1
|
Hacker DL, Lau K, Durrer L, Quinche S, François M, Delisle L, Pojer F. Large-Scale Transient Transfection of Suspension-Adapted Chinese Hamster Ovary Cells for the Production of the Trimeric SARS-CoV-2 Spike Protein. Methods Mol Biol 2025; 2853:7-16. [PMID: 39460911 DOI: 10.1007/978-1-0716-4104-0_2] [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 production and purification of the secreted ectodomain of SARS-CoV-2 spike protein (S protein) were performed by transiently transfecting suspension-adapted Chinese hamster ovary cells (ExpiCHO). The method involved the separate addition of plasmid DNA expressing the S protein and polyethyleneimine to a suspension culture at a density of 5 × 106 cells/mL; and the subsequent addition of dimethyl sulfoxide at 2% (v/v). The transfected ExpiCHO cells were cultivated at 31 °C with agitation by orbital shaking under 5% CO2. On day six post-transfection, the culture was centrifuged, and the supernatant was filtered to remove cells and cell debris. Finally, the secreted recombinant S protein was recovered from the supernatant by a single step of affinity chromatography to the Twin-Strep-Tag of the recombinant S protein.
Collapse
Affiliation(s)
- David L Hacker
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Kelvin Lau
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Laurence Durrer
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Soraya Quinche
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Michael François
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ludovic Delisle
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Florence Pojer
- Protein Production and Structure Core Facility (PTPSP), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
2
|
Gabriel F, Spriestersbach L, Fuhrmann A, Jungnickel KEJ, Mostafavi S, Pardon E, Steyaert J, Löw C. Structural basis of thiamine transport and drug recognition by SLC19A3. Nat Commun 2024; 15:8542. [PMID: 39358356 PMCID: PMC11447181 DOI: 10.1038/s41467-024-52872-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Thiamine (vitamin B1) functions as an essential coenzyme in cells. Humans and other mammals cannot synthesise this vitamin de novo and thus have to take it up from their diet. Eventually, every cell needs to import thiamine across its plasma membrane, which is mainly mediated by the two specific thiamine transporters SLC19A2 and SLC19A3. Loss of function mutations in either of these transporters lead to detrimental, life-threatening metabolic disorders. SLC19A3 is furthermore a major site of drug interactions. Many medications, including antidepressants, antibiotics and chemotherapeutics are known to inhibit this transporter, with potentially fatal consequences for patients. Despite a thorough functional characterisation over the past two decades, the structural basis of its transport mechanism and drug interactions has remained elusive. Here, we report seven cryo-electron microscopy (cryo-EM) structures of the human thiamine transporter SLC19A3 in complex with various ligands. Conformation-specific nanobodies enable us to capture different states of SLC19A3's transport cycle, revealing the molecular details of thiamine recognition and transport. We identify seven previously unknown drug interactions of SLC19A3 and present structures of the transporter in complex with the inhibitors fedratinib, amprolium and hydroxychloroquine. These data allow us to develop an understanding of the transport mechanism and ligand recognition of SLC19A3.
Collapse
Affiliation(s)
- Florian Gabriel
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Lea Spriestersbach
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Antonia Fuhrmann
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Katharina E J Jungnickel
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Siavash Mostafavi
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, 1050, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, 1050, Brussels, Belgium
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany.
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany.
- Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany.
| |
Collapse
|
3
|
Jungnickel KEJ, Guelle O, Iguchi M, Dong W, Kotov V, Gabriel F, Debacker C, Dairou J, McCort-Tranchepain I, Laqtom NN, Chan SH, Ejima A, Sato K, Massa López D, Saftig P, Mehdipour AR, Abu-Remaileh M, Gasnier B, Löw C, Damme M. MFSD1 with its accessory subunit GLMP functions as a general dipeptide uniporter in lysosomes. Nat Cell Biol 2024; 26:1047-1061. [PMID: 38839979 PMCID: PMC11252000 DOI: 10.1038/s41556-024-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
The lysosomal degradation of macromolecules produces diverse small metabolites exported by specific transporters for reuse in biosynthetic pathways. Here we deorphanized the major facilitator superfamily domain containing 1 (MFSD1) protein, which forms a tight complex with the glycosylated lysosomal membrane protein (GLMP) in the lysosomal membrane. Untargeted metabolomics analysis of MFSD1-deficient mouse lysosomes revealed an increase in cationic dipeptides. Purified MFSD1 selectively bound diverse dipeptides, while electrophysiological, isotope tracer and fluorescence-based studies in Xenopus oocytes and proteoliposomes showed that MFSD1-GLMP acts as a uniporter for cationic, neutral and anionic dipeptides. Cryoelectron microscopy structure of the dipeptide-bound MFSD1-GLMP complex in outward-open conformation characterized the heterodimer interface and, in combination with molecular dynamics simulations, provided a structural basis for its selectivity towards diverse dipeptides. Together, our data identify MFSD1 as a general lysosomal dipeptide uniporter, providing an alternative route to recycle lysosomal proteolysis products when lysosomal amino acid exporters are overloaded.
Collapse
Affiliation(s)
| | - Océane Guelle
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France
| | - Miharu Iguchi
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Vadim Kotov
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Florian Gabriel
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Cécile Debacker
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France
| | - Julien Dairou
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Cité, Paris, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Cité, Paris, France
| | - Nouf N Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Sze Ham Chan
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Akika Ejima
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kenji Sato
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - David Massa López
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Bruno Gasnier
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France.
| | - Christian Löw
- Centre for Structural Systems Biology, Hamburg, Germany.
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany.
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany.
| |
Collapse
|
4
|
Loynd C, Singha Roy SJ, Ovalle VJ, Canarelli SE, Mondal A, Jewel D, Ficaretta ED, Weerapana E, Chatterjee A. Electrochemical labelling of hydroxyindoles with chemoselectivity for site-specific protein bioconjugation. Nat Chem 2024; 16:389-397. [PMID: 38082177 PMCID: PMC10932882 DOI: 10.1038/s41557-023-01375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/18/2023] [Indexed: 02/06/2024]
Abstract
Electrochemistry has recently emerged as a powerful approach in small-molecule synthesis owing to its numerous attractive features, including precise control over the fundamental reaction parameters, mild reaction conditions and innate scalability. Even though these advantages also make it an attractive strategy for chemoselective modification of complex biomolecules such as proteins, such applications remain poorly developed. Here we report an electrochemically promoted coupling reaction between 5-hydroxytryptophan (5HTP) and simple aromatic amines-electrochemical labelling of hydroxyindoles with chemoselectivity (eCLIC)-that enables site-specific labelling of full-length proteins under mild conditions. Using genetic code expansion technology, the 5HTP residue can be incorporated into predefined sites of a recombinant protein expressed in either prokaryotic or eukaryotic hosts for subsequent eCLIC labelling. We used the eCLIC reaction to site-specifically label various recombinant proteins, including a full-length human antibody. Furthermore, we show that eCLIC is compatible with strain-promoted alkyne-azide and alkene-tetrazine click reactions, enabling site-specific modification of proteins at two different sites with distinct labels.
Collapse
Affiliation(s)
- Conor Loynd
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | | | - Vincent J Ovalle
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Sarah E Canarelli
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Atanu Mondal
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Delilah Jewel
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Elise D Ficaretta
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Eranthie Weerapana
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, USA.
| |
Collapse
|
5
|
Majumdar S, Desai R, Hans A, Dandekar P, Jain R. From Efficiency to Yield: Exploring Recent Advances in CHO Cell Line Development for Monoclonal Antibodies. Mol Biotechnol 2024:10.1007/s12033-024-01060-6. [PMID: 38363529 DOI: 10.1007/s12033-024-01060-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/29/2023] [Indexed: 02/17/2024]
Abstract
The increasing demand for biosimilar monoclonal antibodies (mAbs) has prompted the development of stable high-producing cell lines while simultaneously decreasing the time required for screening. Existing platforms have proven inefficient, resulting in inconsistencies in yields, growth characteristics, and quality features in the final mAb products. Selecting a suitable expression host, designing an effective gene expression system, developing a streamlined cell line generation approach, optimizing culture conditions, and defining scaling-up and purification strategies are all critical steps in the production of recombinant proteins, particularly monoclonal antibodies, in mammalian cells. As a result, an active area of study is dedicated to expression and optimizing recombinant protein production. This review explores recent breakthroughs and approaches targeted at accelerating cell line development to attain efficiency and consistency in the synthesis of therapeutic proteins, specifically monoclonal antibodies. The primary goal is to bridge the gap between rising demand and consistent, high-quality mAb production, thereby benefiting the healthcare and pharmaceutical industries.
Collapse
Affiliation(s)
- Sarmishta Majumdar
- Department of Biological Science and Biotechnology, Institute of Chemical Technology, Mumbai, 400019, India
| | - Ranjeet Desai
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400019, India
| | - Aakarsh Hans
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400019, India
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400019, India.
| | - Ratnesh Jain
- Department of Biological Science and Biotechnology, Institute of Chemical Technology, Mumbai, 400019, India.
| |
Collapse
|
6
|
Timmins LM, Erickson P, Parekkadan B. Investigating dynamics of lentiviral vector secretion from HEK293T producer cells using a fractionated perfusion system. Biotechnol J 2024; 19:e2300097. [PMID: 37718481 PMCID: PMC11289840 DOI: 10.1002/biot.202300097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/15/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Mammalian cell culture is quickly becoming the go to engineering vehicle to mass produce viral vectors in a manner that is safe, convenient, reproducible, and cost and scale effective. Human embryonic kidney (HEK293) cells, in particular, have been utilized and customized (via differentiated transgene expression, modified culture parameters, addition of cytostatic culture agents) to increase vector yields. However, less attention has been made to understanding innate processes within the cells (such as, immune response, cell cycle, metabolism) themselves to better control or increase viral vector product yield. Accordingly, herein, the variation in viral production was studied from HEK cells over time using a one-way perfusion system and bioreactor to study the impact of external factors on secretion dynamics without retrotransduction. Specifically, the impact of cell density on viral titer, transduction efficiency, and LDH, was studied. Next, we look at the impact of using an inflammatory reporter cell line on viral output, and the secretion dynamics from HEK cells when we use sodium butyrate (cell cycle arrest agent). Lastly, we assess how downregulation of the PDK pathway increases viral titer. Altogether, we investigated the impact of various interventions to increase transient protein expression and viral output from HEK cells in a controlled and measurable environment to ultimately increase the efficiency of HEK cells for downstream clinical applications.
