1
|
Selvaprakash K, Henry M, Ryan D, Meleady P. LC-MS/MS Analysis to Study the Ubiquitin-Modified Proteome of Recombinant Chinese Hamster Ovary Cells. Methods Mol Biol 2025; 2853:191-203. [PMID: 39460922 DOI: 10.1007/978-1-0716-4104-0_13] [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
Ubiquitination is one of the most important post-translational modifications (PTMs) and involves the covalent attachment of ubiquitin to a lysine residue on a target protein. Despite ubiquitination playing a crucial role in regulating cellular processes, the ubiquitinated proteome has not been studied extensively in recombinant Chinese hamster ovary (CHO) cells. Moreover, ubiquitination modification in CHO cells is likely to have an impact on protein function related to the efficient productivity of biopharmaceuticals. In this chapter, we describe a comprehensive protocol for ubiquitin di-Glycine (diGly) peptide enrichment using an immunoprecipitation method from recombinant CHO cell proteins followed by Liquid chromatography-Mass spectrometry (LC-MS) analysis of the ubiquitinated proteome. The methods described are also applicable to differential ubiquitinated proteomic studies.
Collapse
Affiliation(s)
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - David Ryan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
2
|
Zhang J, Du C, Pan Y, Zhang Z, Feng R, Ma M, Wang T. Optimization of a novel expression system for recombinant protein production in CHO cells. Sci Rep 2024; 14:24913. [PMID: 39438721 PMCID: PMC11496728 DOI: 10.1038/s41598-024-76995-6] [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: 06/30/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024] Open
Abstract
Chinese hamster ovary (CHO) cells are common mammalian cell lines for expressing recombinant proteins, yet the expression level of recombinant proteins is still hindered. Vector optimization and cell line modification are the key factors to improve the expression of recombinant proteins. In this study, the vector was optimized by adding the regulatory elements Kozak and Leader to the upstream of target gene to detect the transient and stable expression of recombinant proteins. Results indicated that the expression level of target proteins with the addition of regulatory elements was significantly increased compared with the control group. In addition, the inhibition of apoptotic pathway has great potential to increase recombinant protein production, and Apaf1 protein dependent on the mitochondrial apoptosis pathway plays an important role in this respect. The knockout of apoptotic gene Apaf1 in CHO cells can also increase recombinant protein production. Therefore, the vector was optimized by adding regulatory elements, and the cell line was modified by using CRISPR/Cas9 technology to establish a novel CHO cell expression system, which remarkably improved the expression level of recombinant proteins and laid the foundation for the large-scale production of recombinant proteins.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan, China.
- Xinxiang Medical University, Xinxiang, 453003, China.
| | - Chenyang Du
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan, China
| | - Yue Pan
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhan Zhang
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Ruoyuan Feng
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Mengyao Ma
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan, China.
- Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
3
|
Fu Y, Han Z, Cheng W, Niu S, Wang T, Wang X. Improvement strategies for transient gene expression in mammalian cells. Appl Microbiol Biotechnol 2024; 108:480. [PMID: 39365308 PMCID: PMC11452495 DOI: 10.1007/s00253-024-13315-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Mammalian cells are suitable hosts for producing recombinant therapeutic proteins, with Chinese hamster ovary (CHO) and human embryonic kidney 293 (HEK293) cells being the most commonly used cell lines. Mammalian cell expression system includes stable and transient gene expression (TGE) system, with the TGE system having the advantages of short cycles and simple operation. By optimizing the TGE system, the expression of recombinant proteins has been significantly improved. Here, the TGE system and the detailed and up-to-date improvement strategies of mammalian cells, including cell line, expression vector, culture media, culture processes, transfection conditions, and co-expression of helper genes, are reviewed. KEY POINTS: • Detailed improvement strategies of transient gene expression system of mammalian cells are reviewed • The composition of transient expression system of mammalian cell are summarized • Proposed optimization prospects for transient gene expression systems.
