1
|
Henry M, Selvaprakash K, Meleady P. Phosphopeptide Enrichment and LC-MS/MS Analysis to Study the Phosphoproteome of Recombinant Chinese Hamster Ovary Cells. Methods Mol Biol 2025; 2853:173-189. [PMID: 39460921 DOI: 10.1007/978-1-0716-4104-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
The reversible phosphorylation of proteins on serine, threonine, and tyrosine residues is one of the most important post-translational modifications that regulates many biological processes. There have been relatively few studies on the phosphoproteome of recombinant Chinese hamster ovary (CHO) cells to date despite phosphorylation playing a crucial role in regulating many molecular and cellular processes relevant to bioprocess phenotypes including, for example, transcription, translation, growth, apoptosis, and signal transduction. In this chapter, we provide a protocol for phosphoproteomic analysis of CHO cells using phosphopeptide enrichment with metal oxide affinity chromatography (MOAC) and immobilized metal affinity chromatography (IMAC) techniques, followed by site-specific identification of phosphorylated residues using liquid chromatography mass spectrometry (LC-MS), multistage activation (MSA), and MS3 strategies. This protocol can also be used for quantitative phosphoproteomic analysis using both labeled and label-free approaches.
Collapse
Affiliation(s)
- Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| | | | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
2
|
Kajihara R, Ezaki R, Watanabe T, Ichikawa K, Matsuzaki M, Horiuchi H. Evaluation of expression systems for recombinant protein production in chicken egg bioreactors. Biotechnol J 2024; 19:e2300316. [PMID: 37859508 DOI: 10.1002/biot.202300316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 10/21/2023]
Abstract
Chicken eggs have gained attention as excellent bioreactors because of their genetic modifications. However, the development of chicken egg bioreactors requires a long time from the construction of the production system to the evaluation of the products. Therefore, in this study, a chicken cell line producing ovalbumin (OVA) was established and constructed a system for the rapid evaluation of the production system. First, the EF1α promoter was knocked in upstream of the OVA locus in chicken DF-1 cells for continuous OVA expression. Furthermore, an ideal position at the OVA locus for the insertion of useful protein genes to maximize recombinant protein yield was analyzed and identified. The knocking in the EF1α promoter upstream of exon1 yielded the maximum production of OVA protein was achieved. In addition, Linking a recombinant hFGF2 cDNA to the 5' side of the OVA was found to increase production efficiency. Therefore, an OVA-expressing cell line and an evaluation system for proteins in chicken egg bioreactors was established. The findings may improve the efficiency of chicken expression systems and expand their applications in protein production.
Collapse
Affiliation(s)
- Ryota Kajihara
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Ryo Ezaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Tenkai Watanabe
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Kennosuke Ichikawa
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Mei Matsuzaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Hiroyuki Horiuchi
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
3
|
Masson HO, Karottki KJLC, Tat J, Hefzi H, Lewis NE. From observational to actionable: rethinking omics in biologics production. Trends Biotechnol 2023; 41:1127-1138. [PMID: 37062598 PMCID: PMC10524802 DOI: 10.1016/j.tibtech.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023]
Abstract
As the era of omics continues to expand with increasing ubiquity and success in both academia and industry, omics-based experiments are becoming commonplace in industrial biotechnology, including efforts to develop novel solutions in bioprocess optimization and cell line development. Omic technologies provide particularly valuable 'observational' insights for discovery science, especially in academic research and industrial R&D; however, biomanufacturing requires a different paradigm to unlock 'actionable' insights from omics. Here, we argue the value of omic experiments in biotechnology can be maximized with deliberate selection of omic approaches and forethought about analysis techniques. We describe important considerations when designing and implementing omic-based experiments and discuss how systems biology analysis strategies can enhance efforts to obtain actionable insights in mammalian-based biologics production.
Collapse
Affiliation(s)
- Helen O Masson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Jasmine Tat
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Amgen Inc., Thousand Oaks, CA, USA
| | | | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Meade E, Rowan N, Garvey M. Bioprocessing and the Production of Antiviral Biologics in the Prevention and Treatment of Viral Infectious Disease. Vaccines (Basel) 2023; 11:992. [PMID: 37243096 PMCID: PMC10223144 DOI: 10.3390/vaccines11050992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Emerging, re-emerging and zoonotic viral pathogens represent a serious threat to human health, resulting in morbidity, mortality and potentially economic instability at a global scale. Certainly, the recent emergence of the novel SARS-CoV-2 virus (and its variants) highlighted the impact of such pathogens, with the pandemic creating unprecedented and continued demands for the accelerated production of antiviral therapeutics. With limited effective small molecule therapies available for metaphylaxis, vaccination programs have been the mainstay against virulent viral species. Traditional vaccines remain highly effective at providing high antibody titres, but are, however, slow to manufacture in times of emergency. The limitations of traditional vaccine modalities may be overcome by novel strategies, as outlined herein. To prevent future disease outbreaks, paradigm shift changes in manufacturing and distribution are necessary to advance the production of vaccines, monoclonal antibodies, cytokines and other antiviral therapies. Accelerated paths for antivirals have been made possible due to advances in bioprocessing, leading to the production of novel antiviral agents. This review outlines the role of bioprocessing in the production of biologics and advances in mitigating viral infectious disease. In an era of emerging viral diseases and the proliferation of antimicrobial resistance, this review provides insight into a significant method of antiviral agent production which is key to protecting public health.