Collapse
Affiliation(s)
- Lauren M. Timmins
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Patrick Erickson
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
- Department of Medicine, Rutgers Biomedical Health Sciences, New Brunswick, New Jersey, USA
| |
Collapse
|
7
|
Dübel S. Can antibodies be "vegan"? A guide through the maze of today's antibody generation methods. MAbs 2024; 16:2343499. [PMID: 38634488 PMCID: PMC11028021 DOI: 10.1080/19420862.2024.2343499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024] Open
Abstract
There is no doubt that today's life sciences would look very different without the availability of millions of research antibody products. Nevertheless, the use of antibody reagents that are poorly characterized has led to the publication of false or misleading results. The use of laboratory animals to produce research antibodies has also been criticized. Surprisingly, both problems can be addressed with the same technology. This review charts today's maze of different antibody formats and the various methods for antibody production and their interconnections, ultimately concluding that sequence-defined recombinant antibodies offer a clear path to both improved quality of experimental data and reduced use of animals.
Collapse
Affiliation(s)
- Stefan Dübel
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
8
|
Custódio TF, Guédez G, Löw C. Transient Co-expression of Membrane Protein Complexes in Mammalian Cells. Methods Mol Biol 2024; 2810:11-28. [PMID: 38926270 DOI: 10.1007/978-1-0716-3878-1_2] [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: 06/28/2024]
Abstract
Membrane proteins are essential components of biological membranes with key roles in cellular processes such as nutrient transport, cell communication, signaling, or energy conversion. Due to their crucial functions, membrane proteins and their complexes are often targets for therapeutic interventions. Expression and purification of membrane proteins are often a bottleneck to yield sufficient material for structural studies and further downstream characterization. Taking advantage of the Expi293 expression system for the production of eukaryotic proteins, we present a very efficient and fast protocol for the co-expression of a membrane complex. Here, we use transient transfection to co-express the membrane transporter PHT1 with its adaptor protein TASL. To allow the simultaneous screening of different proteins, constructs, or interaction partners, we make use of the Twin-Strep magnetic system. The protocol can be applied for small-scale screening of any membrane protein alone or co-expressed with interacting partners followed by large-scale production and purification of a potential membrane protein complex.
Collapse
Affiliation(s)
- Tânia F Custódio
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany.
- European Molecular Biology Laboratory (EMBL) Hamburg, Hamburg, Germany.
| | - Gabriela Guédez
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Hamburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany.
- European Molecular Biology Laboratory (EMBL) Hamburg, Hamburg, Germany.
| |
Collapse
|
9
|
Lao T, Farnos O, Bueno A, Alvarez A, Rodríguez E, Palacios J, de la Luz KR, Kamen A, Carpio Y, Estrada MP. Transient Expression in HEK-293 Cells in Suspension Culture as a Rapid and Powerful Tool: SARS-CoV-2 N and Chimeric SARS-CoV-2N-CD154 Proteins as a Case Study. Biomedicines 2023; 11:3050. [PMID: 38002050 PMCID: PMC10669214 DOI: 10.3390/biomedicines11113050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
In a previous work, we proposed a vaccine chimeric antigen based on the fusion of the SARS-CoV-2 N protein to the extracellular domain of the human CD40 ligand (CD154). This vaccine antigen was named N-CD protein and its expression was carried out in HEK-293 stably transfected cells, grown in adherent conditions and serum-supplemented medium. The chimeric protein obtained in these conditions presented a consistent pattern of degradation. The immunization of mice and monkeys with this chimeric protein was able to induce a high N-specific IgG response with only two doses in pre-clinical experiments. In order to explore ways to diminish protein degradation, in the present work, the N and N-CD proteins were produced in suspension cultures and serum-free media following transient transfection of the HEK-293 clone 3F6, at different scales, including stirred-tank controlled bioreactors. The results showed negligible or no degradation of the target proteins. Further, clones stably expressing N-CD were obtained and adapted to suspension culture, obtaining similar results to those observed in the transient expression experiments in HEK-293-3F6. The evidence supports transient protein expression in suspension cultures and serum-free media as a powerful tool to produce in a short period of time high levels of complex proteins susceptible to degradation, such as the SARS-CoV-2 N protein.
Collapse
Affiliation(s)
- Thailin Lao
- Center for Genetic Engineering and Biotechnology, Animal Biotechnology Department, Havana 10600, Cuba; (T.L.)
| | - Omar Farnos
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (O.F.); (A.K.)
| | - Alexi Bueno
- Process Development Department, Center of Molecular Immunology, Havana 11600, Cuba (J.P.); (K.R.d.l.L.)
| | - Anays Alvarez
- Center for Genetic Engineering and Biotechnology, Animal Biotechnology Department, Havana 10600, Cuba; (T.L.)
| | - Elsa Rodríguez
- Center for Genetic Engineering and Biotechnology, Animal Biotechnology Department, Havana 10600, Cuba; (T.L.)
| | - Julio Palacios
- Process Development Department, Center of Molecular Immunology, Havana 11600, Cuba (J.P.); (K.R.d.l.L.)
| | - Kathya Rashida de la Luz
- Process Development Department, Center of Molecular Immunology, Havana 11600, Cuba (J.P.); (K.R.d.l.L.)
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (O.F.); (A.K.)
| | - Yamila Carpio
- Center for Genetic Engineering and Biotechnology, Animal Biotechnology Department, Havana 10600, Cuba; (T.L.)
| | - Mario Pablo Estrada
- Center for Genetic Engineering and Biotechnology, Animal Biotechnology Department, Havana 10600, Cuba; (T.L.)
| |
Collapse
|
10
|
Custódio TF, Killer M, Yu D, Puente V, Teufel DP, Pautsch A, Schnapp G, Grundl M, Kosinski J, Löw C. Molecular basis of TASL recruitment by the peptide/histidine transporter 1, PHT1. Nat Commun 2023; 14:5696. [PMID: 37709742 PMCID: PMC10502012 DOI: 10.1038/s41467-023-41420-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023] Open
Abstract
PHT1 is a histidine /oligopeptide transporter with an essential role in Toll-like receptor innate immune responses. It can act as a receptor by recruiting the adaptor protein TASL which leads to type I interferon production via IRF5. Persistent stimulation of this signalling pathway is known to be involved in the pathogenesis of systemic lupus erythematosus (SLE). Understanding how PHT1 recruits TASL at the molecular level, is therefore clinically important for the development of therapeutics against SLE and other autoimmune diseases. Here we present the Cryo-EM structure of PHT1 stabilized in the outward-open conformation. By combining biochemical and structural modeling techniques we propose a model of the PHT1-TASL complex, in which the first 16 N-terminal TASL residues fold into a helical structure that bind in the central cavity of the inward-open conformation of PHT1. This work provides critical insights into the molecular basis of PHT1/TASL mediated type I interferon production.
Collapse
Affiliation(s)
- Tânia F Custódio
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Maxime Killer
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
- Collaboration for joint PhD degree between EMBL, and Heidelberg University, Faculty of Biosciences, 69120, Heidelberg, Germany
| | - Dingquan Yu
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
- Collaboration for joint PhD degree between EMBL, and Heidelberg University, Faculty of Biosciences, 69120, Heidelberg, Germany
| | - Virginia Puente
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
| | - Daniel P Teufel
- Boehringer Ingelheim Pharma, Birkendorferstraße 65, 88397, Biberach, Germany
| | - Alexander Pautsch
- Boehringer Ingelheim Pharma, Birkendorferstraße 65, 88397, Biberach, Germany
| | - Gisela Schnapp
- Boehringer Ingelheim Pharma, Birkendorferstraße 65, 88397, Biberach, Germany
| | - Marc Grundl
- Boehringer Ingelheim Pharma, Birkendorferstraße 65, 88397, Biberach, Germany
| | - Jan Kosinski
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany.