Collapse
Affiliation(s)
- Yushun Fu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zimeng Han
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wanting Cheng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuaichen Niu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Xiaoyin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
4
|
Zhang C, Fu Y, Zheng W, Chang F, Shen Y, Niu J, Wang Y, Ma X. Enhancing the Antibody Production Efficiency of Chinese Hamster Ovary Cells through Improvement of Disulfide Bond Folding Ability and Apoptosis Resistance. Cells 2024; 13:1481. [PMID: 39273052 PMCID: PMC11394227 DOI: 10.3390/cells13171481] [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: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
The complex structure of monoclonal antibodies (mAbs) expressed in Chinese hamster ovary (CHO) cells may result in the accumulation of unfolded proteins, triggering endoplasmic reticulum (ER) stress and an unfolded protein response (UPR). If the protein folding ability cannot maintain ER homeostasis, the cell will shut down protein translation and ultimately induce apoptosis. We co-overexpressed HsQSOX1b and survivin proteins in the antibody-producing cell line CHO-PAb to obtain a new cell line, CHO-PAb-QS. Compared with CHO-PAb cells, the survival time of CHO-PAb-QS cells in batch culture was extended by 2 days, and the antibody accumulation and productivity were increased by 52% and 45%, respectively. The proportion of (HC-LC)2 was approximately doubled in the CHO-PAb-QS cells, which adapted to the accelerated disulfide bond folding capacity by upregulating the UPR's strength and increasing the ER content. The results of the apoptosis assays indicated that the CHO-PAb-QS cell line exhibited more excellent resistance to apoptosis induced by ER stress. Finally, CHO-PAb-QS cells exhibited mild oxidative stress but did not significantly alter the redox status. This study demonstrated that strategies based on HsQSOX1b and survivin co-overexpression could facilitate protein disulfide bond folding and anti-apoptosis ability, enhancing antibody production efficiency in CHO cell lines.
Collapse
Affiliation(s)
- Chen Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yunhui Fu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Wenyun Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Chang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yue Shen
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Jinping Niu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yangmin Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xingyuan Ma
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
5
|
Adibzadeh S, Amiri S, Barkhordari F, Mowla SJ, Bayat H, Ghanbari S, Faghihi F, Davami F. CHO cell engineering via targeted integration of circular miR-21 decoy using CRISPR/RMCE hybrid system. Appl Microbiol Biotechnol 2024; 108:434. [PMID: 39120640 PMCID: PMC11315787 DOI: 10.1007/s00253-024-13266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
Chinese hamster ovary (CHO) cells, widely acknowledged as the preferred host system for industrial recombinant protein manufacturing, play a crucial role in developing pharmaceuticals, including anticancer therapeutics. Nevertheless, mammalian cell-based biopharmaceutical production methods are still beset by cellular constraints such as limited growth and poor productivity. MicroRNA-21 (miR-21) has a major impact on a variety of malignancies, including glioblastoma multiforme (GBM). However, reduced productivity and growth rate have been linked to miR-21 overexpression in CHO cells. The current study aimed to engineer a recombinant CHO (rCHO) cell using the CRISPR-mediated precise integration into target chromosome (CRIS-PITCh) system coupled with the Bxb1 recombinase-mediated cassette exchange (RMCE) to express a circular miR-21 decoy (CM21D) with five bulged binding sites for miR-21 sponging. Implementing the ribonucleoprotein (RNP) delivery method, a landing pad was inserted into the genome utilizing the CRIS-PITCh technique. Subsequently, the CM21D cassette flanked by Bxb1 attB was then retargeted into the integrated landing pad using the RMCE/Bxb1 system. This strategy raised the targeting efficiency by 1.7-fold, and off-target effects were decreased. The miR-21 target genes (Pdcd4 and Atp11b) noticed a significant increase in expression upon the miR-21 sponging through CM21D. Following the expression of CM21D, rCHO cells showed a substantial decrease in doubling time and a 1.3-fold increase in growth rate. Further analysis showed an increased yield of hrsACE2, a secretory recombinant protein, by 2.06-fold. Hence, we can conclude that sponging-induced inhibition of miR-21 may lead to a growth rate increase that could be linked to increased CHO cell productivity. For industrial cell lines, including CHO cells, an increase in productivity is crucial. The results of our research indicate that CM21D is an auspicious CHO engineering approach. KEY POINTS: • CHO is an ideal host cell line for producing industrial therapeutics manufacturing, and miR-21 is downregulated in CHO cells, which produce recombinant proteins. • The miR-21 target genes noticed a significant increase in expression upon the miR-21 sponging through CM21D. Additionally, sponging of miR-21 by CM21D enhanced the growth rate of CHO cells. • Productivity and growth rate were increased in CHO cells expressing recombinant hrs-ACE2 protein after CM21D knocking in.
