1
|
Gao YP, Lu JT, Zhang HJ, Cui ZM, Guo Y, Zhang X, Wang W, Qiu LL, Wang XY, Wang TY, Jia YL. MAT2A Knockdown Enhances Recombinant Protein Expression in Transgenic CHO Cells Through Regulation of Cell Cycle. Biotechnol Bioeng 2025; 122:1461-1471. [PMID: 40011400 DOI: 10.1002/bit.28962] [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: 12/14/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Chinese hamster ovary (CHO) cells represent the most widely utilized host system for industrial production of high-quality recombinant protein therapeutics. Novel CHO cell line development is achieved through genetic and cellular engineering approaches, effectively addressing limitations such as clonal variation and productivity loss during culture. Previous studies have established that MAT2A inhibition in tumor cells promotes expression of the cyclin-dependent kinase inhibitor p21, inducing antitumor activity. Notably, p21 induction has been shown to enhance recombinant protein expression in CHO cells by triggering cell cycle arrest. In this study, we identified MAT2A as a potential regulatory target, showing significant differential expression in transfected CHO cells with elevated versus diminished recombinant protein production. To investigate this phenomenon, we generated CHO cells with MAT2A knockdown (shMAT2A) and evaluated their recombinant protein output. Results demonstrated that MAT2A silencing enhanced recombiant protein/antibody production by 1.73-/1.70-fold through suppression of CyclinD1, thereby activating p21 and inducing G1 phase arrest. Furthermore, pharmacological inhibition of MAT2A using small molecules increased cell volume, boosted metabolic activity, and improved specific antibody productivity of recombiant protein/antibody production by 1.88-/2.16-fold in transfected CHO cells. These findings advance our understanding of MAT2A-mediated regulatory mechanisms and provide a strategic framework for developing high-efficiency CHO cell expression systems.
Collapse
Affiliation(s)
- Yan-Ping Gao
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Jiang-Tao Lu
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Hui-Jie Zhang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Zhao-Ming Cui
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yang Guo
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Xi Zhang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Wen Wang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Le-Le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Yin Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yan-Long Jia
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Catalán-Tatjer D, Kumar Ganesan S, Martínez-Monje I, Grav LM, Lavado-García J, Nielsen LK. Evaluating Apoptotic Gene Efficiency for CHO Culture Performance Using Targeted Integration. ACS Synth Biol 2025; 14:1414-1424. [PMID: 40268279 PMCID: PMC12090340 DOI: 10.1021/acssynbio.4c00382] [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: 05/31/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
Chinese hamster ovary (CHO) cells have long been the favored platform for producing complex biopharmaceuticals, such as monoclonal antibodies. Cell death is a critical factor in all CHO cultures, dictating the duration until harvest in batch cultures and viable cell density in perfusion. The programmed cell death, or apoptosis, pathway has been widely studied due to its relevance in affecting cell culture performance and the extensive knowledge about its protein-to-protein interaction network. However, clonal variation seen with random integration has confounded results, and it remains unclear which effector genes should be overexpressed. Here, we employed the recombinase-mediated cassette exchange strategy to develop isogenic cell lines expressing one copy of erythropoietin, as a model protein product, and various antiapoptotic genes: bcl-2 from CHO and human origin, bcl-xL from CHO and human origin, mcl-1, and bhrf-1. We tested the generated isogenic cell lines in the presence of sodium butyrate, a well-known apoptotic initiator, in a batch culture. The most promising candidates were cultured in fed-batch in the microbioreactor ambr15 system. The observed phenotype varied significantly depending on the overexpressed gene; therefore, the metabolic differences were further characterized using multiplexed quantitative proteomics. We showed that overexpressing bcl-2 from the CHO origin significantly improved productivity and established a methodology to successfully test candidate genes via targeted integration. This will enable future metabolic engineering strategies to be more comparable and overcome the challenges faced thus far.