- European Molecular Biology Laboratory (EMBL) Hamburg, Notkestraße 85, 22607, Hamburg, Germany.
| |
Collapse
|
11
|
Khalid K, Poh CL. The development of DNA vaccines against SARS-CoV-2. Adv Med Sci 2023; 68:213-226. [PMID: 37364379 PMCID: PMC10290423 DOI: 10.1016/j.advms.2023.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The COVID-19 pandemic exerted significant impacts on public health and global economy. Research efforts to develop vaccines at warp speed against SARS-CoV-2 led to novel mRNA, viral vectored, and inactivated vaccines being administered. The current COVID-19 vaccines incorporate the full S protein of the SARS-CoV-2 Wuhan strain but rapidly emerging variants of concern (VOCs) have led to significant reductions in protective efficacies. There is an urgent need to develop next-generation vaccines which could effectively prevent COVID-19. METHODS PubMed and Google Scholar were systematically reviewed for peer-reviewed papers up to January 2023. RESULTS A promising solution to the problem of emerging variants is a DNA vaccine platform since it can be easily modified. Besides expressing whole protein antigens, DNA vaccines can also be constructed to include specific nucleotide genes encoding highly conserved and immunogenic epitopes from the S protein as well as from other structural/non-structural proteins to develop effective vaccines against VOCs. DNA vaccines are associated with low transfection efficiencies which could be enhanced by chemical, genetic, and molecular adjuvants as well as delivery systems. CONCLUSIONS The DNA vaccine platform offers a promising solution to the design of effective vaccines. The challenge of limited immunogenicity in humans might be solved through the use of genetic modifications such as the addition of nuclear localization signal (NLS) peptide gene, strong promoters, MARs, introns, TLR agonists, CD40L, and the development of appropriate delivery systems utilizing nanoparticles to increase uptake by APCs in enhancing the induction of potent immune responses.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia.
| |
Collapse
|
12
|
Zhou J, Yan GG, Cluckey D, Meade C, Ruth M, Sorm R, Tam AS, Lim S, Petridis C, Lin L, D’Antona AM, Zhong X. Exploring Parametric and Mechanistic Differences between Expi293F TM and ExpiCHO-S TM Cells for Transient Antibody Production Optimization. Antibodies (Basel) 2023; 12:53. [PMID: 37606437 PMCID: PMC10443273 DOI: 10.3390/antib12030053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023] Open
Abstract
Rapidly producing drug-like antibody therapeutics for lead molecule discovery and candidate optimization is typically accomplished by large-scale transient gene expression technologies (TGE) with cultivated mammalian cells. The TGE methodologies have been extensively developed over the past three decades, yet produce significantly lower yields than the stable cell line approach, facing the technical challenge of achieving universal high expression titers for a broad range of antibodies and therapeutics modalities. In this study, we explored various parameters for antibody production in the TGE cell host Expi293FTM and ExpiCHO-STM with the transfection reagents ExpiFectamineTM and polyethylenimine. We discovered that there are significant differences between Expi293FTM and ExpiCHO-STM cells with regards to DNA complex formation time and ratio, complex formation buffers, DNA complex uptake trafficking routes, responses to dimethyl sulfoxide and cell cycle inhibitors, as well as light-chain isotype expression preferences. This investigation mechanistically dissected the TGE processes and provided a new direction for future transient antibody production optimization.
Collapse
|
13
|
Chen C, Wang Z, Sun Z, Li W, Dimitrov DS. Development of an efficient method for selection of stable cell pools for protein expression and surface display with Expi293F cells. Cell Biochem Funct 2023; 41:355-364. [PMID: 36864545 DOI: 10.1002/cbf.3787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/18/2023] [Indexed: 03/04/2023]
Abstract
Compare with transient expression, stable cell lines generally have higher productivity and better quality for protein expression. However, selection of stable cell line is time-consuming and laborious. Here we describe an optimized selection method to achieve high-efficient stable cell pools with Expi293F suspension cells. This method only takes 2-3 weeks to generate stable cell pools with 2- to 10-fold higher productivity than transient gene expression (TGE). In fed-batch culture with Yeastolate, >1 g/L yield was achieved with our KTN0239-IgG stable cell pool in shaker flasks. This method can be also applied to efficiently display proteins on the cell surface.
Collapse
Affiliation(s)
- Chuan Chen
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Zening Wang
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zehua Sun
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| | - Wei Li
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Dimiter S Dimitrov
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Sui S, Wang H, Song J, Tai W. Development of a spermine lipid for transient antibody expression. Bioorg Med Chem 2023; 78:117114. [PMID: 36563514 DOI: 10.1016/j.bmc.2022.117114] [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: 08/18/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Transient expression is the only way to quickly obtain a small scale of antibodies for biomedical research and therapeutic evaluation. The agents for transfecting the suspension cells, e.g. PEI or commercial agents, either lack efficiency or excessively expensive. Herein, a novel spermine-based lipid was developed and fabricated into a cationic liposome for antibody expression. This new transfection agent, designated as sperminoliposome, is feasible, cheap, and highly effective to produce antibodies. Compared to PEI, a 3 times higher yield of antibody was obtained by sperminoliposome during the transient expression of cetuximab in suspension 293F cells. Characterizations confirmed that the expressed antibody is fully functional and eligible for further research. Our study provides an effective tool for the rapid production of antibodies economically and feasibly.
Collapse
Affiliation(s)
- Shaowei Sui
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hao Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiajie Song
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
15
|
Luly KM, Yang H, Lee SJ, Wang W, Ludwig SD, Tarbox HE, Wilson DR, Green JJ, Spangler JB. Poly(Beta-Amino Ester)s as High-Yield Transfection Reagents for Recombinant Protein Production. Int J Nanomedicine 2022; 17:4469-4479. [PMID: 36176585 PMCID: PMC9514136 DOI: 10.2147/ijn.s377371] [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: 06/05/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Transient transfection is an essential tool for recombinant protein production, as it allows rapid screening for expression without stable integration of genetic material into a target cell genome. Poly(ethylenimine) (PEI) is the current gold standard for transient gene transfer, but transfection efficiency and the resulting protein yield are limited by the polymer’s toxicity. This study investigated the use of a class of cationic polymers, poly(beta-amino ester)s (PBAEs), as reagents for transient transfection in comparison to linear 25 kDa PEI, a commonly used transfection reagent. Methods Transfection efficiency and protein production were assessed in human embryonic kidney 293F (HEK) and Chinese hamster ovary-S (CHO) cell suspensions using PBAE-based nanoparticles in comparison to linear 25 kDa PEI. Production of both a cytosolic reporter and secreted antibodies was investigated. Results In both HEK and CHO cells, several PBAEs demonstrated superior transfection efficiency and enhanced production of a cytosolic reporter compared to linear 25 kDa PEI. This result extended to secreted proteins, as a model PBAE increased the production of 3 different secreted antibodies compared to linear 25 kDa PEI at culture scales ranging from 20 to 2000 mL. In particular, non-viral gene transfer using the lead PBAE/plasmid DNA nanoparticle formulation led to robust transfection of mammalian cells across different constructs, doses, volumes, and cell types. Conclusion These results show that PBAEs enhance transfection efficiency and increase protein yield compared to a widespread commercially available reagent, making them attractive candidates as reagents for use in recombinant protein production.
Collapse
Affiliation(s)
- Kathryn M Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huilin Yang
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen J Lee
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Wentao Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth D Ludwig
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Haley E Tarbox
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Neurosurgery and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Repeated Transient Transfection: An Alternative for the Recombinant Production of Difficult-to-Express Proteins Like BMP2. Processes (Basel) 2022. [DOI: 10.3390/pr10061064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Human bone morphogenetic protein 2 (hBMP2) is routinely used in medical applications as an inducer of osteoformation. The recombinant production of BMP2 is typically performed using stable Chinese hamster ovary (CHO) cell lines. However, this process is inefficient, resulting in low product titers. In contrast, transient gene expression (TGE), which also enables the production of recombinant proteins, suffers from short production times and hence limited total product amounts. Here, we show that TGE-based BMP2 production is more efficient in HEKsus than in CHOsus cells. Independently of the cell lines, a bicistronic plasmid co-expressing EGFP and BMP2 facilitated the determination of the transfection efficiency but led to inferior BMP2 titers. Finally, we used a high cell density transient transfection (HCD-TGE) protocol to improve and extend the BMP2 expression by performing four rounds of serial transfections on one pool of HEKsus cells. This repeated transient transfection (RTT) process in HEKsus cells was implemented using EGFP as a reporter gene and further adapted for BMP2 production. The proposed method significantly improves BMP2 production (up to 509 ng/106 cells) by extending the production phase (96–360 h). RTT can be integrated into the seed train and is shown to be compatible with scale-up to the liter range.
Collapse
|
17
|
Johari YB, Scarrott JM, Pohle TH, Liu P, Mayer A, Brown AJ, James DC. Engineering of the CMV promoter for controlled expression of recombinant genes in HEK293 cells. Biotechnol J 2022; 17:e2200062. [PMID: 35482470 DOI: 10.1002/biot.202200062] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/07/2022] [Accepted: 04/23/2022] [Indexed: 11/08/2022]
Abstract
Expression of recombinant genes in HEK293 cells is frequently utilized for production of recombinant proteins and viral vectors. These systems frequently employ the cytomegalovirus (CMV) promoter to drive recombinant gene transcription. However, the mechanistic basis of CMV-mediated transcriptional activation in HEK293 cells is unknown and consequently there are no strategies to engineer CMV for controlled expression of recombinant genes. Extensive bioinformatic analyses of transcription factor regulatory elements (TFREs) within the human CMV sequence and transcription factor mRNAs within the HEK293 transcriptome revealed 80 possible regulatory interactions. Through in vitro functional testing using reporter constructs harboring discrete TFREs or CMV deletion variants we identified key TFRE components and clusters of TFREs (cis-regulatory modules) within the CMV sequence. Our data reveal that CMV activity in HEK293 cells is a function of the promoters various constituent TFREs including AhR:ARNT, CREB, E4F, Sp1, ZBED1, JunB, c-Rel, and NF-κB. We also identified critical Sp1-dependent upstream activator elements near the transcriptional start site that were required for efficient transcription and YY1 and RBP-Jκ binding sites that mediate transrepression. Our study shows for the first time that novel, compact CMV-derived promoters can be engineered that exhibit up to 50% higher transcriptional efficiency (activity per unit DNA sequence) or 14% increase in total activity compared to the wild-type counterpart.
