1
|
Lancho VJ, Eisenhut P, Klanert G, Ivansson D, Jonsson A, Lövgren A, Borth N. Stepwise activation of gene copies results in higher final titers of subclones compared to immediate integration of the full set of active copies. N Biotechnol 2025; 88:89-99. [PMID: 40287132 DOI: 10.1016/j.nbt.2025.04.011] [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: 08/09/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
The increasing demand for production of therapeutic proteins has encouraged both industrial and academic institutions to pursue the development of mammalian expression platforms with high productivities. While protocols for rapid and efficient integration of multiple transgene copies into the genome are available, they require substantial time and resources for screening numerous clones. A contributing factor is the tendency of high producers to disappear from the selected mini-pools due to the stress caused by high productivity without adequate time for adaptation of cellular capacities. Here, we have developed a strategy to stably activate individual copies within an initially repressed multicopy coding cassette harboring 2 GFP-Fc and 2 BFP-Fc genes, each fused to an Fc region for secretion. This toolbox enables gene activation via CRISPR/Cas9-mediated deletion of the repressor elements. Subsequently, producers can be sorted based on increased GFP or BFP fluorescence and assessed by measuring the secreted total Fc protein. We demonstrate that the stepwise activation of initially repressed genes outperforms a control cell line with the same number of genes active from the outset, as evidenced by higher fluorescence signals from GFP and BFP, increased mRNA levels for BFP, GFP, and Fc genes, and enhanced titer of secreted Fc fusion protein. This study demonstrates the ability of cells to adapt to new challenges by modulating both gene expression patterns and channeling of resources to accommodate high production loads.
Collapse
Affiliation(s)
- Víctor Jiménez Lancho
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Graz, Austria; Department of Biotechnology and Food Sciences, BOKU University, Vienna, Austria
| | - Peter Eisenhut
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Graz, Austria; Department of Biotechnology and Food Sciences, BOKU University, Vienna, Austria
| | - Gerald Klanert
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Graz, Austria
| | | | | | | | - Nicole Borth
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Graz, Austria; Department of Biotechnology and Food Sciences, BOKU University, Vienna, Austria.
| |
Collapse
|
2
|
Riedl M, Ruggeri C, Marx N, Borth N. Fantastic genes and where to find them expressed in CHO. Comput Struct Biotechnol J 2025; 27:1407-1415. [PMID: 40242293 PMCID: PMC12002940 DOI: 10.1016/j.csbj.2025.03.050] [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: 12/11/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
The transcriptome of Chinese hamster ovary (CHO) cells plays a crucial role in determining cellular characteristics that are essential for biopharmaceutical applications. RNA-sequencing has been extensively used to profile gene expression patterns, aiming to gain a better understanding of intracellular behavior and mechanisms. Individual datasets, however, do not provide a comprehensive overview and characterization of the CHO cell's transcriptome, such that the fundamental structure of the transcriptome remains unknown. Using 15 RNA-sequencing datasets, encompassing almost 300 samples of various experimental setups, conditions and cell lines, we explore and classify the protein-coding transcriptome of CHO cells. Differences in cell line lineages are found to be the primary source of variation in transcribed genes. By employing a novel approach, we identified the core transcriptome that is ubiquitously expressed in all cell lines and culture conditions, as well as genes that remain entirely non-expressed. Additionally, we identified a set of genes that may be active or inactive depending on different conditions, which are linked to biological processes including translation as well as immune and stress response. Lastly, by integrating chromatin states derived from histone modifications, we provided additional context on the epigenetic level that supports our protein-coding gene classification. Our study offers a comprehensive insight into the CHO cell transcriptome and lays the foundation for future research into cellular adaptation to changing conditions and the development of phenotypes.
Collapse
Affiliation(s)
- Markus Riedl
- Department of Biotechnology, BOKU University, Vienna, Austria
| | | | - Nicolas Marx
- Department of Biotechnology, BOKU University, Vienna, Austria
| | - Nicole Borth
- Department of Biotechnology, BOKU University, Vienna, Austria
| |
Collapse
|
3
|
Xue T, Wang Y, Liu Y, Liu Y, Li C. Establishment and characterization of SSI cell line from Sebastes schlegelii intestine for investigating the immune response to Pathogenic Bacteria. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109993. [PMID: 39481502 DOI: 10.1016/j.fsi.2024.109993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
As a commercially valuable fish species, Sebastes schlegelii faces threats from pathogenic bacteria like Edwardsiella piscicida during aquaculture. The global host range of E. piscicida encompasses various species, yet its pathogenic mechanism remains incompletely elucidated. Cell lines offer invaluable in vitro resources for studying the pathogen pathogenicity. Here, we established and characterized a cell line derived from the intestinal tissue of the S. schlegelii, designated as SSI. SSI has undergone continuous subculturing for over 80 passages, demonstrating robust growth in DMEM supplemented with 10%-20% FBS and 20 μM HEPES at 24°C. Karyotype analysis and 18S rRNA amplification confirm its origin. SSI exhibits high transfection efficiency for exogenous DNA, making it suitable for gene expression and intestinal function analysis. E. piscicida infects SSI cells at low densities without inducing morphological changes within 6 h of infection, suggesting the potential of SSI as an in vitro model for studying E. piscicida pathogenicity. This cell line provides a valuable tool for investigating mucosal immunity and E. piscicida pathogenic mechanisms in marine fish.
Collapse
Affiliation(s)
- Ting Xue
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yanmin Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yiping Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yuping Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
4
|
Splichal RC, Chen K, Walton SP, Chan C. The Role of Endoplasmic Reticulum Stress on Reducing Recombinant Protein Production in Mammalian Cells. Biochem Eng J 2024; 210:109434. [PMID: 39220803 PMCID: PMC11360842 DOI: 10.1016/j.bej.2024.109434] [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] [Indexed: 09/04/2024]
Abstract
Therapeutic recombinant protein production relies on industrial scale culture of mammalian cells to produce active proteins in quantities sufficient for clinical use. The combination of stresses from industrial cell culture environment and recombinant protein production can overwhelm the protein synthesis machinery in the endoplasmic reticulum (ER). This leads to a buildup of improperly folded proteins which induces ER stress. Cells respond to ER stress by activating the Unfolded Protein Response (UPR). To restore proteostasis, ER sensor proteins reduce global protein synthesis and increase chaperone protein synthesis, and if that is insufficient the proteins are degraded. If proteostasis is still not restored, apoptosis is initiated. Increasing evidence suggests crosstalk between ER proteostasis and DNA damage repair (DDR) pathways. External factors (e.g., metabolites) from the cellular environment as well as internal factors (e.g., transgene copy number) can impact genome stability. Failure to maintain genome integrity reduces cell viability and in turn protein production. This review focuses on the association between ER stress and processes that affect protein production and secretion. The processes mediated by ER stress, including inhibition of global protein translation, chaperone protein production, degradation of misfolded proteins, DNA repair, and protein secretion, impact recombinant protein production. Recombinant protein production can be reduced by ER stress through increased autophagy and protein degradation, reduced protein secretion, and reduced DDR response.
Collapse
Affiliation(s)
- R. Chauncey Splichal
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Kevin Chen
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - S. Patrick Walton
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Christina Chan
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI, USA
- Department of Computer Science and Engineering, Michigan State University, MI, USA
- Institute for Quantitative Health Science and Engineering, Division of Medical Devices, Michigan State University, MI, USA
| |
Collapse
|
5
|
Desmurget C, Perilleux A, Souquet J, Borth N, Douet J. Molecular biomarkers identification and applications in CHO bioprocessing. J Biotechnol 2024; 392:11-24. [PMID: 38852681 DOI: 10.1016/j.jbiotec.2024.06.005] [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: 12/18/2023] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Biomarkers are valuable tools in clinical research where they allow to predict susceptibility to diseases, or response to specific treatments. Likewise, biomarkers can be extremely useful in the biomanufacturing of therapeutic proteins. Indeed, constraints such as short timelines and the need to find hyper-productive cells could benefit from a data-driven approach during cell line and process development. Many companies still rely on large screening capacities to develop productive cell lines, but as they reach a limit of production, there is a need to go from empirical to rationale procedures. Similarly, during bioprocessing runs, substrate consumption and metabolism wastes are commonly monitored. None of them possess the ability to predict the culture behavior in the bioreactor. Big data driven approaches are being adapted to the study of industrial mammalian cell lines, enabled by the publication of Chinese hamster and CHO genome assemblies which allowed the use of next-generation sequencing with these cells, as well as continuous proteome and metabolome annotation. However, if these different -omics technologies contributed to the characterization of CHO cells, there is a significant effort remaining to apply this knowledge to biomanufacturing methods. The correlation of a complex phenotype such as high productivity or rapid growth to the presence or expression level of a specific biomarker could save time and effort in the screening of manufacturing cell lines or culture conditions. In this review we will first discuss the different biological molecules that can be identified and quantified in cells, their detection techniques, and associated challenges. We will then review how these markers are used during the different steps of cell line and bioprocess development, and the inherent limitations of this strategy.
Collapse
Affiliation(s)
- Caroline Desmurget
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Arnaud Perilleux
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Jonathan Souquet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Julien Douet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland.
| |
Collapse
|
6
|
Deng H, Zheng S, Li Y, Mo X, Zhao J, Yin J, Shi C, Wang Q, Wang Y. Establishment and characterization of a kidney cell line from hybrid snakehead (male Channa argus × female Channa maculata) and its susceptibility to hybrid snakehead rhabdovirus (HSHRV). Comp Biochem Physiol B Biochem Mol Biol 2024; 273:110971. [PMID: 38621626 DOI: 10.1016/j.cbpb.2024.110971] [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: 12/20/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
Hybrid snakehead (male Channa argus × female Channa maculata) is an emerging fish breed with increasing production levels. However, infection with hybrid snakehead rhabdovirus (HSHRV) critically affects hybrid snakehead farming. In this study, a fish cell line called CAMK, derived from the kidneys of hybrid snakehead, was established and characterized. CAMK cells exhibited the maximum growth rate at 28 °C in Leibovitz's-15 medium supplemented with 10% fetal bovine serum(FBS). Karyotyping revealed diploid chromosomes in 54% of the cells at the 50th passage (2n = 66), and 16S rRNA sequencing validated that CAMK cells originated fromhybrid snakehead, and the detection of kidney-specific antibodies suggested that it originated from kidney. .The culture was free from mycoplasma contamination, and the green fluorescent protein gene was effectively transfected into CAMK cells, indicating their potential use for in vitro gene expression investigations. Furthermore, qRT-PCR and immunofluorescence analysis revealed that HSHRV could replicate in CAMK cells, indicating that the cells were susceptible to the virus. Transmission electron microscopy revealed that the viral particles had bullet-like morphology. The replication efficiency of HSHRV was 107.33 TCID50/mL. Altogether, we successfully established and characterized a kidney cell line susceptible to the virus. These findings provide a valuable reference for further genetic and virological studies.
