ldhA, and adhE in E. coli BL21 (DE3) were firstly knocked out to decrease the generation of succinate, lactate, and ethanol. Then, the genes pfkA and edd were also deleted and bifunctional phosphoketolase and fructose-1,6-bisphosphatase were overexpressed to construct an EP-bifido pathway in E. coli BL21 (DE3) to increase the generation of acetate from glucose. The obtained strain E. coli 5K/pFF can produce 22.89 g/L acetate from 37.5 g/L glucose with a yield of 0.61 g/g glucose. Finally, the ptsG gene in E. coli 5K/pFF was also deleted to make the engineered strain E. coli 6K/pFF to simultaneously utilize glucose and xylose in lignocellulosic hydrolysates. E. coli 6K/pFF can produce 20.09 g/L acetate from corn stover hydrolysate with a yield of 0.52 g/g sugar. CONCLUSION The results presented here provide a promising alternative for acetate production with low cost substrate. Besides acetate production, other biotechnological processes might also be developed for other acetyl-CoA derivatives production with lignocellulose hydrolysate through further metabolic engineering of E. coli 6K/pFF.
Collapse
Affiliation(s)
- Jieni Zhu
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Wei Liu
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Leilei Guo
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Xiaoxu Tan
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Weikang Sun
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Hongxu Zhang
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Hui Zhang
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Wenjia Tian
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Tianyi Jiang
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China
| | - Wensi Meng
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Yidong Liu
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Zhaoqi Kang
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China
| | - Chuanjuan Lü
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China.
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Shandong University, No. 72 Binhai Road, Qingdao, 266237, P. R. China.
| |
Collapse
|
29
|
Mao Y, Zhu J, Zhang Q, Wang G, Fan H, Zhang X, Sun Y, Wang Y. De novo synthesis of 1-phenethylisoquinoline in engineered Escherichia coli. Synth Syst Biotechnol 2024; 10:271-280. [PMID: 39650803 PMCID: PMC11625190 DOI: 10.1016/j.synbio.2024.10.007] [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: 08/17/2024] [Revised: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Phenylethylisoquinoline alkaloids (PIAs) are medicinally important natural products derived from the 1-phenylethylisoquinoline precursor. Heterologous production of the PIAs remains challenging due to the incomplete elucidation of biosynthetic pathway and the lack of proper microbial cell factory designed for precursor enhancement. In this work, an artificial pathway composed of eight enzymes from different species was established for de novo 1-phenylethylisoquinoline biosynthesis in engineered Escherichia coli. The yield of the intermediate 4-hydroxydihydrocinnamaldehyde was optimized through screening various NADP+-dependent 2-alkenal reductases, cofactor regeneration and the site-directed mutagenesis of key residues in ChAER1. Subsequently, incorporation of the modified dopamine pathway into an endogenous reductase-deficient E. coli with high tyrosine yield boosted the production of 1-phenylethylisoquinoline, reaching 402.58 mg/L in a 5L fermenter. Our work lays a foundation for the future large-scale production of high value-added 1-phenylethylisoquinoline-related alkaloids.
Collapse
Affiliation(s)
- Yaping Mao
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiangming Zhu
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qian Zhang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Guangyi Wang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Hongkai Fan
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Xiaowei Zhang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yuwei Sun
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yong Wang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
30
|
Zhou H, Zhang C, Li Z, Xia M, Li Z, Wang Z, Tan GY, Luo Y, Zhang L, Wang W. Systematic development of a highly efficient cell factory for 5-aminolevulinic acid production. Trends Biotechnol 2024; 42:1479-1502. [PMID: 39112275 DOI: 10.1016/j.tibtech.2024.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 11/17/2024]
Abstract
The versatile applications of 5-aminolevulinic acid (5-ALA) across the fields of agriculture, livestock, and medicine necessitate a cost-efficient biomanufacturing process. In this study, we achieved the economic viability of biomanufacturing this compound through a systematic engineering framework. First, we obtained a 5-ALA synthase (ALAS) with superior performance by exploring its natural diversity with divergent evolution. Subsequently, using a genome-scale model, we identified and modified four key targets from distinct pathways in Escherichia coli, resulting in a final enhancement of 5-ALA titers up to 21.82 g/l in a 5-l bioreactor. Furthermore, recognizing that an imbalance of redox equivalents hindered further titer improvement, we developed a dynamic control system that effectively balances redox status and carbon flux. Ultimately, we collaboratively optimized the artificial redox homeostasis system at the transcription level with other cofactors at the feeding level, demonstrating the highest recorded performance to date with a titer of 63.39 g/l for the biomanufacturing of 5-ALA.