Collapse
Affiliation(s)
- David Catalán-Tatjer
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Saravana Kumar Ganesan
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Iván Martínez-Monje
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Lise M. Grav
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Jesús Lavado-García
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Lars K. Nielsen
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
3
|
Budge JD. Mapping cellular processes that determine delivery of plasmid DNA to the nucleus: application in Chinese hamster ovary and human embryonic kidney cells to enhance protein production. Front Bioeng Biotechnol 2025; 13:1466671. [PMID: 40190711 PMCID: PMC11969153 DOI: 10.3389/fbioe.2025.1466671] [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: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 04/09/2025] Open
Abstract
Delivery of DNA into nucleated eukaryotic cells is known as transfection and has been essential in establishing technologies such as recombinant protein production and gene therapy. Considerable research efforts have led to development of a variety of transfection methods for a multitude of applications and cell types. Many methods are efficient in delivering DNA across the plasma membrane but few focus on subsequent delivery into the nucleus, a necessary step in expression of a recombinant transgene, and the cellular processes governing nuclear import of DNA during transfection have proved elusive. Herein, live confocal microscopy was used to track plasmid DNA during transfection of Chinese hamster ovary (CHO) and human embryonic kidney (HEK) cells to map key cellular processes central to nuclear import of DNA showing that there is a strong relationship between events of cell division, promotion of DNA dispersal from endosomes and subsequent nuclear import leading to gene expression. Furthermore, cationic lipid-mediated transfection is more dependent on events of the cell cycle than electroporation to deliver DNA into the nucleus. These findings have informed the design of a method where both CHO and HEK cells are synchronised at G2 phase of the cell cycle followed by timely release enabling cell cycle progression to maximise the frequency of division events immediately after transfection. This led to a 1.2-1.5 fold increase in transfection efficiency for polyethylenimine (PEI) mediated and electroporation transfection respectively. This process enhanced production yields of a monoclonal antibody 4.5 fold in HEK and 18 fold in CHO cells in the first 24 h post transfection. Overall, this study elucidated key cellular processes fundamental to transfection of CHO and HEK cells providing knowledge which can be applied to DNA delivery technologies in a plethora of fields.
Collapse
Affiliation(s)
- James D. Budge
- School of Natural Sciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
4
|
Balassi V, Otto M, Kretzmer C, Petersen A, McLaurin C, Mahadevan J, Gustin J, Borgschulte T, Razafsky D. Improving outcomes in intensified processing via optimization of the cell line development workflow. Biotechnol Prog 2025:e70003. [PMID: 39968655 DOI: 10.1002/btpr.70003] [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: 07/22/2024] [Revised: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
As the industry continues to explore the benefits of continuous and intensified manufacturing, it is important to assure that the cell line development (CLD) workflows in practice today are well suited to generate clones that meet the unique challenges associated with these processes. Most cell lines used in intensified processes are currently developed using traditional fed-batch CLD workflows followed by adaptation of these cell lines to perfusion processes. This method maybe suboptimal as fed-batch CLD workflows select clones which produce high volumetric titers irrespective of cell growth rate and specific productivity (qP). Although sufficient for fed-batch processes, performance of cells derived from this traditional CLD workflow may not be maintained in perfusion processes, where an intricate balance of performance parameters is needed. Until now, a thorough investigation into the effect of the CLD workflow on top clone performance in perfusion processes has not been conducted. Here, we show how the CLD workflow impacts cell performance in both fed-batch and perfusion processes, emphasizing the advantages of adopting a perfusion-specific CLD workflow which includes the use of medium specially designed for expansion and production in a perfusion setting, scale-down models which more accurately simulate perfusion process, and the adoption of perfusion-specific cell line selection criteria. Together, this results in the development of more efficient cell lines, fit for continuous and intensified processing.
Collapse
Affiliation(s)
- Vincent Balassi
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - Mary Otto
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - Corey Kretzmer
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - Amber Petersen
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - Channing McLaurin
- Cell Culture Media and Process Development, MilliporeSigma, Saint Louis, Missouri, USA
| | - Jana Mahadevan
- Cell Culture Media and Process Development, MilliporeSigma, Saint Louis, Missouri, USA
| | - Jason Gustin
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - Trissa Borgschulte
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| | - David Razafsky
- Expression Systems and Novel Biopharmaceutical Materials, MilliporeSigma, Saint Louis, Missouri, USA
| |
Collapse
|
5
|
Galli M, Liu LCY, Sim KH, Kok YJ, Wongtrakul-Kish K, Nguyen-Khuong T, Tate S, Bi X. SWATH-MS insights on sodium butyrate effect on mAbs production and redox homeostasis in CHO cells. AMB Express 2024; 14:140. [PMID: 39718710 DOI: 10.1186/s13568-024-01807-z] [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: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
Sodium butyrate (NaBu), well-known as a histone deacetylase inhibitor and for its capacity to impede cell growth, can enhance the production of a specific protein, such as an antibody, in recombinant Chinese hamster ovary (CHO) cell cultures. In this study, two CHO cell lines, namely K1 and DG44, along with their corresponding mAb-producing lines, K1-Pr and DG44-Pr, were cultivated with or without NaBu. A SWATH-based profiling method was employed to analyze the proteome. Cells cultured in the presence of NaBu exhibited a reduction in mitosis and gene expression, supported by their culture data demonstrating growth inhibition. The presence of NaBu corresponded to upregulation of intracellular trafficking and secretion pathways, aligned with an observed increase in mAb production, and was associated with an elevated glycosylation pathway and a slight alteration in the glycosylation profile of the mAbs. Increased fatty acid oxidation, redox interactions, and lipid biosynthesis were also observed and are likely attributable to the metabolism of NaBu. A comprehensive understanding of the systemic effects of NaBu will facilitate the discovery of strategies to enhance or prolong the productivity of CHO cells.