Collapse
Affiliation(s)
- Yusuf B Johari
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Joseph M Scarrott
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Thilo H Pohle
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Ping Liu
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Ayda Mayer
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Adam J Brown
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK.,Syngensys Ltd., Sheffield, UK
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK.,Syngensys Ltd., Sheffield, UK
| |
Collapse
|
18
|
Zambrano N, Froechlich G, Lazarevic D, Passariello M, Nicosia A, De Lorenzo C, Morelli MJ, Sasso E. High-Throughput Monoclonal Antibody Discovery from Phage Libraries: Challenging the Current Preclinical Pipeline to Keep the Pace with the Increasing mAb Demand. Cancers (Basel) 2022; 14:cancers14051325. [PMID: 35267633 PMCID: PMC8909429 DOI: 10.3390/cancers14051325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Monoclonal antibodies are increasingly used for a broad range of diseases. Rising demand must face with time time-consuming and laborious processes to isolate novel monoclonal antibodies. Next-generation sequencing coupled to phage display provides timely and sustainable high throughput selection strategy to rapidly access novel target. Here, we describe the current NGS-guided strategies to identify potential binders from enriched sub-libraires by applying a user-friendly informatic pipeline to identify and discard false positive clones. Rescue step and strategies to boost mAb yield are also discussed to improve the limiting selection and screening steps. Abstract Monoclonal antibodies are among the most powerful therapeutics in modern medicine. Since the approval of the first therapeutic antibody in 1986, monoclonal antibodies keep holding great expectations for application in a range of clinical indications, highlighting the need to provide timely and sustainable access to powerful screening options. However, their application in the past has been limited by time-consuming and expensive steps of discovery and production. The screening of antibody repertoires is a laborious step; however, the implementation of next-generation sequencing-guided screening of single-chain antibody fragments has now largely overcome this issue. This review provides a detailed overview of the current strategies for the identification of monoclonal antibodies from phage display-based libraries. We also discuss the challenges and the possible solutions to improve the limiting selection and screening steps, in order to keep pace with the increasing demand for monoclonal antibodies.
Collapse
Affiliation(s)
- Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Correspondence: (N.Z.); (E.S.)
| | - Guendalina Froechlich
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Dejan Lazarevic
- Center for Omics Sciences Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (D.L.); (M.J.M.)
| | - Margherita Passariello
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Alfredo Nicosia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Claudia De Lorenzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Marco J. Morelli
- Center for Omics Sciences Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (D.L.); (M.J.M.)
| | - Emanuele Sasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; (G.F.); (M.P.); (A.N.); (C.D.L.)
- CEINGE—Biotecnologie Avanzate s.c. a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Correspondence: (N.Z.); (E.S.)
| |
Collapse
|
19
|
Irani V, Soliman C, Raftis MA, Guy AJ, Elbourne A, Ramsland PA. Expression of monoclonal antibodies for functional and structural studies. METHODS IN MICROBIOLOGY 2022. [DOI: 10.1016/bs.mim.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Kim J, Kim JY, Kim H, Kim E, Park S, Ryu KH, Lee EG. Increasing Transfection Efficiency of Lipoplexes by Modulating Complexation Solution for Transient Gene Expression. Int J Mol Sci 2021; 22:ijms222212344. [PMID: 34830226 PMCID: PMC8619889 DOI: 10.3390/ijms222212344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/16/2022] Open
Abstract
Transient gene expression is a suitable tool for the production of biopharmaceutical candidates in the early stage of development and provides a simple and rapid alternative to the generation of stable cell line. In this study, an efficient transient gene expression methodology using DC-Chol/DOPE cationic liposomes and pDNA in Chinese hamster ovary suspension cells was established through screening of diverse lipoplex formation conditions. We modulated properties of both the liposome formation and pDNA solution, together called complexation solutions. Protein expression and cellular cytotoxicity were evaluated following transfection over the cell cultivation period to select the optimal complexation solution. Changes in hydrodynamic size, polydispersity index, and ζ potential of the liposomes and lipoplexes were analyzed depending on the various pH ranges of the complexation solutions using dynamic light scattering. The transfer of lipoplexes to the cytosol and their conformation were traced using fluorescence analysis until the early period of transfection. As a result, up to 1785 mg/L and 191 mg/L of human Fc protein and immunoglobulin G (bevacizumab), respectively, were successfully produced using acidic liposome formation and alkaline pDNA solutions. We expect that this lipoplex formation in acidic and alkaline complexation solutions could be an effective methodology for a promising gene delivery strategy.
Collapse
Affiliation(s)
- Jaemun Kim
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea; (J.K.); (J.Y.K.)
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Ji Yul Kim
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea; (J.K.); (J.Y.K.)
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Hyeonkyeong Kim
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Eunsil Kim
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Soonyong Park
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Kyoung-Hwa Ryu
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
| | - Eun Gyo Lee
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea; (J.K.); (J.Y.K.)
- Bioprocess Engineering Center, KRIBB, 30 Yeongudanji-ro Ochang-eup, Cheongwon-gu, Cheongju-si 28116, Korea; (H.K.); (E.K.); (S.P.); (K.-H.R.)
- Correspondence: ; Tel.: +82-43-240-6633
| |
Collapse
|
21
|
Killer M, Wald J, Pieprzyk J, Marlovits TC, Löw C. Structural snapshots of human PepT1 and PepT2 reveal mechanistic insights into substrate and drug transport across epithelial membranes. SCIENCE ADVANCES 2021; 7:eabk3259. [PMID: 34730990 PMCID: PMC8565842 DOI: 10.1126/sciadv.abk3259] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The uptake of peptides in mammals plays a crucial role in nutrition and inflammatory diseases. This process is mediated by promiscuous transporters of the solute carrier family 15, which form part of the major facilitator superfamily. Besides the uptake of short peptides, peptide transporter 1 (PepT1) is a highly abundant drug transporter in the intestine and represents a major route for oral drug delivery. PepT2 also allows renal drug reabsorption from ultrafiltration and brain-to-blood efflux of neurotoxic compounds. Here, we present cryogenic electron microscopy (cryo-EM) structures of human PepT1 and PepT2 captured in four different states throughout the transport cycle. The structures reveal the architecture of human peptide transporters and provide mechanistic insights into substrate recognition and conformational transitions during transport. This may support future drug design efforts to increase the bioavailability of different drugs in the human body.
Collapse
Affiliation(s)
- Maxime Killer
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607 Hamburg, Germany
- European Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607 Hamburg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Faculty of Biosciences, Im Neuenheimer Feld 234, D-69120 Heidelberg, Germany
| | - Jiri Wald
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607 Hamburg, Germany
- Institute of Structural and Systems Biology, University Medical Center Hamburg-Eppendorf, Notkestrasse 85, D-22607 Hamburg, Germany
- Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607 Hamburg, Germany
| | - Joanna Pieprzyk
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607 Hamburg, Germany
- European Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607 Hamburg, Germany
| | - Thomas C. Marlovits
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607 Hamburg, Germany
- Institute of Structural and Systems Biology, University Medical Center Hamburg-Eppendorf, Notkestrasse 85, D-22607 Hamburg, Germany
- Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607 Hamburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607 Hamburg, Germany
- European Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607 Hamburg, Germany
- Corresponding author.
| |
Collapse
|
22
|
Kronenberg J, Schrödter K, Noll GA, Twyman RM, Prüfer D, Känel P. The tobacco phosphatidylethanolamine-binding protein NtFT4 simultaneously improves vitality, growth, and protein yield in human cells. Biotechnol Bioeng 2021; 118:3770-3786. [PMID: 34110007 DOI: 10.1002/bit.27853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/30/2021] [Accepted: 06/05/2021] [Indexed: 11/11/2022]
Abstract
The production of biopharmaceutical proteins in mammalian cells by transient expression or stable transformation requires robust and viable cells. Cell line engineering must therefore balance improved cell growth and viability with high productivity. We tested the ability of nonmammalian phosphatidylethanolamine-binding proteins to enhance cell proliferation in monolayers and suspension cultures. The tobacco protein NtFT4 improved the proliferation of multiple human cell lines. Viable cell density is usually impaired by efficient transfection, but we found that the number of HEK-293TNtFT4 cells at the peak of protein expression was twice that of standard HEK-293T cells, and the antibody yield increased by approximately one-third. Improved growth and viability were observed in different cell lines, in different culture media, and also after transient transfection, suggesting the beneficial trait is consistent and transferable. Additional modifications could boost the productivity of high-density HEK-293TNtFT4 cells even further as we showed for a fluorescent marker protein and recombinant antibody expressed in monolayer cultures. The HEK-293TNtFT4 cell line provides a new human model platform that increases cell proliferation, also achieving a fundamental improvement in recombinant protein expression.
Collapse
Affiliation(s)
- Julia Kronenberg
- Department of Functional and Applied Genomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Münster, Germany
| | - Katrin Schrödter
- Department of Functional and Applied Genomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Münster, Germany
| | - Gundula A Noll
- Institute of Plant Biology and Biotechnology, University of Münster, Münster, Germany
| | | | - Dirk Prüfer
- Department of Functional and Applied Genomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Münster, Germany
- Institute of Plant Biology and Biotechnology, University of Münster, Münster, Germany
| | - Philip Känel
- Department of Functional and Applied Genomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Münster, Germany
| |
Collapse
|
23
|
Mangion M, Robert MA, Slivac I, Gilbert R, Gaillet B. Production and Use of Gesicles for Nucleic Acid Delivery. Mol Biotechnol 2021; 64:278-292. [PMID: 34596870 DOI: 10.1007/s12033-021-00389-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 09/08/2021] [Indexed: 12/29/2022]
Abstract
Over-expression of the vesicular stomatitis virus glycoprotein (VSVG) in mammalian cells can induce the formation of VSVG-pseudotyped vesicles (named "gesicles") from membrane budding. Its use as a nucleic acid delivery tool is still poorly documented. Naked-plasmid DNA can be delivered in animal cells with gesicles in presence of hexadimethrine bromide (polybrene). However, little is known about gesicle manufacturing process and conditions to obtain successful nucleic acid delivery. In this study, gesicles production process using polyethylenimine (PEI)-transfected HEK293 cells was developed by defining the VSVG-plasmid concentration, the DNA:PEI mass ratio, and the time of gesicle harvest. Furthermore, parameters described in the literature relevant for nucleic acid delivery such as (i) component concentrations in assembly mixture, (ii) component addition order, (iii) incubation time, and (iv) polybrene concentration were tested by assessing the transfection capacity of the gesicles complexed with a green fluorescent protein (GFP)-coding plasmid. Interestingly, freezing/thawing cycles and storage at + 4 °C, - 20 °C, and - 80 °C did not reduce gesicles' ability to transfer plasmid DNA. Transfection efficiency of 55% and 22% was obtained for HeLa cells and for hard-to-transfect cells such as human myoblasts, respectively. For the first time, gesicles were used for delivery of a large plasmid (18-kb) with 42% of efficiency and for enhanced green fluorescent protein (eGFP) gene silencing with siRNA (up to 60%). In conclusion, gesicles represent attractive bioreagents with great potential to deliver nucleic acids in mammalian cells.
