1
|
Zhang J, Chang K, Tay J, Tiong E, Heng E, Seah T, Lim YW, Peh G, Lim YH, Wong FT, Beh CW. Hyper-porous encapsulation of microbes for whole cell biocatalysis and biomanufacturing. Microb Cell Fact 2025; 24:48. [PMID: 39994799 PMCID: PMC11852520 DOI: 10.1186/s12934-025-02675-3] [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: 12/19/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Biocatalysis using whole cell biotransformation presents an alternative approach to producing complex molecules when compared to traditional synthetic chemical processes. This method offers several advantages, including scalability, self-contained co-factor recycling systems, the use of cost-effective raw materials, and reduced purification costs. Notably, biotransformation using microbial consortia provides benefits over monocultures by enhancing biosynthesis efficiency and productivity through division of labor and a reduction in metabolic burden. However, reliably controlling microbial cell populations within a consortium remains a significant challenge. In this work, we address this challenge through mechanical constraints. We describe the encapsulation and immobilization of cells in a hyper-porous hydrogel block, using methods and materials that are designed to be amenable to industrial scale-up. The porosity of the block provides ample nutrient access to ensure good cell viability, while the mechanical properties of the hydrogel matrix were optimized for Escherichia coli encapsulation, effectively limiting their proliferation while sustaining recombinant protein production. We also demonstrated the potential of this method for achieving stable co-cultivation of microbes by maintaining two different microbial strains spatially in a single porous hydrogel block. Finally, we successfully applied encapsulation to enable biotransformation in a mixed culture. Unlike its non-encapsulated counterpart, encapsulated E. coli expressing RadH halogenase achieved halogenation of the genistein substrate in a co-culture with genistein-producing Streptomyces. Overall, our strategy of controlling microbial cell populations through physical constraints offers a promising approach for engineering synthetic microbial consortia for biotransformation at an industrial scale.
Collapse
Affiliation(s)
- Jingyi Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Keziah Chang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Joyce Tay
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Elaine Tiong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, #07-06, Singapore, 138673, Singapore
| | - Elena Heng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, #07-06, Singapore, 138673, Singapore
| | - Theresa Seah
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore, 117583, Singapore
| | - Yi Wee Lim
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 8 Biomedical Grove, Neuros, #07-01, Singapore, 138665, Singapore
| | - Guangrong Peh
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 8 Biomedical Grove, Neuros, #07-01, Singapore, 138665, Singapore
| | - Yee Hwee Lim
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 8 Biomedical Grove, Neuros, #07-01, Singapore, 138665, Singapore
| | - Fong Tian Wong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, #07-06, Singapore, 138673, Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 8 Biomedical Grove, Neuros, #07-01, Singapore, 138665, Singapore
| | - Cyrus W Beh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| |
Collapse
|
2
|
Wang Y, Zhang G, Zhou Z, Zhang N, Jiang H, Liu Y, Fu T, Zhu Y, Li J. Antitumor Activity of a Bispecific Chimera Targeting EGFR and Met in Gefitinib-Resistant Non-Small Cell Lung Cancer. Adv Healthc Mater 2025; 14:e2402884. [PMID: 39586988 DOI: 10.1002/adhm.202402884] [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: 08/03/2024] [Revised: 09/28/2024] [Indexed: 11/27/2024]
Abstract
Non-small cell lung cancers (NSCLC) frequently acquire resistance to tyrosine kinase inhibitors (TKI) due to epidermal growth factor receptor (EGFR) mutation or activation of the bypass pathway involving mesenchymal-epithelial transition factor (Met). To address this challenge, a bispecific nanobody-aptamer chimera is designed to target mutated EGFR and Met simultaneously to block their cross-talk in NSCLC. The EGFR-Met chimera is cost-effectively engineered using microbial transglutaminase and click chemistry strategies. With enhanced binding affinity toward the target proteins, the as-developed chimera inhibits efficiently the cross-talk between signaling pathways associated with EGFR and Met. This inhibition leads to the suppression of downstream pathways, such as Erk and Akt, and induces upregulation of cell cycle arrest-related proteins, including Rb, p21, and p27. Additionally, the chimera activates the caspase-dependent apoptotic signaling pathway. Consequently, it inhibits cell migration, induces cell death, and causes cell cycle arrest in vitro. Moreover, the chimera exhibits significant antitumor efficacy in drug-resistant xenograft mouse models, showcasing improved tissue penetration and low toxicity. This study accentuates the potential of the bispecific EGFR-Met chimera as a promising therapeutic option for NSCLC resistant to EGFR TKIs.