Collapse
Affiliation(s)
- Huiling Deng
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380; College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai 201306, China.
| | - Shucheng Zheng
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380; Key Laboratory of Marine Pollution, Department of Infectious Diseases and Public Health, Jockey Club School of Animal Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong, China.
| | - Yingying Li
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Xubing Mo
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Jian Zhao
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Jiyuan Yin
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Cunbin Shi
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Qing Wang
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| | - Yingying Wang
- Key Laboratory of Fishery Drug Development,Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China 510380.
| |
Collapse
|
7
|
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
|
8
|
Masuda K, Kubota M, Nakazawa Y, Iwama C, Watanabe K, Ishikawa N, Tanabe Y, Kono S, Tanemura H, Takahashi S, Makino T, Okumura T, Horiuchi T, Nonaka K, Murakami S, Kamihira M, Omasa T. Establishment of a novel cell line, CHO-MK, derived from Chinese hamster ovary tissues for biologics manufacturing. J Biosci Bioeng 2024; 137:471-479. [PMID: 38472071 DOI: 10.1016/j.jbiosc.2024.02.005] [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: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
Chinese hamster ovary (CHO) cells are widely used as a host for producing recombinant therapeutic proteins due to advantages such as human-like post-translational modification, correct protein folding, higher productivity, and a proven track record in biopharmaceutical development. Much effort has been made to improve the process of recombinant protein production, in terms of its yield and productivity, using conventional CHO cell lines. However, to the best of our knowledge, no attempts have been made to acquire new CHO cell lines from Chinese hamster ovary. In this study, we established and characterized a novel CHO cell line, named CHO-MK, derived from freshly isolated Chinese hamster ovary tissues. Some immortalized cell lines were established via sub-culture derived from primary culture, one of which was selected for further development toward a unique expression system design. After adapting serum-free and suspension culture conditions, the resulting cell line exhibited a considerably shorter doubling time (approximately 10 h) than conventional CHO cell lines (approximately 20 h). Model monoclonal antibody (IgG1)-producing cells were generated, and the IgG1 concentration of fed-batch culture reached approximately 5 g/L on day 8 in a 200-L bioreactor. The cell bank of CHO-MK cells was prepared as a new host and assessed for contamination by adventitious agents, with the results indicating that it was free from any such contaminants, including infectious viruses. Taking these findings together, this study showed the potential of CHO-MK cells with a shorter doubling time/process time and enhanced productivity in biologics manufacturing.
Collapse
Affiliation(s)
- Kenji Masuda
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Michi Kubota
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Yuto Nakazawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Chigusa Iwama
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Kazuhiko Watanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Naoto Ishikawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Yumiko Tanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Satoru Kono
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Hiroki Tanemura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Shinichi Takahashi
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Tomohiro Makino
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Takeshi Okumura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Takayuki Horiuchi
- Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Koichi Nonaka
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Sei Murakami
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Masamichi Kamihira
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Omasa
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
9
|
Yamano-Adachi N, Hata H, Nakanishi Y, Omasa T. Effects of genome instability of parental CHO cell clones on chromosome number distribution and recombinant protein production in parent-derived subclones. J Biosci Bioeng 2024; 137:54-63. [PMID: 37981489 DOI: 10.1016/j.jbiosc.2023.10.001] [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: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023]
Abstract
Chinese hamster ovary (CHO) cells are the de facto standard host cells for biopharmaceuticals, and there is great interest in developing methods for constructing stable production cell lines. In this study, clones with a wide chromosome number distribution were selected from isolated antibody-producing strains, and subclones obtained from these clones were evaluated. The transgene copy number varied between the subclones. Even among subclones with similar copy numbers of antibody genes and maintained insertion sites, clones with different productivity were generated. Although the chromosome number distribution differed between these subclones, there was no correlation between the variability in chromosome number after cloning (genome instability) and productivity. Most of the subclones obtained from a parental strain with a wide chromosome number had the same wide chromosome number distribution as the parental strain. Less frequently, cells with less variation (remaining in one distribution) in chromosome number were isolated from cells with a wide chromosome number distribution, from which subclones with less variation in chromosome number were obtained when subcloning was performed again. These results imply that the characteristics of clones with chromosomal instability are inherited by subclones, and thus provide a better understanding of cell line stability/instability.
Collapse
Affiliation(s)
- Noriko Yamano-Adachi
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hirofumi Hata
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuto Nakanishi
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeshi Omasa
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
10
|
Cero C, Shu W, Reese AL, Douglas D, Maddox M, Singh AP, Ali SL, Zhu AR, Katz JM, Pierce AE, Long KT, Nilubol N, Cypess RH, Jacobs JL, Tian F, Cypess AM. Standardized In Vitro Models of Human Adipose Tissue Reveal Metabolic Flexibility in Brown Adipocyte Thermogenesis. Endocrinology 2023; 164:bqad161. [PMID: 37944134 PMCID: PMC11032247 DOI: 10.1210/endocr/bqad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Functional human brown and white adipose tissue (BAT and WAT) are vital for thermoregulation and nutritional homeostasis, while obesity and other stressors lead, respectively, to cold intolerance and metabolic disease. Understanding BAT and WAT physiology and dysfunction necessitates clinical trials complemented by mechanistic experiments at the cellular level. These require standardized in vitro models, currently lacking, that establish references for gene expression and function. We generated and characterized a pair of immortalized, clonal human brown (hBA) and white (hWA) preadipocytes derived from the perirenal and subcutaneous depots, respectively, of a 40-year-old male individual. Cells were immortalized with hTERT and confirmed to be of a mesenchymal, nonhematopoietic lineage based on fluorescence-activated cell sorting and DNA barcoding. Functional assessments showed that the hWA and hBA phenocopied primary adipocytes in terms of adrenergic signaling, lipolysis, and thermogenesis. Compared to hWA, hBA were metabolically distinct, with higher rates of glucose uptake and lactate metabolism, and greater basal, maximal, and nonmitochondrial respiration, providing a mechanistic explanation for the association between obesity and BAT dysfunction. The hBA also responded to the stress of maximal respiration by using both endogenous and exogenous fatty acids. In contrast to certain mouse models, hBA adrenergic thermogenesis was mediated by several mechanisms, not principally via uncoupling protein 1 (UCP1). Transcriptomics via RNA-seq were consistent with the functional studies and established a molecular signature for each cell type before and after differentiation. These standardized cells are anticipated to become a common resource for future physiological, pharmacological, and genetic studies of human adipocytes.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiguo Shu
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amy L Reese
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Diana Douglas
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Michael Maddox
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
- Current Affiliation: Vita Therapeutics, 801 W. Baltimore Street, Suite 301, Baltimore, MD 21201, USA
| | - Ajeet P Singh
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Sahara L Ali
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander R Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline M Katz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly T Long
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, 10 Center Drive, Room 4-5952, Bethesda, MD 20892, USA
| | - Raymond H Cypess
- American Type Culture Collection, 10801 University Blvd, Manassas, VA 20110, USA
| | - Jonathan L Jacobs
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Fang Tian
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
11
|
Pinto J, Ramos JRC, Costa RS, Rossell S, Dumas P, Oliveira R. Hybrid deep modeling of a CHO-K1 fed-batch process: combining first-principles with deep neural networks. Front Bioeng Biotechnol 2023; 11:1237963. [PMID: 37744245 PMCID: PMC10515724 DOI: 10.3389/fbioe.2023.1237963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: Hybrid modeling combining First-Principles with machine learning is becoming a pivotal methodology for Biopharma 4.0 enactment. Chinese Hamster Ovary (CHO) cells, being the workhorse for industrial glycoproteins production, have been the object of several hybrid modeling studies. Most previous studies pursued a shallow hybrid modeling approach based on three-layered Feedforward Neural Networks (FFNNs) combined with macroscopic material balance equations. Only recently, the hybrid modeling field is incorporating deep learning into its framework with significant gains in descriptive and predictive power. Methods: This study compares, for the first time, deep and shallow hybrid modeling in a CHO process development context. Data of 24 fed-batch cultivations of a CHO-K1 cell line expressing a target glycoprotein, comprising 30 measured state variables over time, were used to compare both methodologies. Hybrid models with varying FFNN depths (3-5 layers) were systematically compared using two training methodologies. The classical training is based on the Levenberg-Marquardt algorithm, indirect sensitivity equations and cross-validation. The deep learning is based on the Adaptive Moment Estimation Method (ADAM), stochastic regularization and semidirect sensitivity equations. Results and conclusion: The results point to a systematic generalization improvement of deep hybrid models over shallow hybrid models. Overall, the training and testing errors decreased by 14.0% and 23.6% respectively when applying the deep methodology. The Central Processing Unit (CPU) time for training the deep hybrid model increased by 31.6% mainly due to the higher FFNN complexity. The final deep hybrid model is shown to predict the dynamics of the 30 state variables within the error bounds in every test experiment. Notably, the deep hybrid model could predict the metabolic shifts in key metabolites (e.g., lactate, ammonium, glutamine and glutamate) in the test experiments. We expect deep hybrid modeling to accelerate the deployment of high-fidelity digital twins in the biopharma sector in the near future.
Collapse
Affiliation(s)
- José Pinto
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - João R. C. Ramos
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Rafael S. Costa
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | | | | | - Rui Oliveira
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| |
Collapse
|
12
|
Torres M, Betts Z, Scholey R, Elvin M, Place S, Hayes A, Dickson AJ. Long term culture promotes changes to growth, gene expression, and metabolism in CHO cells that are independent of production stability. Biotechnol Bioeng 2023; 120:2389-2402. [PMID: 37060548 DOI: 10.1002/bit.28399] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 04/16/2023]
Abstract
Phenotypic stability of Chinese hamster ovary (CHO) cells over long term culture (LTC) presents one of the most pressing challenges in the development of therapeutic protein manufacturing processess. However, our current understanding of the consequences of LTC on recombinant (r-) CHO cell lines is still limited, particularly as clonally-derived cell lines present distinct production stability phenotypes. This study evaluated changes of culture performance, global gene expression, and cell metabolism of two clonally-derived CHO cell lines with a stable or unstable phenotype during the LTC (early [EP] vs. late [LP] culture passages). Our findings indicated that LTC altered the behavior of CHO cells in culture, in terms of growth, overall gene expression, and cell metabolism. Regardless whether cells were categorized as stable or unstable in terms of r-protein production, CHO cells at LP presented an earlier decline in cell viability and loss of any observable stationary phase. These changes were parallelled by the upregulation of genes involved in cell proliferation and survival pathways (i.e., MAPK/ERK, PI3K-Akt). Stable and unstable CHO cell lines both showed increased consumption of glucose and amino acids at LP, with a parallel accumulation of greater amounts of lactate and TCA cycle intermediates. In terms of production stability, we found that decreased r-protein production in the unstable cell line directly correlated to the loss in r-gene copy number and r-mRNA expression. Our data revealed that LTC produced ubiquitious effects on CHO cell phenotypes, changes that were rooted in alterations in cell transcriptome and metabolome. Overall, we found that CHO cells adapted their cellular function to proliferation and survival during the LTC, some of these changes may well have limited effects on overall yield or specific productivity of the desired r-product, but they may be critical toward the capacity of cells to handle r-proteins with specific molecular features.