Collapse
Affiliation(s)
- Houming Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chengyu Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zilong Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Menglei Xia
- Metabolism and Fermentation Process Control, College of Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhenghong Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhengduo Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Gao-Yi Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ying Luo
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Weishan Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
31
|
Chang J, Wei X, Liu D, Li Q, Li C, Zhao J, Cheng L, Wang G. Engineering Escherichia coli via introduction of the isopentenol utilization pathway to effectively produce geranyllinalool. Microb Cell Fact 2024; 23:292. [PMID: 39443997 PMCID: PMC11515624 DOI: 10.1186/s12934-024-02563-2] [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: 07/31/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Geranyllinalool, a natural diterpenoid found in plants, has a floral and woody aroma, making it valuable in flavors and fragrances. Currently, its synthesis primarily depends on chemical methods, which are environmentally harmful and economically unsustainable. Microbial synthesis through metabolic engineering has shown potential for producing geranyllinalool. However, achieving efficient synthesis remains challenging owing to the limited availability of terpenoid precursors in microorganisms. Thus, an artificial isopentenol utilization pathway (IUP) was constructed and introduced in Escherichia coli to enhance precursor availability and further improve terpenoid synthesis. RESULTS We first constructed an artificial IUP in E. coli to enhance the supply of precursor geranylgeranyl diphosphate (GGPP) and then screened geranyllinalool synthases from plants to achieve efficient synthesis of geranyllinalool (274.78 ± 2.48 mg/L). To further improve geranyllinalool synthesis, we optimized various cultivation factors, including carbon source, IPTG concentration, and prenol addition and obtained 447.51 ± 6.92 mg/L of geranyllinalool after 72 h of shaken flask fermentation. Moreover, a scaled-up production in a 5-L fermenter was investigated to give 2.06 g/L of geranyllinalool through fed-batch fermentation. To the best of our knowledge, this is the highest reported titer so far. CONCLUSIONS Efficient synthesis of geranyllinalool in E. coli can be achieved through a two-step pathway and optimization of culture conditions. The findings of this study provide valuable insights into the production of other terpenoids in E. coli.
Collapse
Affiliation(s)
- Jin Chang
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Xinduo Wei
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Deyu Liu
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Qian Li
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Chong Li
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Jianguo Zhao
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China
| | - Likun Cheng
- Laboratory of Synthetic Biology, Shandong Binzhou Animal Science and Veterinary Medicine Academy, Research Institution of Veterinarian, No.777 Chang Jiang 5th Road, Binzhou, Shandong Province, 256600, China.
| | - Guanglu Wang
- Laboratory of Biotransformation and Biocatalysis, School of Tobacco Science and Engineering, Zhengzhou University of Light Industry, No.136 Ke Xue Avenue, Zhengzhou, Henan, 450002, People's Republic of China.
| |
Collapse
|
32
|
Hashemabadi M, Sasan HA, Hosseinkhani S, Amandadi M, Samareh Gholami A, Sadeghizadeh M. Intelligent guide RNA: dual toehold switches for modulating luciferase in the presence of trigger RNA. Commun Biol 2024; 7:1344. [PMID: 39420075 PMCID: PMC11487279 DOI: 10.1038/s42003-024-06988-8] [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: 01/05/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
The CRISPR system finds extensive application in molecular biology, but its continuous activity can yield adverse effects. Leveraging programmable CRISPR/Cas9 function via nano-device mediation effectively mitigates these drawbacks. The integration of RNA-sensing platforms into CRISPR thus empowers it as a potent tool for processing internal cell data and modulating gene activity. Here, an intelligent guide RNA-a cis-repressed gRNA synthetic circuit enabling efficient recognition of specific trigger RNAs-is developed. This platform carries two toehold switches and includes an inhibited CrRNA sequence. In this system, the presence of cognate trigger RNA promotes precise binding to the first toehold site, initiating a cascade that releases CrRNA to target a reporter gene (luciferase) in this study. Decoupling the CrRNA segment from the trigger RNA enhances the potential of this genetic logic circuit to respond to specific cellular circumstances, offering promise as a synthetic biology platform.
Collapse
Affiliation(s)
- Mohammad Hashemabadi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Ali Sasan
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mojdeh Amandadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Azadeh Samareh Gholami
- Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
33
|
He F, Liu X, Tang M, Wang H, Wu Y, Liang S. CRISETR: an efficient technology for multiplexed refactoring of biosynthetic gene clusters. Nucleic Acids Res 2024; 52:11378-11393. [PMID: 39271125 PMCID: PMC11472037 DOI: 10.1093/nar/gkae781] [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: 04/01/2024] [Revised: 08/16/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
The efficient refactoring of natural product biosynthetic gene clusters (BGCs) for activating silent BGCs is a central challenge for the discovery of new bioactive natural products. Herein, we have developed a simple and robust CRISETR (CRISPR/Cas9 and RecET-mediated Refactoring) technique, combining clustered regulatory interspaced short palindromic repeats (CRISPR)/Cas9 and RecET, for the multiplexed refactoring of natural product BGCs. By this approach, natural product BGCs can be refactored through the synergistic interaction between RecET-mediated efficient homologous recombination and the CRISPR/Cas9 system. We first performed a proof-of-concept validation of the ability of CRISETR, and CRISETR can achieve simultaneous replacement of four promoter sites and marker-free replacement of single promoter site in natural product BGCs. Subsequently, we applied CRISETR to the promoter engineering of the 74-kb daptomycin BGC containing a large number of direct repeat sequences for enhancing the heterologous production of daptomycin. We used combinatorial design to build multiple refactored daptomycin BGCs with diverse combinations of promoters different in transcriptional strengths, and the yield of daptomycin was improved 20.4-fold in heterologous host Streptomyces coelicolor A3(2). In general, CRISETR exhibits enhanced tolerance to repetitive sequences within gene clusters, enabling efficient refactoring of diverse and complex BGCs, which would greatly accelerate discovery of novel bioactive metabolites present in microorganism.