Collapse
Affiliation(s)
- Ya Wang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Guixi Zhang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Zhilan Zhou
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Ning Zhang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Hang Jiang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yichang Liu
- School of Pharmacy, Nantong University, Nantong, 226019, China
| | - Ting Fu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yingdi Zhu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Juan Li
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| |
Collapse
|
3
|
Ariyoshi R, Matsuzaki T, Sato R, Minamihata K, Hayashi K, Koga T, Orita K, Nishioka R, Wakabayashi R, Goto M, Kamiya N. Engineering the Propeptide of Microbial Transglutaminase Zymogen: Enabling Substrate-Dependent Activation for Bioconjugation Applications. Bioconjug Chem 2024; 35:340-350. [PMID: 38421254 DOI: 10.1021/acs.bioconjchem.3c00544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Microbial transglutaminase (MTG) from Streptomyces mobaraensis is a powerful biocatalytic glue for site-specific cross-linking of a range of biomolecules and synthetic molecules that have an MTG-reactive moiety. The preparation of active recombinant MTG requires post-translational proteolytic digestion of a propeptide that functions as an intramolecular chaperone to assist the correct folding of the MTG zymogen (MTGz) in the biosynthesis. Herein, we report engineered active zymogen of MTG (EzMTG) that is expressed in soluble form in the host Escherichia coli cytosol and exhibits cross-linking activity without limited proteolysis of the propeptide. We found that the saturation mutagenesis of residues K10 or Y12 in the propeptide domain generated several active MTGz mutants. In particular, the K10D/Y12G mutant exhibited catalytic activity comparable to that of mature MTG. However, the expression level was low, possibly because of decreased chaperone activity and/or the promiscuous substrate specificity of MTG, which is potentially harmful to the host cells. The K10R/Y12A mutant exhibited specific substrate-dependent reactivity toward peptidyl substrates. Quantitative analysis of the binding affinity of the mutated propeptides to the active site of MTG suggested an inverse relationship between the binding affinity and the catalytic activity of EzMTG. Our proof-of-concept study provides insights into the design of a new biocatalyst using the MTGz as a scaffold and a potential route to high-throughput screening of EzMTG mutants for bioconjugation applications.
Collapse
Affiliation(s)
- Ryutaro Ariyoshi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Takashi Matsuzaki
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Ryo Sato
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kounosuke Hayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Taisei Koga
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kensei Orita
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Riko Nishioka
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| |
Collapse
|
4
|
Li Z, Xing S, Liu J, Wu X, Zhang S, Ma D, Liu X. Chaperonin co-expression and chemical modification enables production of active microbial transglutaminase from E. coli cytoplasm. Int J Biol Macromol 2023; 253:127355. [PMID: 37838118 DOI: 10.1016/j.ijbiomac.2023.127355] [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: 06/07/2023] [Revised: 09/21/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023]
Abstract
Microbial transglutaminase (MTG) is a usable enzyme for biomacromolecule modification. In the present study, a "molecular chaperonin" strategy was developed to produce MTG in E. coli cytoplasm with high expression level and a "small molecule-mediated chemical modification" strategy was adopted to strip propeptide chaperonin efficiently during purification. Propeptide (Pro) was expressed separately as a chaperonin to facilitate MTG expression in E. coli cytoplasm with a yield up to 300 mg or about 9 kU from 1 L fed-batch culture. Furthermore, small molecular chemicals were applied to interfere the interaction between MTG and Pro. Chemical acetylation was identified as a suitable method to strip Pro resulting in pure MTG with high specific activity up to 49.6 U/mg. The purified acetylated MTG was characterized by MS analysis. The deconvoluted mass and Peptide Sequence Tags analysis confirmed acetylation on amino groups of MTG protein. Finally, the applications of obtained MTG were demonstrated via protein polymerization of bovine serum albumin and PEGylation of human interferon-α2b. Our method provides MTG with high purity and specific activity as well as unique merit with masked amino groups thus avoiding self-polymerization and cross-linking between MTG and substrates.
Collapse
Affiliation(s)
- Zitao Li
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China
| | - Shuang Xing
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China
| | - Jing Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China; School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276826, China
| | - Xiaocong Wu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China
| | - Sichao Zhang
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China
| | - Di Ma
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China
| | - Xianwei Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, Shandong, China.
| |
Collapse
|
5
|
Lerner A, Benzvi C, Vojdani A. Cross-reactivity and sequence similarity between microbial transglutaminase and human tissue antigens. Sci Rep 2023; 13:17526. [PMID: 37845267 PMCID: PMC10579360 DOI: 10.1038/s41598-023-44452-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023] Open
Abstract
Microbial transglutaminase (mTG) is a bacterial survival factor, frequently used as a food additive to glue processed nutrients. As a result, new immunogenic epitopes are generated that might drive autoimmunity. Presently, its contribution to autoimmunity through epitope similarity and cross-reactivity was investigated. Emboss Matcher was used to perform sequence alignment between mTG and various antigens implicated in many autoimmune diseases. Monoclonal and polyclonal antibodies made specifically against mTG were applied to 77 different human tissue antigens using ELISA. Six antigens were detected to share significant homology with mTG immunogenic sequences, representing major targets of common autoimmune conditions. Polyclonal antibody to mTG reacted significantly with 17 out of 77 tissue antigens. This reaction was most pronounced with mitochondrial M2, ANA, and extractable nuclear antigens. The results indicate that sequence similarity and cross-reactivity between mTG and various tissue antigens are possible, supporting the relationship between mTG and the development of autoimmune disorders 150W.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel.