Collapse
Affiliation(s)
- Mauro Torres
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
- Department of Chemical Engineering, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| | - Zeynep Betts
- Department of Biology, Kocaeli University, İzmit, Turkey
| | - Rachel Scholey
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - Mark Elvin
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
- Department of Chemical Engineering, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| | - Svetlana Place
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
- Department of Chemical Engineering, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| | - Andrew Hayes
- Genomic Technologies Core Facility, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alan J Dickson
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
- Department of Chemical Engineering, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Součková K, Jasík M, Sovadinová I, Sember A, Sychrová E, Konieczna A, Bystrý V, Dyková I, Blažek R, Lukšíková K, Pavlica T, Jankásek M, Altmanová M, Žák J, Zbončáková A, Reichard M, Slabý O. From fish to cells: Establishment of continuous cell lines from embryos of annual killifish Nothobranchius furzeri and N. kadleci. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 259:106517. [PMID: 37087860 DOI: 10.1016/j.aquatox.2023.106517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 05/03/2023]
Abstract
There is a growing need of alternative experimental models that avoid or minimize the use of animals due to ethical, economical, and scientific reasons. Surprisingly, the stable embryonic cell lines representing Nothobranchius spp., emerging vertebrate models in aging research, regenerative medicine, ecotoxicology, or genomics, have been not derived so far. This paper reports establishment and deep characterization of ten continuous cell lines from annual killifish embryos of N. furzeri and N. kadleci. The established cell lines exhibited mostly fibroblast- and epithelial-like morphology and steady growth rates with cell doubling time ranging from 27 to 40 h. All cell lines retained very similar characteristics even after continuous subcultivation (more than 100 passages) and extended storage in liquid nitrogen (∼3 years). The cytogenetic analysis of the cell lines revealed a diploid chromosome number mostly equal to 38 elements (i.e., the native chromosome count for both killifish species), with minor but diverse line/passage-specific karyotype changes compared to the patterns observed in non-cultured N. furzeri and N. kadleci somatic cells. Based on transcriptional analysis of marker genes, the cell lines displayed features of an undifferentiated state without signs of senescence even in advanced passages. We confirmed that the cell lines are transfectable and can form viable 3-D spheroids. The applicability of the cell lines for (eco)toxicological surveys was confirmed by assessing the effect of cytotoxic and growth inhibitory agents. Properties of established Nothobranchius embryonic cell lines open new possibilities for the application of this model in various fields of life sciences including molecular mechanisms of aging, karyotype (in)stability or differences in lifespan.
Collapse
Affiliation(s)
- Kamila Součková
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic.
| | - Matej Jasík
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Iva Sovadinová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 602 00, Czech Republic
| | - Alexandr Sember
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov 277 21, Czech Republic
| | - Eliška Sychrová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 602 00, Czech Republic
| | - Anna Konieczna
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Vojtěch Bystrý
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Iva Dyková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic
| | - Radim Blažek
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic; Institute of Vertebrate Biology, Czech Academy of Sciences, Brno 603 00, Czech Republic
| | - Karolína Lukšíková
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov 277 21, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague 128 44, Czech Republic
| | - Tomáš Pavlica
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov 277 21, Czech Republic; Department of Zoology, Faculty of Science, Charles University, Prague 128 44, Czech Republic
| | - Marek Jankásek
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov 277 21, Czech Republic; Department of Zoology, Faculty of Science, Charles University, Prague 128 44, Czech Republic
| | - Marie Altmanová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov 277 21, Czech Republic; Department of Ecology, Faculty of Science, Charles University, Prague 128 44, Czech Republic
| | - Jakub Žák
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic; Institute of Vertebrate Biology, Czech Academy of Sciences, Brno 603 00, Czech Republic
| | - Adriana Zbončáková
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Martin Reichard
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic; Institute of Vertebrate Biology, Czech Academy of Sciences, Brno 603 00, Czech Republic; Department of Ecology and Vertebrate Zoology, University of Łódź, Łódź 90-237, Poland
| | - Ondřej Slabý
- Ondřej Slabý Group, Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno 625 00, Czech Republic
| |
Collapse
|
14
|
de Vos J, Crooijmans RP, Derks MF, Kloet SL, Dibbits B, Groenen MA, Madsen O. Detailed molecular and epigenetic characterization of the pig IPEC-J2 and chicken SL-29 cell lines. iScience 2023; 26:106252. [PMID: 36936794 PMCID: PMC10018572 DOI: 10.1016/j.isci.2023.106252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/05/2022] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The pig IPEC-J2 and chicken SL-29 cell lines are of interest because of their untransformed nature and wide use in functional studies. Molecular characterization of these cell lines is important to gain insight into possible molecular aberrations. The aim of this paper is to provide a molecular and epigenetic characterization of the IPEC-J2 and SL-29 cell lines, a cell-line reference for the FAANG community, and future biomedical research. Whole genome sequencing, gene expression, DNA methylation, chromatin accessibility, and ChIP-seq of four histone marks (H3K4me1, H3K4me3, H3K27ac, H3K27me3) and an insulator (CTCF) are used to achieve these aims. Heteroploidy (aneuploidy) of various chromosomes was observed from whole genome sequencing analysis in both cell lines. Furthermore, higher gene expression for genes located on chromosomes with aneuploidy in comparison to diploid chromosomes was observed. Regulatory complexity of gene expression, DNA methylation, and chromatin accessibility was investigated through an integrative approach.
Collapse
Affiliation(s)
- Jani de Vos
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen 6708PB, the Netherlands
| | | | - Martijn F.L. Derks
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen 6708PB, the Netherlands
| | - Susan L. Kloet
- Human Genetics, Leids Universitair Medisch Centrum, Leiden 2333ZC, the Netherlands
| | - Bert Dibbits
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen 6708PB, the Netherlands
| | - Martien A.M. Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen 6708PB, the Netherlands
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen 6708PB, the Netherlands
| |
Collapse
|
15
|
Sathiyanarayanan A, Yashwanth BS, Pinto N, Thakuria D, Chaudhari A, Gireesh Babu P, Goswami M. Establishment and characterization of a new fibroblast-like cell line from the skin of a vertebrate model, zebrafish (Danio rerio). Mol Biol Rep 2023; 50:19-29. [PMID: 36289143 DOI: 10.1007/s11033-022-08009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/05/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The available fully sequenced genome and genetic similarities compared to humans make zebrafish a prominent in vitro vertebrate model for drug discovery & screening, toxicology, and radiation biology. Zebrafish also possess well developed immune systems which is ideal for studying infectious diseases. Fish skin confers immunity by serving as a physical barrier against the invading pathogens in the aquatic habitat. Therefore in vitro models from the skin tissue of zebrafish help to study the physiology, functional genes in vitro, wound healing, and pathogenicity of microbes. Hence the study aimed to develop and characterize a skin cell line from the wild-type zebrafish Danio rerio. METHODS AND RESULTS A novel cell line designated as DRS (D. rerio skin) was established and characterized from the skin tissue of wild-type zebrafish, D. rerio, by the explant technique. The cells thrived well in the Leibovitz's -15 medium supplemented with 15% FBS and routinely passaged at regular intervals. The DRS cells mainly feature fibroblast-like morphology. The culture conditions of the cells were determined by incubating the cells at varying concentrations of FBS and temperature; the optimum was 15% FBS and 28 °C, respectively. Cells were cryopreserved and revived with 70-75% viability at different passage levels. Two extracellular products from bacterial species Aeromonas hydrophila and Edwardsiella tarda were tested and found toxic to the DRS cells. Mitochondrial genes, namely COI and 16S rRNA PCR amplification and partial sequencing authenticated the species of origin of cells. The modal diploid (2n) chromosome number of the cells was 50. The cell line DRS was found to be free from mycoplasma. The cells were transfected with pMaxGFP plasmid and tested positive for green fluorescence at 24-48 h post-transfection. CONCLUSION The findings from this study thus confirm the usefulness of the developed cell line in bacterial susceptibility and transgene expression studies.
Collapse
Affiliation(s)
- Arjunan Sathiyanarayanan
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - B S Yashwanth
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - Nevil Pinto
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - Dimpal Thakuria
- ICAR-Directorate of Coldwater Fisheries Research, Anusandhan Bhawan, Industrial Area, Bhimtal, 263136, India
| | - Aparna Chaudhari
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India
| | - P Gireesh Babu
- ICAR-National Research Centre on Meat, Chengicherla, Boduppal Post, Hyderabad, 500092, India
| | - Mukunda Goswami
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Panch Marg, Off Yari Road, Versova, Andheri West, Mumbai, Maharashtra, 400061, India.
| |
Collapse
|
16
|
Kretzmer C, Narasimhan RL, Lal RD, Balassi V, Ravellette J, Kotekar Manjunath AK, Koshy JJ, Viano M, Torre S, Zanda VM, Kumravat M, Saldanha KMR, Chandranpillai H, Nihad I, Zhong F, Sun Y, Gustin J, Borgschulte T, Liu J, Razafsky D. De novo assembly and annotation of the CHOZN® GS -/- genome supports high-throughput genome-scale screening. Biotechnol Bioeng 2022; 119:3632-3646. [PMID: 36073082 PMCID: PMC9825924 DOI: 10.1002/bit.28226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/20/2022] [Accepted: 08/28/2022] [Indexed: 01/11/2023]
Abstract
Chinese hamster ovary (CHO) cells have been used as the industry standard for the production of therapeutic monoclonal antibodies for several decades. Despite significant improvements in commercial-scale production processes and media, the CHO cell has remained largely unchanged. Due to the cost and complexity of whole-genome sequencing and gene-editing it has been difficult to obtain the tools necessary to improve the CHO cell line. With the advent of next-generation sequencing and the discovery of the CRISPR/Cas9 system it has become more cost effective to sequence and manipulate the CHO genome. Here, we provide a comprehensive de novo assembly and annotation of the CHO-K1 based CHOZN® GS-/- genome. Using this platform, we designed, built, and confirmed the functionality of a whole genome CRISPR guide RNA library that will allow the bioprocessing community to design a more robust CHO cell line leading to the production of life saving medications in a more cost-effective manner.
Collapse
Affiliation(s)
- Corey Kretzmer
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - Rajagopalan Lakshmi Narasimhan
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Rahul Deva Lal
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Vincent Balassi
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - James Ravellette
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - Ajaya Kumar Kotekar Manjunath
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Jesvin Joy Koshy
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Marta Viano
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Serena Torre
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Valeria M. Zanda
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Mausam Kumravat
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Keith Metelo Raul Saldanha
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Harikrishnan Chandranpillai
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Ifra Nihad
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Fei Zhong
- Life Science Bioinformatics, IT, MilliporeSigmaSt. LouisMissouriUSA
| | - Yi Sun
- Bioinformatics, IT R&D Applications, MilliporeSigmaSt. LouisMissouriUSA
| | - Jason Gustin
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | | | - Jiajian Liu
- Life Science Bioinformatics, IT, MilliporeSigmaSt. LouisMissouriUSA
| | - David Razafsky
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| |
Collapse
|
17
|
Min H, Kim SM, Kim D, Lee S, Lee S, Lee JS. Hybrid cell line development system utilizing site-specific integration and methotrexate-mediated gene amplification in Chinese hamster ovary cells. Front Bioeng Biotechnol 2022; 10:977193. [PMID: 36185448 PMCID: PMC9521551 DOI: 10.3389/fbioe.2022.977193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Site-specific integration has emerged as a promising strategy for streamlined and predictable Chinese hamster ovary (CHO) cell line development (CLD). However, the low specific productivity of the targeted integrants limits their practical application. In this study, we developed a hybrid CLD platform combining site-specific integration of a transgene and dihydrofolate reductase/methotrexate (DHFR/MTX)-mediated gene amplification to generate high-producing recombinant CHO cell lines. We used the CRISPR/Cas9-based recombinase-mediated cassette exchange landing pad platform to integrate the DHFR expression cassette and transgene landing pad into a CHO genomic hot spot, C12orf35 locus, of DHFR-knockout CHO-K1 host cell lines. When subjected to various MTX concentrations up to 1 μM, EGFP-expressing targeted integrants showed a 3.6-fold increase in EGFP expression in the presence of 200 nM MTX, accompanied by an increase in the DHFR and EGFP copy number. A single-step 200 nM MTX amplification increased the specific monoclonal antibody (mAb) productivity (qmAb) of recombinant mAb-producing targeted integrants by 2.8-folds, reaching a qmAb of 9.1–11.0 pg/cell/day. Fluorescence in situ hybridization analysis showed colocalization of DHFR and mAb sequences at the intended chromosomal locations without clear amplified arrays of signals. Most MTX-amplified targeted integrants sustained recombinant mAb production during long-term culture in the absence of MTX, supporting stable gene expression in the amplified cell lines. Our study provides a new CLD platform that increases the productivity of targeted integrants by amplifying the transgene copies.