Collapse
Affiliation(s)
- Elaine Meade
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| | - Neil Rowan
- Bioscience Research Institute, Technical University Shannon Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
5
|
Rahimi A, Karimipoor M, Mahdian R, Alipour A, Hosseini S, Mohammadi M, Kaghazian H, Abbasi A, Shahsavarani H, Shokrgozar MA. Efficient CRISPR/Cas9-Mediated BAX Gene Ablation in CHO Cells To Impair Apoptosis and Enhance Recombinant Protein Production. IRANIAN JOURNAL OF BIOTECHNOLOGY 2023; 21:e3388. [PMID: 37228627 PMCID: PMC10203183 DOI: 10.30498/ijb.2023.343428.3388] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 01/25/2023] [Indexed: 05/27/2023]
Abstract
Background Despite recent advances in recombinant biotherapeutics production using CHO cells, their productivity remains lower than industrial needs, mainly due to apoptosis. Objectives Present study aimed to exploit CRISPR/Cas9 technology to specifically disrupt the BAX gene to attenuate apoptosis in recombinant Chinese hamster's ovary cells producing erythropoietin. Materials and Methods The STRING database was used to identify the key pro-apoptotic genes to be modified by CRISPR/Cas9 technique. The single guide RNAs (sgRNAs) targeting identified gene (BAX) were designed, and CHO cells were then transfected with vectors. Afterward, changes in the expression of the Bax gene and consequent production rates of erythropoietin were investigated in manipulated cells, even in the presence of an apoptosis inducer agent, oleuropein. Results BAX disruption significantly prolonged cell viability and increased proliferation rate in manipulated clones (152%, P-value = 0.0002). This strategy reduced the levels of Bax protein expression in manipulated cells by more than 4.3-fold (P-value <0.0001). The Bax-8 manipulated cells displayed higher threshold tolerance to the stress and consequence apoptosis compared to the control group. Also, they exhibited a higher IC50 compared to the control in the presence of oleuropein (5095 µM.ml-1 Vs. 2505 µM.ml-1). We found a significant increase in recombinant protein production levels in manipulated cells, even in the presence of 1,000 µM oleuropein compared to the control cell line (p-value=0.0002). Conclusions CRISPR/Cas9 assisted BAX gene ablation is promising to improve erythropoietin production in CHO cells via engineering anti-apoptotic genes. Therefore, exploiting genome editing tools such as CRISPR/Cas9 has been proposed to develop host cells that result in a safe, feasible, and robust manufacturing operation with a yield that meets the industrial requirements.
Collapse
Affiliation(s)
- Amirabbas Rahimi
- Laboratory of regenerative medicine and biomedical innovations, National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Reza Mahdian
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Atefeh Alipour
- Department of Nano-Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Saadi Hosseini
- Laboratory of regenerative medicine and biomedical innovations, National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Marzieh Mohammadi
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Hooman Kaghazian
- Department of Research & Development, Production & Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Abdolrahim Abbasi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Ali Shokrgozar
- Laboratory of regenerative medicine and biomedical innovations, National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Garvey M. Non-Mammalian Eukaryotic Expression Systems Yeast and Fungi in the Production of Biologics. J Fungi (Basel) 2022; 8:1179. [PMID: 36354946 PMCID: PMC9692369 DOI: 10.3390/jof8111179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 08/26/2023] Open
Abstract
Biologics have become an important area of medical research generating therapeutics essential for the treatment of many disease states. Biologics are defined as biologically active compounds manufactured by living cells or through biological processes termed bioprocessing. Compared to small molecules which are chemically synthesised they are relatively complex and therapeutically specific molecules. Biologics include hormones, vaccines, blood products, monoclonal antibodies, recombinant therapeutic proteins, enzymes, gene and cellular therapies amongst others. For biologic production prokaryotic and eukaryotic cells (mammalian and non-mammalian) are used as expression systems. Eukaryotic expression systems offer many advantages over prokaryotic based systems. The manufacture of high-quality proteins for human clinical use via recombinant technologies has been achieved in yeast and filamentous fungal systems. Advances in bioprocessing such as genetic engineering, bioreactor design, continuous processing, and quality by design has allowed for increased productivity and higher yield in in these non-mammalian eukaryotic systems with protein translation similar to mammalian systems. The application of eukaryotic expressions systems for the manufacture of biologics of therapeutic importance are described herein.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland; ; Tel.: +353-071-9305529
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
7
|
Komuczki D, Stadermann A, Bentele M, Unsoeld A, Grillari J, Mueller MM, Paul A, Fischer S. High cysteine concentrations in cell culture media lead to oxidative stress and reduced bioprocess performance of recombinant CHO cells. Biotechnol J 2022; 17:e2200029. [PMID: 35876277 DOI: 10.1002/biot.202200029] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022]
Abstract
Cysteine is considered an essential amino acid in the cultivation of Chinese hamster ovary (CHO) cells. An optimized cysteine supply during fed-batch cultivation supports the protein production capacity of recombinant CHO cell lines. However, we observed that CHO production cell lines seeded at low cell densities in chemically defined media enriched with, cysteine greater than 2.5 mM resulted in markedly reduced cell growth during passaging, hampering seed train performance and scale-up. To investigate the underlying mechanism, seeding cell densities and initial cysteine concentrations ranging from low to high cysteine concentrations were varied followed by an analysis of cell culture performance. Additionally, cell cycle analysis, intracellular quantification of reactive oxygen species (ROS) as well as transcriptomic analyses by next-generation sequencing were carried out. Our results demonstrate that CHO cells seeded at low cell densities at high initial cysteine concentrations encountered increased oxidative stress leading to a p21-mediated cell cycle arrest in the G1/S phase. The resulting oxidative stress caused redox imbalance in the endoplasmic reticulum and activation of the unfolded protein response as well as the major antioxidant nuclear factor-like 2 response pathways. We were able to identify potential signature genes related to oxidative stress and the inhibition of the pentose phosphate pathway. Finally, we present that seeding cells at a higher concentration counteract oxidative stress in cysteine-enriched cell culture media. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Daniel Komuczki