- Ariel University, Ariel, Israel.
| | - Carina Benzvi
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| | | |
Collapse
|
6
|
Kolotylo V, Piwowarek K, Kieliszek M. Microbiological transglutaminase: Biotechnological application in the food industry. Open Life Sci 2023; 18:20220737. [PMID: 37791057 PMCID: PMC10543708 DOI: 10.1515/biol-2022-0737] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 10/05/2023] Open
Abstract
Microbial transglutaminases (mTGs) belong to the family of global TGs, isolated and characterised by various bacterial strains, with the first being Streptomyces mobaraensis. This literature review also discusses TGs of animal and plant origin. TGs catalyse the formation of an isopeptide bond, cross-linking the amino and acyl groups. Due to its broad enzymatic activity, TG is extensively utilised in the food industry. The annual net growth in the utilisation of enzymes in the food processing industry is estimated to be 21.9%. As of 2020, the global food enzymes market was valued at around $2.3 billion USD (mTG market was estimated to be around $200 million USD). Much of this growth is attributed to the applications of mTG, benefiting both producers and consumers. In the food industry, TG enhances gelation and modifies emulsification, foaming, viscosity, and water-holding capacity. Research on TG, mainly mTG, provides increasing insights into the wide range of applications of this enzyme in various industrial sectors and promotes enzymatic processing. This work presents the characteristics of TGs, their properties, and the rationale for their utilisation. The review aims to provide theoretical foundations that will assist researchers worldwide in building a methodological framework and furthering the advancement of biotechnology research.
Collapse
Affiliation(s)
- Vitaliy Kolotylo
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences – SGGW, Nowoursynowska 159 C, 02-776Warsaw, Poland
| | - Kamil Piwowarek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences – SGGW, Nowoursynowska 159 C, 02-776Warsaw, Poland
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences – SGGW, Nowoursynowska 159 C, 02-776Warsaw, Poland
| |
Collapse
|
7
|
Khim Chan S, Yi Lai J, Gan CY, Soon Lim T. A Semi-Rational Mutagenesis Approach For Improved Substrate Activity Of Microbial Transglutaminase. Food Chem 2023; 419:136070. [PMID: 37030209 DOI: 10.1016/j.foodchem.2023.136070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
A higher specific activity of microbial transglutaminase (mTGase) is desirable for a broad range of applications ranging from food industry to biotechnology. Three-dimensional docking simulation of mTGase revealed that residues V65, W69, and Y75 were critical for substrate recognition. A semi-rational mutagenesis approach was applied to each residue to generate three separate mini mutant libraries. A high-throughput screening process identified five mutants that demonstrated improved specific activities than the wild type (WT) mTGase were isolated from the Y75 mini mutant library. Mutant Y75L showed approximately 60% increment in specific activity and improved substrate specificity. Conjugation of two heterologous single-chain fragment variable clones to generate a diabody with mutant Y75L was successfully performed and validated. This work demonstrates the successful application of semi-rational mutagenesis coupled with a high-throughput screening approach to identify mTGase mutants with improved specific activities and specificities which are beneficial for protein-protein conjugation.
Collapse
|
8
|
Yamazaki S, Shikida N, Takahashi K, Matsuda Y, Inoue K, Shimbo K, Mihara Y. Lipoate-acid ligase a modification of native antibody: Synthesis and conjugation site analysis. Bioorg Med Chem Lett 2021; 51:128360. [PMID: 34537330 DOI: 10.1016/j.bmcl.2021.128360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/10/2021] [Indexed: 12/23/2022]
Abstract
Bioconjugation is an important chemical biology research focus, especially in the development of methods to produce pharmaceutical bioconjugates and antibody-drug conjugates (ADCs). In this report, an enzyme-catalyzed conjugation method combined with a chemical reaction was used to modify a native antibody under mild reaction conditions. Our investigation revealed that lipoic-acid ligase (LplA) modifies native IgG1 with biased site-specificity. An intact mass analysis revealed that 98.3% of IgG1 was modified by LplA and possessed at least one molecule of octanocic acid. The average number of modifications per antibody was calculated to be 4.6. Peptide mapping analysis revealed that the modified residues were K225, K249 and K363 in the Fc region, and K30, K76 and K136 in the heavy chain and K39/K42, K169, K188 and K190 in the light chain of the Fab region. Careful evaluation including solvent exposed amino acid analysis suggested that these conjugate sites were not only solvent exposed but also biased by the site-specificity of LplA. Furthermore, antibody fragment conjugation may be able to take advantage of this enzymatic approach. This feasibility study serves as a demonstration for preparing enzymatically modified antibodies with conjugation site analysis.