Collapse
Affiliation(s)
- Honggi Min
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Seul Mi Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Dongwoo Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Solhwi Lee
- Department of Applied Chemistry and Biological Engineering, Ajou University, Suwon, South Korea
| | - Sumin Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- Department of Applied Chemistry and Biological Engineering, Ajou University, Suwon, South Korea
- *Correspondence: Jae Seong Lee,
| |
Collapse
|
18
|
Coulet M, Kepp O, Kroemer G, Basmaciogullari S. Metabolic Profiling of CHO Cells during the Production of Biotherapeutics. Cells 2022; 11:cells11121929. [PMID: 35741058 PMCID: PMC9221972 DOI: 10.3390/cells11121929] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/08/2023] Open
Abstract
As indicated by an ever-increasing number of FDA approvals, biotherapeutics constitute powerful tools for the treatment of various diseases, with monoclonal antibodies (mAbs) accounting for more than 50% of newly approved drugs between 2014 and 2018 (Walsh, 2018). The pharmaceutical industry has made great progress in developing reliable and efficient bioproduction processes to meet the demand for recombinant mAbs. Mammalian cell lines are preferred for the production of functional, complex recombinant proteins including mAbs, with Chinese hamster ovary (CHO) cells being used in most instances. Despite significant advances in cell growth control for biologics manufacturing, cellular responses to environmental changes need to be understood in order to further improve productivity. Metabolomics offers a promising approach for developing suitable strategies to unlock the full potential of cellular production. This review summarizes key findings on catabolism and anabolism for each phase of cell growth (exponential growth, the stationary phase and decline) with a focus on the principal metabolic pathways (glycolysis, the pentose phosphate pathway and the tricarboxylic acid cycle) and the families of biomolecules that impact these circuities (nucleotides, amino acids, lipids and energy-rich metabolites).
Collapse
Affiliation(s)
- Mathilde Coulet
- Sanofi R&D, 94400 Vitry-sur-Seine, France;
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France;
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France;
- Institut Universitaire de France, Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, 75006 Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France;
- Institut Universitaire de France, Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, 75006 Paris, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
- Correspondence: (G.K.); (S.B.)
| | | |
Collapse
|
19
|
Improved Titer in Late-Stage Mammalian Cell Culture Manufacturing by Re-Cloning. Bioengineering (Basel) 2022; 9:bioengineering9040173. [PMID: 35447733 PMCID: PMC9030702 DOI: 10.3390/bioengineering9040173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 01/19/2023] Open
Abstract
Improving productivity to reduce the cost of biologics manufacturing and ensure that therapeutics can reach more patients remains a major challenge faced by the biopharmaceutical industry. Chinese hamster ovary (CHO) cell lines are commonly prepared for biomanufacturing by single cell cloning post-transfection and recovery, followed by lead clone screening, generation of a research cell bank (RCB), cell culture process development, and manufacturing of a master cell bank (MCB) to be used in early phase clinical manufacturing. In this study, it was found that an additional round of cloning and clone selection from an established monoclonal RCB or MCB (i.e., re-cloning) significantly improved titer for multiple late phase monoclonal antibody upstream processes. Quality attributes remained comparable between the processes using the parental clones and the re-clones. For two CHO cells expressing different antibodies, the re-clone performance was successfully scaled up at 500-L or at 2000-L bioreactor scales, demonstrating for the first time that the re-clone is suitable for late phase and commercial manufacturing processes for improvement of titer while maintaining comparable product quality to the early phase process.
Collapse
|
20
|
Malm M, Kuo CC, Barzadd MM, Mebrahtu A, Wistbacka N, Razavi R, Volk AL, Lundqvist M, Kotol D, Tegel H, Hober S, Edfors F, Gräslund T, Chotteau V, Field R, Varley PG, Roth RG, Lewis NE, Hatton D, Rockberg J. Harnessing secretory pathway differences between HEK293 and CHO to rescue production of difficult to express proteins. Metab Eng 2022; 72:171-187. [PMID: 35301123 PMCID: PMC9189052 DOI: 10.1016/j.ymben.2022.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/09/2022] [Accepted: 03/10/2022] [Indexed: 10/31/2022]
Abstract
Biologics represent the fastest growing group of therapeutics, but many advanced recombinant protein moieties remain difficult to produce. Here, we identify metabolic engineering targets limiting expression of recombinant human proteins through a systems biology analysis of the transcriptomes of CHO and HEK293 during recombinant expression. In an expression comparison of 24 difficult to express proteins, one third of the challenging human proteins displayed improved secretion upon host cell swapping from CHO to HEK293. Guided by a comprehensive transcriptomics comparison between cell lines, especially highlighting differences in secretory pathway utilization, a co-expression screening of 21 secretory pathway components validated ATF4, SRP9, JUN, PDIA3 and HSPA8 as productivity boosters in CHO. Moreover, more heavily glycosylated products benefitted more from the elevated activities of the N- and O-glycosyltransferases found in HEK293. Collectively, our results demonstrate the utilization of HEK293 for expression rescue of human proteins and suggest a methodology for identification of secretory pathway components for metabolic engineering of HEK293 and CHO.
Collapse
Affiliation(s)
- Magdalena Malm
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Chih-Chung Kuo
- Departments of Pediatrics and Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, 92093, USA
| | - Mona Moradi Barzadd
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Aman Mebrahtu
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Num Wistbacka
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Ronia Razavi
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Anna-Luisa Volk
- 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
| | - David Kotol
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden
| | - Hanna Tegel
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Sophia Hober
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden
| | - Torbjörn Gräslund
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Veronique Chotteau
- Dept. of Industrial Biotechnology, KTH - Royal Institute of Technology, Stockholm, SE-10691, Sweden
| | - Ray Field
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Paul G Varley
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Robert G Roth
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nathan E Lewis
- Departments of Pediatrics and Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, 92093, USA.
| | - Diane Hatton
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Johan Rockberg
- Dept. of Protein Science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden.
| |
Collapse
|
21
|
Spahn PN, Zhang X, Hu Q, Lu H, Hamaker NK, Hefzi H, Li S, Kuo CC, Huang Y, Lee JC, Davis AJ, Ly P, Lee KH, Lewis NE. Restoration of DNA repair mitigates genome instability and increases productivity of Chinese hamster ovary cells. Biotechnol Bioeng 2022; 119:963-982. [PMID: 34953085 PMCID: PMC8821244 DOI: 10.1002/bit.28016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/11/2022]
Abstract
Chinese hamster ovary (CHO) cells are the primary host for manufacturing of therapeutic proteins. However, productivity loss is a major problem and is associated with genome instability, as chromosomal aberrations reduce transgene copy number and decrease protein expression. We analyzed whole-genome sequencing data from 11 CHO cell lines and found deleterious single-nucleotide variants in DNA repair genes. Comparison with primary Chinese hamster cells confirmed DNA repair to be compromised in CHO. Correction of key DNA repair genes by single-nucleotide variant reversal or expression of intact complementary DNAs successfully improved DNA repair and mitigated karyotypic instability. Moreover, overexpression of intact copies of LIG4 and XRCC6 in a CHO cell line expressing secreted alkaline phosphatase mitigated transgene copy loss and improved protein titer retention. These results show that correction of DNA repair genes yields improvements in genome stability in CHO, and provide new opportunities for cell line development for sustainable protein expression.
Collapse
Affiliation(s)
- Philipp N. Spahn
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, San Diego, La Jolla, CA 92093
| | - Xiaolin Zhang
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711
| | - Qing Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Huiming Lu
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nathaniel K. Hamaker
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711
| | - Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Shangzhong Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Yingxiang Huang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Jamie C. Lee
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Anthony J. Davis
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Peter Ly
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kelvin H. Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711, These authors jointly supervised this work: Kelvin H. Lee, , 302-831-0344, Nathan E. Lewis, , 858-997-5844
| | - Nathan E. Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, San Diego, La Jolla, CA 92093, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, These authors jointly supervised this work: Kelvin H. Lee, , 302-831-0344, Nathan E. Lewis, , 858-997-5844
| |
Collapse
|
22
|
Marx N, Eisenhut P, Weinguny M, Klanert G, Borth N. How to train your cell - Towards controlling phenotypes by harnessing the epigenome of Chinese hamster ovary production cell lines. Biotechnol Adv 2022; 56:107924. [PMID: 35149147 DOI: 10.1016/j.biotechadv.2022.107924] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/24/2022]
Abstract
Recent advances in omics technologies and the broad availability of big datasets have revolutionized our understanding of Chinese hamster ovary cells in their role as the most prevalent host for production of complex biopharmaceuticals. In consequence, our perception of this "workhorse of the biopharmaceutical industry" has successively shifted from that of a nicely working, but unknown recombinant protein producing black box to a biological system governed by multiple complex regulatory layers that might possibly be harnessed and manipulated at will. Despite the tremendous progress that has been made to characterize CHO cells on various omics levels, our understanding is still far from complete. The well-known inherent genetic plasticity of any immortalized and rapidly dividing cell line also characterizes CHO cells and can lead to problematic instability of recombinant protein production. While the high mutational frequency has been a focus of CHO cell research for decades, the impact of epigenetics and its role in differential gene expression has only recently been addressed. In this review we provide an overview about the current understanding of epigenetic regulation in CHO cells and discuss its significance for shaping the cell's phenotype. We also look into current state-of-the-art technology that can be applied to harness and manipulate the epigenetic network so as to nudge CHO cells towards a specific phenotype. Here, we revise current strategies on site-directed integration and random as well as targeted epigenome modifications. Finally, we address open questions that need to be investigated to exploit the full repertoire of fine-tuned control of multiplexed gene expression using epigenetic and systems biology tools.