- Institute of Bioprocess Sciences and Engineering (IBSE), University of Natural Resources and Life Sciences, Muthgasse 18, AT-1190, Vienna, Austria.,Upstream Development, Bioprocess and Analytical Development, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Anna Stadermann
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Maximilian Bentele
- Upstream Development, Bioprocess and Analytical Development, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Andreas Unsoeld
- Cell Culture Media Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Johannes Grillari
- Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, AT-1190, Austria.,Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, AT-1200, Austria
| | - Markus M Mueller
- Upstream Development, Bioprocess and Analytical Development, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Albert Paul
- Upstream Development, Bioprocess and Analytical Development, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| | - Simon Fischer
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co.KG, Birkendorfer Strasse 65, D-88397 Biberach a. d. Riss, Germany
| |
Collapse
|
8
|
Park JU, Han HJ, Baik JY. Energy metabolism in Chinese hamster ovary (CHO) cells: Productivity and beyond. KOREAN J CHEM ENG 2022. [DOI: 10.1007/s11814-022-1062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
9
|
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: 1.3] [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
|
10
|
Karottki KJLC, Hefzi H, Li S, Pedersen LE, Spahn PN, Joshi C, Ruckerbauer D, Bort JAH, Thomas A, Lee JS, Borth N, Lee GM, Kildegaard HF, Lewis NE. A metabolic CRISPR-Cas9 screen in Chinese hamster ovary cells identifies glutamine-sensitive genes. Metab Eng 2021; 66:114-122. [PMID: 33813034 DOI: 10.1016/j.ymben.2021.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 12/20/2022]
Abstract
Media and feed optimization have fueled many-fold improvements in mammalian biopharmaceutical production, but genome editing offers an emerging avenue for further enhancing cell metabolism and bioproduction. However, the complexity of metabolism, involving thousands of genes, makes it unclear which engineering strategies will result in desired traits. Here we present a comprehensive pooled CRISPR screen for CHO cell metabolism, including ~16,000 gRNAs against ~2500 metabolic enzymes and regulators. Using this screen, we identified a glutamine response network in CHO cells. Glutamine is particularly important since it is often over-fed to drive increased TCA cycle flux, but toxic ammonia may accumulate. With the screen we found one orphan glutamine-responsive gene with no clear connection to our network. Knockout of this novel and poorly characterized lipase, Abhd11, substantially increased growth in glutamine-free media by altering the regulation of the TCA cycle. Thus, the screen provides an invaluable targeted platform to comprehensively study genes involved in any metabolic trait, and elucidate novel regulators of metabolism.
Collapse
Affiliation(s)
| | - Hooman Hefzi
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, USA; Department of Pediatrics, University of California, San Diego, USA; Department of Bioengineering, University of California, San Diego, USA
| | - Songyuan Li
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Lasse Ebdrup Pedersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Philipp N Spahn
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, USA; Department of Pediatrics, University of California, San Diego, USA
| | - Chintan Joshi
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, USA; Department of Pediatrics, University of California, San Diego, USA
| | - David Ruckerbauer
- Austrian Centre of Industrial Biotechnology, Vienna, Austria; University of Natural Resources and Life Sciences, Vienna, Austria
| | | | - Alex Thomas
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, USA
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Nicole Borth
- Austrian Centre of Industrial Biotechnology, Vienna, Austria; University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gyun Min Lee
- Department of Biological Sciences, Kaist, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 305-701, Republic of Korea
| | | | - Nathan E Lewis
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, USA; Department of Pediatrics, University of California, San Diego, USA; Department of Bioengineering, University of California, San Diego, USA; National Biologics Facility, Technical University of Denmark, Denmark.
| |
Collapse
|
11
|
Schinn SM, Morrison C, Wei W, Zhang L, Lewis NE. Systematic evaluation of parameters for genome-scale metabolic models of cultured mammalian cells. Metab Eng 2021; 66:21-30. [PMID: 33771719 DOI: 10.1016/j.ymben.2021.03.013] [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: 07/31/2020] [Revised: 11/25/2020] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Genome-scale metabolic models describe cellular metabolism with mechanistic detail. Given their high complexity, such models need to be parameterized correctly to yield accurate predictions and avoid overfitting. Effective parameterization has been well-studied for microbial models, but it remains unclear for higher eukaryotes, including mammalian cells. To address this, we enumerated model parameters that describe key features of cultured mammalian cells - including cellular composition, bioprocess performance metrics, mammalian-specific pathways, and biological assumptions behind model formulation approaches. We tested these parameters by building thousands of metabolic models and evaluating their ability to predict the growth rates of a panel of phenotypically diverse Chinese Hamster Ovary cell clones. We found the following considerations to be most critical for accurate parameterization: (1) cells limit metabolic activity to maintain homeostasis, (2) cell morphology and viability change dynamically during a growth curve, and (3) cellular biomass has a particular macromolecular composition. Depending on parameterization, models predicted different metabolic phenotypes, including contrasting mechanisms of nutrient utilization and energy generation, leading to varying accuracies of growth rate predictions. Notably, accurate parameter values broadly agreed with experimental measurements. These insights will guide future investigations of mammalian metabolism.