Collapse
Affiliation(s)
- Fuqiang He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xinpeng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Min Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Haiyi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Yun Wu
- Department of Cell Biology, College of Life Science, Sichuan Normal University, Chengdu, Sichuan, 610101, P.R. China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| |
Collapse
|
34
|
Sun W, Wang S, Tan X, Guo L, Liu W, Tian W, Zhang H, Jiang T, Meng W, Liu Y, Kang Z, Lü C, Gao C, Xu P, Ma C. Production of α-ketoisovalerate with whey powder by systemic metabolic engineering of Klebsiella oxytoca. Microb Cell Fact 2024; 23:264. [PMID: 39367476 PMCID: PMC11452931 DOI: 10.1186/s12934-024-02545-4] [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: 07/20/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Whey, which has high biochemical oxygen demand and chemical oxygen demand, is mass-produced as a major by-product of the dairying industry. Microbial fermentation using whey as the carbon source may convert this potential pollutant into value-added products. This study investigated the potential of using whey powder to produce α-ketoisovalerate, an important platform chemical. RESULTS Klebsiella oxytoca VKO-9, an efficient L-valine producing strain belonging to Risk Group 1 organism, was selected for the production of α-ketoisovalerate. The leucine dehydrogenase and branched-chain α-keto acid dehydrogenase, which catalyzed the reductive amination and oxidative decarboxylation of α-ketoisovalerate, respectively, were inactivated to enhance the accumulation of α-ketoisovalerate. The production of α-ketoisovalerate was also improved through overexpressing α-acetolactate synthase responsible for pyruvate polymerization and mutant acetohydroxyacid isomeroreductase related to α-acetolactate reduction. The obtained strain K. oxytoca KIV-7 produced 37.3 g/L of α-ketoisovalerate from lactose, the major utilizable carbohydrate in whey. In addition, K. oxytoca KIV-7 also produced α-ketoisovalerate from whey powder with a concentration of 40.7 g/L and a yield of 0.418 g/g. CONCLUSION The process introduced in this study enabled efficient α-ketoisovalerate production from low-cost substrate whey powder. Since the key genes for α-ketoisovalerate generation were integrated in genome of K. oxytoca KIV-7 and constitutively expressed, this strain is promising in stable α-ketoisovalerate fermentation and can be used as a chassis strain for α-ketoisovalerate derivatives production.
Collapse
Affiliation(s)
- Weikang Sun
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Shuo Wang
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Xiaoxu Tan
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Leilei Guo
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Wei Liu
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Wenjia Tian
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Hui Zhang
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Tianyi Jiang
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China
| | - Wensi Meng
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Yidong Liu
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Zhaoqi Kang
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Chuanjuan Lü
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Shandong University, NO.72 Binhai Road, Qingdao, 266237, China.
| |
Collapse
|
35
|
Woudstra C, Sørensen AN, Sørensen MCH, Brøndsted L. Strategies for developing phages into novel antimicrobial tailocins. Trends Microbiol 2024; 32:996-1006. [PMID: 38580606 DOI: 10.1016/j.tim.2024.03.003] [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: 12/22/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 04/07/2024]
Abstract
Tailocins are high-molecular-weight bacteriocins produced by bacteria to kill related environmental competitors by binding and puncturing their target. Tailocins are promising alternative antimicrobials, yet the diversity of naturally occurring tailocins is limited. The structural similarities between phage tails and tailocins advocate using phages as scaffolds for developing new tailocins. This article reviews three strategies for producing tailocins: disrupting the capsid-tail junction of phage particles, blocking capsid assembly during phage propagation, and creating headless phage particles synthetically. Particularly appealing is the production of tailocins through synthetic biology using phages with contractile tails as scaffolds to unlock the antimicrobial potential of tailocins.
Collapse
Affiliation(s)
- Cedric Woudstra
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Anders Nørgaard Sørensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Martine C Holst Sørensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Lone Brøndsted
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
36
|
Karbalaei-Heidari HR, Budisa N. Advanced and Safe Synthetic Microbial Chassis with Orthogonal Translation System Integration. ACS Synth Biol 2024; 13:2992-3002. [PMID: 39151168 DOI: 10.1021/acssynbio.4c00437] [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] [Indexed: 08/18/2024]
Abstract
Through the use of CRISPR-assisted transposition, we have engineered a safe Escherichia coli chassis that integrates an orthogonal translation system (OTS) directly into the chromosome. This approach circumvents the limitations and genetic instability associated with conventional plasmid vectors. Precision in genome modification is crucial for the top-down creation of synthetic cells, especially in the orthogonalization of vital cellular processes, such as metabolism and protein translation. Here, we targeted multiple loci in the E. coli chromosome to integrate the OTS simultaneously, creating a synthetic auxotrophic chassis with an altered genetic code to establish a reliable, robust, and safe synthetic protein producer. Our OTS-integrated chassis enabled the site-specific incorporation of m-oNB-Dopa through in-frame amber stop codon readthrough. This allowed for the expression of advanced underwater bioglues containing Dopa-Lysine motifs, which are crucial for wound healing and tissue regeneration. Additionally, we have enhanced the expression process by incorporating scaffold-stabilizing fluoroprolines into bioglues, utilizing our chassis, which has been modified through metabolic engineering (i.e., by introducing proline auxotrophy). We also engineered a synthetic auxotroph reliant on caged Dopa, creating a genetic barrier (genetic firewall) between the synthetic cells and their surroundings, thereby boosting their stability and safety.