Collapse
Affiliation(s)
- Shunsuke Yamazaki
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Natsuki Shikida
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | | | - Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kota Inoue
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kazutaka Shimbo
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Yasuhiro Mihara
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| |
Collapse
|
9
|
Matsuda Y, Seki T, Yamada K, Ooba Y, Takahashi K, Fujii T, Kawaguchi S, Narita T, Nakayama A, Kitahara Y, Mendelsohn BA, Okuzumi T. Chemical Site-Specific Conjugation Platform to Improve the Pharmacokinetics and Therapeutic Index of Antibody-Drug Conjugates. Mol Pharm 2021; 18:4058-4066. [PMID: 34579528 DOI: 10.1021/acs.molpharmaceut.1c00473] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To overcome a lack of selectivity during the chemical modification of native non-engineered antibodies, we have developed a technology platform termed "AJICAP" for the site-specific chemical conjugation of antibodies through the use of a class of IgG Fc-affinity reagents. To date, a limited number of antibody-drug conjugates (ADCs) have been synthesized via this approach, and no toxicological study was reported. Herein, we describe the compatibility and robustness of AJICAP technology, which enabled the synthesis of a wide variety of ADCs. A stability assessment of a thiol-modified antibody synthesized by AJICAP technology indicated no appreciable increase in aggregation or decomposition upon prolonged storage, indicating that the unexpectedly stable thiol intermediate has a great potential intermediate for payload or linker screening or large-scale manufacturing. Payload conjugation with this stable thiol intermediate generated several AJICAP-ADCs. In vivo xenograft studies indicated that the AJICAP-ADCs displayed significant tumor inhibition comparable to benchmark ADC Kadcyla. Furthermore, a rat pharmacokinetic analysis and toxicology study indicated an increase in the maximum tolerated dose, demonstrating an expansion of the AJICAP-ADC therapeutic index, compared with stochastic conjugation technology. This is the first report of the therapeutic index estimation of site-specific ADCs produced by utilizing Fc affinity reagent conjugation. The described site-specific conjugation technology is a powerful platform to enable next-generation ADCs through reduced heterogeneity and enhanced therapeutic index.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan.,Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, California 92121, United States
| | - Takuya Seki
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Kei Yamada
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Yuri Ooba
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Kazutoshi Takahashi
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Tomohiro Fujii
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Sayaka Kawaguchi
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Takahiro Narita
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Akira Nakayama
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Yoshiro Kitahara
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| | - Brian A Mendelsohn
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, California 92121, United States
| | - Tatsuya Okuzumi
- Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-8681, Japan
| |
Collapse
|
10
|
Lerner A, Benzvi C. Microbial Transglutaminase Is a Very Frequently Used Food Additive and Is a Potential Inducer of Autoimmune/Neurodegenerative Diseases. TOXICS 2021; 9:233. [PMID: 34678929 PMCID: PMC8537092 DOI: 10.3390/toxics9100233] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Microbial transglutaminase (mTG) is a heavily used food additive and its industrial transamidated complexes usage is rising rapidly. It was classified as a processing aid and was granted the GRAS (generally recognized as safe) definition, thus escaping full and thorough toxic and safety evaluations. Despite the manufacturers claims, mTG or its cross-linked compounds are immunogenic, pathogenic, proinflammatory, allergenic and toxic, and pose a risk to public health. The enzyme is a member of the transglutaminase family and imitates the posttranslational modification of gluten, by the tissue transglutaminase, which is the autoantigen of celiac disease. The deamidated and transamidated gliadin peptides lose their tolerance and induce the gluten enteropathy. Microbial transglutaminase and its complexes increase intestinal permeability, suppresses enteric protective pathways, enhances microbial growth and gliadin peptide's epithelial uptake and can transcytose intra-enterocytically to face the sub-epithelial immune cells. The present review updates on the potentially detrimental side effects of mTG, aiming to interest the scientific community, induce food regulatory authorities' debates on its safety, and protect the public from the mTG unwanted effects.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel;
- Ariel University, Ariel 40700, Israel
| | - Carina Benzvi
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel;
| |
Collapse
|
11
|
Akbari M, Razavi SH, Kieliszek M. Recent advances in microbial transglutaminase biosynthesis and its application in the food industry. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.02.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
12
|
Multiplexed small molecule impurity monitoring in antibody-based therapeutics by mixed-mode chromatography paired with charged aerosol detection. J Pharm Biomed Anal 2021; 197:113952. [PMID: 33601156 DOI: 10.1016/j.jpba.2021.113952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/23/2021] [Accepted: 01/30/2021] [Indexed: 12/27/2022]
Abstract
With advanced genetic engineering technologies and better understanding of disease biology, antibody-based therapeutics are emerging as promising new generation biopharmaceuticals. These novel antibody formats are carefully designed to possess desired features such as enhanced selectivity. However, their high level of structural complexity with multiple components often leads to long development and complex multi-step manufacturing processes, through which a variety of potential small molecule impurities can be introduced. In this work, an in-process assay was developed in which mixed-mode chromatography coupled with charged aerosol detection was utilized for multiplexed detection of nine reagents commonly used in development and manufacturing of antibody-based therapeutics: isopropyl β-d-1-thiogalactopyranoside, methionine sulfoximine, ampicillin, guanidine, dehydroascorbic acid, glutathione, tris(2-carboxyethyl)phosphine, N-acetyl cysteine, and arginine. This method utilized a mixed-mode column with ion-exchange properties operated in the hydrophilic interaction chromatography mode. Various parameters were systematically optimized and under optimal conditions, the method demonstrated excellent specificity, sensitivity, linearity, precision, accuracy, and was successfully applied to determine residual impurities in multiple samples from antibody-derived molecules.