Collapse
Affiliation(s)
- Song-Min Schinn
- Department of Pediatrics, University of California, San Diego, USA
| | - Carly Morrison
- Pfizer, Biotherapeutics Pharmaceutical Sciences, Andover, MA, USA
| | - Wei Wei
- Pfizer, Biotherapeutics Pharmaceutical Sciences, Andover, MA, USA
| | - Lin Zhang
- Pfizer, Biotherapeutics Pharmaceutical Sciences, Andover, MA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, USA; Department of Bioengineering, University of California, San Diego, USA; Novo Nordisk Foundation Center for Biosustainability at UC, San Diego, USA.
| |
Collapse
|
12
|
Bryan L, Henry M, Kelly RM, Lloyd M, Frye CC, Osborne MD, Clynes M, Meleady P. Global phosphoproteomic study of high/low specific productivity industrially relevant mAb producing recombinant CHO cell lines. CURRENT RESEARCH IN BIOTECHNOLOGY 2021. [DOI: 10.1016/j.crbiot.2021.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
13
|
Development of a non-viral platform for rapid virus-like particle production in Sf9 cells. J Biotechnol 2020; 322:43-53. [DOI: 10.1016/j.jbiotec.2020.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/31/2020] [Accepted: 07/11/2020] [Indexed: 11/21/2022]
|
14
|
Huang Z, Xu J, Yongky A, Morris CS, Polanco AL, Reily M, Borys MC, Li ZJ, Yoon S. CHO cell productivity improvement by genome-scale modeling and pathway analysis: Application to feed supplements. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
15
|
Gutierrez JM, Feizi A, Li S, Kallehauge TB, Hefzi H, Grav LM, Ley D, Baycin Hizal D, Betenbaugh MJ, Voldborg B, Faustrup Kildegaard H, Min Lee G, Palsson BO, Nielsen J, Lewis NE. Genome-scale reconstructions of the mammalian secretory pathway predict metabolic costs and limitations of protein secretion. Nat Commun 2020; 11:68. [PMID: 31896772 PMCID: PMC6940358 DOI: 10.1038/s41467-019-13867-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/22/2019] [Indexed: 01/08/2023] Open
Abstract
In mammalian cells, >25% of synthesized proteins are exported through the secretory pathway. The pathway complexity, however, obfuscates its impact on the secretion of different proteins. Unraveling its impact on diverse proteins is particularly important for biopharmaceutical production. Here we delineate the core secretory pathway functions and integrate them with genome-scale metabolic reconstructions of human, mouse, and Chinese hamster ovary cells. The resulting reconstructions enable the computation of energetic costs and machinery demands of each secreted protein. By integrating additional omics data, we find that highly secretory cells have adapted to reduce expression and secretion of other expensive host cell proteins. Furthermore, we predict metabolic costs and maximum productivities of biotherapeutic proteins and identify protein features that most significantly impact protein secretion. Finally, the model successfully predicts the increase in secretion of a monoclonal antibody after silencing a highly expressed selection marker. This work represents a knowledgebase of the mammalian secretory pathway that serves as a novel tool for systems biotechnology.
Collapse
Affiliation(s)
- Jahir M Gutierrez
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Amir Feizi
- Department of Biology and Biological Engineering, Kemivägen 10, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Shangzhong Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Thomas B Kallehauge
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Lise M Grav
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Daniel Ley
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Department of Systems Biology, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Michael J Betenbaugh
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218-2686, USA
| | - Bjorn Voldborg
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Helene Faustrup Kildegaard
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Gyun Min Lee
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Kemivägen 10, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Mitigating Clonal Variation in Recombinant Mammalian Cell Lines. Trends Biotechnol 2019; 37:931-942. [DOI: 10.1016/j.tibtech.2019.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/27/2022]
|
17
|
Combating viral contaminants in CHO cells by engineering innate immunity. Sci Rep 2019; 9:8827. [PMID: 31222165 PMCID: PMC6586939 DOI: 10.1038/s41598-019-45126-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022] Open
Abstract
Viral contamination in biopharmaceutical manufacturing can lead to shortages in the supply of critical therapeutics. To facilitate the protection of bioprocesses, we explored the basis for the susceptibility of CHO cells to RNA virus infection. Upon infection with certain ssRNA and dsRNA viruses, CHO cells fail to generate a significant interferon (IFN) response. Nonetheless, the downstream machinery for generating IFN responses and its antiviral activity is intact in these cells: treatment of cells with exogenously-added type I IFN or poly I:C prior to infection limited the cytopathic effect from Vesicular stomatitis virus (VSV), Encephalomyocarditis virus (EMCV), and Reovirus-3 virus (Reo-3) in a STAT1-dependent manner. To harness the intrinsic antiviral mechanism, we used RNA-Seq to identify two upstream repressors of STAT1: Gfi1 and Trim24. By knocking out these genes, the engineered CHO cells exhibited activation of cellular immune responses and increased resistance to the RNA viruses tested. Thus, omics-guided engineering of mammalian cell culture can be deployed to increase safety in biotherapeutic protein production among many other biomedical applications.
Collapse
|
18
|
Pohlscheidt M, Kiss R, Gottschalk U. An Introduction to "Recent Trends in the Biotechnology Industry: Development and Manufacturing of Recombinant Antibodies and Proteins". ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:1-8. [PMID: 29748871 DOI: 10.1007/10_2017_39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The production of the first therapeutic proteins in the early 1980s heralded the launch of the biopharmaceuticals industry. The number of approved products has grown year on year over the past three decades to now represent a significant share of the entire pharmaceuticals market. More than 200 therapeutic proteins have been approved, approximately a quarter of which are represented by monoclonal antibodies and their derivatives. In 2016, the list of the top 15 best-selling drugs included more than eight biologics and in 2020 the trend will continue, with more than 50% of the top 20 best-selling drugs predicted to be biologics. From 1986 to 2014 several first-in-class, advance-in-class, and breakthrough designated therapeutic options were approved, with advanced therapies such as immuno-oncology and cell-based therapies being approved for several indications.