Collapse
Affiliation(s)
- Nicolas Marx
- University of Natural Resources and Life Sciences, Vienna, Austria
| | - Peter Eisenhut
- Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Marcus Weinguny
- University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Gerald Klanert
- Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Nicole Borth
- University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria.
| |
Collapse
|
23
|
Pybus LP, Kalsi D, Matthews JT, Hawke E, Barber N, Richer R, Young A, Saunders FL. Coupling picodroplet microfluidics with plate imaging for the rapid creation of biomanufacturing suitable cell lines with high probability and improved multi-step assurance of monoclonality. Biotechnol J 2021; 17:e2100357. [PMID: 34633760 DOI: 10.1002/biot.202100357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND There is an expectation from regulatory agencies that cell lines used in the commercial production of biopharmaceuticals are derived from a single cell progenitor. Traditional methods of single cell cloning include the use of the limiting dilution cloning method which often requires multiple rounds of low cell density cell plating and either microscopic evaluation that wells contain single cells and/or the calculation of a statistically derived probability of monoclonality. METHODS AND RESULTS We have combined the single cell screening, deposition and picodroplet imaging ability of Sphere Fluidics' Cyto-Mine technology with the plate imaging capability of the Solentim Cell Metric to create a novel workflow for the generation of high producing clonal cell lines with both high probability and assurance of monoclonality. The efficiency of three key stages of the process (single cell picodroplet encapsulation, single picodroplet dispensation and single cell settling in the focal plane of the plate imager) was determined and a probability calculation was derived using the Wilson Score Interval method. The combined probability that a single cell is encapsulated into a picodroplet, is deposited into the correct well of a 96-well plate and that a cell settles into the focal plane of the plate imager yields a combined > 99% probability of monoclonality. Furthermore, visual verification of a single cell progenitor is obtained at multiple steps throughout the cloning workflow. CONCLUSION This novel methodology for the rapid creation of high quality clonal cell lines for biomanufacturing purposes has many advantages over more traditional approaches including improved assurance of single cell derivation, integrated imaging capability, assay flexibility, equipment utilization time and in-process cell line segregation.
Collapse
Affiliation(s)
- Leon P Pybus
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Devika Kalsi
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Joe T Matthews
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - Ellie Hawke
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK.,Current Address: Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Nicholas Barber
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Rachel Richer
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Alison Young
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Fay L Saunders
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| |
Collapse
|
24
|
Li GB, Pollard J, Liu R, Stevens RC, Quiroz J, Nelson MC, Manahan M, Murgolo N, Ehrick RS, Wallenstein EJ, Hughes J, Tsao YS, Zhao J, Du Z, Tugcu N, Pollard D. Retrospective assessment of clonality of a legacy cell line by analytical subcloning of the master cell bank. Biotechnol Prog 2021; 38:e3215. [PMID: 34586757 DOI: 10.1002/btpr.3215] [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: 06/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 11/10/2022]
Abstract
In recent years, assurance of clonality of the production cell line has been emphasized by health authorities during review of regulatory submissions. When insufficient assurance of clonality is provided, augmented control strategies may be required for a commercial production process. In this study, we conducted a retrospective assessment of clonality of a legacy cell line through analysis of subclones from the master cell bank (MCB). Twenty-four subclones were randomly selected based on a predetermined acceptance sampling plan. All these subclones share a conserved integration junction, thus providing a high level of assurance that the cell population in the MCB was derived from a single progenitor cell. However, Southern blot analysis indicates that at least four subpopulations possibly exist in the MCB. Additional characterization of these four subpopulations demonstrated that the resulting changes in product quality attributes of some subclones are not related to the genetic heterogeneity observed in Southern blot hybridization. Furthermore, process consistency, process comparability, and analytical comparability have been demonstrated in batches produced across varying manufacturing processes, scales, facilities, cell banks, and cell ages. Finally, process and product consistency together with a high level of assurance of clonal origin of the MCB helped clear the hurdle for regulatory approval without requirement of additional control strategies.
Collapse
Affiliation(s)
- Guanghua Benson Li
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jennifer Pollard
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Ren Liu
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Richard C Stevens
- Genetics and Pharmacogenomics, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jorge Quiroz
- Research CMC Statistics, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Michael C Nelson
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Matthew Manahan
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Nicholas Murgolo
- Genetics and Pharmacogenomics, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Robin S Ehrick
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Eric J Wallenstein
- Biologics Process Development & Commercialization, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jason Hughes
- Global Research IT, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Yung-Shyeng Tsao
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jia Zhao
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Zhimei Du
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Nihal Tugcu
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - David Pollard
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
25
|
Wurm MJ, Wurm FM. Naming CHO cells for bio-manufacturing: Genome plasticity and variant phenotypes of cell populations in bioreactors question the relevance of old names. Biotechnol J 2021; 16:e2100165. [PMID: 34050613 DOI: 10.1002/biot.202100165] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/10/2021] [Accepted: 05/20/2021] [Indexed: 11/06/2022]
Abstract
Chinese Hamster Ovary [CHO] cells are the workhorse for production of modern biopharmaceuticals. They are however immortalized cells with a high propensity for genetic change. Judging from published culture records, CHO cell populations have undergone hundreds of population doublings since their origin in the late 1950s. Different cell populations were established and named from 1 to 3 decades after their generation, such as CHO-Pro-, CHO-K1, CHO-DG44, CHO-S, CHO-DUK, CHO-DXB-11 to indicate origin and certain phenotypic features. These names are commonly used in scientific publications still today. This article discusses the relevance of such names. We argue that they provide a false sense of identity. To substantiate this, we provide the long (and poorly recorded) history of CHO cells as well as their highly complex genetics. Finally, we suggest an alternative naming system for CHO cells which provides more relevant information. While the implementation of a new naming convention will require substantial discussions among members of the relevant community, it should improve interpretation and comparability between laboratories. This, in turn will help scientific communities and industrial users to attain and further the full potential of CHO cells.
Collapse
Affiliation(s)
- Maria J Wurm
- Life Science Faculty, Swiss Federal Institute of Technology Lausanne [EPFL], Lausanne, Switzerland
| | - Florian M Wurm
- Life Science Faculty, Swiss Federal Institute of Technology Lausanne [EPFL], Lausanne, Switzerland
| |
Collapse
|
26
|
Inclusion of maintenance energy improves the intracellular flux predictions of CHO. PLoS Comput Biol 2021; 17:e1009022. [PMID: 34115746 PMCID: PMC8221792 DOI: 10.1371/journal.pcbi.1009022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/23/2021] [Accepted: 04/28/2021] [Indexed: 11/19/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are the leading platform for the production of biopharmaceuticals with human-like glycosylation. The standard practice for cell line generation relies on trial and error approaches such as adaptive evolution and high-throughput screening, which typically take several months. Metabolic modeling could aid in designing better producer cell lines and thus shorten development times. The genome-scale metabolic model (GSMM) of CHO can accurately predict growth rates. However, in order to predict rational engineering strategies it also needs to accurately predict intracellular fluxes. In this work we evaluated the agreement between the fluxes predicted by parsimonious flux balance analysis (pFBA) using the CHO GSMM and a wide range of 13C metabolic flux data from literature. While glycolytic fluxes were predicted relatively well, the fluxes of tricarboxylic acid (TCA) cycle were vastly underestimated due to too low energy demand. Inclusion of computationally estimated maintenance energy significantly improved the overall accuracy of intracellular flux predictions. Maintenance energy was therefore determined experimentally by running continuous cultures at different growth rates and evaluating their respective energy consumption. The experimentally and computationally determined maintenance energy were in good agreement. Additionally, we compared alternative objective functions (minimization of uptake rates of seven nonessential metabolites) to the biomass objective. While the predictions of the uptake rates were quite inaccurate for most objectives, the predictions of the intracellular fluxes were comparable to the biomass objective function. There is an increasing demand for protein pharmaceuticals, especially monoclonal antibodies. Chinese Hamster Ovary (CHO) are currently the leading production host due to their ability to perform human-like post-translational modifications. However, it typically takes several months of trial-and-error approaches to develop a high-producer cell line. Metabolic modelling has the potential to make cell line and process development faster and cheaper by predicting targeted modifications to the cell line genome, cultivation medium or bioprocess. In fact, genome-scale metabolic reconstructions of CHO are already available, and ready for use in cell line development. However, in order to successfully use these models, we need to make sure that they are able to accurately predict metabolic phenotypes. Here we use genome-scale metabolic models of CHO to evaluate the models’ ability to correctly predict intracellular flux distributions. We find that a crucial key ingredient for the correct estimation of central carbon fluxes is the non-growth associated maintenance energy (mATP). We estimated mATP computationally and confirmed it experimentally. Adding this single constraint leads to significantly better predictions of intracellular fluxes, especially in glycolysis and the tricarboxylic acid cycle.
Collapse
|
27
|
Donaldson JS, Dale MP, Rosser SJ. Decoupling Growth and Protein Production in CHO Cells: A Targeted Approach. Front Bioeng Biotechnol 2021; 9:658325. [PMID: 34150726 PMCID: PMC8207133 DOI: 10.3389/fbioe.2021.658325] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/09/2021] [Indexed: 11/28/2022] Open
Abstract
Fed-batch cultures of Chinese Hamster Ovary cells have been used to produce high quantities of biotherapeutics, particularly monoclonal antibodies. However, a growing number of next-generation biotherapeutics, such as bi-specific antibodies and fusion proteins, are difficult to express using standard fed-batch processes. Decoupling cell growth and biotherapeutic production is becoming an increasingly desired strategy for the biomanufacturing industry, especially for difficult-to-express products. Cells are grown to a high cell density in the absence of recombinant protein production (the growth phase), then expression of the recombinant protein is induced and cell proliferation halted (the production phase), usually by combining an inducible gene expression system with a proliferation control strategy. Separating the growth and production phases allows cell resources to be more efficiently directed toward either growth or production, improving growth characteristics and enhancing the production of difficult to express proteins. However, current mammalian cell proliferation control methods rely on temperature shifts and chemical agents, which interact with many non-proliferation pathways, leading to variable impacts on product quality and culture viability. Synthetic biology offers an alternative approach by strategically targeting proliferation pathways to arrest cell growth but have largely remained unused in industrial bioproduction. Due to recent developments in microbial decoupling systems and advances in available mammalian cell engineering tools, we propose that the synthetic biology approach to decoupling growth and production needs revisiting.
Collapse
Affiliation(s)
- James S Donaldson
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew P Dale
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Susan J Rosser
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Langsdorf E, Yu L, Kanevskaia L, Felkner R, Sturner S, McVey D, Khetan A. Retrospective assessment of clonal origin of cell lines. Biotechnol Prog 2021; 37:e3157. [PMID: 33896120 DOI: 10.1002/btpr.3157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 11/08/2022]
Abstract
Cell lines used for the manufacture of recombinant proteins are expected to arise from a single cell as a control strategy to limit variability and ensure consistent protein production. Health authorities require a minimum of two rounds of limiting dilution cloning or its equivalent to meet the requirement of single cell origin. However, many legacy cell lines may not have been generated with process meeting this criteria potentially impeding the path to commercialization. A general monoclonality assessment strategy was developed based on using the site of plasmid integration for a cell's identity. By comparing the identities of subclones from a master cell bank (MCB) to each other and that of the MCB, a probability of monoclonality was established. Two technologies were used for cell identity, Southern blot and a PCR assay based on plasmid-genome junction sequences identified by splinkerette PCR. Southern blot analysis revealed that subclones may have banding patterns that differ from each other and yet indicate monoclonal origin. Splinkerette PCR identifies cellular sequence flanking the point(s) of plasmid integration. The two assays together provide complimentary data for cell identity that enables proper monoclonality assessment and establishes that the three legacy cell lines investigated are all of clonal origin.