Collapse
|
13
|
Matsuda Y, Mendelsohn BA. An overview of process development for antibody-drug conjugates produced by chemical conjugation technology. Expert Opin Biol Ther 2020; 21:963-975. [PMID: 33141625 DOI: 10.1080/14712598.2021.1846714] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: We discuss chemical conjugation strategies for antibody-drug conjugates (ADCs) from an industrial perspective and compare three promising chemical conjugation technologies to produce site-specific ADCs.Areas covered: Currently, nine ADCs are commercially approved and all are produced by chemical conjugation technology. However, seven of these ADCs contain a relatively broad drug distribution, potentially limiting their therapeutic indices. In 2019, the first site-specific ADC was launched on the market by Daiichi-Sankyo. This achievement, and an analysis of clinical trials over the last decade, indicates that current industrial interest in the ADC field is shifting toward site-specific conjugation technologies. From an industrial point of view, we aim to provide guidance regarding established conjugation methodologies that have already been applied to scale-up stages. With an emphasis on highly productive, scalable, and synthetic process robustness, conjugation methodologies for ADC production is discussed herein.Expert opinion: All three chemical conjugation technologies described in this review have various advantages and disadvantages, therefore drug developers can utilize these depending on their biological and/or protein targets. The future landscape of the ADC field is also discussed.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Brian A Mendelsohn
- Process Development & Tech Transfer, Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA 92121, United States
| |
Collapse
|
14
|
Glodowsky AP, Ruberto LA, Martorell MM, Mac Cormack WP, Levin GJ. Cold active transglutaminase from antarctic Penicillium chrysogenum: Partial purification, characterization and potential application in food technology. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2020. [DOI: 10.1016/j.bcab.2020.101807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
15
|
Lu X, Poon TCW, Zhang H. Mass production of active recombinant Chryseobacterium proteolyticum protein glutaminase in Escherichia coli using a sequential dual expression system and one-step purification. IUBMB Life 2020; 72:2391-2399. [PMID: 32827356 DOI: 10.1002/iub.2358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/24/2023]
Abstract
Protein glutaminase (PG) is an enzyme that specifically catalyzes the deamidation of glutamine residues on proteins or peptides, remarkably improving the solubility, emulsification and foaming properties of food proteins and, thereby, conferring great potential in food industry applications. PG is primarily produced from wild strains of Chryseobacterium proteolyticum and the low enzyme production yield restricts large-scale industrial applications. In this context, by evaluating different cleavage site insertions between the pro-region and mature domain of PG as well as different linkers flanking the cleavage site, an E. coli expression and purification protocol has been developed to produce active recombinant PG. To simplify the production workflow, we developed a sequential dual expression system. More than 15 mg of pure and active PG was obtained from 1 L of shaking-flask bacteria culture by one-step nickel affinity chromatography purification. The enzymatic characteristics of the recombinant PG protein were similar to those of native PG. For the deamidation effect of recombinant PG, the deamidation degree (DD) of gliadin reached up to 67% and the solubility increased 84-fold. Thus, this study provides a practical approach to mass producing active PG proteins and investigates its potential applications on food proteins.
Collapse
Affiliation(s)
- Xin Lu
- Pilot Laboratory, Institute of Translational Medicine, Centre for Precision Medicine Research and Training, Faculty of Health sciences, University of Macau, Macau, China.,Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Terence Chuen Wai Poon
- Pilot Laboratory, Institute of Translational Medicine, Centre for Precision Medicine Research and Training, Faculty of Health sciences, University of Macau, Macau, China
| | - Hongmin Zhang
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
16
|
Sato R, Minamihata K, Ariyoshi R, Taniguchi H, Kamiya N. Recombinant production of active microbial transglutaminase in E. coli by using self-cleavable zymogen with mutated propeptide. Protein Expr Purif 2020; 176:105730. [PMID: 32827662 DOI: 10.1016/j.pep.2020.105730] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 10/23/2022]
Abstract
Microbial transglutaminase from Streptomyces mobaraensis (MTG) has been widely used in food industry and also in research and medical applications, since it can site-specifically modify proteins by the cross-linking reaction of glutamine residue and the primary amino group. The recombinant expression system of MTG in E. coli provides better accessibility for the researchers and thus can promote further utilization of MTG. Herein, we report production of active and soluble MTG in E. coli by using a chimeric protein of tobacco etch virus (TEV) protease and MTG zymogen. A chimera of TEV protease and MTG zymogen with native propeptide resulted in active MTG contaminated with cleaved propeptide due to the strong interaction between the propeptide and catalytic domain of MTG. Introduction of mutations of K9R and Y11A to the propeptide facilitated dissociation of the cleaved propeptide from the catalytic domain of MTG and active MTG without any contamination of the propeptide was obtained. The specific activity of the active MTG was 22.7 ± 2.6 U/mg. The successful expression and purification of active MTG by using the chimera protein of TEV protease and MTG zymogen with mutations in the propeptide can advance the use of MTG and the researches using MTG mediated cross-linking reactions.
Collapse
Affiliation(s)
- Ryo Sato
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka, 819-0395, Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka, 819-0395, Japan.
| | - Ryutaro Ariyoshi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka, 819-0395, Japan
| | - Hiromasa Taniguchi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka, 819-0395, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka, 819-0395, Japan; Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744, Motooka, Fukuoka, 819-0395, Japan
| |
Collapse
|
17
|
Doti N, Caporale A, Monti A, Sandomenico A, Selis F, Ruvo M. A recent update on the use of microbial transglutaminase for the generation of biotherapeutics. World J Microbiol Biotechnol 2020; 36:53. [PMID: 32172335 DOI: 10.1007/s11274-020-02829-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/07/2020] [Indexed: 01/12/2023]
Abstract
The recent scientific progresses on the use of enzyme-mediated reactions in organic, non-aqueous and aqueous media have significantly supported the growing demand of new biotechnological and/or pharmacological products. Today, a plethora of microbial enzymes, used as biocatalysts, are available. Among these, microbial transglutaminases (MTGs) are broadly used for their ability to catalyse the formation of an isopeptide bond between the γ-amide group of glutamines and the ε-amino group of lysine. Due to their promiscuity towards primary amine-containing substrates and the more stringent specificity for glutamine-containing peptide sequences, several combined approaches can be tailored for different settings, making MTGs very attractive catalysts for generating protein-protein and protein small molecule's conjugates. The present review offers a recent update on the modifications attainable by MTG-catalysed bioreactions as reported between 2014 and 2019. In particular, we present a detailed and comparative overview on the MTG-based methods for proteins and antibodies engineering, with a particular outlook on the synthesis of homogeneous antibody-drug conjugates.