Collapse
Affiliation(s)
- Setare Adibzadeh
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Farzaneh Barkhordari
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hadi Bayat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, H2W 1R7, Canada
| | - Samaneh Ghanbari
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Davami
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Bauer N, Oberist C, Poth M, Stingele J, Popp O, Ausländer S. Genomic barcoding for clonal diversity monitoring and control in cell-based complex antibody production. Sci Rep 2024; 14:14587. [PMID: 38918509 PMCID: PMC11199663 DOI: 10.1038/s41598-024-65323-7] [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] [Received: 03/25/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Engineered mammalian cells are key for biotechnology by enabling broad applications ranging from in vitro model systems to therapeutic biofactories. Engineered cell lines exist as a population containing sub-lineages of cell clones that exhibit substantial genetic and phenotypic heterogeneity. There is still a limited understanding of the source of this inter-clonal heterogeneity as well as its implications for biotechnological applications. Here, we developed a genomic barcoding strategy for a targeted integration (TI)-based CHO antibody producer cell line development process. This technology provided novel insights about clone diversity during stable cell line selection on pool level, enabled an imaging-independent monoclonality assessment after single cell cloning, and eventually improved hit-picking of antibody producer clones by monitoring of cellular lineages during the cell line development (CLD) process. Specifically, we observed that CHO producer pools generated by TI of two plasmids at a single genomic site displayed a low diversity (< 0.1% RMCE efficiency), which further depends on the expressed molecules, and underwent rapid population skewing towards dominant clones during routine cultivation. Clonal cell lines from one individual TI event demonstrated a significantly lower variance regarding production-relevant and phenotypic parameters as compared to cell lines from distinct TI events. This implies that the observed cellular diversity lies within pre-existing cell-intrinsic factors and that the majority of clonal variation did not develop during the CLD process, especially during single cell cloning. Using cellular barcodes as a proxy for cellular diversity, we improved our CLD screening workflow and enriched diversity of production-relevant parameters substantially. This work, by enabling clonal diversity monitoring and control, paves the way for an economically valuable and data-driven CLD process.
Collapse
Affiliation(s)
- Niels Bauer
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Christoph Oberist
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Poth
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Julian Stingele
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Oliver Popp
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Simon Ausländer
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany.
| |
Collapse
|
7
|
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
|
8
|
Tomris I, van der Woude R, de Paiva Froes Rocha R, Torrents de la Peña A, Ward AB, de Vries RP. Viral envelope proteins fused to multiple distinct fluorescent reporters to probe receptor binding. Protein Sci 2024; 33:e4974. [PMID: 38533540 DOI: 10.1002/pro.4974] [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: 10/25/2023] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Enveloped viruses carry one or multiple proteins with receptor-binding functionalities. Functional receptors can be glycans, proteinaceous, or both; therefore, recombinant protein approaches are instrumental in attaining new insights regarding viral envelope protein receptor-binding properties. Visualizing and measuring receptor binding typically entails antibody detection or direct labeling, whereas direct fluorescent fusions are attractive tools in molecular biology. Here, we report a suite of distinct fluorescent fusions, both N- and C-terminal, for influenza A virus hemagglutinins and SARS-CoV-2 spike RBD. The proteins contained three or six fluorescent protein barrels and were applied directly to cells to assess receptor binding properties.