Collapse
Affiliation(s)
- Mathias Mangion
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada
| | - Marc-André Robert
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.,Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Igor Slivac
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada
| | - Rénald Gilbert
- ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.,Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Bruno Gaillet
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada. .,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada. .,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.
| |
Collapse
|
24
|
Doan CC, Ho NQC, Nguyen TT, Nguyen TPT, Do DG, Hoang NS, Le TL. Enhancement of anti-TNFα monoclonal antibody production in CHO cells through the use of UCOE and DHFR elements in vector construction and the optimization of cell culture media. Prep Biochem Biotechnol 2021; 52:452-470. [PMID: 34427158 DOI: 10.1080/10826068.2021.1963981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Recently, there has been a high demand for anti-tumor necrosis factor-α monoclonal antibodies (mAbTNFα) in the treatment of rheumatoid arthritis and other autoimmune diseases. Thus, efficient strategies and stable high-producing cell lines need to be established to increase antibody production. In this study, we describe an efficient approach to establish a mAbTNFα high-producing clone through the optimization of expression vectors and cell culture media. The ubiquitous chromatin opening element (UCOE) and dihydrofolate reductase (DHFR)-based vectors encoding mAbTNFα were introduced into the CHO-DG44 cells using lipofection. Clones were obtained by selecting transfected cells with G418, amplifying them by treatment with methotrexate, and isolating them by limiting dilution. Different media formulated with commercial feeds and media were also screened to develop an improved medium. The antibody produced by the selected clone was purified, characterized, and compared to standard adalimumab. Using our established protocol, a cell clone obtained from stable mAbTNFα-expressing cell pools showed a 3.8-fold higher antibody titer compared to stable cell pools. Furthermore, the highest antibody yield of selected clones cultured in fed-batch mode using improved medium was 2450 ± 30 µg/mL, which was 13.2-fold higher than that of stable cell pool cultivated in batch mode using a basal medium. The purified antibody had primary chemical and biological characteristics similar to those of adalimumab. Therefore, the use of UCOE and DHFR vectors in combination with the optimization of cell culture media may help in establishing stable and high-producing CHO cell lines for therapeutic antibody production.
Collapse
Affiliation(s)
- Chinh Chung Doan
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam.,Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Nguyen Quynh Chi Ho
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Thi Thuy Nguyen
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Thi Phuong Thao Nguyen
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam.,Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Dang Giap Do
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Nghia Son Hoang
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam.,Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Thanh Long Le
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam.,Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| |
Collapse
|
25
|
Abaandou L, Quan D, Shiloach J. Affecting HEK293 Cell Growth and Production Performance by Modifying the Expression of Specific Genes. Cells 2021; 10:cells10071667. [PMID: 34359846 PMCID: PMC8304725 DOI: 10.3390/cells10071667] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
The HEK293 cell line has earned its place as a producer of biotherapeutics. In addition to its ease of growth in serum-free suspension culture and its amenability to transfection, this cell line’s most important attribute is its human origin, which makes it suitable to produce biologics intended for human use. At the present time, the growth and production properties of the HEK293 cell line are inferior to those of non-human cell lines, such as the Chinese hamster ovary (CHO) and the murine myeloma NSO cell lines. However, the modification of genes involved in cellular processes, such as cell proliferation, apoptosis, metabolism, glycosylation, secretion, and protein folding, in addition to bioprocess, media, and vector optimization, have greatly improved the performance of this cell line. This review provides a comprehensive summary of important achievements in HEK293 cell line engineering and on the global engineering approaches and functional genomic tools that have been employed to identify relevant genes for targeted engineering.
Collapse
Affiliation(s)
- Laura Abaandou
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
- Department of Chemistry and Biochemistry, College of Science, George Mason University, Fairfax, VA 22030, USA
| | - David Quan
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
| | - Joseph Shiloach
- Biotechnology Core Laboratory National Institutes of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; (L.A.); (D.Q.)
- Correspondence:
| |
Collapse
|
26
|
Budge JD, Young RJ, Smales CM. Engineering of Chinese Hamster Ovary Cells With NDPK-A to Enhance DNA Nuclear Delivery Combined With EBNA1 Plasmid Maintenance Gives Improved Exogenous Transient Reporter, mAb and SARS-CoV-2 Spike Protein Expression. Front Bioeng Biotechnol 2021; 9:679448. [PMID: 34150735 PMCID: PMC8212061 DOI: 10.3389/fbioe.2021.679448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
Transient gene expression (TGE) in mammalian cells is a method of rapidly generating recombinant protein material for initial characterisation studies that does not require time-consuming processes associated with stable cell line construction. High TGE yields are heavily dependent on efficient delivery of plasmid DNA across both the plasma and nuclear membranes. Here, we harness the protein nucleoside diphosphate kinase (NDPK-A) that contains a nuclear localisation signal (NLS) to enhance DNA delivery into the nucleus of CHO cells. We show that co-expression of NDPK-A during transient expression results in improved transfection efficiency in CHO cells, presumably due to enhanced transportation of plasmid DNA into the nucleus via the nuclear pore complex. Furthermore, introduction of the Epstein Barr Nuclear Antigen-1 (EBNA-1), a protein that is capable of inducing extrachromosomal maintenance, when coupled with complementary oriP elements on a transient plasmid, was utilised to reduce the effect of plasmid dilution. Whilst there was attenuated growth upon introduction of the EBNA-1 system into CHO cells, when both NDPK-A nuclear import and EBNA-1 mediated technologies were employed together this resulted in enhanced transient recombinant protein yields superior to those generated using either approach independently, including when expressing the complex SARS-CoV-2 spike (S) glycoprotein.
Collapse
Affiliation(s)
- James D Budge
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Robert J Young
- R&D Cell Engineering Group, Lonza Biologics, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Christopher Mark Smales
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
27
|
Schmitt MG, White RN, Barnard GC. Development of a high cell density transient CHO platform yielding mAb titers greater than 2 g/L in only 7 days. Biotechnol Prog 2021; 36:e3047. [PMID: 33411420 DOI: 10.1002/btpr.3047] [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: 05/21/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022]
Abstract
We developed a simple transient Chinese Hamster Ovary expression platform. Titers for a random panel of 20 clinical monoclonal antibodies (mAbs) ranged from 0.6 to 2.7 g/L after 7 days. Two factors were the key in obtaining these high titers. First, we utilized an extremely high starting cell density (20 million cells/ml), and then arrested further cell growth by employing mild hypothermic conditions (32°C). Second, we performed a 6-variable Design of Experiments to find optimal concentrations of plasmid DNA (coding DNA), boost DNA (DNA encoding the XBP1S transcription factor), transfection reagent (polyethylenimine [PEI]), and nutrient feed amounts. High coding DNA concentrations (12.5 mg/L) were found to be optimal. We therefore diluted expensive coding DNA with inexpensive inert filler DNA (herring sperm DNA). Reducing the coding DNA concentration by 70% from 12.5 to 3.75 mg/L did not meaningfully reduce mAb titers. Titers for the same panel of 20 clinical mAbs ranged from 0.7 to 2.2 g/L after reducing the coding DNA concentration to 3.75 mg/L. Finally, we found that titer and product quality attributes were similar for a clinical mAb (rituximab) expressed at very different scales (volumes ranging from 3 ml to 2 L).
Collapse
Affiliation(s)
- Matthew G Schmitt
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Technology Center, Indianapolis, Indiana, USA
| | - Regina N White
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Technology Center, Indianapolis, Indiana, USA
| | - Gavin C Barnard
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Technology Center, Indianapolis, Indiana, USA
| |
Collapse
|
28
|
Enhancing transient protein expression in HEK-293 cells by briefly exposing the culture to DMSO. J Neurosci Methods 2020; 350:109058. [PMID: 33359979 DOI: 10.1016/j.jneumeth.2020.109058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Transient expression of proteins in mammalian cells is a key technique for many functional and structural studies of human and higher eukaryotic genes as well as for the production of recombinant protein therapeutics. Maximizing the expression efficiency to achieve a higher expression yield is desirable and may be even critical when, for instance, an expressed protein must be characterized at the single-cell level. NEW METHODS Our goal was to develop a simple method by which protein expression yield in human embryonic kidney (HEK)-293 cells could be enhanced with a brief treatment of dimethyl sulfoxide (DMSO) solution. RESULTS By expressing green fluorescent protein (GFP) as a reporter protein using the calcium phosphate transfection method and imaging a large population of cells, we found that a 5-min exposure of 10 % DMSO to HEK-293 cells, 4 h after transfection of the protein of interest, leads to ∼1.6-fold increase in the expression yield without causing any appreciable cytotoxicity. By expressing an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and separately a kainate receptor in HEK-293 cells and measuring glutamate-induced whole-cell current response, we also found that such a brief DMSO treatment did not affect channel activity. CONCLUSION This method is simple, efficient and inexpensive to use for enhancing transient transfection yield in HEK-293 cells.