Collapse
Affiliation(s)
| | - Robert Kiss
- Biogen International GmbH, International Manufacturing, Zug, Switzerland
| | - Uwe Gottschalk
- Biogen International GmbH, International Manufacturing, Zug, Switzerland
| |
Collapse
|
19
|
Zhu J, Hatton D. New Mammalian Expression Systems. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:9-50. [PMID: 28585079 DOI: 10.1007/10_2016_55] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are an increasing number of recombinant antibodies and proteins in preclinical and clinical development for therapeutic applications. Mammalian expression systems are key to enabling the production of these molecules, and Chinese hamster ovary (CHO) cell platforms continue to be central to delivery of the stable cell lines required for large-scale production. Increasing pressure on timelines and efficiency, further innovation of molecular formats and the shift to new production systems are driving developments of these CHO cell line platforms. The availability of genome and transcriptome data coupled with advancing gene editing tools are increasing the ability to design and engineer CHO cell lines to meet these challenges. This chapter aims to give an overview of the developments in CHO expression systems and some of the associated technologies over the past few years.
Collapse
Affiliation(s)
- Jie Zhu
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Diane Hatton
- MedImmune, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
20
|
Pristovšek N, Nallapareddy S, Grav LM, Hefzi H, Lewis NE, Rugbjerg P, Hansen HG, Lee GM, Andersen MR, Kildegaard HF. Systematic Evaluation of Site-Specific Recombinant Gene Expression for Programmable Mammalian Cell Engineering. ACS Synth Biol 2019; 8:758-774. [PMID: 30807689 DOI: 10.1021/acssynbio.8b00453] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many branches of biology depend on stable and predictable recombinant gene expression, which has been achieved in recent years through targeted integration of the recombinant gene into defined integration sites. However, transcriptional levels of recombinant genes in characterized integration sites are controlled by multiple components of the integrated expression cassette. Lack of readily available tools has inhibited meaningful experimental investigation of the interplay between the integration site and the expression cassette components. Here we show in a systematic manner how multiple components contribute to final net expression of recombinant genes in a characterized integration site. We develop a CRISPR/Cas9-based toolbox for construction of mammalian cell lines with targeted integration of a landing pad, containing a recombinant gene under defined 5' proximal regulatory elements. Generated site-specific recombinant cell lines can be used in a streamlined recombinase-mediated cassette exchange for fast screening of different expression cassettes. Using the developed toolbox, we show that different 5' proximal regulatory elements generate distinct and robust recombinant gene expression patterns in defined integration sites of CHO cells with a wide range of transcriptional outputs. This approach facilitates the generation of user-defined and product-specific gene expression patterns for programmable mammalian cell engineering.
Collapse
Affiliation(s)
- Nuša Pristovšek
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Saranya Nallapareddy
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Lise Marie Grav
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Hooman Hefzi
- Departments of Pediatrics and Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, La Jolla, California 92093, United States
| | - Nathan E. Lewis
- Departments of Pediatrics and Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, La Jolla, California 92093, United States
| | - Peter Rugbjerg
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Henning Gram Hansen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Gyun Min Lee
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
- Department of Biological Sciences, KAIST, 291 Daehak-ro,
Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Mikael Rørdam Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 223, 2800 Kgs. Lyngby, Denmark
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
21
|
Zeng H, Yang A. Quantification of proteomic and metabolic burdens predicts growth retardation and overflow metabolism in recombinant Escherichia coli. Biotechnol Bioeng 2019; 116:1484-1495. [PMID: 30712260 DOI: 10.1002/bit.26943] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/17/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
Abstract
Escherichia coli has been the host organism most frequently investigated for efficient recombinant protein production. However, the production of a foreign protein in recombinant E. coli often leads to growth deterioration and elevated secretion of acetic acid. Such observed phenomena have been widely linked with cell stress responses and metabolic burdens originated particularly from the increased energy demand. In this study, flux balance analysis and dynamic flux balance analysis were applied to investigate the observed growth physiology of recombinant E. coli, incorporating the proteome allocation theory and an adjustable maintenance energy level (ATPM) to capture the proteomic and energetic burdens introduced by recombinant protein synthesis. Model predictions of biomass growth, substrate consumption, acetate excretion, and protein production with two different strains were in good agreement with the experimental data, indicating that the constraint on the available proteomic resource and the change in ATPM might be important contributors governing the growth physiology of recombinant strains. The modeling framework developed in this work, currently with several limitations to overcome, offers a starting point for the development of a practical, model-based tool to guide metabolic engineering decisions for boosting recombinant protein production.