Collapse
Affiliation(s)
- N Doti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| | - A Caporale
- Institute of Crystallography, CNR (IC-CNR), c/o Area Science Park s.s. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Alessandra Monti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABIF), University L. Vanvitelli, Via Vivaldi, 43, 80100, Caserta, Italy
| | - A Sandomenico
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy
| | - Fabio Selis
- BioVIIIx R&D, Via B. Brin, 59C, 80142, Naples, Italy
| | - M Ruvo
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| |
Collapse
|
18
|
Duarte LS, Barsé LQ, Dalberto PF, da Silva WTS, Rodrigues RC, Machado P, Basso LA, Bizarro CV, Ayub MAZ. Cloning and expression of the Bacillus amyloliquefaciens transglutaminase gene in E. coli using a bicistronic vector construction. Enzyme Microb Technol 2020; 134:109468. [DOI: 10.1016/j.enzmictec.2019.109468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
|
19
|
Duarte L, Matte CR, Bizarro CV, Ayub MAZ. Transglutaminases: part I-origins, sources, and biotechnological characteristics. World J Microbiol Biotechnol 2020; 36:15. [PMID: 31897837 DOI: 10.1007/s11274-019-2791-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/20/2019] [Indexed: 12/17/2022]
Abstract
The transglutaminases form a large family of intracellular and extracellular enzymes that catalyze cross-links between protein molecules. Transglutaminases crosslinking properties are widely applied to various industrial processes, to improve the firmness, viscosity, elasticity, and water-holding capacity of products in the food and pharmaceutical industries. However, the extremely high costs of obtaining transglutaminases from animal sources have prompted scientists to search for new sources of these enzymes. Therefore, research has been focused on producing transglutaminases by microorganisms, which may present wider scope of use, based on enzyme-specific characteristics. In this review, we present an overview of the literature addressing the origins, types, reactions, and general characterizations of this important enzyme family. A second review will deal with transglutaminases applications in the area of food industry, medicine, pharmaceuticals and biomaterials, as well as applications in the textile and leather industries.
Collapse
Affiliation(s)
- Lovaine Duarte
- Biotechnology, Bioprocess, and Biocatalysis Group, Food Science and Technology Institute, Federal University of Rio Grande Do Sul, Av. Bento Gonçalves 9500, PO Box 15090, Porto Alegre, RS, 91501-970, Brazil
| | - Carla Roberta Matte
- Biotechnology, Bioprocess, and Biocatalysis Group, Food Science and Technology Institute, Federal University of Rio Grande Do Sul, Av. Bento Gonçalves 9500, PO Box 15090, Porto Alegre, RS, 91501-970, Brazil
| | - Cristiano Valim Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), 92A Building at TECNOPUC, 4592 Bento Gonçalves Avenue, Porto Alegre, 90650-001, Brazil
| | - Marco Antônio Záchia Ayub
- Biotechnology, Bioprocess, and Biocatalysis Group, Food Science and Technology Institute, Federal University of Rio Grande Do Sul, Av. Bento Gonçalves 9500, PO Box 15090, Porto Alegre, RS, 91501-970, Brazil.
| |
Collapse
|
20
|
Site Selective Antibody-Oligonucleotide Conjugation via Microbial Transglutaminase. Molecules 2019; 24:molecules24183287. [PMID: 31509944 PMCID: PMC6767100 DOI: 10.3390/molecules24183287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
Nucleic Acid Therapeutics (NATs), including siRNAs and AntiSense Oligonucleotides (ASOs), have great potential to drug the undruggable genome. Targeting siRNAs and ASOs to specific cell types of interest has driven dramatic improvement in efficacy and reduction in toxicity. Indeed, conjugation of tris-GalNAc to siRNAs and ASOs has shown clinical efficacy in targeting diseases driven by liver hepatocytes. However, targeting non-hepatic diseases with oligonucleotide therapeutics has remained problematic for several reasons, including targeting specific cell types and endosomal escape. Monoclonal antibody (mAb) targeting of siRNAs and ASOs has the potential to deliver these drugs to a variety of specific cell and tissue types. However, most conjugation strategies rely on random chemical conjugation through lysine or cysteine residues resulting in conjugate heterogeneity and a distribution of Drug:Antibody Ratios (DAR). To produce homogeneous DAR-2 conjugates with two siRNAs per mAb, we developed a novel two-step conjugation procedure involving microbial transglutaminase (MTGase) tagging of the antibody C-terminus with an azide-functionalized linker peptide that can be subsequently conjugated to dibenzylcyclooctyne (DBCO) bearing oligonucleotides through azide-alkyne cycloaddition. Antibody-siRNA (and ASO) conjugates (ARCs) produced using this strategy are soluble, chemically defined targeted oligonucleotide therapeutics that have the potential to greatly increase the number of targetable cell types.