Collapse
Affiliation(s)
- Hamid Reza Karbalaei-Heidari
- Laboratory for Chemical Synthetic Biology and Xenobiology, Department of Chemistry, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba, Canada R3T 2N2
| | - Nediljko Budisa
- Laboratory for Chemical Synthetic Biology and Xenobiology, Department of Chemistry, University of Manitoba, 144 Dysart Road, Winnipeg, Manitoba, Canada R3T 2N2
| |
Collapse
|
37
|
Sørensen AN, Woudstra C, Kalmar D, Poppeliers J, Lavigne R, Sørensen MCH, Brøndsted L. The branched receptor-binding complex of Ackermannviridae phages promotes adaptive host recognition. iScience 2024; 27:110813. [PMID: 39310758 PMCID: PMC11414711 DOI: 10.1016/j.isci.2024.110813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/02/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Bacteriophages can encode multiple receptor-binding proteins, allowing them to recognize diverse receptors for infecting different strains. Ackermannviridae phages recognize various polysaccharides as receptors by encoding multiple tail spike proteins (TSPs), forming a branched complex. We aimed to mimic the evolution of the TSP complex by studying the acquisition of TSPs without disrupting the complex's functionality. Using kuttervirus S117 as a backbone, we demonstrated that acquiring tsp genes from Kuttervirus and Agtrevirus phages within the Ackermannviridae family led to altered host recognition. A fifth TSP was designed to interact with the branched complex and expand host recognition even further. Interestingly, the acquisition of tsp5 resulted in a recombination event between tsp4 and tsp5 or deletion of tsp3 and truncation of tsp4 genes. Our study provides insight into the development of the branched TSP complex, enabling Ackermannviridae phages to adapt to different hosts.
Collapse
Affiliation(s)
- Anders Nørgaard Sørensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Denmark
| | - Cedric Woudstra
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Denmark
| | - Dorottya Kalmar
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Denmark
| | - Jorien Poppeliers
- Laboratory of Gene Technology, KU Leuven, Kasteelpark Arenberg 21 Box 2462, 3001 Heverlee, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Kasteelpark Arenberg 21 Box 2462, 3001 Heverlee, Belgium
| | | | - Lone Brøndsted
- Department of Veterinary and Animal Sciences, University of Copenhagen, Stigbøjlen 4, 1870 Frederiksberg C, Denmark
| |
Collapse
|
38
|
Xu X, Meng Y, Su B, Lin J. Development of whole cell biocatalytic system for asymmetric synthesis of esomeprazole with enhancing coenzyme biosynthesis pathway. Enzyme Microb Technol 2024; 179:110469. [PMID: 38878426 DOI: 10.1016/j.enzmictec.2024.110469] [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: 05/20/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 08/07/2024]
Abstract
Esomeprazole is the most popular proton pump inhibitor for treating gastroesophageal reflux disease. Previously, a phenylacetone monooxygenase mutant LnPAMOmu15 (LM15) was obtained by protein engineering for asymmetric synthesis of esomeprazole using pyrmetazole as substrate. To scale up the whole cell asymmetric synthesis of esomeprazole and reduce the cost, in this work, an Escherichia coli whole-cell catalyst harboring LM15 and formate dehydrogenase from Burkholderia stabilis 15516 (BstFDH) were constructed through optimized gene assembly patterns. CRISPR/Cas9 mediated insertion of Ptrc promoter in genome was done to enhance the expression of key genes to increase the cellular NADP supply in the whole cell catalyst, by which the amount of externally added NADP+ for the asymmetric synthesis of esomeprazole decreased to 0.05 mM from 0.3 mM for reducing the cost. After the optimization of reaction conditions in the reactor, the scalable synthesis of esomeprazole was performed using the efficient LM15-BstFDH whole-cell as catalyst, which showed the highest reported space-time yield of 3.28 g/L/h with 50 mM of pyrmetazole loading. Isolation procedure was conducted to obtain esomeprazole sodium of 99.55 % purity and > 99.9 % ee with 90.1 % isolation yield. This work provides the basis for production of enantio-pure esomeprazole via cost-effective whole cell biocatalysis.
Collapse
Affiliation(s)
- Xinqi Xu
- Institute of Enzyme Catalysis and Synthetic Biotechnology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 360105, China
| | - Yaping Meng
- Institute of Enzyme Catalysis and Synthetic Biotechnology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 360105, China
| | - Bingmei Su
- Institute of Enzyme Catalysis and Synthetic Biotechnology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 360105, China.
| | - Juan Lin
- Institute of Enzyme Catalysis and Synthetic Biotechnology, College of Biological Science and Engineering, Fuzhou University, Fuzhou 360105, China.
| |
Collapse
|
39
|
Sun X, Zhang H, Jia Y, Li J, Jia M. CRISPR-Cas9-based genome-editing technologies in engineering bacteria for the production of plant-derived terpenoids. ENGINEERING MICROBIOLOGY 2024; 4:100154. [PMID: 39629108 PMCID: PMC11611024 DOI: 10.1016/j.engmic.2024.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 12/06/2024]
Abstract
Terpenoids are widely used as medicines, flavors, and biofuels. However, the use of these natural products is largely restricted by their low abundance in native plants. Fortunately, heterologous biosynthesis of terpenoids in microorganisms offers an alternative and sustainable approach for efficient production. Various genome-editing technologies have been developed for microbial strain construction. Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 9 (Cas9) is the most commonly used system owing to its outstanding efficiency and convenience in genome editing. In this review, the basic principles of CRISPR-Cas9 systems are briefly introduced and their applications in engineering bacteria for the production of plant-derived terpenoids are summarized. The aim of this review is to provide an overview of the current developments of CRISPR-Cas9-based genome-editing technologies in bacterial engineering, concluding with perspectives on the challenges and opportunities of these technologies.