Collapse
Affiliation(s)
- Hong Zeng
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Kaushik P, Henry M, Clynes M, Meleady P. The Expression Pattern of the Phosphoproteome Is Significantly Changed During the Growth Phases of Recombinant CHO Cell Culture. Biotechnol J 2018; 13:e1700221. [DOI: 10.1002/biot.201700221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/13/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Prashant Kaushik
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| |
Collapse
|
23
|
Fouladiha H, Marashi SA, Shokrgozar MA, Farokhi M, Atashi A. Applications of a metabolic network model of mesenchymal stem cells for controlling cell proliferation and differentiation. Cytotechnology 2018; 70:331-338. [PMID: 28980092 PMCID: PMC5809662 DOI: 10.1007/s10616-017-0148-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from several tissues of adults. In addition, MSCs have the potential of differentiation into several cell types. Therefore, MSCs are very useful in stem cell therapy and regenerative medicine. MSCs have also been used as gene or protein carriers. As a result, maintaining MSCs in a desirable metabolic state has been the subject of several studies. Here, we used a genome scale metabolic network model of bone marrow derived MSCs for exploring the metabolism of these cells. We analyzed metabolic fluxes of the model in order to find ways of increasing stem cell proliferation and differentiation. Consequently, the experimental results were in consistency with computational results. Therefore, analyzing metabolic models was proven to be a promising field in biomedical researches of stem cells.
Collapse
Affiliation(s)
- Hamideh Fouladiha
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
24
|
Stolfa G, Smonskey MT, Boniface R, Hachmann AB, Gulde P, Joshi AD, Pierce AP, Jacobia SJ, Campbell A. CHO-Omics Review: The Impact of Current and Emerging Technologies on Chinese Hamster Ovary Based Bioproduction. Biotechnol J 2017; 13:e1700227. [PMID: 29072373 DOI: 10.1002/biot.201700227] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023]
Abstract
CHO cells are the most prevalent platform for modern bio-therapeutic production. Currently, there are several CHO cell lines used in bioproduction with distinct characteristics and unique genotypes and phenotypes. These differences limit advances in productivity and quality that can be achieved by the most common approaches to bioprocess optimization and cell line engineering. Incorporating omics-based approaches into current bioproduction processes will complement traditional methodologies to maximize gains from CHO engineering and bioprocess improvements. In order to highlight the utility of omics technologies in CHO bioproduction, the authors discuss current applications as well as limitations of genomics, transcriptomics, proteomics, metabolomics, lipidomics, fluxomics, glycomics, and multi-omics approaches and the potential they hold for the future of bioproduction. Multiple omics approaches are currently being used to improve CHO bioprocesses; however, the application of these technologies is still limited. As more CHO-omic datasets become available and integrated into systems models, the authors expect significant gains in product yield and quality. While individual omics technologies provide incremental improvements in bioproduction, the authors will likely see the most significant gains by applying multi-omics and systems biology approaches to individual CHO cell lines.
Collapse
Affiliation(s)
- Gino Stolfa
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | | | - Ryan Boniface
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | | | - Paul Gulde
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | - Atul D Joshi
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | - Anson P Pierce
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | - Scott J Jacobia
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| | - Andrew Campbell
- Bioproduction R&D, Thermo Fisher Scientific, Grand Island, USA
| |
Collapse
|
25
|
Use of CellNetAnalyzer in biotechnology and metabolic engineering. J Biotechnol 2017; 261:221-228. [DOI: 10.1016/j.jbiotec.2017.05.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/28/2017] [Accepted: 05/03/2017] [Indexed: 01/28/2023]
|
26
|
Huang Z, Lee DY, Yoon S. Quantitative intracellular flux modeling and applications in biotherapeutic development and production using CHO cell cultures. Biotechnol Bioeng 2017; 114:2717-2728. [DOI: 10.1002/bit.26384] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 06/07/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Zhuangrong Huang
- Department of Chemical Engineering, University of Massachusetts Lowell; One University Avenue; Lowell Massachusetts
| | - Dong-Yup Lee
- Department of Chemical and Biomolecular Engineering; National University of Singapore; Singapore Singapore
- Bioprocessing Technology Institute; Agency for Science, Technology and Research (A*STAR); Singapore Singapore
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell; One University Avenue; Lowell Massachusetts
| |
Collapse
|
27
|
Zahrl RJ, Peña DA, Mattanovich D, Gasser B. Systems biotechnology for protein production in Pichia pastoris. FEMS Yeast Res 2017; 17:4093073. [DOI: 10.1093/femsyr/fox068] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/22/2017] [Indexed: 12/31/2022] Open
|
28
|
Richelle A, Lewis NE. Improvements in protein production in mammalian cells from targeted metabolic engineering. ACTA ACUST UNITED AC 2017; 6:1-6. [PMID: 29104947 DOI: 10.1016/j.coisb.2017.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bioprocess optimization has yielded powerful clones for biotherapeutic production. However, new genomic technologies allow more targeted approaches to cell line development. Here we review efforts to enhance protein production in mammalian cells through metabolic engineering. Most efforts aimed to reduce toxic byproducts accumulation to enhance protein productivity. However, recent work highlights the possibility of regulating other desirable traits (e.g., apoptosis and glycosylation) by targeting central metabolism since these processes are interconnected. Therefore, as we further detail the pathways underlying cell growth and protein production and deploy diverse algorithms for their analysis, opportunities will arise to move beyond simple cell line designs and facilitate cell engineering strategies with complex combinations of genes that together underlie a phenotype of interest.