Collapse
Affiliation(s)
- Ilhan Tomris
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Roosmarijn van der Woude
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Rebeca de Paiva Froes Rocha
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Alba Torrents de la Peña
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| |
Collapse
|
9
|
Cui ZM, Feng YY, Gao YP, Wang HT, Lu JT, Guo JL, Xu HY, Qiu LL, Wang TY, Jia YL. Overexpression of YTHDF3 increases the specific productivity of the recombinant protein in CHO cells by promoting the translation process. Biotechnol J 2024; 19:e2400078. [PMID: 38651251 DOI: 10.1002/biot.202400078] [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: 02/08/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Due to their high-quality characteristics, Chinese hamster ovary (CHO) cells have become the most widely used and reliable host cells for the production of recombinant therapeutic proteins in the biomedical field. Previous studies have shown that the m6A reader YTHDF3, which contains the YTH domain, can affect a variety of biological processes by regulating the translation and stability of target mRNAs. This study investigates the effect of YTHDF3 on transgenic CHO cells. The results indicate that stable overexpression of YTHDF3 significantly enhances recombinant protein expression without affecting host cell growth. Transcriptome sequencing indicated that several genes, including translation initiation factor, translation extension factor, and ribosome assembly factor, were upregulated in CHO cells overexpressing YTHDF3. In addition, cycloheximide experiments confirmed that YTHDF3 enhanced transgene expression by promoting translation in CHO cells. In conclusion, the findings in this study provide a novel approach for mammalian cell engineering to increase protein productivity by regulating m6A.
Collapse
Affiliation(s)
- Zhao-Ming Cui
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ying-Ying Feng
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yan-Ping Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hai-Tong Wang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jiang-Tao Lu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Jia-Liang Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hong-Yan Xu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Le-le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yan-Long Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
10
|
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
|
11
|
Rudometova NB, Fando AA, Kisakova LA, Kisakov DN, Borgoyakova MB, Litvinova VR, Yakovlev VA, Tigeeva EV, Vahitov DI, Sharabrin SV, Shcherbakov DN, Evseenko VI, Ivanova KI, Gudymo AS, Ilyicheva TN, Marchenko VY, Ilyichev AA, Rudometov AP, Karpenko LI. Immunogenic and Protective Properties of Recombinant Hemagglutinin of Influenza A (H5N8) Virus. Vaccines (Basel) 2024; 12:143. [PMID: 38400127 PMCID: PMC10893068 DOI: 10.3390/vaccines12020143] [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: 11/30/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
In this study, we characterized recombinant hemagglutinin (HA) of influenza A (H5N8) virus produced in Chinese hamster ovary cells (CHO-K1s). Immunochemical analysis showed that the recombinant hemagglutinin was recognized by the serum of ferrets infected with influenza A (H5N8) virus, indicating that its antigenic properties were retained. Two groups of Balb/c mice were immunized with intramuscular injection of recombinant hemagglutinin or propiolactone inactivated A/Astrakhan/3212/2020 (H5N8) influenza virus. The results demonstrated that both immunogens induced a specific antibody response as determined by ELISA. Virus neutralization assay revealed that sera of immunized animals were able to neutralize A/turkey/Stavropol/320-01/2020 (H5N8) influenza virus-the average neutralizing titer was 2560. Immunization with both recombinant HA/H5 hemagglutinin and inactivated virus gave 100% protection against lethal H5N8 virus challenge. This study shows that recombinant HA (H5N8) protein may be a useful antigen candidate for developing subunit vaccines against influenza A (H5N8) virus with suitable immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Nadezhda B. Rudometova
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Anastasia A. Fando
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Lyubov A. Kisakova
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Denis N. Kisakov
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Mariya B. Borgoyakova
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Victoria R. Litvinova
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Vladimir A. Yakovlev
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Elena V. Tigeeva
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Danil I. Vahitov
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Sergey V. Sharabrin
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Dmitriy N. Shcherbakov
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Veronika I. Evseenko
- Institute of Solid State Chemistry and Mechanochemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Novosibirsk Region, Russia;
| | - Ksenia I. Ivanova
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Andrei S. Gudymo
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Tatiana N. Ilyicheva
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Vasiliy Yu. Marchenko
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Alexander A. Ilyichev
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Andrey P. Rudometov
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| | - Larisa I. Karpenko
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology «Vector», Rospotrebnadzor, Koltsovo 630559, Novosibirsk Region, Russia (L.A.K.); (D.N.K.); (M.B.B.); (V.R.L.); (E.V.T.); (D.I.V.); (S.V.S.); (D.N.S.); (K.I.I.); (A.S.G.); (T.N.I.); (V.Y.M.); (A.A.I.); (A.P.R.); (L.I.K.)
