1
|
Duan H, Tang C, Han S, Yang N, Wang S, Gao F, Zhou Y, Tong G, Zhao K, Li L. Single-B-cell cloning and recombinant antibodies generation to analyze the antigenicity of porcine reproductive and respiratory syndrome virus nonstructural protein 12. Vet Microbiol 2025; 304:110506. [PMID: 40203677 DOI: 10.1016/j.vetmic.2025.110506] [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: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
The prevalence and variation of porcine reproductive and respiratory syndrome virus (PRRSV) in China are increasing. The rapid preparation of essential antibodies will effectively reveal the antigenicity, epitopes, and intracellular distribution of viral proteins. Single-B-cell antibody technology is a novel method for screening diverse functional monoclonal antibodies (mAbs). Herein, we successfully expressed PRRSV nonstructural protein 12 (Nsp12) in suspension-cultured Chinese hamster ovary (CHO) cells. Using single-B-cell antibody technology, we utilized fluorescence-activated cell sorting to collect individual immune B cells and prepared single-cell reverse transcription-polymerase chain reaction to clone the variable region of immunoglobulin heavy chain (IgH) and immunoglobulin light chain (IgK). Two recombinant mAbs were generated via transient transfection of CHO cells with the corresponding expression plasmids of IgH and IgK. A novel linear epitope (104YEFTGNGEDW113) of Nsp12 was identified using mAb1N14. This epitope was conserved in lineages 1, 5, and 8 of PRRSV-2 and was located on the surface of the Nsp12 spatial structure. The amino acid mutation in Nsp12 of lineage 3 PRRSV-2 affected the antigenicity of this linear epitope. A conserved conformational epitope was identified using mAb2S18, and the spatial structure of Nsp12 showed high similarity between PRRSV-1 and different lineages of PRRSV-2. During PRRSV infection, Nsp12 was distributed in the cytoplasm and accumulated in the nucleus. Overall, antigenicity analysis and novel epitope identification contributed to the in-depth exploration of the biological function of Nsp12 and will facilitate the development of detection assays and antiviral strategies.
Collapse
Affiliation(s)
- Hongyong Duan
- College of Life Science, Henan Normal University, Xinxiang 453007, PR China; Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chaozhi Tang
- College of Life Science, Henan Normal University, Xinxiang 453007, PR China
| | - Song Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Nan Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Shumao Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Fei Gao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Kuan Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, PR China.
| | - Liwei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
2
|
Yang Z, Wang X, Luo S, Li H, Xu J, Liang L, He Z, Wang G, Wu Z, Zhong N, Xiang H, Zhang Z, Guo C, Zhang Y, Yan F. Efficient production of recombinant human FVII in CHO cells using the piggyBac transposon system. Protein Expr Purif 2025; 229:106666. [PMID: 39848303 DOI: 10.1016/j.pep.2025.106666] [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: 11/11/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 01/25/2025]
Abstract
As an important coagulation factor, activated coagulation factor VII (FVIIa) is mainly used to treat the bleeding of hemophilia patients who have developed inhibitory antibodies against FVIII and FIX conventional treatment. Recombinant human factor VII (rhFVII) produced in mammalian cell lines have been developed as the most important resource of FVIIa. However, cell lines express rhFVII protein derived from an exogenous expression vector at a lower level than most other proteins. In the current study, we have shown efficient rhFVII production in CHO cell lines using piggyBac (PB) transposon system. rhFVII is successfully expressed in fed-batch culture of CHO cells, and the expression of rhFVII up to 100 mg/L. Moreover, the purified secreted rhFVII was determined by SDS-PAGE and Western Blot. The coagulation activity was determined by the chromogenic Activity ELISA kit. In conclusion, this study has demonstrated that the piggyBac transposon system can be used for an efficient production of recombinant FVII.
Collapse
Affiliation(s)
- Zhen Yang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Xueyun Wang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Shan Luo
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Hui Li
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Jiangbo Xu
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Linlin Liang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Zhimin He
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Guangyuan Wang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Zhuobin Wu
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Nan Zhong
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Haijun Xiang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Zhan Zhang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China
| | - Caiping Guo
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China.
| | - Yunjia Zhang
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China; Shenzhen Innovation Pharmaceutical Engineering Laboratory for Recombinant Plasma Proteins, Shenzhen, 518107, PR China.
| | - Fei Yan
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen, 518107, PR China.
| |
Collapse
|
3
|
Shi Y, Wan Y, Wang Y, Fang K, Yang J, Lu Y, Xie X, Pan J, Gao D, Wang H, Qu H. Quantitative 1H NMR optimization for high-throughput metabolite analysis in industrial bioprocess monitoring. Anal Bioanal Chem 2025:10.1007/s00216-025-05845-9. [PMID: 40167598 DOI: 10.1007/s00216-025-05845-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Quantitative 1H NMR (1H qNMR) is an ideal tool for bioprocess monitoring because it can comprehensively detect and quantify diverse metabolites that significantly influence bioprocess performance. However, the long experiment time associated with the 1H qNMR, due to the long longitudinal relaxation time (T1) of some metabolites, does not meet the requirements for high-throughput analysis. We developed a high-throughput 1H qNMR method for bioprocess analysis using a short relaxation delay (D1) to reduce analytical time and a correction factor (k) to compensate for incomplete relaxation. A total of 27 metabolites were quantified using spectral deconvolution via a peak fitting algorithm and MCR-ALS. Methodological validation results indicated that the precision and accuracy of the developed qNMR method were consistently high across different D1 values, with LOQs ranging from 0.008 to 0.13 mM and LODs ranging from 0.024 to 0.38 mM. Notably, a longer D1 value generally resulted in lower LODs and LOQs for most metabolites. A D1 value of 4 s was optimal for balancing analysis time and performance. The method is broadly applicable for bioprocess monitoring and control, offering valuable guidance for optimizing CHO cell culture processes and improving yield.
Collapse
Affiliation(s)
- Yingting Shi
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiang Wan
- BioRay Pharmaceutical Co., Ltd., Taizhou, 318000, China
| | - Yiru Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Kerui Fang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiayu Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuting Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyuan Xie
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianyang Pan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dong Gao
- BioRay Pharmaceutical Co., Ltd., Taizhou, 318000, China
| | - Haibin Wang
- BioRay Pharmaceutical Co., Ltd., Taizhou, 318000, China.
| | - Haibin Qu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
4
|
Bryant EE, Gong D, Guo C, Garces F, Hubert R, Chen I. An Arrayed CRISPR Screen Identifies Knockout Combinations Improving Antibody Productivity in HEK293 Cells. ACS Synth Biol 2025; 14:855-866. [PMID: 40014422 DOI: 10.1021/acssynbio.4c00772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Mammalian cells are used to express complex biologics, such as multispecific antibodies. While multispecifics enable promising new strategies for treating human disease, their production at high expression titer and purity can be challenging. To understand how cells respond to antibody and multispecific expression, five molecules were selected for bulk RNA sequencing (RNA-seq) early after the transfection of a human embryonic kidney 293 (HEK293) host. All five molecules shared a differential expression signature of secretory and protein folding stresses, but this signature was stronger for molecules with low titer. We then designed an arrayed CRISPR knockout screen of 206 differentially expressed target genes and 223 literature-motivated targets to identify knockouts that affect antibody productivity. Eight novel knockout targets were identified that increased expression titers by 20-80%. Notably, seven of these top eight hits were from the differentially expressed set of candidate-gene knockouts. The top knockout target, HIST2H3C, showed evidence for additivity with five other hits, including a knockout combination that increased the titer of a difficult-to-express antibody by up to 100%. Findings for both HIST2H3C and INHBE knockout targets generalized to an alternate HEK293 host expressing an additional antibody and a multispecific host with no meaningful impact on product purity. Thus, we propose HIST2H3C and INHBE disruption as a promising and novel strategy for host-cell engineering to improve antibody and multispecific productivity.
Collapse
Affiliation(s)
- Eric Edward Bryant
- Amgen R&D Postdoctoral Fellows Program, Thousand Oaks, California 91320, United States
- Large Molecule Discovery & Research Data Science, Amgen Research, Thousand Oaks, California 91320, United States
| | - Danyang Gong
- Large Molecule Discovery & Research Data Science, Amgen Research, Thousand Oaks, California 91320, United States
| | - Cai Guo
- Large Molecule Discovery & Research Data Science, Amgen Research, Thousand Oaks, California 91320, United States
| | - Fernando Garces
- Large Molecule Discovery & Research Data Science, Amgen Research, Thousand Oaks, California 91320, United States
| | - René Hubert
- Large Molecule Discovery & Research Data Science, Amgen Research, Thousand Oaks, California 91320, United States
| | - Irwin Chen
- Large Molecule Discovery & Research Data Science, Amgen Research, South San Francisco, California 94080, United States
| |
Collapse
|
5
|
Shin SW, Kim SH, Gasselin A, Lee GM, Lee JS. Comprehensive genome-scale CRISPR knockout screening of CHO cells. Sci Data 2025; 12:71. [PMID: 39814846 PMCID: PMC11735622 DOI: 10.1038/s41597-025-04438-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Chinese hamster ovary (CHO) cells play a pivotal role in the production of recombinant therapeutics. In the present study, we conducted a genome-scale pooled CRISPR knockout (KO) screening using a virus-free, recombinase-mediated cassette exchange-based platform in CHO-K1 host and CHO-K1 derived recombinant cells. Genome-wide guide RNA (gRNA) amplicon sequencing data were generated from cell libraries, as well as short- and long-term KO libraries, and validated through phenotypic assessment and gRNA read count distribution. Additionally, we obtained gRNA amplicon sequencing data from the highly productive recombinant cell populations. By analyzing these datasets, essential genes involved in cell fitness as well as functional target genes associated with therapeutic protein production can be identified. Collectively, our next-generation sequencing datasets, derived from a robust and reliable CRISPR screening method, provide valuable insights into CHO genomic functions, advancing the development of next-generation CHO factories.
Collapse
Affiliation(s)
- Sung Wook Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Aghiles Gasselin
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
6
|
Chang X, Gao J, Yang J, Ma Y, Song G. The Chinese hamster as an excellent experimental animal model. Exp Anim 2025; 74:1-15. [PMID: 39198205 PMCID: PMC11742471 DOI: 10.1538/expanim.24-0029] [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: 03/22/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024] Open
Abstract
Hamsters are valuable rodent models that are distinct from mice and rats. Currently, the main hamster species used for experimental research are the Syrian golden hamster and Chinese hamster, in addition to hamster species from other countries. Chinese hamsters are small, easy to run and feed, and inexpensive. They are prominent species found only in China and are part of the experimental animal resources of Chinese specialty. Chinese hamsters are distinguished by a black stripe on their back, short tail, pair of easily retractable cheek pouches, and pair of large drooping testes in males with 22 chromosomes. Due to their unique anatomical structure and biological features, Chinese hamsters have been used as a model in biomedical research. Moreover, the breeding and use of Chinese hamsters was comprehensively studied in 1958, with significant breakthroughs. We present a thorough review of the current developments and applications of Chinese hamsters and support the use of this species as a suitable and innovative experimental research model. With the success of Chinese hamster transgenic technology, this species will become more commonly employed in biological and medical research in the future.