Collapse
|
29
|
Lu XA, He T, Han Z, Ding Y, Zhao L, Liu G, De Smet F, Huang X, Chen D, Qi F, Zhao X. Production of lentiviral vectors in suspension cells using low proportion of supercoiled circular plasmid DNA. Cytotechnology 2020; 72:10.1007/s10616-020-00433-4. [PMID: 33123933 PMCID: PMC7695760 DOI: 10.1007/s10616-020-00433-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/16/2020] [Indexed: 10/23/2022] Open
Abstract
The supercoiled circular (SC) topology form of plasmid DNA has been regarded to be advantageous over open circular or linearized analogue in transfection and expression efficiency, and therefore are largely demanded in the biopharmaceutical manufacturing. However, production of high-purity SC plasmid DNA would result in high manufacturing cost. The effect of SC proportion in plasmid DNA on the quality of packaged lentiviral vectors has never been reported. In this study, we established an efficient system for production of high-titer lentiviral vectors using suspension HEK293SF cells in serum-free media, and the lentiviral titer was not associated with the proportion of SC plasmid DNA. Plasmids DNA with different proportion of SC, open-circular, and linearized forms were prepared using the thermal denaturation method, and were transfected to adherent HEK293T or suspension HEK293SF cells for packaging of lentiviral vectors. The titer of lentiviral vectors from HEK293T cells, but not from HEK293SF cells, was significantly impaired when the proportion of SC plasmid DNA decreased from 60-80% to 30-40%. Further decrease of SC plasmid proportion to 3% led to a dramatic reduction of lentiviral titer no matter the packaging cell line was. However, lentiviral vectors from HEK293SF cells still showed a high titer even when the proportion of SC plasmid DNA was 3%. This study demonstrated that extremely high proportion of SC plasmid DNA was not required for packaging of high-titer lentiviral vector in HEK293SF cells, at least under our manufacturing process.
Collapse
Affiliation(s)
- Xin-An Lu
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Ting He
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of PLA General Hospital, 100048, Beijing, China
| | - Yanping Ding
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Liang Zhao
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Guanghua Liu
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Floris De Smet
- Sartorius Stedim North America Inc., 565 Johnson Avenue, Bohemia, New York, 11716, USA
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Danqing Chen
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China
| | - Feifei Qi
- Immunochina Pharmaceuticals Co., Ltd, 100089, Beijing, China.
| | - Xiangyu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China.
| |
Collapse
|
30
|
RCAN1 Inhibits BACE2 Turnover by Attenuating Proteasome-Mediated BACE2 Degradation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1920789. [PMID: 32566665 PMCID: PMC7293731 DOI: 10.1155/2020/1920789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/22/2020] [Indexed: 11/23/2022]
Abstract
Amyloid-β protein (Aβ) is the main component of neuritic plaques, the pathological hallmark of Alzheimer's disease (AD). β-site APP cleaving enzyme 1 (BACE1) is a major β-secretase contributing to Aβ generation. β-site APP cleaving enzyme 2 (BACE2), the homolog of BACE1, is not only a θ-secretase but also a conditional β-secretase. Previous studies showed that regulator of calcineurin 1 (RCAN1) is markedly increased by AD and promotes BACE1 expression. However, the role of RCAN1 in BACE2 regulation remains elusive. Here, we showed that RCAN1 increases BACE2 protein levels. Moreover, RCAN1 inhibits the turnover of BACE2 protein. Furthermore, RCAN1 attenuates proteasome-mediated BACE2 degradation, but not lysosome-mediated BACE2 degradation. Taken together, our work indicates that RCAN1 inhibits BACE2 turnover by attenuating proteasome-mediated BACE2 degradation. It advances our understanding of BACE2 regulation and provides a potential mechanism of BACE2 dysregulation in AD.
Collapse
|
31
|
Park SJ, Kim J, Kang S, Cha HJ, Shin H, Park J, Jang YS, Woo JS, Won C, Min DH. Discovery of direct-acting antiviral agents with a graphene-based fluorescent nanosensor. SCIENCE ADVANCES 2020; 6:eaaz8201. [PMID: 32523995 PMCID: PMC7259931 DOI: 10.1126/sciadv.aaz8201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/31/2020] [Indexed: 05/04/2023]
Abstract
Direct-acting agents against viral components are considered as the most promising candidates for the successful antiviral therapeutics. To date, no direct-acting drugs exist for the treatment against dengue virus (DV) infection, which can develop into life-threatening diseases. RNA-dependent RNA polymerase (RdRp), an RNA virus-specific enzyme highly conserved among various viral families, has been known as the broad-range antiviral drug target. Here, we developed an RNA-based graphene biosensor system [RNA nano-graphene oxide system (RANGO)] to enable the fluorescence-based quantitative analysis of the RdRp enzyme activity. We used the RANGO system to a high-throughput chemical screening to identify novel direct-acting antiviral drug candidates targeting DV RdRp from the FDA-approved small-molecule library. RANGO accelerated the massive selection of drug candidates. We found that one of the selected hit compounds, montelukast, showed antiviral activity in vitro and in vivo by directly inhibiting replication of DV and thus relieved related symptoms.
Collapse
Affiliation(s)
- Se-Jin Park
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jungho Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, Republic of Korea
| | - Seounghun Kang
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung Jin Cha
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science (IBS), Seoul National University, Seoul 08826, Republic of Korea
| | - Hojeong Shin
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jisang Park
- Department of Bioactive Material Sciences and Institute of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Yong-Suk Jang
- Department of Bioactive Material Sciences and Institute of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae-Sung Woo
- Center for RNA Research, Institute for Basic Science (IBS), Seoul National University, Seoul 08826, Republic of Korea
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Cheolhee Won
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, Republic of Korea
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
32
|
Qiu K, Liang W, Wang S, Kong T, Wang X, Li C, Wang Z, Wu Y. BACE2 degradation is mediated by both the proteasome and lysosome pathways. BMC Mol Cell Biol 2020; 21:13. [PMID: 32160867 PMCID: PMC7066761 DOI: 10.1186/s12860-020-00260-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/05/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease is the most common neurodegenerative disease in the elderly. Amyloid-β protein (Aβ) is the major component of neuritic plaques which are the hallmark of AD pathology. β-site APP cleaving enzyme 1 (BACE1) is the major β-secretase contributing to Aβ generation. β-site APP-cleaving enzyme 2 (BACE2), the homolog of BACE1, might play a complex role in the pathogenesis of Alzheimer's disease as it is not only a θ-secretase but also a conditional β-secretase. Dysregulation of BACE2 is observed in Alzheimer's disease. However, the regulation of BACE2 is less studied compared with BACE1, including its degradation pathways. In this study, we investigated the turnover rates and degradation pathways of BACE2 in both neuronal cells and non-neuronal cells. RESULTS Both lysosomal inhibition and proteasomal inhibition cause a time- and dose-dependent increase of transiently overexpressed BACE2 in HEK293 cells. The half-life of transiently overexpressed BACE2 protein is approximately 6 h. Moreover, the half-life of endogenous BACE2 protein is approximately 4 h in both HEK293 cells and mouse primary cortical neurons. Furthermore, both lysosomal inhibition and proteasomal inhibition markedly increases endogenous BACE2 in HEK293 cells and mouse primary cortical neurons. CONCLUSIONS This study demonstrates that BACE2 is degraded by both the proteasome and lysosome pathways in both neuronal and non-neuronal cells at endogenous level and in transient overexpression system. It indicates that BACE2 dysregulation might be mediated by the proteasomal and lysosomal impairment in Alzheimer's disease. This study advances our understanding of the regulation of BACE2 and provides a potential mechanism of its dysregulation in Alzheimer's disease.
Collapse
Affiliation(s)
- Kaixin Qiu
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Tingting Kong
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
| | - Xin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Chunyan Li
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China.
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China.
| |
Collapse
|
33
|
Anisimov RL, Ershova OA, Ershov AV, Filatova MA, Katorkin SA, Simonov VM. Recombinant β-Glucocerebrosidase specific immunoaffinity ligands selected from phage-displayed combinatorial scFv libraries. Protein Expr Purif 2020; 170:105573. [PMID: 31981620 DOI: 10.1016/j.pep.2020.105573] [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: 12/04/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 11/19/2022]
Abstract
Antibodies specific to β-Glucocerebrosidase were selected from phage displayed naïve scFv libraries. Biopannings were performed against recombinant human protein β-Glucocerebrosidase immobilized on polystyrene surface, specific phages were eluted with 50% ethylene glycol in citrate buffer (pH 6.0). Several specific binders were discovered and converted to full-size hIgG1 antibodies leading to highly stable binders with dissociation constants (Kd) in the range 10-40 nM. The antibodies were used further as ligands for affinity chromatography, where efficient and selective recovery of biologically active β-Glucocerebrosidase from cultured media of Chinese hamster ovary cells was demonstrated. β-Glucocerebrosidase was purified to nearly homogeneous state and had specific activity comparable to the commercially available preparations (40-44 U/mg protein). The obtained immunoaffinity sorbents have high capacity and can be easily regenerated.