Collapse
Affiliation(s)
- Erik Langsdorf
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Le Yu
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Lioudmila Kanevskaia
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Roland Felkner
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Stephen Sturner
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Duncan McVey
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Anurag Khetan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| |
Collapse
|
29
|
Baik JY, Han HJ, Lee KH. DNA Double-Strand Breaks Affect Chromosomal Rearrangements during Methotrexate-Mediated Gene Amplification in Chinese Hamster Ovary Cells. Pharmaceutics 2021; 13:pharmaceutics13030376. [PMID: 33809068 PMCID: PMC8000239 DOI: 10.3390/pharmaceutics13030376] [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: 01/18/2021] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Methotrexate (MTX)-mediated gene amplification has been widely used in Chinese hamster ovary (CHO) cells for the biomanufacturing of therapeutic proteins. Although many studies have reported chromosomal instability and extensive chromosomal rearrangements in MTX-mediated gene-amplified cells, which may be associated with cell line instability issues, the mechanisms of chromosomal rearrangement formation remain poorly understood. We tested the impact of DNA double-strand breaks (DSBs) on chromosomal rearrangements using bleomycin, a DSB-inducing reagent. Bleomycin-treated CHO-DUK cells, which are one of the host cell lines deficient in dihydrofolate reductase (Dhfr) activity, exhibited a substantial number of cells containing radial formations or non-radial formations with chromosomal rearrangements, suggesting that DSBs may be associated with chromosomal rearrangements. To confirm the causes of DSBs during gene amplification, we tested the effects of MTX treatment and the removal of nucleotide base precursors on DSB formation in Dhfr-deficient (i.e., CHO-DUK) and Dhfr-expressing (i.e., CHO-K1) cells. Immunocytochemistry demonstrated that MTX treatment did not induce DSBs per se, but a nucleotide shortage caused by the MTX-mediated inhibition of Dhfr activity resulted in DSBs. Our data suggest that a nucleotide shortage caused by MTX-mediated Dhfr inhibition in production cell lines is the primary cause of a marked increase in DSBs, resulting in extensive chromosomal rearrangements after gene amplification processes.
Collapse
Affiliation(s)
- Jong Youn Baik
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
- Department of Biological Engineering, Inha University, Incheon 22212, Korea;
- Correspondence: (J.Y.B.); (K.H.L.); Tel.: +82-32-860-7513 (J.Y.B.); +1-302-831-0344 (K.H.L.)
| | - Hye-Jin Han
- Department of Biological Engineering, Inha University, Incheon 22212, Korea;
| | - Kelvin H. Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
- Correspondence: (J.Y.B.); (K.H.L.); Tel.: +82-32-860-7513 (J.Y.B.); +1-302-831-0344 (K.H.L.)
| |
Collapse
|
30
|
Turilova VI, Goryachaya TS, Yakovleva TK. Chinese hamster ovary cell line DXB-11: chromosomal instability and karyotype heterogeneity. Mol Cytogenet 2021; 14:11. [PMID: 33596973 PMCID: PMC7888135 DOI: 10.1186/s13039-021-00528-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chinese hamster ovary cell lines, also known as CHO cells, represent a large family of related, yet quite different, cell lines which are metabolic mutants derived from the original cell line, CHO-ori. Dihydrofolate reductase-deficient DXB-11 cell line, one of the first CHO derivatives, serves as the host cell line for the production of therapeutic proteins. It is generally assumed that DXB-11 is identical to DUKX or CHO-DUK cell lines, but, to our knowledge, DXB-11 karyotype has not been described yet. RESULTS Using differential staining approaches (G-, C-banding and Ag-staining), we presented DXB-11 karyotype and revealed that karyotypes of DXB-11 and CHO-DUK cells have a number of differences. Although the number of chromosomes is equal-20 in each cell line-DXB-11 has normal chromosomes of the 1st and 5th pairs as well as an intact chromosome 8. Besides, in DXB-11 line, chromosome der(Z9) includes the material of chromosomes X and 6, whereas in CHO-DUK it results from the translocation of chromosomes 1 and 6. Ag-positive nucleolar organizer regions were revealed in the long arms of chromosome del(4)(q11q12) and both chromosome 5 homologues, as well as in the short arms of chromosomes 8 and add(8)(q11). Only 19 from 112 (16.96%) DXB-11 cells display identical chromosome complement accepted as the main structural variant of karyotype. The karyotype heterogeneity of all the rest of cells (93, 83.04%) occurs due to clonal and nonclonal additional structural rearrangements of chromosomes. Estimation of the frequency of chromosome involvement in these rearrangements allowed us to reveal that chromosomes 9, der(X)t(X;3;4), del(2)(p21p23), del(2)(q11q22) /Z2, der(4) /Z7, add(6)(p11) /Z8 are the most stable, whereas mar2, probably der(10), is the most unstable chromosome. A comparative analysis of our own and literary data on CHO karyotypes allowed to designate conservative chromosomes, both normal and rearranged, that remain unchanged in different CHO cell lines, as well as variable chromosomes that determine the individuality of karyotypes of CHO derivatives. CONCLUSION DXB-11and CHO-DUK cell lines differ in karyotypes. The revealed differential instability of DXB-11 chromosomes is likely not incidental and results in karyotype heterogeneity of cell population.
Collapse
Affiliation(s)
- Victoria I Turilova
- Laboratory of Cell Morphology, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky ave., 4, St Petersburg, Russia, 194064.
| | - Tatyana S Goryachaya
- Centre of Cell Technologies, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky ave., 4, St Petersburg, Russia, 194064
| | - Tatiana K Yakovleva
- Laboratory of Cell Morphology, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky ave., 4, St Petersburg, Russia, 194064
| |
Collapse
|
31
|
Weinguny M, Klanert G, Eisenhut P, Lee I, Timp W, Borth N. Subcloning induces changes in the DNA-methylation pattern of outgrowing Chinese hamster ovary cell colonies. Biotechnol J 2021; 16:e2000350. [PMID: 33484505 DOI: 10.1002/biot.202000350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Chinese hamster ovary (CHO) cells are the most extensively used mammalian production system for biologics intended for use in humans. A critical step in the establishment of production cell lines is single cell cloning, with the objective of achieving high productivity and product quality. Despite general use, knowledge of the effects of this process is limited. Importantly, single cell cloned cells display a wide array of observed phenotypes, which so far was attributed to the instability and variability of the CHO genome. In this study we present data indicating that the emergence of diverse phenotypes during single cell cloning is associated with changes in DNA methylation patterns and transcriptomes that occur during the subcloning process. The DNA methylation pattern of each analyzed subclone, randomly picked from all outgrowing clones of the experiment, had unique changes preferentially found in regulatory regions of the genome such as enhancers, and de-enriched in actively transcribed sequences (not including the respective promoters), indicating that these changes resulted in adaptations of the relative gene expression pattern. The transcriptome of each subclone also had a significant number of individual changes. These results indicate that epigenetic regulation is a hidden, but important player in cell line development with a major role in the establishment of high performing clones with improved characteristics for bioprocessing.
Collapse
Affiliation(s)
- Marcus Weinguny
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Gerald Klanert
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Peter Eisenhut
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Isac Lee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, USA
| | - Nicole Borth
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| |
Collapse
|
32
|
O’Brien SA, Hu WS. Cell culture bioprocessing - the road taken and the path forward. Curr Opin Chem Eng 2020; 30:100663. [PMID: 33391982 PMCID: PMC7773285 DOI: 10.1016/j.coche.2020.100663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cell culture processes are used to produce the vast majority of protein therapeutics, valued at over US$180 billion per annum worldwide. For more than a decade now, these processes have become highly productive. To further enhance capital efficiency, there has been an increase in the adoption of disposable apparatus and continuous processing, as well as a greater exploration of in-line sensing, various -omic tools, and cell engineering to enhance process controllability and product quality consistency. These feats in cell culture processing for protein biologics will help accelerate the bioprocess advancements for virus and cell therapy applications.
Collapse
Affiliation(s)
- Sofie A. O’Brien
- Department of Biomedical Engineering and Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455-0132 USA
| | - Wei-Shou Hu
- Department of Biomedical Engineering and Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455-0132 USA
| |
Collapse
|
33
|
|
34
|
Bevan S, Schoenfelder S, Young RJ, Zhang L, Andrews S, Fraser P, O'Callaghan PM. High-resolution three-dimensional chromatin profiling of the Chinese hamster ovary cell genome. Biotechnol Bioeng 2020; 118:784-796. [PMID: 33095445 PMCID: PMC7894165 DOI: 10.1002/bit.27607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022]
Abstract
Chinese hamster ovary (CHO) cell lines are the pillars of a multibillion‐dollar biopharmaceutical industry producing recombinant therapeutic proteins. The effects of local chromatin organization and epigenetic repression within these cell lines result in unpredictable and unstable transgene expression following random integration. Limited knowledge of the CHO genome and its higher order chromatin organization has thus far impeded functional genomics approaches required to tackle these issues. Here, we present an integrative three‐dimensional (3D) map of genome organization within the CHOK1SV® 10E9 cell line in conjunction with an improved, less fragmented CHOK1SV 10E9 genome assembly. Using our high‐resolution chromatin conformation datasets, we have assigned ≈90% of sequence to a chromosome‐scale genome assembly. Our genome‐wide 3D map identifies higher order chromatin structures such as topologically associated domains, incorporates our chromatin accessibility data to enhance the identification of active cis‐regulatory elements, and importantly links these cis‐regulatory elements to target promoters in a 3D promoter interactome. We demonstrate the power of our improved functional annotation by evaluating the 3D landscape of a transgene integration site and two phenotypically different cell lines. Our work opens up further novel genome engineering targets, has the potential to inform vital improvements for industrial biotherapeutic production, and represents a significant advancement for CHO cell line development.