Collapse
Affiliation(s)
- Xin Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Haobin Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuping Jia
- Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
| | - Jingyi Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meirong Jia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
40
|
Su B, Yang W, Zhou Y, Lin J. Efficiently manufacturing ectoine via metabolic engineering and protein engineering of L-2,4-diaminobutyrate transaminase. Int J Biol Macromol 2024; 275:133612. [PMID: 38960226 DOI: 10.1016/j.ijbiomac.2024.133612] [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: 04/26/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Ectoine, so-called tetrahydropyrimidine, is an important osmotic adjustment solute and widely applied in cosmetics and protein protectant. Some attempts have been made to improve the ectoine productivity. However, the strains with both high ectoine production capacity and high glucose conversion were still absent so far. Aim to construct a strain for efficiently producing ectoine, ectoine synthetic gene cluster ectABC from Pseudomonas stutzeri was overexpressed in E. coli BL21 (DE3). The ection production was improved by 382 % (ectoine titer increased from 1.73 g/L to 8.33 g/L) after the rational design of rate-limiting enzyme L-2,4-diaminobutyrate transaminase EctBps (protein engineering) combined with the metabolic engineering that focused on the enrichment and conversion of precursors. The final strain YW20 was applied to overproduce ectoine in fed-batch fermentation and yield 68.9 g/L of ectoine with 0.88 g/L/h of space-time yield and the highest glucose conversion reported [34 % (g/g)]. From the fermentation broth, ectoine was purified with 99.7 % purity and 79.8 % yield. This study successfully provided an engineered strain as well as an efficient method for the industrial bio-synthesis and preparation of ectoine.
Collapse
Affiliation(s)
- Bingmei Su
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; Institute of Enzyme Catalysis and Synthetic Biotechnology, Fuzhou University, Fuzhou 350108, China
| | - Wen Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Yi Zhou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Juan Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; Institute of Enzyme Catalysis and Synthetic Biotechnology, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
41
|
Gong Z, Zhang W, Chen J, Li J, Tan T. Upcycling CO2 into succinic acid via electrochemical and engineered Escherichia coli. BIORESOURCE TECHNOLOGY 2024; 406:130956. [PMID: 38871229 DOI: 10.1016/j.biortech.2024.130956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Converting CO2 into value-added chemicals still remains a grand challenge. Succinic acid has long been considered as one of the top building block chemicals. This study reported efficiently upcycling CO2 into succinic acid by combining between electrochemical and engineered Escherichia coli. In this process, the Cu-organic framework catalyst was synthesized for electrocatalytic CO2-to-ethanol conversion with high Faradaic efficiency (FE, 84.7 %) and relative purity (RP, 95 wt%). Subsequently, an engineered E. coli with efficiently assimilating CO2-derived ethanol to produce succinic acid was constructed by combining computational design and metabolic engineering, and the succinic acid titer reached 53.8 mM with the yield of 0.41 mol/mol, which is 82 % of the theoretical yield. This study effort to link the two processes of efficient ethanol synthesis by electrocatalytic CO2 and succinic acid production from CO2-derived ethanol, paving a way for the production of succinic acid and other value-added chemicals by converting CO2 into ethanol.
Collapse
Affiliation(s)
- Zhijin Gong
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wei Zhang
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiayao Chen
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jingchuan Li
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Tianwei Tan
- National Energy R&D Center for Biorefinery, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
42
|
Liu M, Li Z, Huang J, Yan J, Zhao G, Zhang Y. OptoLacI: optogenetically engineered lactose operon repressor LacI responsive to light instead of IPTG. Nucleic Acids Res 2024; 52:8003-8016. [PMID: 38860425 PMCID: PMC11260447 DOI: 10.1093/nar/gkae479] [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: 03/09/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
Optogenetics' advancement has made light induction attractive for controlling biological processes due to its advantages of fine-tunability, reversibility, and low toxicity. The lactose operon induction system, commonly used in Escherichia coli, relies on the binding of lactose or isopropyl β-d-1-thiogalactopyranoside (IPTG) to the lactose repressor protein LacI, playing a pivotal role in controlling the lactose operon. Here, we harnessed the light-responsive light-oxygen-voltage 2 (LOV2) domain from Avena sativa phototropin 1 as a tool for light control and engineered LacI into two light-responsive variants, OptoLacIL and OptoLacID. These variants exhibit direct responsiveness to light and darkness, respectively, eliminating the need for IPTG. Building upon OptoLacI, we constructed two light-controlled E. coli gene expression systems, OptoE.coliLight system and OptoE.coliDark system. These systems enable bifunctional gene expression regulation in E. coli through light manipulation and show superior controllability compared to IPTG-induced systems. We applied the OptoE.coliDark system to protein production and metabolic flux control. Protein production levels are comparable to those induced by IPTG. Notably, the titers of dark-induced production of 1,3-propanediol (1,3-PDO) and ergothioneine exceeded 110% and 60% of those induced by IPTG, respectively. The development of OptoLacI will contribute to the advancement of the field of optogenetic protein engineering, holding substantial potential applications across various fields.
Collapse
Affiliation(s)
- Meizi Liu
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300308, China
| | - Zuhui Li
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
- School of Biological Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jianfeng Huang
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| | - Junjun Yan
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Guoping Zhao
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yanfei Zhang
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| |
Collapse
|
43
|
McDonald ND, Antoshak EE. Towards a Yersinia pestis lipid A recreated in an Escherichia coli scaffold genome. Access Microbiol 2024; 6:000723.v3. [PMID: 39130741 PMCID: PMC11316592 DOI: 10.1099/acmi.0.000723.v3] [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: 10/11/2023] [Accepted: 06/26/2024] [Indexed: 08/13/2024] Open
Abstract
Synthetic biology and genome engineering capabilities have facilitated the utilization of bacteria for a myriad of applications, ranging from medical treatments to biomanufacturing of complex molecules. The bacterial outer membrane, specifically the lipopolysaccharide (LPS), plays an integral role in the physiology, pathogenesis, and serves as a main target of existing detection assays for Gram-negative bacteria. Here we use CRISPR/Cas9 recombineering to insert Yersinia pestis lipid A biosynthesis genes into the genome of an Escherichia coli strain expressing the lipid IVa subunit. We successfully inserted three genes: kdsD, lpxM, and lpxP into the E. coli genome and demonstrated their expression via reverse transcription PCR (RT-PCR). Despite observing expression of these genes, analytical characterization of the engineered strain's lipid A structure via MALDI-TOF mass spectrometry indicated that the Y. pestis lipid A was not recapitulated in the E. coli background. As synthetic biology and genome engineering technologies advance, novel applications and utilities for the detection and treatments of dangerous pathogens like Yersinia pestis will continue to be developed.