Collapse
Affiliation(s)
- R L Anisimov
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia.
| | - O A Ershova
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia
| | - A V Ershov
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia
| | - M A Filatova
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia
| | - S A Katorkin
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia
| | - V M Simonov
- Generium LLC International Biotechnology Center, Volginsky, Vladimirskaya oblast, 601125, Russia
| |
Collapse
|
34
|
Antibody glycosylation: impact on antibody drug characteristics and quality control. Appl Microbiol Biotechnol 2020; 104:1905-1914. [DOI: 10.1007/s00253-020-10368-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/02/2020] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
35
|
Reese H, Bordelon T, Shanahan C, Crapanzano M, Sly J, Menegatti S. Novel peptoid-based adsorbents for purifying IgM and IgG from polyclonal and recombinant sources. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1137:121909. [DOI: 10.1016/j.jchromb.2019.121909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022]
|
36
|
Lee Y, Kim H, Kim E, Park S, Ryu KH, Lee EG. Rational design of transient gene expression process with lipoplexes for high-level therapeutic protein production in HEK293 cells. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Establishment of DHFR-deficient HEK293 cells for high yield of therapeutic glycoproteins. J Biosci Bioeng 2019; 128:487-494. [DOI: 10.1016/j.jbiosc.2019.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 01/21/2023]
|
38
|
Chen P, Demirji J, Ivleva VB, Horwitz J, Schwartz R, Arnold F. The transient expression of CHIKV VLP in large stirred tank bioreactors. Cytotechnology 2019; 71:1079-1093. [PMID: 31560090 DOI: 10.1007/s10616-019-00346-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/19/2019] [Indexed: 01/06/2023] Open
Abstract
Transient gene expression (TGE) bioprocesses have been difficult to scale up in large stirred tank bioreactors with volumes of more than 1.5 L. Low production levels are often observed, but the causes have not been investigated (Gutierrez-Granados et al. in Crit Rev Biotechnol 38:918-940, 2018). Chikungunya Virus-like particle (VLP), expressed by DNA-PEI transient transfection, is a representative case study for these difficulties. Clinical materials were produced in shake flasks, but the process suffered when transferred to large stirred tank bioreactors. The resulting process was not operationally friendly nor cost effective. In this study, a systematic approach was used to investigate the root causes of the poor scale up performance. The transfection conditions were first screened in ambr® 15 high throughput mini bioreactors then examined in 3 L stirred-tank systems. The studies found that production level was negatively correlated with inoculum cell growth status (P < 0.05). The pH range, DNA and PEI levels, order of the reagent addition, and gas-sparging systems were also studied and found to affect process performance. Further hydromechanical characterizations (Re, energy dissipation rates, and P/V, etc.) of shake flasks, ambr® 15, and 3-L stirred tank systems were performed. Overall, the study discovered that the shear stress (caused by a microsparger) and PEI toxicity together were the root causes of scale-up failure. Once the microsparger was replaced by a macrosparger, the scale-up was successful.
Collapse
Affiliation(s)
- Peifeng Chen
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA.
| | - Jacob Demirji
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA
| | - Vera B Ivleva
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd, Gaithersburg, MD, 20878, USA
| | - Joe Horwitz
- Amicus Therapeutics, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | | | - Frank Arnold
- Tunnell Consulting, 900 E. 8th Ave, King of Prussia, PA, 19406, USA
| |
Collapse
|
39
|
Lee C, Hyun Jo D, Hwang GH, Yu J, Kim JH, Park SE, Kim JS, Kim JH, Bae S. CRISPR-Pass: Gene Rescue of Nonsense Mutations Using Adenine Base Editors. Mol Ther 2019; 27:1364-1371. [PMID: 31164261 PMCID: PMC6698196 DOI: 10.1016/j.ymthe.2019.05.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/08/2023] Open
Abstract
A nonsense mutation is a substitutive mutation in a DNA sequence that causes a premature termination during translation and produces stalled proteins, resulting in dysfunction of a gene. Although it usually induces severe genetic disorders, there are no definite methods for inducing read through of premature termination codons (PTCs). Here, we present a targeted tool for bypassing PTCs, named CRISPR-pass, that uses CRISPR-mediated adenine base editors. CRISPR-pass, which should be applicable to 95.5% of clinically significant nonsense mutations in the ClinVar database, rescues protein synthesis in patient-derived fibroblasts, suggesting potential clinical utility.
Collapse
Affiliation(s)
- Choongil Lee
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea; Center for Genome Engineering, Institute for Basic Science, Seoul 08826, South Korea
| | - Dong Hyun Jo
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea
| | - Gue-Ho Hwang
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
| | - Jihyeon Yu
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea; Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea
| | - Jin Hyoung Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea
| | - Se-Eun Park
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
| | - Jin-Soo Kim
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea; Center for Genome Engineering, Institute for Basic Science, Seoul 08826, South Korea
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea; Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, South Korea.
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea; Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
40
|
Andrews NP, Boeckman JX, Manning CF, Nguyen JT, Bechtold H, Dumitras C, Gong B, Nguyen K, van der List D, Murray KD, Engebrecht J, Trimmer JS. A toolbox of IgG subclass-switched recombinant monoclonal antibodies for enhanced multiplex immunolabeling of brain. eLife 2019; 8:43322. [PMID: 30667360 PMCID: PMC6377228 DOI: 10.7554/elife.43322] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/21/2019] [Indexed: 11/17/2022] Open
Abstract
Generating recombinant monoclonal antibodies (R-mAbs) from mAb-producing hybridomas offers numerous advantages that increase the effectiveness, reproducibility, and transparent reporting of research. We report here the generation of a novel resource in the form of a library of recombinant R-mAbs validated for neuroscience research. We cloned immunoglobulin G (IgG) variable domains from cryopreserved hybridoma cells and input them into an integrated pipeline for expression and validation of functional R-mAbs. To improve efficiency over standard protocols, we eliminated aberrant Sp2/0-Ag14 hybridoma-derived variable light transcripts using restriction enzyme treatment. Further, we engineered a plasmid backbone that allows for switching of the IgG subclasses without altering target binding specificity to generate R-mAbs useful in simultaneous multiplex labeling experiments not previously possible. The method was also employed to rescue IgG variable sequences and generate functional R-mAbs from a non-viable cryopreserved hybridoma. All R-mAb sequences and plasmids will be archived and disseminated from open source suppliers. The immune system fights off disease-causing microbes using antibodies: Y-shaped proteins that each bind to a specific foreign molecule. Indeed, these proteins bind so tightly and so specifically that they can pick out a single target in a complex mixture of different molecules. This property also makes them useful in research. For example, neurobiologists can use antibodies to mark target proteins in thin sections of brain tissue. This reveals their position inside brain cells, helping to link the structure of the brain to the roles the different parts of this structure perform. To use antibodies in this way, scientists need to be able to produce them in large quantities without losing their target specificity. The most common way to do this is with cells called hybridomas. A hybridoma is a hybrid of an antibody-producing immune cell and a cancer cell, and it has properties of both. From the immune cell, it inherits the genes to make a specific type of antibody. From the cancer cell, it inherits the ability to go on dividing forever. In theory, hybridomas should be immortal antibody factories, but they have some limitations. They are expensive to keep alive, hard to transport between labs, and their genes can be unstable. Problems can creep into their genetic code, halting their growth or changing the targets their antibodies recognize. When this happens, scientists can lose vital research tools. Instead of keeping the immune cells alive, an alternative approach is to make recombinant antibodies. Rather than store the whole cell, this approach just stores the parts of the genes that encode antibody target-specificity. Andrews et al. set out to convert a valuable toolbox of neuroscience antibodies into recombinant form. This involved copying the antibody genes from a large library of preserved hybridoma cells. However, many hybridomas also carry genes that produce non-functional antibodies. A step in the process removed these DNA sequences, ensuring that only working antibodies made it into the final library. Using frozen cells made it possible to recover antibody genes from hybridoma cells that could no longer grow. The recombinant DNA sequences provide a permanent record of useful antibodies. Not only does this prevent the loss of research tools, it is also much more shareable than living cells. Modifications to the DNA sequences in the library allow for the use of many antibodies at once. This could help when studying the interactions between different molecules in the brain. Toolkits like these could also make it easier to collaborate, and to reproduce data gathered by different researchers around the world.
Collapse
Affiliation(s)
- Nicolas P Andrews
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Justin X Boeckman
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Colleen F Manning
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Joe T Nguyen
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Hannah Bechtold
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Camelia Dumitras
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Belvin Gong
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Kimberly Nguyen
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Deborah van der List
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - Karl D Murray
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, United States.,Department of Physiology and Membrane Biology, University of California, Davis, United States
| |
Collapse
|
41
|
Callegari A, Sieben C, Benke A, Suter DM, Fierz B, Mazza D, Manley S. Single-molecule dynamics and genome-wide transcriptomics reveal that NF-kB (p65)-DNA binding times can be decoupled from transcriptional activation. PLoS Genet 2019; 15:e1007891. [PMID: 30653501 PMCID: PMC6353211 DOI: 10.1371/journal.pgen.1007891] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/30/2019] [Accepted: 12/12/2018] [Indexed: 12/27/2022] Open
Abstract
Transcription factors (TFs) regulate gene expression in both prokaryotes and eukaryotes by recognizing and binding to specific DNA promoter sequences. In higher eukaryotes, it remains unclear how the duration of TF binding to DNA relates to downstream transcriptional output. Here, we address this question for the transcriptional activator NF-κB (p65), by live-cell single molecule imaging of TF-DNA binding kinetics and genome-wide quantification of p65-mediated transcription. We used mutants of p65, perturbing either the DNA binding domain (DBD) or the protein-protein transactivation domain (TAD). We found that p65-DNA binding time was predominantly determined by its DBD and directly correlated with its transcriptional output as long as the TAD is intact. Surprisingly, mutation or deletion of the TAD did not modify p65-DNA binding stability, suggesting that the p65 TAD generally contributes neither to the assembly of an "enhanceosome," nor to the active removal of p65 from putative specific binding sites. However, TAD removal did reduce p65-mediated transcriptional activation, indicating that protein-protein interactions act to translate the long-lived p65-DNA binding into productive transcription.