| |
Collapse
|
12
|
Zhao Y, Li H, Fan Z, Wang T. Effect of Host Cell Protein on Chinese Hamster Ovary Recombinant Protein Production and its Removal Strategies: A Mini Review. Curr Pharm Biotechnol 2024; 25:665-675. [PMID: 37594091 DOI: 10.2174/1389201024666230818112633] [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: 04/05/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
Chinese hamster ovary cells are the main expression system for recombinant therapeutic proteins. During the production of these proteins, certain host cell proteins are secreted, broken down, and released by host cells in the culture along with the proteins of interest. These host cell proteins are often difficult to remove during the downstream purification process, and thus affect the quality, safety, and effectiveness of recombinant protein biopharmaceutical products and increase the production cost of recombinant therapeutic proteins. Therefore, host cell protein production must be reduced as much as possible during the production process and eliminated during purification. This article reviews the harm caused by host cell proteins in the production of recombinant protein drugs using Chinese hamster ovary cell, factors affecting host cell proteins, the monitoring and identification of these proteins, and methods to reduce their type and quantity in the final product.
Collapse
Affiliation(s)
- Yaru Zhao
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Recombinant Pharmaceutical Protein Expression System, Xinxiang Medical University, Xinxiang, China
| | - He Li
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Recombinant Pharmaceutical Protein Expression System, Xinxiang Medical University, Xinxiang, China
| | - Zhenlin Fan
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Recombinant Pharmaceutical Protein Expression System, Xinxiang Medical University, Xinxiang, China
| | - Tianyun Wang
- Henan International Joint Laboratory of Recombinant Pharmaceutical Protein Expression System, Xinxiang Medical University, Xinxiang, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
13
|
Rahimpour A, Mosallaei M, Pourghazi F, Tabatabaee SH, Hoseinpoor R, Pourmaleki E, Soosanabadi M. Development of an Expression Vector Engineering Strategy Based on tDNA Insulator Element for the Stable Expression of Vascular Endothelial Growth Factor Receptor-Fc Fusion Protein. Monoclon Antib Immunodiagn Immunother 2023; 42:140-144. [PMID: 37624609 DOI: 10.1089/mab.2023.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023] Open
Abstract
During the past decades, tremendous advances have occurred in manufacturing recombinant therapeutic proteins in Chinese hamster ovary (CHO) cells. Nevertheless, the production of stable high-producing cell lines has remained a major obstacle in the development process of the CHO cell line. It has been shown that genomic regulatory elements can promote cell line development efficiency by improving transgenes' productivity and stability. Such elements include insulators, ubiquitous chromatin opening elements, scaffold/matrix attachment regions, and antirepressors. In addition, tDNA elements are shown to act as insulators in mammalian cells. This study examines the effect of the tDNA insulator on stable expression of a vascular endothelial growth factor receptor-Fc fusion protein.