Collapse
Affiliation(s)
- Nathan D. McDonald
- United States Army Combat Capabilities Development Command Chemical Biological Center, 8908 Guard St. E3831, Gunpowder, MD 21010, USA
| | - Erin E. Antoshak
- United States Army Combat Capabilities Development Command Chemical Biological Center, 8908 Guard St. E3831, Gunpowder, MD 21010, USA
- Excet Inc. 6225 Brandon Ave #360, Springfield, VA 22150, USA
| |
Collapse
|
44
|
Meng H, Köbbing S, Blank LM. Establishing a straightforward I-SceI-mediated recombination one-plasmid system for efficient genome editing in P. putida KT2440. Microb Biotechnol 2024; 17:e14531. [PMID: 39031514 PMCID: PMC11258999 DOI: 10.1111/1751-7915.14531] [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: 01/24/2024] [Accepted: 07/04/2024] [Indexed: 07/22/2024] Open
Abstract
Pseudomonas putida has become an increasingly important chassis for producing valuable bioproducts. This development is not least due to the ever-improving genetic toolbox, including gene and genome editing techniques. Here, we present a novel, one-plasmid design of a critical genetic tool, the pEMG/pSW system, guaranteeing one engineering cycle to be finalized in 3 days. The pEMG/pSW system proved in the last decade to be valuable for targeted genome engineering in Pseudomonas, as it enables the deletion of large regions of the genome, the integration of heterologous gene clusters or the targeted generation of point mutations. Here, to expedite genetic engineering, two alternative plasmids were constructed: (1) The sacB gene from Bacillus subtilis was integrated into the I-SceI expressing plasmid pSW-2 as a counterselection marker to accelerated plasmid curing; (2) double-strand break introducing gene I-sceI and sacB counterselection marker were integrated into the backbone of the original pEMG vector, named pEMG-RIS. The single plasmid of pEMG-RIS allows rapid genome editing despite the low transcriptional activity of a single copy of the I-SceI encoding gene. Here, the usability of the pEMG-RIS is shown in P. putida KT2440 by integrating an expression cassette including an msfGFP gene in 3 days. In addition, a large fragment of 12.1 kb was also integrated. In summary, we present an updated pEMG/pSW genome editing system that allows efficient and rapid genome editing in P. putida. All plasmids designed in this study will be available via the Addgene platform.
Collapse
Affiliation(s)
- Hao Meng
- iAMB—Institute of Applied Microbiology, ABBt—Aachen Biology and BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Sebastian Köbbing
- iAMB—Institute of Applied Microbiology, ABBt—Aachen Biology and BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Lars M. Blank
- iAMB—Institute of Applied Microbiology, ABBt—Aachen Biology and BiotechnologyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
45
|
Zhang H, You J, Pan X, Hu Y, Zhang Z, Zhang X, Zhang W, Rao Z. Genomic and biological insights of bacteriophages JNUWH1 and JNUWD in the arms race against bacterial resistance. Front Microbiol 2024; 15:1407039. [PMID: 38989022 PMCID: PMC11233448 DOI: 10.3389/fmicb.2024.1407039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
The coevolution of bacteria and bacteriophages has created a great diversity of mechanisms by which bacteria fight phage infection, and an equivalent diversity of mechanisms by which phages subvert bacterial immunity. Effective and continuous evolution by phages is necessary to deal with coevolving bacteria. In this study, to better understand the connection between phage genes and host range, we examine the isolation and genomic characterization of two bacteriophages, JNUWH1 and JNUWD, capable of infecting Escherichia coli. Sourced from factory fermentation pollutants, these phages were classified within the Siphoviridae family through TEM and comparative genomic analysis. Notably, the phages exhibited a viral burst size of 500 and 1,000 PFU/cell, with latent periods of 15 and 20 min, respectively. They displayed stability over a pH range of 5 to 10, with optimal activity at 37°C. The complete genomes of JNUWH1 and JNUWD were 44,785 bp and 43,818 bp, respectively. Phylogenetic analysis revealed their close genetic relationship to each other. Antibacterial assays demonstrated the phages' ability to inhibit E. coli growth for up to 24 h. Finally, through laboratory-driven adaptive evolution, we successfully identified strains for both JNUWH1 and JNUWD with mutations in receptors specifically targeting lipopolysaccharides (LPS) and the lptD gene. Overall, these phages hold promise as additives in fermentation products to counter E. coli, offering potential solutions in the context of evolving bacterial resistance.