Collapse
|
21
|
Enzymatic activity and thermoresistance of improved microbial transglutaminase variants. Amino Acids 2019; 52:313-326. [DOI: 10.1007/s00726-019-02764-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/17/2019] [Indexed: 01/31/2023]
|
22
|
Bioengineering of microbial transglutaminase for biomedical applications. Appl Microbiol Biotechnol 2019; 103:2973-2984. [PMID: 30805670 DOI: 10.1007/s00253-019-09669-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
Microbial transglutaminase (mTGase) is commonly known in the food industry as meat glue due to its incredible ability to "glue" meat proteins together. Aside from being widely exploited in the meat processing industries, mTGase is also widely applied in other food and textile industries by catalysing the formation of isopeptide bonds between peptides or protein substrates. The advancement of technology has opened up new avenues for mTGase in the field of biomedical engineering. Efforts have been made to study the structural properties of mTGase in order to gain an in-depth understanding of the structure-function relationship. This review highlights the developments in mTGase engineering together with its role in biomedical applications including biomaterial fabrication for tissue engineering and biotherapeutics.
Collapse
|
23
|
Hilberg V, Avrutina O, Ebenig A, Yanakieva D, Meckel T, Biesalski M, Kolmar H. Light-Controlled Chemoenzymatic Immobilization of Proteins towards Engineering of Bioactive Papers. Chemistry 2019; 25:1746-1751. [DOI: 10.1002/chem.201804889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Indexed: 01/14/2023]
Affiliation(s)
- Valentina Hilberg
- Institut für Organische Chemie und Biochemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
- Merck Lab@; Technische Universität Darmstadt; Alarich-Weiss-Strasse 8 64287 Darmstadt Germany
| | - Olga Avrutina
- Institut für Organische Chemie und Biochemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
| | - Aileen Ebenig
- Institut für Organische Chemie und Biochemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
| | - Desislava Yanakieva
- Institut für Organische Chemie und Biochemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
- Merck Lab@; Technische Universität Darmstadt; Alarich-Weiss-Strasse 8 64287 Darmstadt Germany
| | - Tobias Meckel
- Institut für Technische und Makromolekulare Chemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 8 64287 Darmstadt Germany
- Merck Lab@; Technische Universität Darmstadt; Alarich-Weiss-Strasse 8 64287 Darmstadt Germany
| | - Markus Biesalski
- Institut für Technische und Makromolekulare Chemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 8 64287 Darmstadt Germany
| | - Harald Kolmar
- Institut für Organische Chemie und Biochemie; Technische Universität Darmstadt; Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
| |
Collapse
|
24
|
Wang L, Yu B, Wang R, Xie J. Biotechnological routes for transglutaminase production: Recent achievements, perspectives and limits. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
25
|
Yu T, Lin M, Wan J, Cao X. Molecular interaction mechanisms in reverse micellar extraction of microbial transglutaminase. J Chromatogr A 2017; 1511:25-36. [PMID: 28697931 DOI: 10.1016/j.chroma.2017.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/23/2017] [Accepted: 07/03/2017] [Indexed: 11/17/2022]
Abstract
Reverse micellar extraction is an efficient and economical alternative for protein purification. In this study, microbial transglutaminase (MTGase) from crude materials was purified using reverse micellar extraction, and the molecular interaction mechanism in reverse micellar extraction of MTGase was explored. By using a molecular simulation study, the interaction mechanism of forward extraction was investigated. The molecular simulation results reveal the interaction of MTGase-water-surfactant is the major driving force for the forward extraction. Further, the effect of ionic strength on molecular interactions in backward extraction was investigated using 1H low-field nuclear magnetic resonance (LF-NMR) and circular dichroism (CD) spectra. In backward extraction, the interactions between water and the other two molecules (MTGase and surfactant molecules) are enhanced while the interactions between target molecules (MTGase) and the other two molecules (water and surfactant molecules) are weakened as the ionic strength increases. Moreover, the effect of size exclusion on backward extraction was also investigated. The results demonstrate size exclusion has limit effect at high ionic strength, and the weakened interaction of MTGase-water-surfactant is the main reason causing the release of the target molecules in backward extraction. This work might provide valuable reference to the MTGase purification and downstream processing.
Collapse
Affiliation(s)
- Tingting Yu
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Mingxiang Lin
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Junfen Wan
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Xuejun Cao
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
26
|
Steffen W, Ko FC, Patel J, Lyamichev V, Albert TJ, Benz J, Rudolph MG, Bergmann F, Streidl T, Kratzsch P, Boenitz-Dulat M, Oelschlaegel T, Schraeml M. Discovery of a microbial transglutaminase enabling highly site-specific labeling of proteins. J Biol Chem 2017; 292:15622-15635. [PMID: 28751378 PMCID: PMC5612097 DOI: 10.1074/jbc.m117.797811] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/26/2017] [Indexed: 12/21/2022] Open
Abstract
Microbial transglutaminases (MTGs) catalyze the formation of Gln–Lys isopeptide bonds and are widely used for the cross-linking of proteins and peptides in food and biotechnological applications (e.g. to improve the texture of protein-rich foods or in generating antibody-drug conjugates). Currently used MTGs have low substrate specificity, impeding their biotechnological use as enzymes that do not cross-react with nontarget substrates (i.e. as bio-orthogonal labeling systems). Here, we report the discovery of an MTG from Kutzneria albida (KalbTG), which exhibited no cross-reactivity with known MTG substrates or commonly used target proteins, such as antibodies. KalbTG was produced in Escherichia coli as soluble and active enzyme in the presence of its natural inhibitor ammonium to prevent potentially toxic cross-linking activity. The crystal structure of KalbTG revealed a conserved core similar to other MTGs but very short surface loops, making it the smallest MTG characterized to date. Ultra-dense peptide array technology involving a pool of 1.4 million unique peptides identified specific recognition motifs for KalbTG in these peptides. We determined that the motifs YRYRQ and RYESK are the best Gln and Lys substrates of KalbTG, respectively. By first reacting a bifunctionalized peptide with the more specific KalbTG and in a second step with the less specific MTG from Streptomyces mobaraensis, a successful bio-orthogonal labeling system was demonstrated. Fusing the KalbTG recognition motif to an antibody allowed for site-specific and ratio-controlled labeling using low label excess. Its site specificity, favorable kinetics, ease of use, and cost-effective production render KalbTG an attractive tool for a broad range of applications, including production of therapeutic antibody-drug conjugates.