Collapse
Affiliation(s)
- Azam Rahimpour
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Mosallaei
- Personalized Medicine and Genometabolomics Research Center, Hope Generation Foundation, Tehran, Iran
| | - Farzad Pourghazi
- Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Hassan Tabatabaee
- Department of Life Science Engineering, Faculty of New Sciences and Technology, University of Tehran, Tehran, Iran
| | - Reyhaneh Hoseinpoor
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Es'hagh Pourmaleki
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Soosanabadi
- Department of Medical Genetics, Semnan University of Medical Sciences, Semnan, Iran
- Department of Genetics, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
14
|
Hashemi N, Tabatabaee SH, Shams F, Rahimpour A, Kazemi B, Rajabibazl M, Ranjbari J. Overexpression of SIRT6 alleviates apoptosis and enhances cell viability and monoclonal antibody expression in CHO-K1 cells. Mol Biol Rep 2023:10.1007/s11033-023-08483-5. [PMID: 37286776 DOI: 10.1007/s11033-023-08483-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/24/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Chinese hamster ovary (CHO) cells are the most predominantly utilized host for the production of monoclonal antibodies (mAbs) and other complex glycoproteins. A major challenge in the process of CHO cell culture is the occurrence of cell death following different stressful conditions, which hinders the production yield. Engineering genes involved in pathways related to cell death is a remarkable strategy to delay apoptosis, improve cell viability and enhance productivity. SIRT6 is a stress-responsive protein that regulates DNA repair, maintains genome integrity, and is critical for longevity and cell survival in organisms. METHODS AND RESULTS In this study, SIRT6 was stably overexpressed in CHO-K1 cells and the impact of its expression on apoptosis related gene expression profile, viability, apoptosis, and mAb productivity was investigated. While a significant increase was observed in Bcl-2 mRNA level, caspase-3 and Bax mRNA levels were decreased in the SIRT6 engineered cells compared to the parental CHO-K1 cells. Moreover, improved cell viability and decreased rate of apoptotic progression was observed in a SIRT6-derived clone in comparision to the CHO-K1 cells during 5 days of batch culture. anti-CD52 IgG1 mAb titers were improved up to 1.7- and 2.8-fold in SIRT6-derived clone during transient and stable expression, respectively. CONCLUSIONS This study indicates the positive effects of SIRT6 overexpression on cell viability and anti-CD52 IgG1 mAb expression in CHO-K1 cells. Further studies are needed to examine the potential of SIRT6-engineered host cells for the production of recombinant biotherapeutics in industrial settings.
Collapse
Affiliation(s)
- Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayed Hassan Tabatabaee
- Department of Life Science Engineering, Faculty of New Sciences and Technology, University of Tehran, Tehran, Iran
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Meade E, Rowan N, Garvey M. Bioprocessing and the Production of Antiviral Biologics in the Prevention and Treatment of Viral Infectious Disease. Vaccines (Basel) 2023; 11:992. [PMID: 37243096 PMCID: PMC10223144 DOI: 10.3390/vaccines11050992] [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: 03/22/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Emerging, re-emerging and zoonotic viral pathogens represent a serious threat to human health, resulting in morbidity, mortality and potentially economic instability at a global scale. Certainly, the recent emergence of the novel SARS-CoV-2 virus (and its variants) highlighted the impact of such pathogens, with the pandemic creating unprecedented and continued demands for the accelerated production of antiviral therapeutics. With limited effective small molecule therapies available for metaphylaxis, vaccination programs have been the mainstay against virulent viral species. Traditional vaccines remain highly effective at providing high antibody titres, but are, however, slow to manufacture in times of emergency. The limitations of traditional vaccine modalities may be overcome by novel strategies, as outlined herein. To prevent future disease outbreaks, paradigm shift changes in manufacturing and distribution are necessary to advance the production of vaccines, monoclonal antibodies, cytokines and other antiviral therapies. Accelerated paths for antivirals have been made possible due to advances in bioprocessing, leading to the production of novel antiviral agents. This review outlines the role of bioprocessing in the production of biologics and advances in mitigating viral infectious disease. In an era of emerging viral diseases and the proliferation of antimicrobial resistance, this review provides insight into a significant method of antiviral agent production which is key to protecting public health.