Collapse
Affiliation(s)
- Xiaoqi Chang
- The Laboratory Animal Center, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
- Department of Basic Medical Sciences, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
| | - Jiping Gao
- The Laboratory Animal Center, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
- Department of Basic Medical Sciences, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
| | - Junting Yang
- The Laboratory Animal Center, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
- Department of Basic Medical Sciences, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
| | - Yunhui Ma
- The Laboratory Animal Center, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
- Department of Basic Medical Sciences, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
| | - Guohua Song
- The Laboratory Animal Center, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
- Department of Basic Medical Sciences, Shanxi Medical University, No 56 Xinjian South Road, Taiyuan 030001, P.R. China
| |
Collapse
|
7
|
Felix MN, Waerner T, Lakatos D, Reisinger B, Fischer S, Garidel P. Polysorbates degrading enzymes in biotherapeutics - a current status and future perspectives. Front Bioeng Biotechnol 2025; 12:1490276. [PMID: 39867473 PMCID: PMC11760601 DOI: 10.3389/fbioe.2024.1490276] [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: 09/02/2024] [Accepted: 12/02/2024] [Indexed: 01/28/2025] Open
Abstract
Polysorbates, in particular polysorbate (PS) 20 and 80, are the most commonly used surfactants for stabilising biotherapeutics produced by biotechnological processes. PSs are derived from ethoxylated sorbitan (a derivative of sorbitol) esterified with fatty acids of varying chain length and degree of saturation. In the past, these surfactants have been reported to have specific liabilities. Chemical (oxidations and hydrolyses) and enzymatic degradations have been reported to affect the stability of PS in drug products. Specifically, the presence of trace amounts (sub-ppm) of certain host cell proteins (HCPs) can induce enzymatic PS degradation, which can lead to the release of free fatty acids during storage over time. Enzymatic polysorbate degradation may impair the functionality of the surfactant in stabilising therapeutic proteins, leading to the formation of visible and/or sub-visible particles in biopharmaceutical drug products. This review summarises the enzymes currently known to be involved in the degradation of polysorbate in mammalian biotechnological processes for therapeutic proteins. In recent years, advanced analytical methods have been developed to qualify and quantify the PS-degrading enzymes. Most of these assays are based on mass spectrometry with a preceding HCP enrichment approach. Efforts were made to measure the enzyme activity and correlate it with observed PS degradation. The impact on drug product quality attributes, including fatty acid solubility and phase separation, up to the formation of visible particles, and the potential induction of protein and protein/fatty acid mixed particles as well as the sensitivity of specific PS quality towards enzymatic degradation, was considered. Various drug substance (DS) mitigation strategies related to the occurrence of PS degrading enzymes are discussed as amongst them the generation of stable HCP knockout cell lines, which are also carefully analysed. The underlying opinion article reflects the undergoing discussions related to PS degrading enzymes and focusses on (i) impact on drug product, (ii) analytics for identification/quantification (characterisation) of the PS degrading enzymes, (iii) enzyme activity (iv) currently identified enzymes, and (v) potential mitigation strategies to avoid enzymatic PS degradation during DS manufacturing.
Collapse
Affiliation(s)
- Marius Nicolaus Felix
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Thomas Waerner
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Daniel Lakatos
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Bernd Reisinger
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Simon Fischer
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Patrick Garidel
- Pharmaceutical Development Biologicals, TIP, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| |
Collapse
|
8
|
Grav LM, Rojek JB, la Cour Karottki KJ, Lee JS, Kildegaard HF. Application of CRISPR/Cas9 Genome Editing to Improve Recombinant Protein Production in CHO Cells. Methods Mol Biol 2025; 2853:49-69. [PMID: 39460914 DOI: 10.1007/978-1-0716-4104-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Genome editing has become an important aspect of Chinese hamster ovary (CHO) cell line engineering for improving the production of recombinant protein therapeutics. Currently, the engineering focus is directed toward expanding product diversity while controlling and improving product quality and yields. In this chapter, we present our protocol for using the genome editing tool Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) to knock out engineering target genes in CHO cells. As an example, we describe how to knock out the glutamine synthetase (GS) gene, which increases the selection efficiency of the GS-mediated gene amplification system.
Collapse
Affiliation(s)
- Lise Marie Grav
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| | - Johan Blatt Rojek
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
9
|
Schlossbauer P, Klingler F, Burkhart M, Leroux AC, Hesse F, Otte K. MiRNA Chaining for Efficient Stable Overexpression to Improve Protein Quantity and Quality in CHO Cells. Methods Mol Biol 2025; 2853:85-101. [PMID: 39460916 DOI: 10.1007/978-1-0716-4104-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
MicroRNAs (miRNAs), small noncoding RNAs with a length of about 22 nucleotides, harbor the potential to be powerful tools for the genetic engineering of production cell lines like Chinese hamster ovary (CHO) cells. Their ability to regulate multiple targets at once and their potential to fine-tune effect strengths contrast with classical engineering approaches. However, most studies of miRNAs rely on transiently flooding the cells with miRNA mimics. Since this approach is not suitable for long-term cultivation in a bioprocess, stable overexpression of miRNAs becomes more and more important for the biotech industry. Here, the user might be confronted with insufficient overexpression of the miRNA of interest. In this chapter, we present a method for the generation of stable CHO cell lines expressing a miRNA from a plasmid-based system containing multiple copies of the miRNA, allowing tuning of overexpression and regulation.
Collapse
Affiliation(s)
- Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Florian Klingler
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Madina Burkhart
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | | | - Friedemann Hesse
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Biberach, Germany.
| |
Collapse
|
10
|
Baek M, Kim CL, Kim SH, la Cour Karottki KJ, Hefzi H, Grav LM, Pedersen LE, Lewis NE, Lee JS, Lee GM. Unraveling productivity-enhancing genes in Chinese hamster ovary cells via CRISPR activation screening using recombinase-mediated cassette exchange system. Metab Eng 2025; 87:11-20. [PMID: 39566816 DOI: 10.1016/j.ymben.2024.11.009] [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: 07/08/2024] [Revised: 10/14/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
Chinese hamster ovary (CHO) cells, which are widely used for therapeutic protein production, have been genetically manipulated to enhance productivity. Nearly half of the genes in CHO cells are silenced, which are promising targets for CHO cell engineering. To identify novel gene targets among the silenced genes that can enhance productivity, we established a genome-wide clustered regularly interspaced short palindromic repeats activation (CRISPRa) screening platform for bispecific antibody (bsAb)-producing CHO (CHO-bsAb) cells with 110,979 guide RNAs (gRNAs) targeting 13,812 silenced genes using a virus-free recombinase-mediated cassette exchange-based gRNA integration method. Using this platform, we performed a fluorescence-activated cell sorting-based cold-capture assay to isolate cells with high fluorescence intensity, which is indicative of high specific bsAb productivity (qbsAb), and identified 90 significantly enriched genes. To verify the screening results, 14 high-scoring candidate genes were individually activated in CHO-bsAb cells via CRISPRa. Among these, 10 genes demonstrated enhanced fluorescence intensity of CHO-bsAb cells in the cold-capture assay when activated. Furthermore, the overexpression of the identified novel gene target Syce3 in CHO-bsAb cells resulted in a 1.4- to 1.9-fold increase in the maximum bsAb concentration, owing to improved qbsAb and specific growth rate. Thus, this virus-free CRISPRa screening platform is a potent tool for identifying novel engineering targets in CHO cells to improve bsAb production.
Collapse
Affiliation(s)
- Minhye Baek
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Che Lin Kim
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | | | - Hooman Hefzi
- Departments of Pediatrics and Bioengineering, University of California, San Diego, USA
| | - Lise Marie Grav
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lasse Ebdrup Pedersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Nathan E Lewis
- Departments of Pediatrics and Bioengineering, University of California, San Diego, USA
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
11
|
Wu Y, Ji X, Yang Y, Wu B. Discovery of a fully human antibody to the proximal membrane terminus of MUC1 based on a B-cell high-throughput screening technique. Int Immunopharmacol 2024; 142:113204. [PMID: 39317052 DOI: 10.1016/j.intimp.2024.113204] [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: 06/19/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Mucin 1 plays an important role in tumor signaling and is overexpressed in adenocarcinoma and the digestive system. Many antibodies have been developed against MUC1 targets. Previously developed antibodies were mainly directed against distal membrane-terminal MUC1-N, but distal membrane-terminal MUC1-N is shed during cell growth and therefore binds to antibodies developed against tandem repeat sequences and becomes ineffective. Here, we provide a simple and rapid method for preparing antibodies targeting the proximal membrane end of MUC1. Immunological target antigens were designed based on Biocytogen Renlite KO mice. With the help of B-cell high-throughput screening technology, we rapidly screened and prepared fully human antibodies with human-macaque cross-reactivity, high affinity, high specificity, and endocytosis. Using this method, we screened 40 antibodies with human-monkey cross-reactivity, which specifically recognized breast cancer cell lines with human and monkey affinities ranging from (1.04E-07-2.91E-09). Of these, the antibodies with germline genes IGHV4-59*01 and IGHV3-30*03 had nanomolar affinities, with high endocytosis effects in breast cancer cells. Ab.07 (IGHV3-30*03) coupled with monomethyl auristatin E (MMAE) showed good anti-tumor activity in different tumor cells. In summary, we describe a method for designing and producing excellent antibodies that can be assembled into antibody-drug conjugates and bispecific antibodies by proximal-membrane-end immunization and B-cell high-throughput screening that can rapidly generate high-quality antibodies.
Collapse
Affiliation(s)
- Yilin Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226133, China
| | - Xin Ji
- Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China.
| | - Yi Yang
- Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226133, China.
| | - Bo Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
12
|
Zhang J, Wang L, Zhang X, Sun Q, Zhang J. Matrix attachment regions enhance transgene expression by manipulating position-dependent effects in stably transfected CHO-K1 cells. Biochem Cell Biol 2024; 102:526-534. [PMID: 39029107 DOI: 10.1139/bcb-2023-0337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
We previously found that the position of matrix attachment regions (MARs) within the vector significantly affects its ability to enhance transgenic expression in the recombinant protein production. This study aims to systematically investigate the position-dependent impacts of MAR on transgene expression. We observed a significant increase in enhanced green fluorescent protein (eGFP) expression levels in stably transfected CHO-K1 cells with either MAR 1-68 or MAR X-29 when MARs located upstream of the promoter. This increase was especially evident with MAR flanked the expression cassette. Concurrently, a substantial increase was observed in the percentage of eGFP-expressing cells, with 97.8% and 96.0% in MAR-containing constructs versus 73.7% in MAR-absent constructs. Further analysis of erythropoietin (EPO) expression revealed that constructs with flanking MARs induced the highest EPO productivity. Bioinformatics analysis revealed that certain specific transcription factors are important in modulating the transcription process. In conclusion, vectors harboring both MARs around the expression cassette constitute an optimal construct for enhanced recombinant protein production in CHO-K1 cells. This insight underscores the importance of strategic MAR incorporation in vector design for optimized recombinant protein expression.
Collapse
Affiliation(s)
- Jihong Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Lin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Xi Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Qiuli Sun
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Junhe Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
13
|
Wang Q, Shi S, Liu S, Ye S. A user-friendly fluorescent biosensor for precise lactate detection and quantification in vitro. Chem Commun (Camb) 2024; 60:12884-12887. [PMID: 39404007 DOI: 10.1039/d4cc04925j] [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: 11/02/2024]
Abstract
As a critical metabolite, the standardization of lactate quantification is increasingly crucial. Therefore, we developed LaconicSF, a lactate-responsive biosensor exhibiting exceptional specificity in lactate detection. LaconicSF enables efficient lactate quantification in CHO cell culture medium and holds potential as a user-friendly detection tool for lactate quantification in vitro.
Collapse
Affiliation(s)
- Qiwei Wang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China.
| | - Sai Shi
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China.
| | - Si Liu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China.
| | - Sheng Ye
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China.