Collapse
Affiliation(s)
- Hengwei Zhang
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Jiajia You
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Xuewei Pan
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Yanglu Hu
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Zan Zhang
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Xian Zhang
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Weiguo Zhang
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zhiming Rao
- Key Laboratory of Industrial Biotechnology of the Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| |
Collapse
|
46
|
Sun M, Gao J, Tang H, Wu T, Ma Q, Zhang S, Zuo Y, Li Q. Increasing CRISPR/Cas9-mediated gene editing efficiency in T7 phage by reducing the escape rate based on insight into the survival mechanism. Acta Biochim Biophys Sin (Shanghai) 2024; 56:937-944. [PMID: 38761011 PMCID: PMC11294054 DOI: 10.3724/abbs.2024030] [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: 12/13/2023] [Accepted: 02/18/2024] [Indexed: 05/20/2024] Open
Abstract
Bacteriophages have been used across various fields, and the utilization of CRISPR/Cas-based genome editing technology can accelerate the research and applications of bacteriophages. However, some bacteriophages can escape from the cleavage of Cas protein, such as Cas9, and decrease the efficiency of genome editing. This study focuses on the bacteriophage T7, which is widely utilized but whose mechanism of evading the cleavage of CRISPR/Cas9 has not been elucidated. First, we test the escape rates of T7 phage at different cleavage sites, ranging from 10 -2 to 10 -5. The sequencing results show that DNA point mutations and microhomology-mediated end joining (MMEJ) at the target sites are the main causes. Next, we indicate the existence of the hotspot DNA region of MMEJ and successfully reduce MMEJ events by designing targeted sites that bypass the hotspot DNA region. Moreover, we also knock out the ATP-dependent DNA ligase 1. 3 gene, which may be involved in the MMEJ event, and the frequency of MMEJ at 4. 3 is reduced from 83% to 18%. Finally, the genome editing efficiency in T7 Δ 1. 3 increases from 20% to 100%. This study reveals the mechanism of T7 phage evasion from the cleavage of CRISPR/Cas9 and demonstrates that the special design of editing sites or the deletion of key gene 1. 3 can reduce MMEJ events and enhance gene editing efficiency. These findings will contribute to advancing CRISPR/Cas-based tools for efficient genome editing in phages and provide a theoretical foundation for the broader application of phages.
Collapse
Affiliation(s)
- Mingjun Sun
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Jie Gao
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Hongjie Tang
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Ting Wu
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Qinqin Ma
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Suyi Zhang
- Luzhou Laojiao CoLtdLuzhou646000China
- National Engineering Research Center of Solid-State BrewingLuzhou646000China
| | - Yong Zuo
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| | - Qi Li
- College of Life SciencesSichuan Normal UniversityChengdu610101China
| |
Collapse
|
47
|
Zhao W, Guo Y. Increasing the efficiency of gene editing with CRISPR-Cas9 via concurrent expression of the Beta protein. Int J Biol Macromol 2024; 270:132431. [PMID: 38759853 DOI: 10.1016/j.ijbiomac.2024.132431] [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: 02/13/2024] [Revised: 04/03/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Escherichia coli has emerged as an important host for the production of biopharmaceuticals or other industrially relevant molecules. An efficient gene editing tool is indispensable for ensuring high production levels and optimal release of target products. However, in Escherichia coli, the CRISPR-Cas9 system has been shown to achieve gene modifications with relatively low frequency. Large-scale PCR screening is required, hindering the identification of positive clones. The beta protein, which weakly binds to single-stranded DNA but tightly associates with complementary strand annealing products, offers a promising solution to this issue. In the present study, we describe a targeted and continuous gene editing strategy for the Escherichia coli genome. This strategy involves the coexpression of the beta protein alongside the CRISPR-Cas9 system, enabling a variety of genome modifications such as gene deletion and insertion with an efficiency exceeding 80 %. The integrity of beta proteins is essential for the CRISPR-Cas9/Beta-based gene editing system. In this work, the deletion of either the N- or C-terminal domain significantly impaired system efficiency. Overall, our findings established the CRISPR-Cas9/Beta system as a suitable gene editing tool for various applications in Escherichia coli.
Collapse
Affiliation(s)
- Weiyu Zhao
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, China; School of Economics and Management, Tongji University, No. 1239 Siping Road, Shanghai 200092, China; Institute of Logistics Science and Engineering, Shanghai Maritime University, 1550 Haigang Avenue, Shanghai 201306, China
| | - Yanan Guo
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, China; Department of Biology, Georgia State University, Atlanta, GA 30303, United States of America.
| |
Collapse
|
48
|
Wang Y, Chen E, Wang Y, Sun X, Dong Q, Chen P, Zhang C, Yang J, Sun Y. Biosynthesis of mannose from glucose via constructing phosphorylation-dephosphorylation reactions in Escherichia coli. Enzyme Microb Technol 2024; 177:110427. [PMID: 38518553 DOI: 10.1016/j.enzmictec.2024.110427] [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: 12/08/2023] [Revised: 02/20/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024]
Abstract
d-mannose has been widely used in food, medicine, cosmetic, and food-additive industries. To date, chemical synthesis or enzymatic conversion approaches based on iso/epimerization reactions for d-mannose production suffered from low conversion rate due to the reaction equilibrium, necessitating intricate separation processes for obtaining pure products on an industrial scale. To circumvent this challenge, this study showcased a new approach for d-mannose synthesis from glucose through constructing a phosphorylation-dephosphorylation pathway in an engineered strain. Specifically, the gene encoding phosphofructokinase (PfkA) in glycolytic pathway was deleted in Escherichia coli to accumulate fructose-6-phosphate (F6P). Additionally, one endogenous phosphatase, YniC, with high specificity to mannose-6-phosphate, was identified. In ΔpfkA strain, a recombinant synthetic pathway based on mannose-6-phosphate isomerase and YniC was developed to direct F6P to mannose. The resulting strain successfully produced 25.2 g/L mannose from glucose with a high conversion rate of 63% after transformation for 48 h. This performance surpassed the 15% conversion rate observed with 2-epimerases. In conclusion, this study presents an efficient method for achieving high-yield mannose synthesis from cost-effective glucose.