Collapse
Affiliation(s)
- Mauro Galli
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Lillian Chia-Yi Liu
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Kae Hwan Sim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Katherine Wongtrakul-Kish
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Terry Nguyen-Khuong
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | | | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
- Duke-NUS Medical School, 8 College Rd, Singapore, 169857, Singapore.
- Singapore Institute of Technology, 10 Dover Dr, Singapore, 138683, Singapore.
| |
Collapse
|
6
|
Jarusintanakorn S, Mastrobattista E, Yamabhai M. Ectoine enhances recombinant antibody production in Chinese hamster ovary cells by promoting cell cycle arrest. N Biotechnol 2024; 83:56-65. [PMID: 38945523 DOI: 10.1016/j.nbt.2024.06.006] [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: 02/07/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Chinese hamster ovary (CHO) cells represent the most preferential host cell system for therapeutic monoclonal antibody (mAb) production. Enhancing mAb production in CHO cells can be achieved by adding chemical compounds that regulate the cell cycle and cell survival pathways. This study investigated the impact of ectoine supplementation on mAb production in CHO cells. The results showed that adding ectoine at a concentration of 100 mM on the 3rd day of cultivation improved mAb production by improving cell viability and extending the culture duration. RNA sequencing analysis revealed differentially expressed genes associated with cell cycle regulation, cell proliferation, and cellular homeostasis, in particular promotion of cell cycle arrest, which was then confirmed by flow cytometry analysis. Ectoine-treated CHO cells exhibited an increase in the number of cells in the G0/G1 phase. In addition, the cell diameter was also increased. These findings support the hypothesis that ectoine enhances mAb production in CHO cells through mechanisms involving cell cycle arrest and cellular homeostasis. Overall, this study highlights the potential of ectoine as a promising supplementation strategy to enhance mAb production not only in CHO cells but also in other cell lines.
Collapse
Affiliation(s)
- Salinthip Jarusintanakorn
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447, Sri-Ayuthaya Road, Rachathevi, Bangkok 10400, Thailand
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands.
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
7
|
Madabhushi SR, Chakravarty T, Kasza T, Padellan M, Atieh TB, Gupta B. Enhancing protein productivities in CHO cells through adenosine uptake modulation - Novel insights into cellular growth and productivity regulation. N Biotechnol 2024; 83:163-174. [PMID: 39151888 DOI: 10.1016/j.nbt.2024.08.500] [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: 05/21/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Maximizing production potential of recombinant proteins such as monoclonal antibodies (mAbs) in Chinese Hamster Ovary (CHO) cells is a key enabler of reducing cost of goods of biologics. In this study, we explored various strategies to utilize adenosine mediated effects in biologics manufacturing processes. Results show that supplementation of adenosine increases specific productivity by up to two-fold while also arresting cell growth. Introducing adenosine in intensified perfusion processes in a biphasic manner significantly enhanced overall productivity. Interestingly, adenosine effect was observed to be dependent on the cell growth state. Using specific receptor antagonists and inhibitors, we identified that ENTs (primarily Slc29a1) mediate the uptake of adenosine in CHO cell cultures. Transcriptomics data showed an inverse correlation between Slc29a1 expression levels and peak viable cell densities. Data suggests that in fed-batch cultures, adenosine can be produced extracellularly. Blocking Slc29a1 using ENT inhibitors such as DZD and DP alone or in combination with CD73 inhibitor, PSB12379, resulted in a twofold increase in peak viable cell densities as well as productivities in fed batch - a novel strategy that can be applied to biologics manufacturing processes. This is the first study that suggests that adenosine production/accumulation in CHO cell cultures can potentially regulate the transition of CHO cells from exponential to stationary phase. We also demonstrate strategies to leverage this regulatory mechanism to maximize the productivity potential of biologics manufacturing processes.