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| |
Collapse
|
14
|
Selvaprakash K, Sideri C, Henry M, Efeoglu E, Ryan D, Meleady P. Characterization of the Ubiquitin-Modified Proteome of Recombinant Chinese Hamster Ovary Cells in Response to Endoplasmic Reticulum Stress. Biotechnol J 2024; 19:e202400413. [PMID: 39623727 PMCID: PMC11612545 DOI: 10.1002/biot.202400413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 12/06/2024]
Abstract
Chinese hamster ovary (CHO) cells remain the most widely used host cell line for biotherapeutics production. Despite their widespread use, understanding endoplasmic reticulum (ER) stress conditions in recombinant protein production remains limited, often creating bottlenecks preventing improved production titers and product quality. Ubiquitination not only targets substrates (e.g., misfolded proteins) for proteasome degradation but also has important regulatory control functions including cell cycle regulation, translation, apoptosis, autophagy, etc. and hence is likely to be central to understanding and controlling the productivity of recombinant biotherapeutics. This study aimed to uncover differentially expressed ubiquitinated proteins following artificial induction of ER-stress in recombinant CHO cells. CHO cells were treated with the stress inducer tunicamycin and the proteasome inhibitor MG132, followed by LC-MS/MS proteomic analysis. We identified >4000 ubiquitinated peptides from CHO-DP12 cells under ER stress conditions and proteasome inhibition. Moreover, data analysis showed altered abundance levels of >900 ubiquitinated proteins under the combination of ER stress and proteasome inhibition compared to untreated controls. Gene Ontology (GO) analysis of these ubiquitinated proteins resulted in a significant enrichment of key pathways involving the proteasome, protein processing in the ER, N-glycan biosynthesis, and ubiquitin-mediated proteolysis. ER stress response proteins such as GRP78, HSP90B1, ATF6, HERPUD1, and PDIA4 were found to be highly ubiquitinated and exhibited a significant increase in abundance following induction of ER-stress conditions. This study broadens our comprehension of the roles played by protein ubiquitination in CHO cell stress responses, potentially revealing targets for tailored cell line engineering aimed at enhancing stress tolerance and production efficiency.
Collapse
Affiliation(s)
| | - Christiana‐Kondylo Sideri
- Life Sciences InstituteDublin City UniversityDublinIreland
- School of BiotechnologyDublin City UniversityDublinIreland
| | - Michael Henry
- Life Sciences InstituteDublin City UniversityDublinIreland
| | - Esen Efeoglu
- Life Sciences InstituteDublin City UniversityDublinIreland
| | - David Ryan
- Life Sciences InstituteDublin City UniversityDublinIreland
- School of BiotechnologyDublin City UniversityDublinIreland
| | - Paula Meleady
- Life Sciences InstituteDublin City UniversityDublinIreland
- School of BiotechnologyDublin City UniversityDublinIreland
- SSPC the SFI Research Centre for PharmaceuticalsDublin City UniversityDublinIreland
| |
Collapse
|
15
|
Riquelme-Guzmán C, Stout AJ, Kaplan DL, Flack JE. Unlocking the potential of cultivated meat through cell line engineering. iScience 2024; 27:110877. [PMID: 39351194 PMCID: PMC11440241 DOI: 10.1016/j.isci.2024.110877] [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] [Indexed: 10/04/2024] Open
Abstract
Cultivated meat has the potential to revolutionize food production, but its progress is hindered by fundamental shortcomings of mammalian cells with respect to industrial-scale bioprocesses. Here, we discuss the essential role of cell line engineering in overcoming these limitations, highlighting the balance between the benefits of enhanced cellular traits and the associated regulatory and consumer acceptance challenges. We believe that careful selection of cell engineering strategies, including both genetic and non-genetic modifications, can address this trade-off and is essential to advancing the field.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Andrew J Stout
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
- Deco Labs, Inc., Boston, MA, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Joshua E Flack
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
16
|
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] [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
|
17
|
Shin SW, Min H, Kim J, Lee JS. A precise and sustainable doxycycline-inducible cell line development platform for reliable mammalian cell engineering with gain-of-function mutations. Metab Eng 2024; 86:12-28. [PMID: 39242074 DOI: 10.1016/j.ymben.2024.09.004] [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: 05/28/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
For mammalian synthetic biology research, multiple orthogonal and tunable gene expression systems have been developed, among which the tetracycline (Tet)-inducible system is a key tool for gain-of-function mutations. Precise and long-lasting regulation of genetic circuits is necessary for the effective use of these systems in genetically engineered stable cell lines. However, current cell line development strategies, which depend on either random or site-specific integration along with antibiotic selection, are unpredictable and unsustainable, limiting their widespread use. To overcome these issues, we aimed to establish a Robust Overexpression via Site-specific integration of Effector (ROSE) system, a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9-mediated streamlined Tet-On3G-inducible master cell line (MCL) development platform. ROSE MCLs equipped with a landing pad facilitated the transcriptional regulation of various effector genes via recombinase-mediated cassette exchange. Long-term investigation revealed that the modular design of genetic payloads and integration sites significantly affected the induction capacity and stability, with ROSE MCLs exhibiting exceptional induction performance. To demonstrate the versatility of our platform, we explored its efficiency for the precise regulation of selection stringency, manufacturing of therapeutic antibodies with tunable expression levels and timing, and transcription factor engineering. Overall, this study demonstrated the effectiveness and reliability of the ROSE platform, highlighting its potential for various biological and biotechnological applications.
Collapse
Affiliation(s)
- Sung Wook Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Honggi Min
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Jiwon Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea; Advanced College of Bio-convergence Engineering, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
18
|
Gopalakrishnan S, Johnson W, Valderrama-Gomez MA, Icten E, Tat J, Lay F, Diep J, Gomez N, Stevens J, Schlegel F, Rolandi P, Kontoravdi C, Lewis NE. Multi-omic characterization of antibody-producing CHO cell lines elucidates metabolic reprogramming and nutrient uptake bottlenecks. Metab Eng 2024; 85:94-104. [PMID: 39047894 DOI: 10.1016/j.ymben.2024.07.009] [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: 07/06/2023] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the phenotypic diversity and metabolic capabilities of industrially relevant manufacturing cell lines is critical to bioprocess optimization and cell line development. Metabolic capabilities of production hosts limit nutrient and resource channeling into desired cellular processes and can have a profound impact on productivity. These limitations cannot be directly inferred from measured data such as spent media concentrations or transcriptomics. Here, we present an integrated multi-omic analysis pipeline combining exo-metabolomics, transcriptomics, and genome-scale metabolic network analysis and apply it to three antibody-producing Chinese Hamster Ovary cell lines to identify reprogramming features associated with high-producing clones and metabolic bottlenecks limiting product formation in an industrial bioprocess. Analysis of individual datatypes revealed a decreased nitrogenous byproduct secretion in high-producing clones and the topological changes in peripheral metabolic pathway expression associated with phase shifts. An integrated omics analysis in the context of the genome-scale metabolic model elucidated the differences in central metabolism and identified amino acid utilization bottlenecks limiting cell growth and antibody production that were not evident from exo-metabolomics or transcriptomics alone. Thus, we demonstrate the utility of a multi-omics characterization in providing an in-depth understanding of cellular metabolism, which is critical to efforts in cell engineering and bioprocess optimization.
Collapse
Affiliation(s)
| | | | | | | | - Jasmine Tat
- Process Development Amgen, USA; Department of Bioengineering, University of California San Diego, USA
| | | | | | | | | | | | | | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, UK
| | - Nathan E Lewis
- Department of Pediatrics, University of California San Diego, USA; Department of Bioengineering, University of California San Diego, USA.
| |
Collapse
|
19
|
Schlossbauer P, Naumann L, Klingler F, Burkhart M, Handrick R, Korff K, Neusüß C, Otte K, Hesse F. Stable overexpression of native and artificial miRNAs for the production of differentially fucosylated antibodies in CHO cells. Eng Life Sci 2024; 24:2300234. [PMID: 38845814 PMCID: PMC11151017 DOI: 10.1002/elsc.202300234] [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: 08/08/2023] [Revised: 03/04/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Cell engineering strategies typically rely on energy-consuming overexpression of genes or radical gene-knock out. Both strategies are not particularly convenient for the generation of slightly modulated phenotypes, as needed in biosimilar development of for example differentially fucosylated monoclonal antibodies (mAbs). Recently, transiently transfected small noncoding microRNAs (miRNAs), known to be regulators of entire gene networks, have emerged as potent fucosylation modulators in Chinese hamster ovary (CHO) production cells. Here, we demonstrate the applicability of stable miRNA overexpression in CHO production cells to adjust the fucosylation pattern of mAbs as a model phenotype. For this purpose, we applied a miRNA chaining strategy to achieve adjustability of fucosylation in stable cell pools. In addition, we were able to implement recently developed artificial miRNAs (amiRNAs) based on native miRNA sequences into a stable CHO expression system to even further fine-tune fucosylation regulation. Our results demonstrate the potential of miRNAs as a versatile tool to control mAb fucosylation in CHO production cells without adverse side effects on important process parameters.
Collapse
Affiliation(s)
- Patrick Schlossbauer
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| | | | - Florian Klingler
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| | - Madina Burkhart
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| | - René Handrick
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| | | | | | - Kerstin Otte
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| | - Friedemann Hesse
- Institute for Applied BiotechnologyUniversity of Applied Sciences BiberachBiberachGermany
| |
Collapse
|
20
|
Curry E, Sedelnikova S, Rafferty J, Hulley M, Pohle M, Muir G, Brown A. Expanding the RNA polymerase biocatalyst solution space for mRNA manufacture. Biotechnol J 2024; 19:e2400012. [PMID: 39031865 PMCID: PMC11475235 DOI: 10.1002/biot.202400012] [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: 01/05/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 07/22/2024]
Abstract
All mRNA products are currently manufactured in in vitro transcription (IVT) reactions that utilize single-subunit RNA polymerase (RNAP) biocatalysts. Although it is known that discrete polymerases exhibit highly variable bioproduction phenotypes, including different relative processivity rates and impurity generation profiles, only a handful of enzymes are generally available for mRNA biosynthesis. This limited RNAP toolbox restricts strategies to design and troubleshoot new mRNA manufacturing processes, which is particularly undesirable given the continuing diversification of mRNA product lines toward larger and more complex molecules. Herein, we describe development of a high-throughput RNAP screening platform, comprising complementary in silico and in vitro testing modules, that enables functional characterization of large enzyme libraries. Utilizing this system, we identified eight novel sequence-diverse RNAPs, with associated active cognate promoters, and subsequently validated their performance as recombinant enzymes in IVT-based mRNA production processes. By increasing the number of available characterized functional RNAPs by more than 130% and providing a platform to rapidly identify further potentially useful enzymes, this work significantly expands the RNAP biocatalyst solution space for mRNA manufacture, thereby enhancing the capability for application-specific and molecule-specific optimization of both product yield and quality.
Collapse
Affiliation(s)
- Edward Curry
- Department of Chemical and Biological EngineeringUniversity of SheffieldSheffieldUK
| | | | - John Rafferty
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | | | - Melinda Pohle
- Department of Chemical and Biological EngineeringUniversity of SheffieldSheffieldUK
| | - George Muir
- Department of Chemical and Biological EngineeringUniversity of SheffieldSheffieldUK
| | - Adam Brown
- Department of Chemical and Biological EngineeringUniversity of SheffieldSheffieldUK
| |
Collapse
|
21
|
Wang W, Liu C, Zhang X, Yan J, Zhang J, You S, Su R, Qi W. Time-resolved fluoroimmunoassay for Aspergillus detection based on anti-galactomannan monoclonal antibody from stable cell line. Anal Biochem 2024; 689:115494. [PMID: 38403258 DOI: 10.1016/j.ab.2024.115494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Invasive Aspergillosis is a high-risk illness with a high death rate in immunocompromised people due to a lack of early detection and timely treatment. Based on immunology study, we achieved an efficient production of anti-galactomannan antibody by Chinese hamster ovary (CHO) cells and applied it to time-resolved fluoroimmunoassay for Aspergillus galactomannan detection. We first introduced dual promoter expression vector into CHO host cells, and then applied a two-step screening strategy to screen the stable cell line by methionine sulfoximine pressurization. After amplification and fermentation, antibody yield reached 4500 mg/L. Then we conjugated the antibodies with fluorescent microspheres to establish a double antibody sandwich time-resolved fluoroimmunoassay, which was compared with the commercial Platelia™ Aspergillus Ag by clinical serum samples. The preformed assay could obtain the results in less than 25 min, with a limit of detection for galactomannan of approximately 1 ng/mL. Clinical results of the two methods showed that the overall percent agreement was 97.7% (95% CI: 96.6%-98.4%) and Cohen's kappa coefficient was 0.94. Overall, the assay is highly consistent with commercial detection, providing a more sensitive and effective method for the rapid diagnosis of invasive aspergillosis.