Collapse
Affiliation(s)
- Yuyao Wang
- Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, China; National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Enhui Chen
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanfei Wang
- Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, China; National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Xinming Sun
- Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, China; National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Qianzhen Dong
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Chen
- Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, China; National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Chenglin Zhang
- Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, China
| | - Jiangang Yang
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| | - Yuanxia Sun
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
49
|
Chai R, Guo J, Geng Y, Huang S, Wang H, Yao X, Li T, Qiu L. The Influence of Homologous Arm Length on Homologous Recombination Gene Editing Efficiency Mediated by SSB/CRISPR-Cas9 in Escherichia coli. Microorganisms 2024; 12:1102. [PMID: 38930484 PMCID: PMC11205466 DOI: 10.3390/microorganisms12061102] [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: 05/19/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
The precise editing of genes mediated by CRISPR-Cas9 necessitates the application of donor DNA with appropriate lengths of homologous arms and fragment sizes. Our previous development, SSB/CRISPR-Cas9, has demonstrated high efficiency in homologous recombination and non-homologous end joining gene editing within bacteria. In this study, we optimized the lengths and sizes of homologous arms of the donor DNA within this system. Two sets of donor DNA constructs were generated: one set comprised donors with only 10-100 bp homologous arms, while the other set included donors with homologous arms ranging from 10-100 bp, between which was a tetracycline resistance expression cassette (1439 bp). These donor constructs were transformed into Escherichia coli MG1655 cells alongside pCas-SSB/pTargetF-lacZ. Notably, when the homologous arms ranged from 10 to 70 bp, the transformation efficiency of non-selectable donors was significantly higher than that of selectable donors. However, within the range of 10-100 bp homologous arm lengths, the homologous recombination rate of selectable donors was significantly higher than that of non-selectable donors, with the gap narrowing as the homologous arm length increased. For selectable donor DNA with homologous arm lengths of 10-60 bp, the homologous recombination rate increased linearly, reaching a plateau when the homologous arm length was between 60-100 bp. Conversely, for non-selectable donor DNA, the homologous recombination rate increased linearly with homologous arm lengths of 10-90 bp, plateauing at 90-100 bp. Editing two loci simultaneously with 100 bp homologous arms, whether selectable or non-selectable, showed no difference in transformation or homologous recombination rates. Editing three loci simultaneously with 100 bp non-selectable homologous arms resulted in a 45% homologous recombination rate. These results suggest that efficient homologous recombination gene editing mediated by SSB/CRISPR-Cas9 can be achieved using donor DNA with 90-100 bp non-selectable homologous arms or 60-100 bp selectable homologous arms.
Collapse
Affiliation(s)
- Ran Chai
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
- College of Life Sciences, Henan Agricultural University, Key Laboratory of Enzyme Engineering of Agricultural Microbiology, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
| | - Jiaxiang Guo
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
| | - Yue Geng
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
| | - Shuai Huang
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
| | - Haifeng Wang
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
| | - Xinding Yao
- School of Environmental Engineering, Yellow River Conservancy Technical Institute, Henan Engineering Technology Research Center of Green Coating Materials, Kaifeng 475004, China; (R.C.)
| | - Tao Li
- College of Applied Engineering, Henan University of Science and Technology, Sanmenxia 472000, China
| | - Liyou Qiu
- College of Life Sciences, Henan Agricultural University, Key Laboratory of Enzyme Engineering of Agricultural Microbiology, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
| |
Collapse
|
50
|
Feng D, Zhang H, Li Z, Li Y, Yan J, Zhang Y, Yang Y. Categorization of the effects of E. coli LF82 and mutants lacking the chuT and shuU genes on survival, the transcriptome, and metabolome in germ-free honeybee. FEBS Open Bio 2024; 14:756-770. [PMID: 38403884 PMCID: PMC11073505 DOI: 10.1002/2211-5463.13776] [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: 09/19/2023] [Revised: 12/28/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
The precise etiology of inflammatory bowel diseases (IBDs) remains elusive. The Escherichia coli strain LF82 (LF82) is known to be associated with IBD, and we hypothesized that this association may be related to the chuT and shuU genes. Here we constructed a germ-free (GF) honeybee model to investigate the effects of LF82 chuT and shuU genes on the honeybee intestine and their mechanisms. The chuT and shuU gene deletion strains LF82∆chuT and LF82∆shuU were generated by CRISPR-Cas9. These strains, together with nonpathogenic E. coli MG1655 (MG1655) and wildtype LF82, were allowed to colonize the guts of GF honeybees to establish single bacterial colonization models. Intestinal permeability was assessed following the administration of a sterile Brilliant Blue (FCF) solution. Comprehensive transcriptomic and metabolomic analyses of intestinal samples indicated that MG1655 had few disadvantageous effects on honeybees. Conversely, colonization with LF82 and its gene-deletion mutants provoked pronounced activation of genes associated with innate immune pathways, stimulated defensive responses, and induced expression of genes associated with inflammation, oxidative stress, and glycosaminoglycan degradation. Crucially, the LF82∆chuT and LF82∆shuU strains perturbed host heme and iron regulation, as well as tryptophan metabolism. These findings suggest that the deletion of chuT and shuU genes in E. coli LF82 may alleviate intestinal inflammation by partially modulating tryptophan catabolism. Our study proposes that targeting iron uptake mechanisms could be a potential strategy to mitigate the virulence of IBD-associated bacteria.
Collapse
Affiliation(s)
- Dongping Feng
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Hujun Zhang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhengpeng Li
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yiyuan Li
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Jingshuang Yan
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yunsheng Yang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|