Collapse
Affiliation(s)
- Anne Richelle
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.,Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Nathan E Lewis
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.,Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| |
Collapse
|
29
|
Henry M, Power M, Kaushik P, Coleman O, Clynes M, Meleady P. Differential Phosphoproteomic Analysis of Recombinant Chinese Hamster Ovary Cells Following Temperature Shift. J Proteome Res 2017; 16:2339-2358. [DOI: 10.1021/acs.jproteome.6b00868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Michael Henry
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Power
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Prashant Kaushik
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Orla Coleman
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular
Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
30
|
Guzmán-Trampe S, Ceapa CD, Manzo-Ruiz M, Sánchez S. Synthetic biology era: Improving antibiotic’s world. Biochem Pharmacol 2017; 134:99-113. [DOI: 10.1016/j.bcp.2017.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
|
31
|
Farzaneh M, Hassani SN, Mozdziak P, Baharvand H. Avian embryos and related cell lines: A convenient platform for recombinant proteins and vaccine production. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201600598] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/25/2017] [Accepted: 03/09/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
| | - Paul Mozdziak
- Graduate Physiology Program; Campus Box 7608/321 Scott Hall; Raleigh NC USA
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
- Department of Developmental Biology; University of Science and Culture; Tehran Iran
| |
Collapse
|
32
|
Hansen HG, Pristovšek N, Kildegaard HF, Lee GM. Improving the secretory capacity of Chinese hamster ovary cells by ectopic expression of effector genes: Lessons learned and future directions. Biotechnol Adv 2017; 35:64-76. [DOI: 10.1016/j.biotechadv.2016.11.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/12/2016] [Accepted: 11/28/2016] [Indexed: 12/12/2022]
|
33
|
Henry M, Coleman O, Clynes M, Meleady P. Phosphopeptide Enrichment and LC-MS/MS Analysis to Study the Phosphoproteome of Recombinant Chinese Hamster Ovary Cells. Methods Mol Biol 2017; 1603:195-208. [PMID: 28493132 DOI: 10.1007/978-1-4939-6972-2_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The reversible phosphorylation of proteins on serine, threonine, and tyrosine residues is one of the most important post-translational modifications that regulates many biological processes. The phosphoproteome has not been studied in any great detail in recombinant Chinese hamster ovary (CHO) cells to date despite phosphorylation playing a crucial role in regulating many molecular and cellular processes relevant to bioprocess phenotypes including, for example, transcription, translation, growth, apoptosis, and signal transduction. In this chapter, we provide a protocol for the phosphoproteomic analysis of Chinese hamster ovary cells using phosphopeptide enrichment with metal oxide affinity chromatography (MOAC) and immobilized metal affinity chromatography (IMAC) techniques, followed by site-specific identification of phosphorylated residues using LC-MS (MS2 and MS3) strategies.
Collapse
Affiliation(s)
- Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Orla Coleman
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
34
|
Hefzi H, Ang KS, Hanscho M, Bordbar A, Ruckerbauer D, Lakshmanan M, Orellana CA, Baycin-Hizal D, Huang Y, Ley D, Martinez VS, Kyriakopoulos S, Jiménez NE, Zielinski DC, Quek LE, Wulff T, Arnsdorf J, Li S, Lee JS, Paglia G, Loira N, Spahn PN, Pedersen LE, Gutierrez JM, King ZA, Lund AM, Nagarajan H, Thomas A, Abdel-Haleem AM, Zanghellini J, Kildegaard HF, Voldborg BG, Gerdtzen ZP, Betenbaugh MJ, Palsson BO, Andersen MR, Nielsen LK, Borth N, Lee DY, Lewis NE. A Consensus Genome-scale Reconstruction of Chinese Hamster Ovary Cell Metabolism. Cell Syst 2016; 3:434-443.e8. [PMID: 27883890 PMCID: PMC5132346 DOI: 10.1016/j.cels.2016.10.020] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/16/2016] [Accepted: 10/21/2016] [Indexed: 12/22/2022]
Abstract
Chinese hamster ovary (CHO) cells dominate biotherapeutic protein production and are widely used in mammalian cell line engineering research. To elucidate metabolic bottlenecks in protein production and to guide cell engineering and bioprocess optimization, we reconstructed the metabolic pathways in CHO and associated them with >1,700 genes in the Cricetulus griseus genome. The genome-scale metabolic model based on this reconstruction, iCHO1766, and cell-line-specific models for CHO-K1, CHO-S, and CHO-DG44 cells provide the biochemical basis of growth and recombinant protein production. The models accurately predict growth phenotypes and known auxotrophies in CHO cells. With the models, we quantify the protein synthesis capacity of CHO cells and demonstrate that common bioprocess treatments, such as histone deacetylase inhibitors, inefficiently increase product yield. However, our simulations show that the metabolic resources in CHO are more than three times more efficiently utilized for growth or recombinant protein synthesis following targeted efforts to engineer the CHO secretory pathway. This model will further accelerate CHO cell engineering and help optimize bioprocesses.