Collapse
Affiliation(s)
- Wenjun Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China
| | - Chunlong Liu
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Dynamiker Biotechnology (Tianjin) Co., Ltd, PR China
| | - Xuemei Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China
| | - Jun Yan
- Dynamiker Biotechnology (Tianjin) Co., Ltd, PR China
| | - Jiaxing Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China.
| | - Shengping You
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China.
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin, 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, PR China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin, 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, PR China
| |
Collapse
|
22
|
LeBarre JP, Chu W, Altern SH, Kocot AJ, Bhandari D, Barbieri E, Sly J, Crapanzano M, Cramer SM, Phillips M, Roush D, Carbonell R, Boi C, Menegatti S. Mixed-mode size-exclusion silica resin for polishing human antibodies in flow-through mode. J Chromatogr A 2024; 1720:464772. [PMID: 38452560 DOI: 10.1016/j.chroma.2024.464772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/07/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
The polishing step in the downstream processing of therapeutic antibodies removes residual impurities from Protein A eluates. Among the various classes of impurities, antibody fragments are especially challenging to remove due to the broad biomolecular diversity generated by a multitude of fragmentation patterns. The current approach to fragment removal relies on ion exchange or mixed-mode adsorbents operated in bind-and-gradient-elution mode. However, fragments that bear strong similarity to the intact product or whose biophysical features deviate from the ensemble average can elude these adsorbents, and the lack of a chromatographic technology enabling robust antibody polishing is recognized as a major gap in downstream bioprocessing. Responding to this challenge, this study introduces size-exclusion mixed-mode (SEMM) silica resins as a novel chromatographic adsorbent for the capture of antibody fragments irrespective of their biomolecular features. The pore diameter of the silica beads features a narrow distribution and is selected to exclude monomeric antibodies, while allowing their fragments to access the pores where they are captured by the mixed-mode ligands. The static and dynamic binding capacity of the adsorbent ranged respectively between 30-45 and 25-33 gs of antibody fragments per liter of resin. Selected SEMM-silica resins also demonstrated the ability to capture antibody aggregates, which adsorb on the outer layer of the beads. Optimization of the SEMM-silica design and operation conditions - namely, pore size (10 nm) and ligand composition (quaternary amine and alkyl chain) as well as the linear velocity (100 cm/h), ionic strength (5.7 mS/cm), and pH (7) of the mobile phase - afforded a significant reduction of both fragments and aggregates, resulting into a final antibody yield up to 80% and monomeric purity above 97%.
Collapse
Affiliation(s)
- Jacob P LeBarre
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Scott H Altern
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Andrew J Kocot
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Dipendra Bhandari
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Eduardo Barbieri
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Jae Sly
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Michael Crapanzano
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Steven M Cramer
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | | | - David Roush
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, Roush Biopharma Panacea, 20 Squire Terrace, Colts Neck, NJ, 07033, USA
| | - Ruben Carbonell
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Cristiana Boi
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA; Department of Civil, Chemical Environmental and Materials Engineering, University of Bologna, Via Terracini 28, 40131, Bologna, Italy
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA.
| |
Collapse
|
23
|
Roshanmehr F, Abdoli S, Bazi Z, Jari M, Shahbazi M. Enhancing the productivity and proliferation of CHO-K1 cells by oncoprotein YAP (Yes-associated protein). Appl Microbiol Biotechnol 2024; 108:285. [PMID: 38573360 PMCID: PMC10994876 DOI: 10.1007/s00253-024-13122-5] [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: 12/19/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
CHO cells are extensively employed in biological drug industry to manufacture therapeutic proteins. Nevertheless, production of biopharmaceuticals faces obstacles such as limited growth and inadequate productivity. Employing host cell engineering techniques for CHO cells serves as a valuable approach to address the constraints encountered in biologics manufacturing. Despite advancements, most techniques focus on specific genes to address individual cellular challenges. The significance of YAP, transcriptional co-activator, cannot be overstated due to its involvement in regulating organ size and tumor formation. YAP's influence extends to various cellular processes and is regulated by kinase cascade in the Hippo pathway, which phosphorylates serine residues in specific LATS recognition motifs. Activation of YAP has been observed to impact both the size and quantity of cells. This research investigates the effects of YAP5SA on proliferation, apoptosis, and productivity in CHO-K1 cells. YAP5SA, with mutations in all five LATS-target sites, is selected for its heightened activity and resistance to repression through the Hippo-LATS1/2 kinase signaling pathway. Plasmid harboring YAP5SA was transfected into EPO-CHO and the influence of YAP5SA overexpression was investigated. According to our findings, transfection of EPO-CHO cells with YAP5SA exhibited a substantial enhancement in CHO cell productivity, resulting in a 3-fold increase in total protein and EPO, as well as a 1.5-fold increase in specific productivity. Additionally, it significantly contributes in augmenting viability, size, and proliferation. Overall, the findings of this study exemplify the potential of utilizing YAP5SA to impact particular cellular mechanisms, thereby presenting an avenue for customizing cells to fulfill production demands. KEY POINTS: • YAP5SA in CHO cells boosts growth, reduces apoptosis, and significantly improves productivity. • YAP5SA regulates genes involved in proliferation, survival, and mTOR activation. • YAP5SA increases productivity by improving cell cycle, c-MYC expression, and mTOR pathway.
Collapse
Affiliation(s)
- Farnaz Roshanmehr
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Bazi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Jari
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
- Arya Tina Gene (ATG), Biopharmaceutical Company, Gorgan, Iran.
| |
Collapse
|
24
|
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
|
25
|
Eisenhut P, Marx N, Borsi G, Papež M, Ruggeri C, Baumann M, Borth N. Manipulating gene expression levels in mammalian cell factories: An outline of synthetic molecular toolboxes to achieve multiplexed control. N Biotechnol 2024; 79:1-19. [PMID: 38040288 DOI: 10.1016/j.nbt.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/06/2023] [Accepted: 11/26/2023] [Indexed: 12/03/2023]
Abstract
Mammalian cells have developed dedicated molecular mechanisms to tightly control expression levels of their genes where the specific transcriptomic signature across all genes eventually determines the cell's phenotype. Modulating cellular phenotypes is of major interest to study their role in disease or to reprogram cells for the manufacturing of recombinant products, such as biopharmaceuticals. Cells of mammalian origin, for example Chinese hamster ovary (CHO) and Human embryonic kidney 293 (HEK293) cells, are most commonly employed to produce therapeutic proteins. Early genetic engineering approaches to alter their phenotype have often been attempted by "uncontrolled" overexpression or knock-down/-out of specific genetic factors. Many studies in the past years, however, highlight that rationally regulating and fine-tuning the strength of overexpression or knock-down to an optimum level, can adjust phenotypic traits with much more precision than such "uncontrolled" approaches. To this end, synthetic biology tools have been generated that enable (fine-)tunable and/or inducible control of gene expression. In this review, we discuss various molecular tools used in mammalian cell lines and group them by their mode of action: transcriptional, post-transcriptional, translational and post-translational regulation. We discuss the advantages and disadvantages of using these tools for each cell regulatory layer and with respect to cell line engineering approaches. This review highlights the plethora of synthetic toolboxes that could be employed, alone or in combination, to optimize cellular systems and eventually gain enhanced control over the cellular phenotype to equip mammalian cell factories with the tools required for efficient production of emerging, more difficult-to-express biologics formats.
Collapse
Affiliation(s)
- Peter Eisenhut
- Austrian Centre of Industrial Biotechnology (acib GmbH), Muthgasse 11, 1190 Vienna, Austria
| | - Nicolas Marx
- BOKU University of Natural Resources and Life Sciences, Institute of Animal Cell Technology and Systems Biology, Muthgasse 18, 1190 Vienna, Austria.
| | - Giulia Borsi
- BOKU University of Natural Resources and Life Sciences, Institute of Animal Cell Technology and Systems Biology, Muthgasse 18, 1190 Vienna, Austria
| | - Maja Papež
- Austrian Centre of Industrial Biotechnology (acib GmbH), Muthgasse 11, 1190 Vienna, Austria; BOKU University of Natural Resources and Life Sciences, Institute of Animal Cell Technology and Systems Biology, Muthgasse 18, 1190 Vienna, Austria
| | - Caterina Ruggeri
- BOKU University of Natural Resources and Life Sciences, Institute of Animal Cell Technology and Systems Biology, Muthgasse 18, 1190 Vienna, Austria
| | - Martina Baumann
- Austrian Centre of Industrial Biotechnology (acib GmbH), Muthgasse 11, 1190 Vienna, Austria
| | - Nicole Borth
- Austrian Centre of Industrial Biotechnology (acib GmbH), Muthgasse 11, 1190 Vienna, Austria; BOKU University of Natural Resources and Life Sciences, Institute of Animal Cell Technology and Systems Biology, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
26
|
Jerabek T, Weiß L, Fahrion H, Zeh N, Raab N, Lindner B, Fischer S, Otte K. In pursuit of a minimal CHO genome: Establishment of large-scale genome deletions. N Biotechnol 2024; 79:100-110. [PMID: 38154614 DOI: 10.1016/j.nbt.2023.12.007] [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: 10/20/2023] [Revised: 11/27/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Chinese hamster ovary (CHO) cells are the most commonly used mammalian cell line for the production of complex therapeutic glycoproteins. As CHO cells have evolved as part of a multicellular organism, they harbor many cellular functions irrelevant for their application as production hosts in industrial bioprocesses. Consequently, CHO cells have been the target for numerous genetic engineering efforts in the past, but a tailored host cell chassis holistically optimized for its specific task in a bioreactor is still missing. While the concept of genome reduction has already been successfully applied to bacterial production cells, attempts to create higher eukaryotic production hosts exhibiting reduced genomes have not been reported yet. Here, we present the establishment and application of a large-scale genome deletion strategy for targeted excision of large genomic regions in CHO cells. We demonstrate the feasibility of genome reduction in CHO cells using optimized CRISPR/Cas9 based experimental protocols targeting large non-essential genomic regions with high efficiency. Achieved genome deletions of non-essential genetic regions did not introduce negative effects on bioprocess relevant parameters, although we conducted the largest reported genomic excision of 864 kilobase pairs in CHO cells so far. The concept presented serves as a directive to accelerate the development of a significantly genome-reduced CHO host cell chassis paving the way for a next generation of CHO cell factories.