Collapse
Affiliation(s)
| | | | - Tomas Kasza
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Malik Padellan
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Balrina Gupta
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| |
Collapse
|
8
|
Fu Y, Han Z, Cheng W, Niu S, Wang T, Wang X. Improvement strategies for transient gene expression in mammalian cells. Appl Microbiol Biotechnol 2024; 108:480. [PMID: 39365308 PMCID: PMC11452495 DOI: 10.1007/s00253-024-13315-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: 07/09/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Mammalian cells are suitable hosts for producing recombinant therapeutic proteins, with Chinese hamster ovary (CHO) and human embryonic kidney 293 (HEK293) cells being the most commonly used cell lines. Mammalian cell expression system includes stable and transient gene expression (TGE) system, with the TGE system having the advantages of short cycles and simple operation. By optimizing the TGE system, the expression of recombinant proteins has been significantly improved. Here, the TGE system and the detailed and up-to-date improvement strategies of mammalian cells, including cell line, expression vector, culture media, culture processes, transfection conditions, and co-expression of helper genes, are reviewed. KEY POINTS: • Detailed improvement strategies of transient gene expression system of mammalian cells are reviewed • The composition of transient expression system of mammalian cell are summarized • Proposed optimization prospects for transient gene expression systems.
Collapse
Affiliation(s)
- Yushun Fu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zimeng Han
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wanting Cheng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuaichen Niu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Xiaoyin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
9
|
Han MM, Wang HT, Zhang HJ, Lu JT, Guo JL, Qiu LL, Zhang X, Wang XY, Wang TY, Jia YL. A novel dual-epigenetic inhibitor enhances recombinant monoclonal antibody expression in CHO cells. Appl Microbiol Biotechnol 2024; 108:467. [PMID: 39292268 PMCID: PMC11411004 DOI: 10.1007/s00253-024-13302-3] [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: 04/11/2024] [Revised: 08/20/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Epigenetic regulation plays a central role in the regulation of a number of cellular processes such as proliferation, differentiation, cell cycle, and apoptosis. In particular, small molecule epigenetic modulators are key elements that can effectively influence gene expression by precisely regulating the epigenetic state of cells. To identify useful small-molecule regulators that enhance the expression of recombinant proteins in Chinese hamster ovary (CHO) cells, we examined a novel dual-HDAC/LSD1 inhibitor I-4 as a supplement for recombinant CHO cells. Treatment with 2 μM I-4 was most effective in increasing monoclonal antibody production. Despite cell cycle arrest at the G1/G0 phase, which inhibits cell growth, the addition of the inhibitor at 2 µM to monoclonal antibody-expressing CHO cell cultures resulted in a 1.94-fold increase in the maximal monoclonal antibody titer and a 2.43-fold increase in specific monoclonal antibody production. In addition, I-4 significantly increased the messenger RNA levels of the monoclonal antibody and histone H3 acetylation and methylation levels. We also investigated the effect on HDAC-related isoforms and found that interference with the HDAC5 gene increased the monoclonal antibody titer by 1.64-fold. The results of this work provide an effective method of using epigenetic regulatory strategies to enhance the expression of recombinant proteins in CHO cells. KEY POINTS: • HDAC/LSD1 dual-target small molecule inhibitor can increase the expression level of recombinant monoclonal antibodies in CHO cells. • By affecting the acetylation and methylation levels of histones in CHO cells and downregulating HDAC5, the production of recombinant monoclonal antibodies increased. • It provides an effective pathway for applying epigenetic regulation strategies to enhance the expression of recombinant proteins.