Collapse
Affiliation(s)
- Wojtek Steffen
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany,
| | - Fu Chong Ko
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany
| | - Jigar Patel
- Roche Sequencing, NimbleGen, Madison, Wisconsin 53719, and
| | | | | | - Jörg Benz
- F. Hoffmann-La Roche Ltd., pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Markus G Rudolph
- F. Hoffmann-La Roche Ltd., pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Frank Bergmann
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany
| | - Thomas Streidl
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany
| | - Peter Kratzsch
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany
| | | | | | - Michael Schraeml
- From Roche Diagnostics GmbH, CPS, Nonnenwald 2, 82377 Penzberg, Germany
| |
Collapse
|
27
|
Shi N, Xu H, Guo K, Kang C, Zhang W, Zhang Y, Zhang L, Tan J. Extraction of microbial transglutaminase from Amycolatopsis sp. fermentation broth using aqueous two-phase system. KOREAN J CHEM ENG 2017. [DOI: 10.1007/s11814-017-0105-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Javitt G, Ben-Barak-Zelas Z, Jerabek-Willemsen M, Fishman A. Constitutive expression of active microbial transglutaminase in Escherichia coli and comparative characterization to a known variant. BMC Biotechnol 2017; 17:23. [PMID: 28245818 PMCID: PMC5331659 DOI: 10.1186/s12896-017-0339-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/15/2017] [Indexed: 01/11/2023] Open
Abstract
Background Microbial transglutaminase (mTG) is a robust enzyme catalyzing the formation of an isopeptide bond between glutamine and lysine residues. It has found use in food applications, pharmaceuticals, textiles, and biomedicine. Overexpression of soluble and active mTG in E. coli has been limited due to improper protein folding and requirement for proteolytic cleavage of the pro-domain. Furthermore, to integrate mTG more fully industrially and academically, thermostable and solvent-stable variants may be imperative. Results A novel expression system constitutively producing active mTG was designed. Wild-type (WT) mTG and a S2P variant had similar expression levels, comparable to previous studies. Kinetic constants were determined by a glutamate dehydrogenase-coupled assay, and the S2P variant showed an increased affinity and a doubled enzyme efficiency towards Z-Gln-Gly. The melting temperature (Tm) of the WT was determined by intrinsic fluorescence measurements to be 55.8 ± 0.1 °C and of the S2P variant to be 56.3 ± 0.4 °C and 45.5 ± 0.1 °C, showing a moderately different thermostability profile. Stability in water miscible organic solvents was determined for both the WT and S2P variant. Of the solvents tested, incubation of mTG in isopropanol for 24 h at 4 °C showed the strongest stabilizing effect with mTG retaining 61 and 72% activity for WT and S2P respectively in 70% isopropanol. Both enzymes also showed an increased initial activity in the presence of organic solvents with the highest activity increase in 40% DMSO. Nevertheless, both enzymes were inactivated in 70% of all organic solvents tested. Conclusions A constitutive expression system of active mTG in E. coli without downstream proteolytic cleavage processing was used for overexpression and characterization. High throughput techniques for testing thermostability and kinetics were useful in streamlining analysis and could be used in the future for quickly identifying beneficial mutants. Hitherto untested thermostability and stability of mTG in organic solvents was evaluated, which can pave the way for use of the enzyme in novel applications and processes.
Collapse
Affiliation(s)
- Gabe Javitt
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zohar Ben-Barak-Zelas
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | | | - Ayelet Fishman
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
29
|
Li S, Ding Z, Cao X. Separation of transglutaminase by thermo-responsive affinity precipitation using l-thyroxin as ligand. SPRINGERPLUS 2016; 5:37. [PMID: 26835219 PMCID: PMC4713401 DOI: 10.1186/s40064-016-1680-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/06/2016] [Indexed: 01/10/2023]
Abstract
Transglutaminase (TGase) is widely used in the food industry. In this study, TGase was purified by affinity precipitation using l-thyroxin, coupled to a thermo-responsive polymer (PNBN), as an affinity ligand. The lower critical solution temperature and recovery of the affinity polymer were 31.0 °C and 99.6 %, respectively. The optimal adsorption condition was 0.02 mol/L phosphate buffer (pH 5.0). The recoveries 99.01 % (protein) and 98.85 % (activity) were obtained by 0.2 mol/L Gly-NaOH buffer (pH 10.0) as the elution agent. Circular dichroism spectroscopy and FortéBio Octet system were used to explore the interactions between l-thyroxin and TGase. The results show that l-thyroxin is suitable for affinity precipitation of TGase. The purity of the final product was verified using sodium dodecyl sulfate polyacrylamide gel electrophoresis.
Collapse
Affiliation(s)
- Sipeng Li
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Zhaoyang Ding
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Xuejun Cao
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, Shanghai, 200237 China
| |
Collapse
|