Collapse
Affiliation(s)
- Tobias Jerabek
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany.
| | - Linus Weiß
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Hannah Fahrion
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Nikolas Zeh
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany; Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Nadja Raab
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Benjamin Lindner
- Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Simon Fischer
- Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Kerstin Otte
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| |
Collapse
|
27
|
Jerabek T, Burkhart M, Goetz S, Greck B, Menthe A, Neef R, Otte K. Inefficient transcription is a production bottleneck for artificial therapeutic BiTE® proteins. N Biotechnol 2024; 79:91-99. [PMID: 38154615 DOI: 10.1016/j.nbt.2023.12.008] [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: 11/10/2023] [Revised: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
Antibodies are potent biopharmaceuticals used to treat severe diseases, including cancers. During the past decade, more complex modalities have been developed including bispecific T-cell engager (BiTE®) molecules, e.g. by Amgen. However, non-natural and complex molecule formats often prove to be difficult-to-express (DTE), which is the case for BiTE® molecules. Due to the growing importance of multispecific modalities such as half-life extended (HLE) BiTE® and HLE dual-targeting bispecific T-cell engager (dBiTE) molecules, this artificial class of therapeutic proteins was investigated for molecular bottlenecks in stable production cell lines, by analyzing all relevant steps of recombinant protein production. As a result, drastically reduced intracellular BiTE® molecule-encoding mRNA levels were identified as a potential production bottleneck. Using in vitro transcription (IVT), the transcription rate of the BiTE® molecule-encoding mRNA was identified as the root cause for reduced amounts of intracellular mRNA. In an attempt to improve the transcription rate of a BiTE® molecule, it could be demonstrated that the artificial and special structure of the BiTE® molecule was not the rate limiting step for reduced IVT rate. However, modulation of the primary DNA sequence led to significant improvement of IVT rate. The analyses presented provide insight into the HLE BiTE® / HLE d(BiTE®) class of DTE proteins and perhaps into other classes of DTE proteins, and therefore may lead to identification of further production bottlenecks and optimization strategies to overcome manufacturability challenges associated with various complex therapeutics.
Collapse
Affiliation(s)
- Tobias Jerabek
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400 Biberach an der Riss, Germany.
| | - Madina Burkhart
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400 Biberach an der Riss, Germany
| | - Selina Goetz
- Process Development, Amgen Research (Munich) GmbH, Staffelseestraße 2, 81477 Munich, Germany
| | - Benedikt Greck
- Process Development, Amgen Research (Munich) GmbH, Staffelseestraße 2, 81477 Munich, Germany
| | - Anika Menthe
- Process Development, Amgen Research (Munich) GmbH, Staffelseestraße 2, 81477 Munich, Germany
| | - Ruediger Neef
- Process Development, Amgen Research (Munich) GmbH, Staffelseestraße 2, 81477 Munich, Germany
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400 Biberach an der Riss, Germany
| |
Collapse
|
28
|
Wang X, Xu J, Guo Q, Li Z, Cao J, Fu R, Xu M, Zhao X, Wang F, Zhang X, Dong T, Li X, Qian W, Hou S, Ji L, Zhang D, Guo H. Improving product quality and productivity of an antibody-based biotherapeutic using inverted frustoconical shaking bioreactors. Front Bioeng Biotechnol 2024; 12:1352098. [PMID: 38585708 PMCID: PMC10995296 DOI: 10.3389/fbioe.2024.1352098] [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: 12/07/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
The Chinese hamster ovarian (CHO) cells serve as a common choice in biopharmaceutical production, traditionally cultivated in stirred tank bioreactors (STRs). Nevertheless, the pursuit of improved protein quality and production output for commercial purposes demand exploration into new bioreactor types. In this context, inverted frustoconical shaking bioreactors (IFSB) present unique physical properties distinct from STRs. This study aims to compare the production processes of an antibody-based biotherapeutic in both bioreactor types, to enhance production flexibility. The findings indicate that, when compared to STRs, IFSB demonstrates the capability to produce an antibody-based biotherapeutic with either comparable or enhanced bioprocess performance and product quality. IFSB reduces shear damage to cells, enhances viable cell density (VCD), and improves cell state at a 5-L scale. Consequently, this leads to increased protein expression (3.70 g/L vs 2.56 g/L) and improved protein quality, as evidenced by a reduction in acidic variants from 27.0% to 21.5%. Scaling up the culture utilizing the Froude constant and superficial gas velocity ensures stable operation, effective mixing, and gas transfer. The IFSB maintains a high VCD and cell viability at both 50-L and 500-L scales. Product expression levels range from 3.0 to 3.6 g/L, accompanied by an improved acidic variants attribute of 20.6%-22.7%. The IFSB exhibits superior productivity and product quality, underscoring its potential for incorporation into the manufacturing process for antibody-based biotherapeutics. These results establish the foundation for IFSB to become a viable option in producing antibody-based biotherapeutics for clinical and manufacturing applications.
Collapse
Affiliation(s)
- Xuekun Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Jin Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qingcheng Guo
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Taizhou Mabtech Pharmaceuticals Co., Ltd., Taizhou, China
| | - Zhenhua Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, Shanghai Zhangjiang Biotechnology Co., Ltd., Shanghai, China
| | - Jiawei Cao
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, Shanghai Zhangjiang Biotechnology Co., Ltd., Shanghai, China
| | - Rongrong Fu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Mengjiao Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Xiang Zhao
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Fugui Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Xinmeng Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
| | - Taimin Dong
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
| | - Xu Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
| | - Weizhu Qian
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shen Hou
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lusha Ji
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dapeng Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huaizu Guo
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
- NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, Shanghai Zhangjiang Biotechnology Co., Ltd., Shanghai, China
| |
Collapse
|
29
|
Liu HN, Wang XY, Zou Y, Wu WB, Lin Y, Ji BY, Wang TY. Synthetic enhancers including TFREs improve transgene expression in CHO cells. Heliyon 2024; 10:e26901. [PMID: 38468921 PMCID: PMC10926067 DOI: 10.1016/j.heliyon.2024.e26901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
The human cytomegalovirus major immediate early gene (CMV) promoter is currently the most preferred promoter for recombinant therapeutic proteins (RTPs) production in CHO cells. To enhance the production of RTPs, five synthetic enhancers including multiple transcription factor regulatory elements (TFREs) were evaluated to enhance recombinant protein level in transient and stably transfected CHO cells. Compared with the control, four elements can enhance the report genes expression under both two transfected states. Further, the function of these four enhancers on human serum albumin (HSA) were investigated. We found that the transient expression can increase by up to 1.5 times, and the stably expression can maximum increase by up to 2.14 times. The enhancement of transgene expression was caused by the boost of their corresponding mRNA levels. Transcriptomics analysis was performed and found that transcriptional activation and cell cycle regulation genes were involved. In conclusion, optimization of enhancers in the CMV promoter could increase the production yield of transgene in transfected CHO cells, which has significance for developing high-yield CHO cell expression system.
Collapse
Affiliation(s)
- Hui-Ning Liu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- SanQuan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Xiao-Yin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Zou
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Wen-Bao Wu
- Shanghai Immunocan Biotech Co., LTD, Shanghai 200000, China
| | - Yan Lin
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
| | - Bo-Yu Ji
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
30
|
Jari M, Abdoli S, Bazi Z, Shamsabadi FT, Roshanmehr F, Shahbazi M. Enhancing protein production and growth in chinese hamster ovary cells through miR-107 overexpression. AMB Express 2024; 14:16. [PMID: 38302631 PMCID: PMC10834913 DOI: 10.1186/s13568-024-01670-y] [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: 10/12/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
Chinese Hamster Ovary (CHO) cells are widely employed as host cells for biopharmaceutical production. The manufacturing of biopharmaceuticals poses several challenges, including restricted growth potential and inadequate productivity of the host cells. MicroRNAs play a crucial role in regulating gene expression and are considered highly promising tools for cell engineering to enhance protein production. Our study aimed to evaluate the effects of miR-107, which is recognized as an onco-miR, on erythropoietin-producing CHO cells (CHO-hEPO). To assess the impact of miR-107 on CHO cells, a DNA plasmid containing miR-107 was introduced to CHO-hEPO cells through transfection. Cell proliferation and viability were assessed using the trypan blue dye exclusion method. Cell cycle analysis was conducted by utilizing propidium iodide (PI) staining. The quantification of EPO was determined using an immunoassay test. Moreover, the impact of miR-107 on the expression of downstream target genes was evaluated using qRT-PCR. Our findings highlight and underscore the substantial impact of transient miR-107 overexpression, which led to a remarkable 2.7-fold increase in EPO titers and a significant 1.6-fold increase in the specific productivity of CHO cells (p < 0.01). Furthermore, this intervention resulted in significant enhancements in cell viability and growth rate (p < 0.05). Intriguingly, the overexpression of miR‑107 was linked to the downregulation of LATS2, PTEN, and TSC1 genes while concurrently driving upregulation in transcript levels of MYC, YAP, mTOR, and S6K genes within transgenic CHO cells. In conclusion, this study collectively underscores the feasibility of utilizing cancer-associated miRNAs as a powerful tool for CHO cell engineering. However, more in-depth exploration is warranted to unravel the precise molecular intricacies of miR-107's effects in the context of CHO cells.
Collapse
Affiliation(s)
- Maryam Jari
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Bazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Tash Shamsabadi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farnaz Roshanmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran
- Department of Medical Biotechnology School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Shastkola Road, Falsafi Complex, Gorgan, Zip code: 4934174611, Iran.
- AryaTina Gene (ATG) Biopharmaceutical Company Gorgan, Gorgan, Iran.
| |
Collapse
|
31
|
Rahman MR, Kawabe Y, Suzuki K, Chen S, Amamoto Y, Kamihira M. Inducible transgene expression in CHO cells using an artificial transcriptional activator with estrogen-binding domain. Biotechnol J 2024; 19:e2300362. [PMID: 38161242 DOI: 10.1002/biot.202300362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Biopharmaceuticals, including therapeutic antibodies, are rapidly growing products in the pharmaceutical market. Mammalian cells, such as Chinese hamster ovary (CHO) cells, are widely used as production hosts because recombinant antibodies require complex three-dimensional structures modified with sugar chains. Recombinant protein production using mammalian cells is generally performed with cell growth. In this study, we developed a technology that controls cell growth and recombinant protein production to induce recombinant protein production with predetermined timing. Expression of green fluorescent protein (GFP) gene and a single-chain antibody fused with the Fc-region of the human IgG1 (scFv-Fc) gene can be induced and mediated by the estrogen receptor-based artificial transcription factor Gal4-ERT2-VP16 and corresponding inducer drugs. We generated CHO cells using an artificial gene expression system. The addition of various concentrations of inducer drugs to the culture medium allowed control of proliferation and transgene expression of the engineered CHO cells. Use of 4-hydroxytamoxifen, an antagonist of estrogen, as an inducing agent yielded high gene expression at a concentration more than 10-fold lower than that of β-estradiol. When scFv-Fc was produced under inducing conditions, continuous production was possible for more than 2 weeks while maintaining high specific productivity (57 pg cell-1 day-1 ). This artificial gene expression control system that utilizes the estrogen response of estrogen receptors can be an effective method for inducible production of biopharmaceuticals.
Collapse
Affiliation(s)
- Md Rashidur Rahman
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Kozumi Suzuki
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Satoshi Chen
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Yuki Amamoto
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
32
|
Thalén NB, Barzadd MM, Lundqvist M, Rodhe J, Andersson M, Bidkhori G, Possner D, Su C, Nilsson J, Eisenhut P, Malm M, Karlsson A, Vestin J, Forsberg J, Nordling E, Mardinoglu A, Volk AL, Sandegren A, Rockberg J. Tuning of CHO secretional machinery improve activity of secreted therapeutic sulfatase 150-fold. Metab Eng 2024; 81:157-166. [PMID: 38081506 DOI: 10.1016/j.ymben.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 10/12/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Rare diseases are, despite their name, collectively common and millions of people are affected daily of conditions where treatment often is unavailable. Sulfatases are a large family of activating enzymes related to several of these diseases. Heritable genetic variations in sulfatases may lead to impaired activity and a reduced macromolecular breakdown within the lysosome, with several severe and lethal conditions as a consequence. While therapeutic options are scarce, treatment for some sulfatase deficiencies by recombinant enzyme replacement are available. The recombinant production of such sulfatases suffers greatly from both low product activity and yield, further limiting accessibility for patient groups. To mitigate the low product activity, we have investigated cellular properties through computational evaluation of cultures with varying media conditions and comparison of two CHO clones with different levels of one active sulfatase variant. Transcriptome analysis identified 18 genes in secretory pathways correlating with increased sulfatase production. Experimental validation by upregulation of a set of three key genes improved the specific enzymatic activity at varying degree up to 150-fold in another sulfatase variant, broadcasting general production benefits. We also identified a correlation between product mRNA levels and sulfatase activity that generated an increase in sulfatase activity when expressed with a weaker promoter. Furthermore, we suggest that our proposed workflow for resolving bottlenecks in cellular machineries, to be useful for improvements of cell factories for other biologics as well.