Collapse
Affiliation(s)
- Stephen Bevan
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Stefan Schoenfelder
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Robert J Young
- R&D Cell Engineering, Lonza Biologics, Little Chesterford, UK
| | - Lin Zhang
- Cell Line Development, World Wide Pharmaceutical Sciences, BioTherapeutics Research and Development, Pfizer Inc., Andover, Massachusetts, USA
| | - Simon Andrews
- Bioinformatics Facility, The Babraham Institute, Cambridge, UK
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | | |
Collapse
|
35
|
Dhiman H, Campbell M, Melcher M, Smith KD, Borth N. Predicting favorable landing pads for targeted integrations in Chinese hamster ovary cell lines by learning stability characteristics from random transgene integrations. Comput Struct Biotechnol J 2020; 18:3632-3648. [PMID: 33304461 PMCID: PMC7710658 DOI: 10.1016/j.csbj.2020.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 01/06/2023] Open
Abstract
Chinese Hamster Ovary (CHO) cell lines are considered to be the preferred platform for the production of biotherapeutics, but issues related to expression instability remain unresolved. In this study, we investigated potential causes for an unstable phenotype by comparing cell lines that express stably to such that undergo loss in titer across 10 passages. Factors related to transgene integrity and copy number as well as the genomic profile around the integration sites were analyzed. Horizon Discovery CHO-K1 (HD-BIOP3) derived production cell lines selected for phenotypes with low, medium or high copy number, each with stable and unstable transgene expression, were sequenced to capture changes at genomic and transcriptomic levels. The exact sites of the random integration events in each cell line were also identified, followed by profiling of the genomic, transcriptomic and epigenetic patterns around them. Based on the information deduced from these random integration events, genomic loci that potentially favor reliable and stable transgene expression were reported for use as targeted transgene integration sites. By comparing stable vs unstable phenotypes across these parameters, we could establish that expression stability may be controlled at three levels: 1) Good choice of integration site, 2) Ensuring integrity of transgene and observing concatemerization pattern after integration, and 3) Checking for potential stress related cellular processes. Genome wide favorable and unfavorable genomic loci for targeted transgene integration can be browsed at https://www.borthlabchoresources.boku.ac.at/
Collapse
Affiliation(s)
- Heena Dhiman
- University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | | | - Michael Melcher
- University of Natural Resources and Life Sciences, Vienna, Austria
| | | | - Nicole Borth
- University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Centre of Industrial Biotechnology, Vienna, Austria
| |
Collapse
|
36
|
Evolution from adherent to suspension: systems biology of HEK293 cell line development. Sci Rep 2020; 10:18996. [PMID: 33149219 PMCID: PMC7642379 DOI: 10.1038/s41598-020-76137-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 10/22/2020] [Indexed: 01/28/2023] Open
Abstract
The need for new safe and efficacious therapies has led to an increased focus on biologics produced in mammalian cells. The human cell line HEK293 has bio-synthetic potential for human-like production attributes and is currently used for manufacturing of several therapeutic proteins and viral vectors. Despite the increased popularity of this strain we still have limited knowledge on the genetic composition of its derivatives. Here we present a genomic, transcriptomic and metabolic gene analysis of six of the most widely used HEK293 cell lines. Changes in gene copy and expression between industrial progeny cell lines and the original HEK293 were associated with cellular component organization, cell motility and cell adhesion. Changes in gene expression between adherent and suspension derivatives highlighted switching in cholesterol biosynthesis and expression of five key genes (RARG, ID1, ZIC1, LOX and DHRS3), a pattern validated in 63 human adherent or suspension cell lines of other origin.
Collapse
|
37
|
Edwards CE, Yount BL, Graham RL, Leist SR, Hou YJ, Dinnon KH, Sims AC, Swanstrom J, Gully K, Scobey TD, Cooley MR, Currie CG, Randell SH, Baric RS. Swine acute diarrhea syndrome coronavirus replication in primary human cells reveals potential susceptibility to infection. Proc Natl Acad Sci U S A 2020; 117:26915-26925. [PMID: 33046644 PMCID: PMC7604506 DOI: 10.1073/pnas.2001046117] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Zoonotic coronaviruses represent an ongoing threat, yet the myriads of circulating animal viruses complicate the identification of higher-risk isolates that threaten human health. Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered, highly pathogenic virus that likely evolved from closely related HKU2 bat coronaviruses, circulating in Rhinolophus spp. bats in China and elsewhere. As coronaviruses cause severe economic losses in the pork industry and swine are key intermediate hosts of human disease outbreaks, we synthetically resurrected a recombinant virus (rSADS-CoV) as well as a derivative encoding tomato red fluorescent protein (tRFP) in place of ORF3. rSADS-CoV replicated efficiently in a variety of continuous animal and primate cell lines, including human liver and rectal carcinoma cell lines. Of concern, rSADS-CoV also replicated efficiently in several different primary human lung cell types, as well as primary human intestinal cells. rSADS-CoV did not use human coronavirus ACE-2, DPP4, or CD13 receptors for docking and entry. Contemporary human donor sera neutralized the group I human coronavirus NL63, but not rSADS-CoV, suggesting limited human group I coronavirus cross protective herd immunity. Importantly, remdesivir, a broad-spectrum nucleoside analog that is effective against other group 1 and 2 coronaviruses, efficiently blocked rSADS-CoV replication in vitro. rSADS-CoV demonstrated little, if any, replicative capacity in either immune-competent or immunodeficient mice, indicating a critical need for improved animal models. Efficient growth in primary human lung and intestinal cells implicate SADS-CoV as a potential higher-risk emerging coronavirus pathogen that could negatively impact the global economy and human health.
Collapse
Affiliation(s)
- Caitlin E Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Rachel L Graham
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yixuan J Hou
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Kenneth H Dinnon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Amy C Sims
- Chemical and Biological Signatures Division, Pacific Northwest National Laboratory, Richland, WA 99354
| | - Jesica Swanstrom
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Kendra Gully
- Department of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Trevor D Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Michelle R Cooley
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Caroline G Currie
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599;
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
38
|
Hilliard W, Lee KH. Systematic identification of safe harbor regions in the CHO genome through a comprehensive epigenome analysis. Biotechnol Bioeng 2020; 118:659-675. [PMID: 33049068 DOI: 10.1002/bit.27599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/07/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
The Chinese hamster ovary (CHO) cell lines that are used to produce commercial quantities of therapeutic proteins commonly exhibit a decrease in productivity over time in culture, a phenomenon termed production instability. Random integration of the transgenes encoding the protein of interest into locations in the CHO genome that are vulnerable to genetic and epigenetic instability often causes production instability through copy number loss and silencing of expression. Several recent publications have shown that these cell line development challenges can be overcome by using site-specific integration (SSI) technology to insert the transgenes at genomic loci, often called "hotspots," that are transcriptionally permissive and have enhanced stability relative to the rest of the genome. However, extensive characterization of the CHO epigenome is needed to identify hotspots that maintain their desirable epigenetic properties in an industrial bioprocess environment and maximize transcription from a single integrated transgene copy. To this end, the epigenomes and transcriptomes of two distantly related cell lines, an industrially relevant monoclonal antibody-producing cell line and its parental CHO-K1 host, were characterized using high throughput chromosome conformation capture and RNAseq to analyze changes in the epigenome that occur during cell line development and associated changes in system-wide gene expression. In total, 10.9% of the CHO genome contained transcriptionally permissive three-dimensional chromatin structures with enhanced genetic and epigenetic stability relative to the rest of the genome. These safe harbor regions also showed good agreement with published CHO epigenome data, demonstrating that this method was suitable for finding genomic regions with epigenetic markers of active and stable gene expression. These regions significantly reduce the genomic search space when looking for CHO hotspots with widespread applicability and can guide future studies with the goal of maximizing the potential of SSI technology in industrial production CHO cell lines.
Collapse
Affiliation(s)
- William Hilliard
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
39
|
Kuhn A, Le Fourn V, Fisch I, Mermod N. Genome-wide analysis of single nucleotide variants allows for robust and accurate assessment of clonal derivation in cell lines used to produce biologics. Biotechnol Bioeng 2020; 117:3628-3638. [PMID: 32779734 PMCID: PMC7818110 DOI: 10.1002/bit.27534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/22/2020] [Accepted: 07/29/2020] [Indexed: 11/11/2022]
Abstract
A clonally derived (or “monoclonal”) cell line is a cell population derived from a single progenitor cell. Clonally derived cell lines are required for many biotechnological applications. For instance, recombinant mammalian cells used to produce therapeutic proteins are expected by regulatory authorities to be clonally derived. Assurance of clonal derivation (or “clonality”) is usually obtained from the characterization of the procedure used for cell cloning, for instance by assessing the success rate of single‐cell sorting but not by assessing the cell line itself. We have developed a method to assess clonal derivation directly from the genetic makeup of cells. The genomic test of clonality is based on whole‐genome sequencing and statistical analysis of single nucleotide variants. This approach quantifies the clonal fractions present in nonclonal samples and it provides a measure of the probability that a cell line is derived from a single cell. Upon experimental validation of the test, we show that it is highly accurate and that it can robustly detect minor clonal fractions of as little as 1% of the cell population. Moreover, we find that it is applicable to various cell line development protocols. This approach can simplify development protocols and shorten timelines while ensuring clonal derivation with high confidence.
Collapse
Affiliation(s)
- Alexandre Kuhn
- Department of Fundamental Microbiology, Institute of Biotechnology, University of Lausanne, Switzerland.,Selexis SA, Geneva, Switzerland
| | | | | | - Nicolas Mermod
- Department of Fundamental Microbiology, Institute of Biotechnology, University of Lausanne, Switzerland
| |
Collapse
|
40
|
Weinguny M, Klanert G, Eisenhut P, Jonsson A, Ivansson D, Lövgren A, Borth N. Directed evolution approach to enhance efficiency and speed of outgrowth during single cell subcloning of Chinese Hamster Ovary cells. Comput Struct Biotechnol J 2020; 18:1320-1329. [PMID: 32612755 PMCID: PMC7306589 DOI: 10.1016/j.csbj.2020.05.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/16/2020] [Accepted: 05/17/2020] [Indexed: 01/08/2023] Open
Abstract
Chinese Hamster Ovary (CHO) cells are the working horse of the pharmaceutical industry. To obtain high producing cell clones and to satisfy regulatory requirements single cell cloning is a necessary step in cell line development. However, it is also a tedious, labor intensive and expensive process. Here we show an easy way to enhance subclonability using subcloning by single cell sorting itself as the selection pressure, resulting in improved subcloning performance of three different host cell lines. These improvements in subclonability also lead to an enhanced cellular growth behavior during standard batch culture. RNA-seq was performed to shed light on the underlying mechanisms, showing that there is little overlap in differentially expressed genes or associated pathways between the cell lines, each finding their individual strategy for optimization. However, in all three cell lines pathways associated with the extracellular matrix were found to be enriched, indicating that cells struggle predominantly with their microenvironment and possibly lack of cell-to-cell contact. The observed small overlap may hint that there are multiple ways for a cell line to achieve a certain phenotype due to numerous genetic and subsequently metabolic redundancies.
Collapse
Key Words
- CHO
- CHO cells
- CHO, Chinese hamster ovary
- Cell line development
- Cell sorting
- Chinese Hamster Ovary Cells
- CoI, clusters of interest
- DE, directed evolved
- Directed Evolution
- ECM, extracellular matrix
- ES, enrichment score
- FACS
- FACS, fluorescent-activated cell sorting
- Fluorescent-activated cell sorting
- GSEA, gene set analysis
- Growth enhancement
- Growth improvement
- LDC, limiting dilution cloning
- NES, negative enrichment score
- PC, principal component
- PCA, principal component analysis
- POI, product of interest
- RNA Sequencing
- RNA-Seq
- RNA-Seq, RNA sequencing
- SCC, single cell cloning
- Single Cell Cloning
- Single Cell Subcloning
- Subcloning
- lfcSE, logfoldstandard error
Collapse
Affiliation(s)
- Marcus Weinguny
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Gerald Klanert
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Peter Eisenhut
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | | | | | | | - Nicole Borth
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
- Corresponding author at: Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
41
|
Abstract
Following the success of and the high demand for recombinant protein-based therapeutics during the last 25 years, the pharmaceutical industry has invested significantly in the development of novel treatments based on biologics. Mammalian cells are the major production systems for these complex biopharmaceuticals, with Chinese hamster ovary (CHO) cell lines as the most important players. Over the years, various engineering strategies and modeling approaches have been used to improve microbial production platforms, such as bacteria and yeasts, as well as to create pre-optimized chassis host strains. However, the complexity of mammalian cells curtailed the optimization of these host cells by metabolic engineering. Most of the improvements of titer and productivity were achieved by media optimization and large-scale screening of producer clones. The advances made in recent years now open the door to again consider the potential application of systems biology approaches and metabolic engineering also to CHO. The availability of a reference genome sequence, genome-scale metabolic models and the growing number of various “omics” datasets can help overcome the complexity of CHO cells and support design strategies to boost their production performance. Modular design approaches applied to engineer industrially relevant cell lines have evolved to reduce the time and effort needed for the generation of new producer cells and to allow the achievement of desired product titers and quality. Nevertheless, important steps to enable the design of a chassis platform similar to those in use in the microbial world are still missing. In this review, we highlight the importance of mammalian cellular platforms for the production of biopharmaceuticals and compare them to microbial platforms, with an emphasis on describing novel approaches and discussing still open questions that need to be resolved to reach the objective of designing enhanced modular chassis CHO cell lines.