Collapse
Affiliation(s)
- Ming-Ming Han
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Hai-Tong Wang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Hui-Jie Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jiang-Tao Lu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jia-Liang Guo
- Junji College, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Le-Le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xi Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiao-Yin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Yan-Long Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
10
|
Ghobadian H, Roshanzamir K, Mohammadhasan KA, Ostadi H, Zati Keikha R, Dolatkhah Baghan M, Talebkhan Y, Torkashvand F. Optimization of Culture Conditions to Improve Follicle-Stimulating Hormone Production by CHO-DG44 Cells in Serum-Free Medium. IRANIAN BIOMEDICAL JOURNAL 2024; 28:282-96. [PMID: 39901854 PMCID: PMC11829159 DOI: 10.61186/ibj.4160] [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: 03/11/2024] [Accepted: 05/27/2024] [Indexed: 02/05/2025]
Abstract
Background In the present study, we attempted to adapt an adherent and serum-dependent Chinese hamster ovary DG44 cell line to a serum-free suspension culture and optimize the culture condition to achieve a higher yield of recombinant human follicle stimulating hormone (r-hFSH) with acceptable quality. This approach helps to mitigate the risks associated with blood-borne pathogens, reduces lot-to-lot variability, and lowers costs, making it suitable for industrial processing and scale-up. Methods The cell adaptation was performed using different chemically defined SFM. This process was followed by optimization through statistical experimental design, focusing on selected physicochemical parameters, including chemical supplementation of the medium and temperature shift. Both small- and large-scale cultures were conducted to test the reproducibility of the optimized condition. The expressed protein was evaluated for comparability with the standard molecule according to the Pharmacopeia guidelines. Results response surface methodology (RSM) analysis indicated that supplementation of the culture medium with galactose and sodium butyrate (NaBu), along with a temperature downshift, were the main parameters leading to increased cell viability (10%), r-hFSH level (96%), and more importantly, the glycosylation content (49%) of r-hFSH compared to the control condition. As r-hFSH isoforms generated during in vivo post-translational modifications typically exhibit different serum/plasma half-lives and bioactivity due to their incorporated sialic acid content/glycosylation, further optimizations of r-hFSH production are necessary to enhance its biological activity. In this study, following a primary screening of the studied parameters, optimization of culture conditions based on selected parameters resulted in enhanced quality and quantity of the produced r-hFSH. However, further examination is necessary before transitioning to industrial production.
Collapse
Affiliation(s)
- Hanna Ghobadian
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Khashayar Roshanzamir
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Kouhi Abdolabadi Mohammadhasan
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Hadi Ostadi
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Reza Zati Keikha
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Mohammad Dolatkhah Baghan
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Yeganeh Talebkhan
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
11
|
Chakrabarti L, Savery J, Mpindi JP, Klover J, Li L, Zhu J. Simplifying stable CHO cell line generation with high probability of monoclonality by using microfluidic dispensing as an alternative to fluorescence activated cell sorting. Biotechnol Prog 2024; 40:e3441. [PMID: 38462762 DOI: 10.1002/btpr.3441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 03/12/2024]
Abstract
Single cell cloning is a critical step for cell line development (CLD) for therapeutic protein production, with proof of monoclonality being compulsorily sought in regulatory filings. Among the different single cell deposition technologies, we found that fluorescence activated cell sorting (FACS) offers high probability of monoclonality and can allow selective enrichment of the producer cells. However, FACS instruments are expensive and resource-intensive, have a large footprint, require highly skilled operators and take hours for setup, thereby complicating the cell line generation process. With the aim of finding an easy-to-use alternative to FACS, we identified a flow cytometry-based microfluidic cell dispenser, which presents a single cell sorting solution for biopharmaceutical CLD. The microfluidic cell dispenser is small, budget-friendly, easy-to-use, requires lower-cost consumables, permits flow cytometry-enabled multiparametric target cell enrichment and offers fast and gentle single cell dispensing into multiwell plates. Following comprehensive evaluation, we found that single cell deposition by the microfluidic cell dispenser resulted in >99% probability of monoclonality for production cell lines. Moreover, the clonally derived producer cell lines generated from the microfluidic cell dispenser demonstrated comparable or improved growth profiles and production capability compared to the FACS derived cell lines. Taken together, microfluidic cell dispensing can serve as a cost-effective, efficient and convenient alternative to FACS, simplifying the biopharmaceutical CLD platform with significant reductions in both scientist time and running costs.