Collapse
Affiliation(s)
- Niklas Berndt Thalén
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Mona Moradi Barzadd
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Magnus Lundqvist
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | | | - Gholamreza Bidkhori
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden; AIVIVO Ltd. Unit 25, Bio-innovation centre, Cambridge Science park, Cambridge, UK
| | | | - Chao Su
- SOBI AB, Tomtebodavägen 23A, Stockholm, Sweden
| | | | - Peter Eisenhut
- ACIB - Austrian Centre of Industrial Biotechnology, Krenngasse 37, 8010 Graz, Austria; BOKU - University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, 1190, Austria
| | - Magdalena Malm
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Alice Karlsson
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | | | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden
| | - Anna-Luisa Volk
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | - Johan Rockberg
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden.
| |
Collapse
|
33
|
Hashizume T, Ying BW. Challenges in developing cell culture media using machine learning. Biotechnol Adv 2024; 70:108293. [PMID: 37984683 DOI: 10.1016/j.biotechadv.2023.108293] [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: 07/01/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Microbial and mammalian cells are widely used in the food, pharmaceutical, and medical industries. Developing or optimizing culture media is essential to improve cell culture performance as a critical technology in cell culture engineering. Methodologies for media optimization have been developed to a great extent, such as the approaches of one-factor-at-a-time (OFAT) and response surface methodology (RSM). The present review introduces the emerging machine learning (ML) technology in cell culture engineering by combining high-throughput experimental technologies to develop highly efficient and effective culture media. The commonly used ML algorithms and the successful applications of employing ML in medium optimization are summarized. This review highlights the benefits of ML-assisted medium development and guides the selection of the media optimization method appropriate for various cell culture purposes.
Collapse
Affiliation(s)
- Takamasa Hashizume
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan
| | - Bei-Wen Ying
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan.
| |
Collapse
|
34
|
Clarke EC. Considerations for Glycoprotein Production. Methods Mol Biol 2024; 2762:329-351. [PMID: 38315375 DOI: 10.1007/978-1-0716-3666-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
This chapter is intended to provide insights for researchers aiming to choose an appropriate expression system for the production of recombinant glycoproteins. Producing glycoproteins is complex, as glycosylation patterns are determined by the availability and abundance of specific enzymes rather than a direct genetic blueprint. Furthermore, the cell systems often employed for protein production are evolutionarily distinct, leading to significantly different glycosylation when utilized for glycoprotein production. The selection of an appropriate production system depends on the intended applications and desired characteristics of the protein. Whether the goal is to produce glycoproteins mimicking native conditions or to intentionally alter glycan structures for specific purposes, such as enhancing immunogenicity in vaccines, understanding glycosylation present in the different systems and in different growth conditions is essential. This chapter will cover Escherichia coli, baculovirus/insect cell systems, Pichia pastoris, as well as different mammalian cell culture systems including Chinese hamster ovary (CHO) cells, human endothelial kidney (HEK) cell lines, and baby hamster kidney (BHK) cells.
Collapse
Affiliation(s)
- Elizabeth C Clarke
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
35
|
Sorourian S, Behzad Behbahani A, Forouzanfar M, Jafarinia M, Safari F. Time and Cost-Effective Genome Editing Protocol for Simultaneous Caspase 8 Associated Protein 2 Gene Knock in/out in Chinese Hamster Ovary Cells Using CRISPR-Cas9 System. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3714. [PMID: 38827341 PMCID: PMC11139449 DOI: 10.30498/ijb.2024.398567.3714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/27/2023] [Indexed: 06/04/2024]
Abstract
Background CHO cells are preferred for producing biopharmaceuticals, and genome editing technologies offer opportunities to enhance recombinant protein production. Targeting apoptosis-related genes, such as Caspases 8-Associated Protein 2 (CASP8AP2), improves CHO cell viability and productivity. Integrating robust strategies with the CRISPR-Cas9 system enables its application in CHO cell engineering. Objectives This study was performed to develop a cost-effective protocol using the CRISPR-Cas9 system combined with the HITI strategy for simultaneous CASP8AP2 gene deletion/insertion in CHO cells and to assess its impact on cell viability and protein expression. Materials and Methods We developed an efficient protocol for CHO cell engineering by combining CRISPR/Cas9 with the HITI strategy. Two distinct sgRNA sequences were designed to target the 3' UTR region of the CASP8AP2 gene using CHOPCHOP software. The gRNAs were cloned into PX459 and PX460-1 vectors and transfected into CHO cells using the cost-effective PEI reagent. A manual selection system was employed to streamline the process of single-cell cloning. MTT assays assessed gene silencing and cell viability at 24, 48, and 72 hours. Flow cytometry evaluated protein expression in CASP8AP2-silenced CHO cells. Results The study confirmed the robustness of combining CRISPR-Cas9 with the HITI strategy, achieving a high 60% efficiency in generating knockout clones. PEI transfection successfully delivered the constructs to nearly 65% of the clones, with the majority being homozygous. The protocol proved feasible for resource-limited labs, requiring only an inverted fluorescent microscope. CASP8AP2 knockout (CHO-KO) cells exhibited significantly extended cell viability compared to CHO-K1 cells when treated with NaBu, with IC50 values of 7.28 mM and 14.25 mM at 48 hours, respectively (P-value 24 hours ≤ 0.0001, 48 hours ≤ 0.0001, P-value 72 hours = 0.0007). CHO CASP8AP2-silenced cells showed a 1.3-fold increase in JRed expression compared to native cells. Conclusions CRISPR-Cas9 and HITI strategy was used to efficiently engineer CHO cells for simultaneous CASP8AP2 gene deletion/insertion, which improved cell viability and protein expression.
Collapse
Affiliation(s)
- Soofia Sorourian
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Abbas Behzad Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Forouzanfar
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Mojtaba Jafarinia
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
36
|
Martínez VS, Rodriguez K, McCubbin T, Tong J, Mahler S, Shave E, Baker K, Munro TP, Marcellin E. Amino acid degradation pathway inhibitory by-products trigger apoptosis in CHO cells. Biotechnol J 2024; 19:e2300338. [PMID: 38375561 DOI: 10.1002/biot.202300338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 02/21/2024]
Abstract
Chinese hamster ovary (CHO) cells are widely used to produce complex biopharmaceuticals. Improving their productivity is necessary to fulfill the growing demand for such products. One way to enhance productivity is by cultivating cells at high densities, but inhibitory by-products, such as metabolite derivatives from amino acid degradation, can hinder achieving high cell densities. This research examines the impact of these inhibitory by-products on high-density cultures. We cultured X1 and X2 CHO cell lines in a small-scale semi-perfusion system and introduced a mix of inhibitory by-products on day 10. The X1 and X2 cell lines were chosen for their varied responses to the by-products; X2 was susceptible, while X1 survived. Proteomics revealed that the X2 cell line presented changes in the proteins linked to apoptosis regulation, cell building block synthesis, cell growth, DNA repair, and energy metabolism. We later used the AB cell line, an apoptosis-resistant cell line, to validate the results. AB behaved similar to X1 under stress. We confirmed the activation of apoptosis in X2 using a caspase assay. This research provides insights into the mechanisms of cell death triggered by inhibitory by-products and can guide the optimization of CHO cell culture for biopharmaceutical manufacturing.
Collapse
Affiliation(s)
- Verónica S Martínez
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
| | - Karen Rodriguez
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
| | - Timothy McCubbin
- Queensland Metabolomics and Proteomics (Q-MAP), The University of Queensland, St Lucia, Queensland, Australia
| | - Junjie Tong
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
| | - Stephen Mahler
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
| | - Evan Shave
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- Patheon, by Thermo Fisher Scientific, Woolloongabba, Queensland, Australia
| | - Kym Baker
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- Patheon, by Thermo Fisher Scientific, Woolloongabba, Queensland, Australia
| | - Trent P Munro
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- National Biologics Facility, The University of Queensland, St Lucia, Queensland, Australia
| | - Esteban Marcellin
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- Queensland Metabolomics and Proteomics (Q-MAP), The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
37
|
Meng P, Wei Y, Liang M, Yuan W, Zhu L, Sun J, Huang J, Zhu J. Fusion with CTP increases the stability of recombinant neuritin. Protein Expr Purif 2023; 212:106344. [PMID: 37567400 DOI: 10.1016/j.pep.2023.106344] [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: 06/10/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Neuritin is a vital neurotrophin that plays an essential role in recovery from nerve injury and neurodegenerative diseases and may become a new target for treating these conditions. However, improving neuritin protein stability is an urgent problem. In this study, to obtain active and stable neuritin proteins, we added a carboxyl-terminal peptide (CTP) sequence containing four O-linked glycosylation sites to the C-terminus of neuritin and cloned it into the Chinese hamster ovary (CHO) expression system. The neuritin-CTP protein was purified using a His-Tag purification strategy after G418 screening of stable high-expression cell lines. Ultimately, we obtained neuritin-CTP protein with a purity >90%. Functional analyses showed that the purified neuritin-CTP protein promoted the neurite outgrowth of PC12 cells, and stability experiments showed that neuritin stability was increased by adding CTP. These results indicate that neuritin protein-CTP fusion effectively increases stability without affecting secretion and activity. This study offers a sound strategy for improving the stability of neuritin protein and provides material conditions for further study of the function of neuritin.
Collapse
Affiliation(s)
- Pingping Meng
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Yu Wei
- The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Mengjie Liang
- Department of Clinical Laboratory, Hospital of Xinjiang Production and Construction Corps/Second Affiliated Hospital, Medical School of Shihezi University, Urumqi, Xinjiang, 832000, China
| | - Wumei Yuan
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Liyan Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jiawei Sun
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
38
|
Huang Z, Habib A, Zhao G, Ding X. CRISPR-Cas9 Mediated Stable Expression of Exogenous Proteins in the CHO Cell Line through Site-Specific Integration. Int J Mol Sci 2023; 24:16767. [PMID: 38069090 PMCID: PMC10706275 DOI: 10.3390/ijms242316767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Chinese hamster ovary (CHO) cells are a popular choice in biopharmaceuticals because of their beneficial traits, including high-density suspension culture, safety, and exogenously produced proteins that closely resemble natural proteins. Nevertheless, a decline in the expression of exogenous proteins is noted as culture time progresses. This is a consequence of foreign gene recombination into chromosomes by random integration. The current investigation employs CRISPR-Cas9 technology to integrate foreign genes into a particular chromosomal location for sustained expression. Results demonstrate the successful integration of enhanced green fluorescent protein (EGFP) and human serum albumin (HSA) near base 434814407 on chromosome NC_048595.1 of CHO-K1 cells. Over 60 successive passages, monoclonal cell lines were produced that consistently expressed all relevant external proteins without discernible variation in expression levels. In conclusion, the CHO-K1 cell locus, NC_048595.1, proves an advantageous locus for stable exogenous protein expression. This study provides a viable approach to establishing a CHO cell line capable of enduring reliable exogenous protein expression.