Collapse
|
42
|
Faulty Cell Lines Can Befuddle Clinical Pharmacology Decision-Making. J Clin Pharmacol 2020; 60:563-565. [DOI: 10.1002/jcph.1569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
|
43
|
Welch JT, Arden NS. Considering “clonality”: A regulatory perspective on the importance of the clonal derivation of mammalian cell banks in biopharmaceutical development. Biologicals 2019; 62:16-21. [DOI: 10.1016/j.biologicals.2019.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022] Open
|
44
|
Möller J, Bhat K, Riecken K, Pörtner R, Zeng AP, Jandt U. Process-induced cell cycle oscillations in CHO cultures: Online monitoring and model-based investigation. Biotechnol Bioeng 2019; 116:2931-2943. [PMID: 31342512 DOI: 10.1002/bit.27124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 01/04/2023]
Abstract
The influence of process strategies on the dynamics of cell population heterogeneities in mammalian cell culture is still not well understood. We recently found that the progression of cells through the cell cycle causes metabolic regulations with variable productivities in antibody-producing Chimese hamster ovary (CHO) cells. On the other hand, it is so far unknown how bulk cultivation conditions, for example, variable nutrient concentrations depending on process strategies, can influence cell cycle-derived population dynamics. In this study, process-induced cell cycle synchronization was assessed in repeated-batch and fed-batch cultures. An automated flow cytometry set-up was developed to measure the cell cycle distribution online, using antibody-producing CHO DP-12 cells transduced with the cell cycle-specific fluorescent ubiquitination-based cell cycle indicator (FUCCI) system. On the basis of the population-resolved model, feeding-induced partial self-synchronization was predicted and the results were evaluated experimentally. In the repeated-batch culture, stable cell cycle oscillations were confirmed with an oscillating G1 phase distribution between 41% and 72%. Furthermore, oscillations of the cell cycle distribution were simulated and determined in a (bolus) fed-batch process with up to 25 × 1 0 6 cells/ml. The cell cycle synchronization arose with pulse feeding only and ceased with continuous feeding. Both simulated and observed oscillations occurred at higher frequencies than those observable based on regular (e.g., daily) sample analysis, thus demonstrating the need for high-frequency online cell cycle analysis. In summary, we showed how experimental methods combined with simulations enable the improved assessment of the effects of process strategies on the dynamics of cell cycle-dependent population heterogeneities. This provides a novel approach to understand cell cycle regulations, control cell population dynamics, avoid inadvertently induced oscillations of cell cycle distributions and thus to improve process stability and efficiency.
Collapse
Affiliation(s)
- Johannes Möller
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Krathika Bhat
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Kristoffer Riecken
- Department of Stem Cell Transplantation, Research Department Cell and Gene Therapy, University Medical Centre (UMC) Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Pörtner
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - An-Ping Zeng
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Uwe Jandt
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| |
Collapse
|
45
|
Tharmalingam T, Barkhordarian H, Tejeda N, Daris K, Yaghmour S, Yam P, Lu F, Goudar C, Munro T, Stevens J. Characterization of phenotypic and genotypic diversity in subclones derived from a clonal cell line. Biotechnol Prog 2019; 34:613-623. [PMID: 29882350 PMCID: PMC6099272 DOI: 10.1002/btpr.2666] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/09/2018] [Indexed: 12/19/2022]
Abstract
Regulatory guidelines require the sponsors to provide assurance of clonality of the production cell line, and when such evidence is not available, additional studies are typically required to further ensure consistent long‐term manufacturing of the product. One potential approach to provide such assurance of clonal derivation of a production cell line is to characterize subclones generated from the original cell line and assess their phenotypic and genotypic similarity with the hypothesis that cell lines derived from a clonal bank will share performance, productivity and product quality characteristics. In this study, a production cell line that was cloned by a validated FACS approach coupled with day 0 imaging for verification of single‐cell deposition was subcloned using validated FACS and imaging methods. A total of 46 subclones were analyzed for growth, productivity, product quality, copy number, and integration site analysis. Significant diversity in cell growth, protein productivity, product quality attributes, and copy number was observed between the subclones, despite stability of the parent clone over time. The diversity in protein productivity and quality of the subclones were reproduced across time and production scales, suggesting that the resulting population post sub‐cloning originating from a single cell is stable but with unique properties. Overall, this work demonstrates that the characteristics of isolated subclones are not predictive of a clonally derived parental clone. Consequently, the analysis of subclones may not be an effective approach to demonstrate clonal origin of a cell bank. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:613–623, 2018
Collapse
Affiliation(s)
- Tharmala Tharmalingam
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Hedieh Barkhordarian
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Nicole Tejeda
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Kristi Daris
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Sam Yaghmour
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Pheng Yam
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Fang Lu
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Chetan Goudar
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Trent Munro
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| | - Jennitte Stevens
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320
| |
Collapse
|
46
|
Amann T, Schmieder V, Faustrup Kildegaard H, Borth N, Andersen MR. Genetic engineering approaches to improve posttranslational modification of biopharmaceuticals in different production platforms. Biotechnol Bioeng 2019; 116:2778-2796. [PMID: 31237682 DOI: 10.1002/bit.27101] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/27/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
The number of approved biopharmaceuticals, where product quality attributes remain of major importance, is increasing steadily. Within the available variety of expression hosts, the production of biopharmaceuticals faces diverse limitations with respect to posttranslational modifications (PTM), while different biopharmaceuticals demand different forms and specifications of PTMs for proper functionality. With the growing toolbox of genetic engineering technologies, it is now possible to address general as well as host- or biopharmaceutical-specific product quality obstacles. In this review, we present diverse expression systems derived from mammalians, bacteria, yeast, plants, and insects as well as available genetic engineering tools. We focus on genes for knockout/knockdown and overexpression for meaningful approaches to improve biopharmaceutical PTMs and discuss their applicability as well as future trends in the field.
Collapse
Affiliation(s)
- Thomas Amann
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Valerie Schmieder
- acib GmbH-Austrian Centre of Industrial Biotechnology, Graz, Austria.,Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nicole Borth
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Mikael Rørdam Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
47
|
Human Pluripotent Stem Cells: Applications and Challenges for Regenerative Medicine and Disease Modeling. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 171:189-224. [PMID: 31740987 DOI: 10.1007/10_2019_117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, human pluripotent stem (hPS) cells have started to emerge as a potential tool with application in fields such as regenerative medicine, disease modeling, and drug screening. In particular, the ability to differentiate human-induced pluripotent stem (hiPS) cells into different cell types and to mimic structures and functions of a specific target organ, resourcing to organoid technology, has introduced novel model systems for disease recapitulation while offering a powerful tool to provide a faster and reproducible approach in the process of drug discovery. All these technologies are expected to improve the overall quality of life of the humankind. Here, we highlight the main applications of hiPS cells and the main challenges associated with the translation of hPS cell derivatives into clinical settings and other biomedical applications, such as the costs of the process and the ability to mimic the complexity of the in vivo systems. Moreover, we focus on the bioprocessing approaches that can be applied towards the production of high numbers of cells as well as their efficient differentiation into the final product and further purification.
Collapse
|
48
|
Bandyopadhyay AA, O’Brien SA, Zhao L, Fu HY, Vishwanathan N, Hu WS. Recurring genomic structural variation leads to clonal instability and loss of productivity. Biotechnol Bioeng 2019; 116:41-53. [PMID: 30144379 PMCID: PMC7058117 DOI: 10.1002/bit.26823] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/13/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
Abstract
Chinese hamster ovary cells, commonly used in the production of therapeutic proteins, are aneuploid. Their chromosomes bear structural abnormality and undergo changes in structure and number during cell proliferation. Some production cell lines are unstable and lose their productivity over time in the manufacturing process and during the product's life cycle. To better understand the link between genomic structural changes and productivity stability, an immunoglobulin G producing cell line was successively single-cell cloned to obtain subclones that retained or lost productivity, and their genomic features were compared. Although each subclone started with a single karyotype, the progeny quickly diversified to a population with a distribution of chromosome numbers that is not distinctive from the parent and among subclones. The comparative genomic hybridization (CGH) analysis showed that the extent of copy variation of gene coding regions among different subclones stayed at levels of a few percent. Genome regions that were prone to loss of copies, including one with a product transgene integration site, were identified in CGH. The loss of the transgene copy was accompanied by loss of transgene transcript level. Sequence analysis of the host cell and parental producing cell showed prominent structural variations within the regions prone to loss of copies. Taken together, we demonstrated the transient nature of clonal homogeneity in cell line development and the retention of a population distribution of chromosome numbers; we further demonstrated that structural variation in the transgene integration region caused cell line instability. Future cell line development may target the transgene into structurally stable regions.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue SE, Minneapolis, MN 55455-0132 USA
| |
Collapse
|
49
|
|
50
|
Semple SL, Vo NTK, Poynter SJ, Li M, Heath DD, DeWitte-Orr SJ, Dixon B. Extracellular dsRNA induces a type I interferon response mediated via class A scavenger receptors in a novel Chinook salmon derived spleen cell line. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:93-101. [PMID: 30118734 DOI: 10.1016/j.dci.2018.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 06/08/2023]
Abstract
Despite increased global interest in Chinook salmon aquaculture, little is known of their viral immune defenses. This study describes the establishment and characterization of a continuous cell line derived from Chinook salmon spleen, CHSS, and its use in innate immune studies. Optimal growth was seen at 14-18 °C when grown in Leibovitz's L-15 media with 20% fetal bovine serum. DNA analyses confirmed that CHSS was Chinook salmon and genetically different from the only other available Chinook salmon cell line, CHSE-214. Unlike CHSE-214, CHSS could bind extracellular dsRNA, resulting in the rapid and robust expression of antiviral genes. Receptor/ligand blocking assays confirmed that class A scavenger receptors (SR-A) facilitated dsRNA binding and subsequent gene expression. Although both cell lines expressed three SR-A genes: SCARA3, SCARA4, and SCARA5, only CHSS appeared to have functional cell-surface SR-As for dsRNA. Collectively, CHSS is an excellent cell model to study dsRNA-mediated innate immunity in Chinook salmon.
Collapse
Affiliation(s)
- S L Semple
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, N2L 3G1, Canada
| | - N T K Vo
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, N2L 3G1, Canada
| | - S J Poynter
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, N2L 3G1, Canada
| | - M Li
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, N2L 3G1, Canada
| | - D D Heath
- Great Lakes Institute of Environmental Research, University of Windsor, 2990 Riverside Drive, West Windsor, Ontario, N9C 1A2, Canada
| | - S J DeWitte-Orr
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario, N2L 3C5, Canada; Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario, N2L 3C5, Canada
| | - B Dixon
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|