Collapse
Affiliation(s)
- Elaine Meade
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| | - Neil Rowan
- Bioscience Research Institute, Technical University Shannon Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
16
|
Enhanced Production of ECM Proteins for Pharmaceutical Applications Using Mammalian Cells and Sodium Heparin Supplementation. Pharmaceutics 2022; 14:pharmaceutics14102138. [PMID: 36297573 PMCID: PMC9609459 DOI: 10.3390/pharmaceutics14102138] [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: 07/31/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
The yields of soluble ECM proteins recombinantly produced with mammalian cells can be significantly enhanced by exploiting the stabilizing properties of heparin. Here, we propose a simple and straightforward scalable protocol for the mammalian cell production of ECM proteins with affinity for heparin, using heparin as a supplement. As proof of concept, we have demonstrated the high-level expression of four biomedically relevant human enzymes such as carboxypeptidase Z (CPZ), carboxypeptidase A6 (CPA6), beta-galactoside alpha-2,6-sialyltransferase 2 (ST6GAL1) and thrombin-activable fibrinolysis inhibitor (TAFI). We found a strong linear correlation between the isoelectric point (pI) of a protein and the improvement in protein expression levels upon heparin addition, providing a reference for selecting novel protein targets that would benefit from heparin supplementation. Finally, we demonstrated the compatibility of this approach with a three-step purification strategy that includes an initial heparin affinity purification step. Using CPZ as a representative example, we performed a preparative purification of this enzyme. The purified protein is enzymatically active and can be used for pharmaceutical applications as well as for high-throughput functional and structural studies.
Collapse
|
17
|
de Pinho Favaro MT, Atienza-Garriga J, Martínez-Torró C, Parladé E, Vázquez E, Corchero JL, Ferrer-Miralles N, Villaverde A. Recombinant vaccines in 2022: a perspective from the cell factory. Microb Cell Fact 2022; 21:203. [PMID: 36199085 PMCID: PMC9532831 DOI: 10.1186/s12934-022-01929-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
The last big outbreaks of Ebola fever in Africa, the thousands of avian influenza outbreaks across Europe, Asia, North America and Africa, the emergence of monkeypox virus in Europe and specially the COVID-19 pandemics have globally stressed the need for efficient, cost-effective vaccines against infectious diseases. Ideally, they should be based on transversal technologies of wide applicability. In this context, and pushed by the above-mentioned epidemiological needs, new and highly sophisticated DNA-or RNA-based vaccination strategies have been recently developed and applied at large-scale. Being very promising and effective, they still need to be assessed regarding the level of conferred long-term protection. Despite these fast-developing approaches, subunit vaccines, based on recombinant proteins obtained by conventional genetic engineering, still show a wide spectrum of interesting potentialities and an important margin for further development. In the 80's, the first vaccination attempts with recombinant vaccines consisted in single structural proteins from viral pathogens, administered as soluble plain versions. In contrast, more complex formulations of recombinant antigens with particular geometries are progressively generated and explored in an attempt to mimic the multifaceted set of stimuli offered to the immune system by replicating pathogens. The diversity of recombinant antimicrobial vaccines and vaccine prototypes is revised here considering the cell factory types, through relevant examples of prototypes under development as well as already approved products.
Collapse
Affiliation(s)
- Marianna Teixeira de Pinho Favaro
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jan Atienza-Garriga
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
| | - Carlos Martínez-Torró
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
| | - José Luis Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, 08193, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193, Barcelona, Spain.
| |
Collapse
|