Collapse
Affiliation(s)
- Zhipeng Huang
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Arslan Habib
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Guoping Zhao
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaoming Ding
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
39
|
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
|
40
|
Li L, Qiao S, Wang S, Liu J, Zhao K, Zhou Y, Li G, Jiang Y, Liu C, Tong G, Tong W, Gao F. Expression of ASFV p17 in CHO cells and identification of one novel epitope using a monoclonal antibody. Virus Res 2023; 336:199194. [PMID: 37579847 PMCID: PMC10470389 DOI: 10.1016/j.virusres.2023.199194] [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: 05/17/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
As a highly pathogenic large DNA virus, African swine fever virus (ASFV) has huge particles and numerous encoded proteins. At present, few of the existing studies on ASFV proteins have investigated the function of p17. Specific antibodies against p17 to promote the development of prevention techniques against African swine fever (ASF) are urgently needed. Herein, we successfully expressed ASFV p17 in CHO cells using a suspension culture system and generated a monoclonal antibody (mAb) against p17. The mAb recognized a novel linear epitope (8LLSHNLSTREGIK20) and exhibited specific reactivity, which was conducive to the identification of ectopically expressed p17, the recombinant porcine reproductive and respiratory syndrome virus expressing p17, and the ASFV-SY18. The epitope was conservative among genotype I and genotype II ASFV strains. Overall, the mAb against p17 revealed efficient detection and promising application prospects, making it a useful tool for future vaccine research on ASF. Determination of the conserved linear epitope of p17 would contribute to the in-depth exploration of the biological function of ASFV antigen protein.
Collapse
Affiliation(s)
- Liwei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Sina Qiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Shumao Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Jiachen Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Kuan Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Guoxin Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Changlong Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Wu Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, China.
| | - Fei Gao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
41
|
Song B, Wei W, Liu X, Huang Y, Zhu S, Yi L, Eerdunfu, Ding H, Zhao M, Chen J. Recombinant Porcine Interferon-α Decreases Pseudorabies Virus Infection. Vaccines (Basel) 2023; 11:1587. [PMID: 37896991 PMCID: PMC10610829 DOI: 10.3390/vaccines11101587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Interferon (IFN) is a cell-secreted cytokine possessing biological activities including antiviral functioning, immune regulation, and others. Interferon-alpha (IFN-α) mainly derives from plasmacytoid dendritic cells, which activate natural killer cells and regulate immune responses. IFN-α responds to the primary antiviral mechanism in the innate immune system, which can effectively cure acute infectious diseases. Pseudorabies (PR) is an acute infectious disease caused by pseudorabies virus (PRV). The clinical symptoms of PRV are as follows: reproductive dysfunction among pregnant sows and high mortality rates among piglets. These pose a severe threat to the swine industry. Related studies show that IFN-α has broad applications in preventing and treating viral diseases. Therefore, a PRV mouse model using artificial infection was established in this study to explore the pathogenic effect of IFN-α on PRV. We designed a sequence with IFN-α4 (M28623, Genbank) and cloned it on the lentiviral vector. CHO-K1 cells were infected and identified using WB and RT-PCR; a CHO-K1 cell line with a stable expression of the recombinant protein PoIFN-α was successfully constructed. H&E staining and virus titer detection were used to investigate the recombinant protein PoIFN-α's effect on PR in BALB/c mice. The results show that the PoIFN-α has a preventive and therapeutic impact on PR. In conclusion, the recombinant protein can alleviate symptoms and reduce the replication of PRV in vivo.
Collapse
Affiliation(s)
- Bowen Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Wenkang Wei
- Agro-Biological Gene Research Center, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China;
| | - Xueyi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Yaoyao Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Shuaiqi Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Eerdunfu
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
- Agro-Biological Gene Research Center, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China;
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (B.S.); (X.L.); (Y.H.); (S.Z.); (L.Y.); (H.D.); (M.Z.)
| |
Collapse
|
42
|
Yang S, Braczkowski R, Chen SH, Busse R, Li Y, Fabri L, Bekard IB. Scalability of Sartobind ® Rapid A Membrane for High Productivity Monoclonal Antibody Capture. MEMBRANES 2023; 13:815. [PMID: 37887987 PMCID: PMC10608304 DOI: 10.3390/membranes13100815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023]
Abstract
Improved upstream titres in therapeutic monoclonal antibody (mAb) production have shifted capacity constraints to the downstream process. The consideration of membrane-based chromatographic devices as a debottlenecking option is gaining increasing attention with the recent introduction of high-capacity bind and elute membranes. We have evaluated the performance and scalability of the Sartobind® Rapid A affinity membrane (1 mL) for high-productivity mAb capture. For scalability assessment, a 75 mL prototype device was used to process 100 L of clarified cell culture harvest (CH) on a novel multi-use rapid cycling chromatography system (MU-RCC). MabSelect™ PrismA (4.7 mL) was used as a benchmark comparator for Protein A (ProtA) resin studies. Results show that in addition to a productivity gain of >10×, process and product quality attributes were either improved or comparable to the benchmark. Concentrations of eluate pools were 7.5× less than that of the benchmark, with the comparatively higher bulk volume likely to cause handling challenges at process scale. The MU-RCC system is capable of membrane operation at pilot scale with comparable product quality profile to the 1 mL device. The Sartobind® Rapid A membrane is a scalable alternative to conventional ProtA resin chromatography for the isolation and purification of mAbs from harvested cell culture media.
Collapse
Affiliation(s)
- Sabrina Yang
- CSL Innovation Pty Ltd., 655 Elizabeth Street, Melbourne, VIC 3000, Australia
| | - Ryszard Braczkowski
- CSL Innovation Pty Ltd., 655 Elizabeth Street, Melbourne, VIC 3000, Australia
| | - Shih-Hsun Chen
- CSL Innovation Pty Ltd., 655 Elizabeth Street, Melbourne, VIC 3000, Australia
| | - Ricarda Busse
- Sartorius Stedim Biotech GmbH, August-Spindler-Strasse 11, 37079 Goettingen, Germany
| | - Yudhi Li
- Sartorius Stedim Singapore Pte Ltd., 30 Pasir Panjang Rd., #06-31A/32, Singapore 117440, Singapore
| | - Louis Fabri
- CSL Innovation Pty Ltd., 655 Elizabeth Street, Melbourne, VIC 3000, Australia
| | | |
Collapse
|
43
|
Martinez-Navajas G, Ceron-Hernandez J, Simon I, Lupiañez P, Diaz-McLynn S, Perales S, Modlich U, Guerrero JA, Martin F, Sevivas T, Lozano ML, Rivera J, Ramos-Mejia V, Tersteeg C, Real PJ. Lentiviral gene therapy reverts GPIX expression and phenotype in Bernard-Soulier syndrome type C. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:75-92. [PMID: 37416759 PMCID: PMC10320622 DOI: 10.1016/j.omtn.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023]
Abstract
Bernard-Soulier syndrome (BSS) is a rare congenital disease characterized by macrothrombocytopenia and frequent bleeding. It is caused by pathogenic variants in three genes (GP1BA, GP1BB, or GP9) that encode for the GPIbα, GPIbβ, and GPIX subunits of the GPIb-V-IX complex, the main platelet surface receptor for von Willebrand factor, being essential for platelet adhesion and aggregation. According to the affected gene, we distinguish BSS type A1 (GP1BA), type B (GP1BB), or type C (GP9). Pathogenic variants in these genes cause absent, incomplete, or dysfunctional GPIb-V-IX receptor and, consequently, a hemorrhagic phenotype. Using gene-editing tools, we generated knockout (KO) human cellular models that helped us to better understand GPIb-V-IX complex assembly. Furthermore, we developed novel lentiviral vectors capable of correcting GPIX expression, localization, and functionality in human GP9-KO megakaryoblastic cell lines. Generated GP9-KO induced pluripotent stem cells produced platelets that recapitulated the BSS phenotype: absence of GPIX on the membrane surface and large size. Importantly, gene therapy tools reverted both characteristics. Finally, hematopoietic stem cells from two unrelated BSS type C patients were transduced with the gene therapy vectors and differentiated to produce GPIX-expressing megakaryocytes and platelets with a reduced size. These results demonstrate the potential of lentiviral-based gene therapy to rescue BSS type C.
Collapse
Affiliation(s)
- Gonzalo Martinez-Navajas
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Jorge Ceron-Hernandez
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Iris Simon
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Pablo Lupiañez
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
| | - Sofia Diaz-McLynn
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Sonia Perales
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Ute Modlich
- Department of Gene and Cell Therapy, Institute of Regenerative Medicine, University of Zürich, Wagistrasse 12, 8952 Schlieren-Zürich, Switzerland
| | - Jose A. Guerrero
- Department of Haematology, University of Cambridge, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Francisco Martin
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, Avenida Ilustracion S/n, 18016 Granada, Spain
| | - Teresa Sevivas
- Serviço de Sangue, Medicina Transfusional e Imunohemoterapia Do Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria L. Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, U765 Murcia, Spain
| | - Jose Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, U765 Murcia, Spain
- Grupo Español de Alteraciones Plaquetarias Congénitas (GEAPC), Madrid, Spain
| | - Veronica Ramos-Mejia
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Pedro J. Real
- GENyO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, PTS, Granada, Avenida de la Ilustracion 114, 18016 Granada, Spain
- University of Granada, Department of Biochemistry and Molecular Biology I, Faculty of Science, Avenida Fuentenueva S/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| |
Collapse
|
44
|
Sacco SA, McAtee Pereira AG, Trenary I, Smith KD, Betenbaugh MJ, Young JD. Overexpression of peroxisome proliferator-activated receptor γ co-activator-1⍺ (PGC-1⍺) in Chinese hamster ovary cells increases oxidative metabolism and IgG productivity. Metab Eng 2023; 79:108-117. [PMID: 37473833 DOI: 10.1016/j.ymben.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/17/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Chinese hamster ovary (CHO) cells are used extensively to produce protein therapeutics, such as monoclonal antibodies (mAbs), in the biopharmaceutical industry. MAbs are large proteins that are energetically demanding to synthesize and secrete; therefore, high-producing CHO cell lines that are engineered for maximum metabolic efficiency are needed to meet increasing demands for mAb production. Previous studies have identified that high-producing cell lines possess a distinct metabolic phenotype when compared to low-producing cell lines. In particular, it was found that high mAb production is correlated to lactate consumption and elevated TCA cycle flux. We hypothesized that enhancing flux through the mitochondrial TCA cycle and oxidative phosphorylation would lead to increased mAb productivities and final titers. To test this hypothesis, we overexpressed peroxisome proliferator-activated receptor γ co-activator-1⍺ (PGC-1⍺), a gene that promotes mitochondrial metabolism, in an IgG-producing parental CHO cell line. Stable cell pools overexpressing PGC-1⍺ exhibited increased oxygen consumption, indicating increased mitochondrial metabolism, as well as increased mAb specific productivity compared to the parental line. We also performed 13C metabolic flux analysis (MFA) to quantify how PGC-1⍺ overexpression alters intracellular metabolic fluxes, revealing not only increased TCA cycle flux, but global upregulation of cellular metabolic activity. This study demonstrates the potential of rationally engineering the metabolism of industrial cell lines to improve overall mAb productivity and to increase the abundance of high-producing clones in stable cell pools.