Collapse
Affiliation(s)
- Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kok Siong Ang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore; Bioprocessing Technology Institute, Agency for Science, Technology and Research (A(∗)STAR), 20 Biopolis Way, 06-01, Centros, Singapore 138668, Singapore
| | - Michael Hanscho
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190 Vienna, Austria; Austrian Centre of Industrial Biotechnology, 1190 Vienna, Austria
| | - Aarash Bordbar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - David Ruckerbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190 Vienna, Austria; Austrian Centre of Industrial Biotechnology, 1190 Vienna, Austria
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A(∗)STAR), 20 Biopolis Way, 06-01, Centros, Singapore 138668, Singapore
| | - Camila A Orellana
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Corner College and Cooper Roads (Building 75), Brisbane, QLD 4072, Australia
| | - Deniz Baycin-Hizal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yingxiang Huang
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla 92093, CA, USA
| | - Daniel Ley
- Department of Systems Biology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Veronica S Martinez
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Corner College and Cooper Roads (Building 75), Brisbane, QLD 4072, Australia
| | - Sarantos Kyriakopoulos
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A(∗)STAR), 20 Biopolis Way, 06-01, Centros, Singapore 138668, Singapore
| | - Natalia E Jiménez
- Centre for Biotechnology and Bioengineering, Department of Chemical Engineering and Biotechnology, University of Chile, Santiago 8370456, Chile; MATHomics, Center for Mathematical Modeling; Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago 8370456, Chile
| | - Daniel C Zielinski
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lake-Ee Quek
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Corner College and Cooper Roads (Building 75), Brisbane, QLD 4072, Australia
| | - Tune Wulff
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Johnny Arnsdorf
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Shangzhong Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jae Seong Lee
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Giuseppe Paglia
- Center for Systems Biology, University of Iceland, 101 Reykjavik, Iceland
| | - Nicolas Loira
- Centre for Biotechnology and Bioengineering, Department of Chemical Engineering and Biotechnology, University of Chile, Santiago 8370456, Chile; MATHomics, Center for Mathematical Modeling; Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago 8370456, Chile
| | - Philipp N Spahn
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lasse E Pedersen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jahir M Gutierrez
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zachary A King
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anne Mathilde Lund
- Department of Systems Biology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Harish Nagarajan
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla 92093, CA, USA
| | - Alex Thomas
- Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla 92093, CA, USA
| | - Alyaa M Abdel-Haleem
- Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Computational Bioscience Research Centre, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Juergen Zanghellini
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190 Vienna, Austria; Austrian Centre of Industrial Biotechnology, 1190 Vienna, Austria
| | - Helene F Kildegaard
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Bjørn G Voldborg
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Ziomara P Gerdtzen
- Centre for Biotechnology and Bioengineering, Department of Chemical Engineering and Biotechnology, University of Chile, Santiago 8370456, Chile; MATHomics, Center for Mathematical Modeling; Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago 8370456, Chile
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mikael R Andersen
- Department of Systems Biology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Lars K Nielsen
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Corner College and Cooper Roads (Building 75), Brisbane, QLD 4072, Australia
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190 Vienna, Austria; Austrian Centre of Industrial Biotechnology, 1190 Vienna, Austria.
| | - Dong-Yup Lee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore; Bioprocessing Technology Institute, Agency for Science, Technology and Research (A(∗)STAR), 20 Biopolis Way, 06-01, Centros, Singapore 138668, Singapore.
| | - Nathan E Lewis
- Novo Nordisk Foundation Center for Biosustainability at the School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
35
|
Chiang AW, Li S, Spahn PN, Richelle A, Kuo CC, Samoudi M, Lewis NE. Modulating carbohydrate-protein interactions through glycoengineering of monoclonal antibodies to impact cancer physiology. Curr Opin Struct Biol 2016; 40:104-111. [PMID: 27639240 DOI: 10.1016/j.sbi.2016.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/08/2016] [Accepted: 08/29/2016] [Indexed: 01/05/2023]
Abstract
Diverse glycans on proteins impact cell and organism physiology, along with drug activity. Since many protein-based biotherapeutics are glycosylated and these glycans have biological activity, there is a desire to engineer glycosylation for recombinant protein-based biotherapeutics. Engineered glycosylation can impact the recombinant protein efficacy and also influence many cell pathways by first changing glycan-protein interactions and consequently modulating disease physiologies. However, its complexity is enormous. Recent advances in glycoengineering now make it easier to modulate protein-glycan interactions. Here, we discuss how engineered glycans contribute to therapeutic monoclonal antibodies (mAbs) in the treatment of cancers, how these glycoengineered therapeutic mAbs affect the transformed phenotypes and downstream cell pathways. Furthermore, we suggest how systems biology can help in the next generation mAb glycoengineering process by aiding in data analysis and guiding engineering efforts to tailor mAb glycan and ultimately drug efficacy, safety and affordability.
Collapse
Affiliation(s)
- Austin Wt Chiang
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Shangzhong Li
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Philipp N Spahn
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Anne Richelle
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Chih-Chung Kuo
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Mojtaba Samoudi
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA.
| |
Collapse
|
36
|
Golabgir A, Gutierrez JM, Hefzi H, Li S, Palsson BO, Herwig C, Lewis NE. Quantitative feature extraction from the Chinese hamster ovary bioprocess bibliome using a novel meta-analysis workflow. Biotechnol Adv 2016; 34:621-633. [DOI: 10.1016/j.biotechadv.2016.02.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 02/21/2016] [Accepted: 02/28/2016] [Indexed: 01/01/2023]
|
37
|
Ritter A, Rauschert T, Oertli M, Piehlmaier D, Mantas P, Kuntzelmann G, Lageyre N, Brannetti B, Voedisch B, Geisse S, Jostock T, Laux H. Disruption of the gene C12orf35
leads to increased productivities in recombinant CHO cell lines. Biotechnol Bioeng 2016; 113:2433-42. [DOI: 10.1002/bit.26009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/08/2016] [Accepted: 05/09/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Anett Ritter
- Novartis Institutes for BioMedical Research; Basel Switzerland
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| | | | - Mevion Oertli
- Novartis Institutes for BioMedical Research; Basel Switzerland
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| | - Daniel Piehlmaier
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| | - Panagiotis Mantas
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| | | | - Nadine Lageyre
- Novartis Institutes for BioMedical Research; Basel Switzerland
| | | | - Bernd Voedisch
- Novartis Institutes for BioMedical Research; Basel Switzerland
| | - Sabine Geisse
- Novartis Institutes for BioMedical Research; Basel Switzerland
| | - Thomas Jostock
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| | - Holger Laux
- Novartis Pharma AG; Integrated Biologics Profiling Unit, Werk Klybeck Postfach CH-4002; Basel Switzerland
| |
Collapse
|
38
|
|