Collapse
Affiliation(s)
- Lina Chakrabarti
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, USA
| | - James Savery
- Machine Learning & AI, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - John Patrick Mpindi
- Biostatistics, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Judith Klover
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, USA
| | - Lina Li
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, USA
| | - Jie Zhu
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, USA
| |
Collapse
|
12
|
Zhang X, Wang Y, Yi D, Zhang C, Ning B, Fu Y, Jia Y, Wang T, Wang X. Synergistic promotion of transient transgene expression in CHO cells by PDI/XBP-1s co-transfection and mild hypothermia. Bioprocess Biosyst Eng 2024; 47:557-565. [PMID: 38416261 DOI: 10.1007/s00449-024-02987-5] [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: 09/02/2023] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
Transient gene expression system is an important tool for rapid production of recombinant proteins in Chinese hamster ovary (CHO) cells. However, their low productivity is the main hurdle to overcome. An effective approach through which to obtain high protein yield involves targeting transcriptional, post-transcriptional events (PTEs), and culture conditions. Here, we investigated the effects of protein disulfide isomerase (PDI) and spliced X-box binding protein 1 (XBP-1s) co-overexpression combined with mild hypothermia on the transient yields of recombinant proteins in CHO cells. The results showed that the gene of interest (GOI) and the PDI/XBP-1s helper vector at a co-transfection ratio of 10:1 could obviously increase transient expression level of recombinant protein in CHO cells. However, PDI/XBP-1s overexpression had no significance effect on the mRNA levels of the recombinant protein, suggesting that it targeted PTEs. Moreover, the increased production was due to the enhancing of cell specific productivity, not related to cell growth, viability, and cell cycle. In addition, combined PDI/XBP-1s co-overexpression and mild hypothermia could further improve Adalimumab expression, compared to the control/37 °C and PDI/XBP-1s/37 °C, the Adalimumab volume yield of PDI/XBP-1s/33 °C increased by 203% and 142%, respectively. Mild hypothermia resulted in 3.52- and 2.33-fold increase in the relative mRNA levels of PDI and XBP-1s, respectively. In conclusion, the combination of PDI/XBP-1s overexpression and culture temperature optimization can achieve higher transient expression of recombinant protein, which provides a synergetic strategy to improve transient production of recombinant protein in CHO cells.
Collapse
Affiliation(s)
- Xi Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yaokun Wang
- The School of Medical Humanities, Xinxiang Medical University, Xinxiang, 453003, China
| | - Dandan Yi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chi Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Binhuan Ning
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yushun Fu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yanlong Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Xiaoyin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
13
|
Nelson L, Veling M, Farhangdoust F, Cai X, Huhn S, Soloveva V, Chang M. Transcriptomics and cell painting analysis reveals molecular and morphological features associated with fed-batch production performance in CHO recombinant clones. Biotechnol Bioeng 2023; 120:3177-3190. [PMID: 37555462 DOI: 10.1002/bit.28518] [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: 04/29/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 08/10/2023]
Abstract
Stable, highly productive mammalian cells are critical for manufacturing affordable and effective biological medicines. Establishing a rational design of optimal biotherapeutic expression systems requires understanding how cells support the high demand for efficient biologics production. To that end, we performed transcriptomics and high-throughput imaging studies to identify putative genes and morphological features that underpin differences in antibody productivity among clones from a Chinese hamster ovary cell line. During log phase growth, we found that the expression of genes involved in biological processes related to cellular morphology varied significantly between clones with high specific productivity (qP > 35 pg/cell/day) and low specific productivity (qP < 20 pg/cell/day). At Day 10 of a fed-batch production run, near peak viable cell density, differences in gene expression related to metabolism, epigenetic regulation, and proliferation became prominent. Furthermore, we identified a subset of genes whose expression predicted overall productivity, including glutathione synthetase (Gss) and lactate dehydrogenase A (LDHA). Finally, we demonstrated the feasibility of cell painting coupled with high-throughput imaging to assess the morphological properties of intracellular organelles in relation to growth and productivity in fed-batch production. Our efforts lay the groundwork for systematic elucidation of clone performance using a multiomics approach that can guide future process design strategies.
Collapse
Affiliation(s)
| | | | | | - Xuezhu Cai
- Merck & Co., Inc., Rahway, New Jersey, USA
| | - Steve Huhn
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | | |
Collapse
|
14
|
Misorin AK, Chernyshova DO, Karbyshev MS. State-of-the-Art Approaches to Heterologous Expression of Bispecific Antibodies Targeting Solid Tumors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1215-1231. [PMID: 37770390 DOI: 10.1134/s0006297923090031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/09/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023]
Abstract
Bispecific antibodies (bsAbs) are some of the most promising biotherapeutics due to the versatility provided by their structure and functional features. bsAbs simultaneously bind two antigens or two epitopes on the same antigen. Moreover, they are capable of directing immune effector cells to cancer cells and delivering various compounds (radionuclides, toxins, and immunologic agents) to the target cells, thus offering a broad spectrum of clinical applications. Current review is focused on the technologies used in bsAb engineering, current progress and prospects of these antibodies, and selection of various heterologous expression systems for bsAb production. We also discuss the platforms development of bsAbs for the therapy of solid tumors.
Collapse
|