Collapse
Affiliation(s)
- Sarah A Sacco
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin D Smith
- Pharmaceutical Development and Manufacturing Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
45
|
Novak N, Baumann M, Friss A, Cairns V, DeMaria C, Borth N. LncRNA analysis of mAb producing CHO clones reveals marker and engineering potential. Metab Eng 2023; 78:26-40. [PMID: 37196898 DOI: 10.1016/j.ymben.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a potential new cell line engineering tool for improvement of yield and stability of CHO cells. In this study, we performed RNA sequencing of mAb producer CHO clones to study the lncRNA and protein coding transcriptome in relation to productivity. First, a robust linear model was used to identify genes correlating to productivity. To unravel specific patterns in expression of these genes, we employed weighted gene coexpression analysis (WGCNA) to find coexpressed modules, looking both for lncRNAs and coding genes. There was little overlap in the genes associated with productivity between the two products studied, possibly due to the difference in absolute range of productivity between the two mAbs. Therefore, we focused on the product with higher productivity and stronger candidate lncRNAs. To evaluate their potential as engineering targets, these candidate lncRNAs were transiently overexpressed or deleted by stable CRISPR Cas9 knock out both in a high and a low productivity subclone. We found that the thus achieved expression level of the identified lncRNAs, as confirmed by qPCR, does correlate well to productivity, so that they represent good markers that may be used for early clone selection. Additionally, we found that the deletion of one tested lncRNA region decreased viable cell density (VCD), prolonged culture time and increased cell size, final titer and specific productivity per cell. These results demonstrate the feasibility and usefulness of engineering lncRNA expression in production cell lines.
Collapse
Affiliation(s)
- Neža Novak
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Martina Baumann
- ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Amy Friss
- Sanofi Biopharmaceutics Development, Framingham, MA, USA
| | - Victor Cairns
- Sanofi Biopharmaceutics Development, Framingham, MA, USA
| | | | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria.
| |
Collapse
|
46
|
Analysis of the in vitro function and internalization ability of a humanized EGFR antibody AE01 expressed by Chinese hamster ovary cells. Protein Expr Purif 2023; 206:106243. [PMID: 36754125 DOI: 10.1016/j.pep.2023.106243] [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: 09/16/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
The primary objective of this study was to obtain humanized EGFR antibody and to study it in vitro binding and endocytosis to A431 epidermoid carcinoma cells overexpressing EGFR. Firstly, humanized anti-EGFR AE01 was stably expressed in CHO system. The expression of AE01 was detected by SDS-PAGE and Western blot. The binding and endocytosis of AE01 were detected by flow cytometry and immunofluorescence assay. The results showed that: (1) Pure humanized AE01 was prepared, (2) AE01 specifically binds to A431 cells on the cell surface (EGFR-positive), but not binds to NIH 3T3 cells (EGFR-negative), (3) AE01 can effectively inhibit the proliferation of A431 cells, and (4) AE01 binds to A431 cell surface triggered internalization. The antibody is expected to be a candidate molecule for EGFR overexpressed cancer cell targeted therapeutic vectors.
Collapse
|
47
|
Alejandra WP, Miriam Irene JP, Fabio Antonio GS, Patricia RGR, Elizabeth TA, Juan Pablo AA, Rebeca GV. Production of monoclonal antibodies for therapeutic purposes: A review. Int Immunopharmacol 2023; 120:110376. [PMID: 37244118 DOI: 10.1016/j.intimp.2023.110376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
Monoclonal antibodies (mAbs) have been used in the development of immunotherapies that target a variety of diseases, such as cancer, autoimmune diseases, and even viral infections; they play a key role in immunization and are expected after vaccination. However, some conditions do not promote the development of neutralizing antibodies. Production and use of mAbs, generated in biofactories, represent vast potential as aids in immunological responses when the organism cannot produce them on their own, these convey unique specificity by recognizing and targeting specific antigen. Antibodies can be defined as heterotetrametric glycoproteins of symmetric nature, and they participate as effector proteins in humoral responses. Additionally, there are different types of mAbs (murine, chimeric, humanized, human, mAbs as Antibody-drug conjugates and bispecific mAbs) discussed in the present work. When these molecules are produced in vitro as mAbs, several common techniques, such as hybridomas or phage display are used. There are several preferred cell lines that function as biofactories, for the production of mAbs, the selection of which rely on the variation of adaptability, productivity and both phenotypic and genotypic shifts. After the cell expression systems and culture techniques are used, there are diverse specialized downstream processes to achieve desired yield and isolation as well as product quality and characterization. Novel perspectives regarding these protocols represent a potential improvement for mAbs high-scale production.
Collapse
Affiliation(s)
- Waller-Pulido Alejandra
- Tecnologico de Monterrey, School of Engineering and Science, Ave. General Ramon Corona 2514, 45138 Zapopan, Jalisco, Mexico
| | - Jiménez-Pérez Miriam Irene
- Tecnologico de Monterrey, School of Medicine and Health Science, Ave. General Ramon Corona 2514, 45138 Zapopan, Jalisco, Mexico
| | - Gonzalez-Sanchez Fabio Antonio
- Tecnologico de Monterrey, School of Engineering and Science, Ave. General Ramon Corona 2514, 45138 Zapopan, Jalisco, Mexico
| | | | | | - Aleman-Aguilar Juan Pablo
- Tecnologico de Monterrey, School of Medicine and Health Science, Ave. General Ramon Corona 2514, 45138 Zapopan, Jalisco, Mexico.
| | - Garcia-Varela Rebeca
- Tecnologico de Monterrey, School of Engineering and Science, Ave. General Ramon Corona 2514, 45138 Zapopan, Jalisco, Mexico.
| |
Collapse
|
48
|
Feser CJ, Williams JM, Lammers DT, Bingham JR, Eckert MJ, Tolar J, Osborn MJ. Engineering Human Cells Expressing CRISPR/Cas9-Synergistic Activation Mediators for Recombinant Protein Production. Int J Mol Sci 2023; 24:8468. [PMID: 37239814 PMCID: PMC10218281 DOI: 10.3390/ijms24108468] [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: 03/07/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant engineering for protein production commonly employs plasmid-based gene templates for introduction and expression of genes in a candidate cell system in vitro. Challenges to this approach include identifying cell types that can facilitate proper post-translational modifications and difficulty expressing large multimeric proteins. We hypothesized that integration of the CRISPR/Cas9-synergistic activator mediator (SAM) system into the human genome would be a powerful tool capable of robust gene expression and protein production. SAMs are comprised of a "dead" Cas9 (dCas9) linked to transcriptional activators viral particle 64 (VP64), nuclear factor-kappa-B p65 subunit (p65), and heat shock factor 1 (HSF1) and are programmable to single or multiple gene targets. We integrated the components of the SAM system into human HEK293, HKB11, SK-HEP1, and HEP-g2 cells using coagulation factor X (FX) and fibrinogen (FBN) as proof of concept. We observed upregulation of mRNA in each cell type with concomitant protein expression. Our findings demonstrate the capability of human cells stably expressing SAM for user-defined singleplex and multiplex gene targeting and highlight their broad potential utility for recombinant engineering as well as transcriptional modulation across networks for basic, translational, and clinical modeling and applications.
Collapse
Affiliation(s)
- Colby J. Feser
- Department of Pediatrics, Division of Blood and Marrow Transplantation, MMC 366 Mayo, 8366A, 420 Delaware Street SE, Minneapolis, MN 55455, USA; (C.J.F.); (J.T.)
| | - James M. Williams
- Department of General Surgery, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA 98431, USA; (J.M.W.); (D.T.L.); (J.R.B.); (M.J.E.)
| | - Daniel T. Lammers
- Department of General Surgery, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA 98431, USA; (J.M.W.); (D.T.L.); (J.R.B.); (M.J.E.)
| | - Jason R. Bingham
- Department of General Surgery, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA 98431, USA; (J.M.W.); (D.T.L.); (J.R.B.); (M.J.E.)
| | - Matthew J. Eckert
- Department of General Surgery, Madigan Army Medical Center, 9040 Jackson Ave, Tacoma, WA 98431, USA; (J.M.W.); (D.T.L.); (J.R.B.); (M.J.E.)
- Department of Surgery, University of North Carolina, 160 Dental Circle, Chapel Hill, NC 27599, USA
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, MMC 366 Mayo, 8366A, 420 Delaware Street SE, Minneapolis, MN 55455, USA; (C.J.F.); (J.T.)
| | - Mark J. Osborn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, MMC 366 Mayo, 8366A, 420 Delaware Street SE, Minneapolis, MN 55455, USA; (C.J.F.); (J.T.)
| |
Collapse
|
49
|
Bazaz M, Adeli A, Azizi M, Karimipoor M, Mahboudi F, Davoudi N. Overexpression of miR-32 in Chinese hamster ovary cells increases production of Fc-fusion protein. AMB Express 2023; 13:45. [PMID: 37160545 PMCID: PMC10170017 DOI: 10.1186/s13568-023-01540-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/22/2023] [Indexed: 05/11/2023] Open
Abstract
The demand for industrial genetically modified host cells were increased with the growth of the biopharmaceutical market. Numerous studies on improving host cell productivity have shown that altering host cell growth and viability through genetic engineering can increase recombinant protein production. During the last decades, it was demonstrated that overexpression or downregulation of some microRNAs in Chinese Hamster Ovary (CHO) cells as the host cell in biopharmaceutical manufacturing, can improve their productivity. The selection of microRNA targets has been based on their previously identified role in human cancers. MicroRNA-32 (miR-32), which is conserved between humans and hamsters (Crisetulus griseus), was shown to play a role in the regulation of cell proliferation and apoptosis in some human cancers. In this study, we investigated the effect of miR-32 overexpression on the productivity of CHO-VEGF-trap cells. Our results indicated that stable overexpression of miR-32 could dramatically increase the productivity of CHO cells by 1.8-fold. It also significantly increases cell viability, batch culture longevity, and cell growth. To achieve these results, following the construction of a single clone producing an Fc-fusion protein, we transfected cells with a pLexJRed-miR-32 plasmid to stably produce the microRNA and evaluate the impact of mir-32 overexpression on cell productivity, growth and viability in compare with scrambled control. Our findings highlight the application of miRNAs as engineering tools and indicated that miR-32 could be a target for engineering CHO cells to increase cell productivity.
Collapse
Affiliation(s)
- Masoume Bazaz
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Adeli
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Azizi
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Karimipoor
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Freidoun Mahboudi
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Noushin Davoudi
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
50
|
Grindes L, Florimond C, Ribault S, Raymond C, Dieryck W, Corbin C, Joucla G. Weak promoters to drive selection marker expression: improvement of cell line development process for therapeutic protein production in CHO-K1 cells. J Biotechnol 2023; 369:43-54. [PMID: 37149043 DOI: 10.1016/j.jbiotec.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Chinese Hamster Ovary cells have been widely used as host cells for production of recombinant therapeutic molecules. Cell line development is a decisive step, which must be carried out with an efficient process. In particular, degree of selection stringency is an important parameter for identification of rare, high-producing cell lines. In the CHOZN® CHO K1 platform, selection of top-producing clones is based on puromycin resistance, whose expression is driven by Simian Virus 40 Early (SV40E) promoter. In this study, novel promoters have been identified to drive expression of selection marker. Decrease of transcriptional activity compared to SV40E promoter was confirmed by RT-qPCR. Selection stringency was increased, as seen by decreased surviving rate of transfected mini-pools and longer recovery duration of transfected bulk pools. Several promoters led to a 1.5-fold increase of maximum titer and a 1.3-fold increase of mean specific productivity of the monoclonal antibody over the clone generation. Expression level was maintained stable over long term cultivation. Finally, productivity increase was confirmed on several monoclonal antibodies and fusion proteins. Lowering the strength of promoter for expression of selective pressure resistance is an efficient strategy to increase selection stringency, which can be applied on industrial CHO-based cell line development platforms.
Collapse
Affiliation(s)
- Lucie Grindes
- Process Development Department, Merck Biodevelopment, Martillac, France; Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France.
| | - Camille Florimond
- Process Development Department, Merck Biodevelopment, Martillac, France
| | - Sébastien Ribault
- Process Development Department, Merck Biodevelopment, Martillac, France
| | - Céline Raymond
- Process Development Department, Merck Biodevelopment, Martillac, France
| | - Wilfrid Dieryck
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France
| | - Cyrielle Corbin
- Process Development Department, Merck Biodevelopment, Martillac, France
| | - Gilles Joucla
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France
| |
Collapse
|