Collapse
Affiliation(s)
- Andrea Callegari
- Institute of Physics, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Current address: EMBL Heidelberg, Germany
| | - Christian Sieben
- Institute of Physics, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Swiss National Centre for Competence in Research (NCCR) in Chemical Biology
| | - Alexander Benke
- Institute of Physics, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - David M Suter
- UPSUTER, The Institute of Bioengineering (IBI), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Beat Fierz
- Swiss National Centre for Competence in Research (NCCR) in Chemical Biology.,Institute of Chemical Sciences and Engineering (ISIC), School of Basic Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Davide Mazza
- Fondazione CEN, European Center for Nanomedicine Milano, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute Milano, Italy
| | - Suliana Manley
- Institute of Physics, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,UPSUTER, The Institute of Bioengineering (IBI), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
42
|
Zhong X, Ma W, Meade CL, Tam AS, Llewellyn E, Cornell R, Cote K, Scarcelli JJ, Marshall JK, Tzvetkova B, Figueroa B, DiNino D, Sievers A, Lee C, Guo J, Mahan E, Francis C, Lam K, D'Antona AM, Zollner R, Zhu HL, Kriz R, Somers W, Lin L. Transient CHO expression platform for robust antibody production and its enhanced N-glycan sialylation on therapeutic glycoproteins. Biotechnol Prog 2018; 35:e2724. [PMID: 30299005 DOI: 10.1002/btpr.2724] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/24/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022]
Abstract
Large-scale transient expression in mammalian cells is a rapid protein production technology often used to shorten overall timelines for biotherapeutics drug discovery. In this study we demonstrate transient expression in a Chinese hamster ovary (CHO) host (ExpiCHO-S™) cell line capable of achieving high recombinant antibody expression titers, comparable to levels obtained using human embryonic kidney (HEK) 293 cells. For some antibodies, ExpiCHO-S™ cells generated protein materials with better titers and improved protein quality characteristics (i.e., less aggregation) than those from HEK293. Green fluorescent protein imaging data indicated that ExpiCHO-S™ displayed a delayed but prolonged transient protein expression process compared to HEK293. When therapeutic glycoproteins containing non-Fc N-linked glycans were expressed in transient ExpiCHO-S™, the glycan pattern was unexpectedly found to have few sialylated N-glycans, in contrast to glycans produced within a stable CHO expression system. To improve N-glycan sialylation in transient ExpiCHO-S™, we co-transfected galactosyltransferase and sialyltransferase genes along with the target genes, as well as supplemented the culture medium with glycan precursors. The authors have demonstrated that co-transfection of glycosyltransferases combined with medium addition of galactose and uridine led to increased sialylation content of N-glycans during transient ExpiCHO-S™ expression. These results have provided a scientific basis for developing a future transient CHO system with N-glycan compositions that are similar to those profiles obtained from stable CHO protein production systems. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 35: e2724, 2019.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Weijun Ma
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Caryl L Meade
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Amy S Tam
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Eliza Llewellyn
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Richard Cornell
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Kaffa Cote
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - John J Scarcelli
- Cell Line Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Jeffrey K Marshall
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Boriana Tzvetkova
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Bruno Figueroa
- Bioprocessing Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Dana DiNino
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Annette Sievers
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Christopher Lee
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Jane Guo
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Evan Mahan
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Christopher Francis
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Khetemenee Lam
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Aaron M D'Antona
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Richard Zollner
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Hongli L Zhu
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Ron Kriz
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Will Somers
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Laura Lin
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| |
Collapse
|
43
|
Barb AW, Falconer DJ, Subedi GP. The Preparation and Solution NMR Spectroscopy of Human Glycoproteins Is Accessible and Rewarding. Methods Enzymol 2018; 614:239-261. [PMID: 30611426 DOI: 10.1016/bs.mie.2018.08.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The majority of proteins excreted by human cells and borne at the cell surface are modified with carbohydrates. Glycoproteins mediate a wide range of processes and adopt fundamental roles in many diseases. The carbohydrates covalently attached to proteins during maturation in the cell directly impact protein structure and function as integral and indispensable components. However, the ability to study the structure of glycoproteins to high resolution was historically limited by technical barriers including a limited availability of appropriate recombinant protein expression platforms, limited methods to generate compositional homogeneity, and difficulties analyzing glycoprotein composition. Furthermore, glycoproteins and in particular the glycan moieties themselves often exhibit a high degree of conformational heterogeneity. Solution NMR spectroscopy is a powerful tool to study biological macromolecules that is capable of characterizing mobile elements of molecules with atomic-level resolution. Methods to express glycoproteins, incorporate stable isotope labels, and analyze glycoproteins have recently opened new avenues to prepare and investigate glycoproteins. These methods are accessible to many laboratories with experience expressing and purifying proteins from prokaryotic expression hosts.
Collapse
Affiliation(s)
- Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States.
| | - Daniel J Falconer
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Ganesh P Subedi
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|
44
|
Silva MM, Gomes-Alves P, Rosa S, Simão D, Inácio JM, Peixoto C, Serra M, Belo JA, Alves PM. Full-length human CCBE1 production and purification: leveraging bioprocess development for high quality glycosylation attributes and functionality. J Biotechnol 2018; 285:6-14. [PMID: 30165116 DOI: 10.1016/j.jbiotec.2018.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/21/2018] [Accepted: 08/26/2018] [Indexed: 11/25/2022]
Abstract
Collagen and calcium-binding EGF domain-1 (CCBE1) is a secreted protein critical for lymphatic/cardiac vascular development and regeneration. However, the low efficient production of the recombinant full-length CCBE1 (rCCBE1) has been a setback for functional studies and therapeutic applications using this protein. The main goal of this work was to implement a robust bioprocess for efficient production of glycosylated rCCBE1. Different bioprocess strategies were combined with proteomic tools for process/product characterization, evaluating the impact of process parameters on cell performance, rCCBE1 production and quality. We have shown that rCCBE1 volumetric yield was positively correlated with higher cell density at transfection (HDT), and under these conditions the secreted protein presented a mature glycosylated profile (complex N-glycans). Mild hypothermia was also applied to HDT condition that resulted in enhanced cell viability; however an enrichment of immature rCCBE1 variants was detected. Mass spectrometry-based tools allowed the identification of rCCBE1 peptides confirming protein identity in the affinity chromatography enriched product. rCCBE1 biological activity was validated by in vitro angiogenesis assay, where enhanced vessel formation was observed. Herein, we report a step forward in the production and characterization of human glycosylated rCCBE1, amenable for in vitro and in vivo studies to explore its regenerative therapeutic potential.
Collapse
Affiliation(s)
- Marta M Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; Stem Cells and Development Laboratory, CEDOC, NOVA Medical School, Lisboa, Portugal
| | - Patrícia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sara Rosa
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Daniel Simão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - José M Inácio
- Stem Cells and Development Laboratory, CEDOC, NOVA Medical School, Lisboa, Portugal
| | - Cristina Peixoto
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - José A Belo
- Stem Cells and Development Laboratory, CEDOC, NOVA Medical School, Lisboa, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
45
|
Sun L, Ishihara M, Middleton DR, Tiemeyer M, Avci FY. Metabolic labeling of HIV-1 envelope glycoprotein gp120 to elucidate the effect of gp120 glycosylation on antigen uptake. J Biol Chem 2018; 293:15178-15194. [PMID: 30115684 DOI: 10.1074/jbc.ra118.004798] [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: 07/09/2018] [Revised: 08/12/2018] [Indexed: 12/21/2022] Open
Abstract
The glycan shield on the envelope glycoprotein gp120 of HIV-1 has drawn immense attention as a vulnerable site for broadly neutralizing antibodies and for its significant impact on host adaptive immune response to HIV-1. Glycosylation sites and glycan composition/structure at each site on gp120 along with the interactions of gp120 glycan shield with broadly neutralizing antibodies have been extensively studied. However, a method for directly and selectively tracking gp120 glycans has been lacking. Here, we integrate metabolic labeling and click chemistry technology with recombinant gp120 expression to demonstrate that gp120 glycans could be specifically labeled and directly detected. Selective labeling of gp120 by N-azidoacetylmannosamine (ManNAz) and N-azidoacetylgalactosamine (GalNAz) incorporation into the gp120 glycan shield was characterized by MS of tryptic glycopeptides. By using metabolically labeled gp120, we investigated the impact of gp120 glycosylation on its interaction with host cells and demonstrated that oligomannose enrichment and sialic acid deficiency drastically enhanced gp120 uptake by bone marrow-derived dendritic cells. Collectively, our data reveal an effective labeling and detection method for gp120, serving as a tool for functional characterization of the gp120 glycans and potentially other glycosylated proteins.
Collapse
Affiliation(s)
- Lina Sun
- From the Department of Biochemistry and Molecular Biology, Center for Molecular Medicine and
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Dustin R Middleton
- From the Department of Biochemistry and Molecular Biology, Center for Molecular Medicine and
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Fikri Y Avci
- From the Department of Biochemistry and Molecular Biology, Center for Molecular Medicine and .,Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
46
|
Yuan J, W. Xu W, Jiang S, Yu H, Fai Poon H. The Scattered Twelve Tribes of HEK293. ACTA ACUST UNITED AC 2018. [DOI: 10.13005/bpj/1414] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Their ease of growth and transfection makes HEK293 cells a common cell culture in academic research. In addition, high transfection efficiency of HEK293 cells enable production of exogenous proteins or viruses for pharmaceutical and biomedical research purposes. Recently, HEK293 cells has gained attention due to it is versatility for transfection experiments, particularly the propagation of adenoviral-based and retroviral-based vectors during CART-T bioprocess. Since traceability is critical to pharmaceutical manufacturing process, we provide a mini review to clarify the historical development and intent use of different variants of HEK293 cells. This review should provide a key reference for the HEK293 variants’ historical and developmental background.
Collapse
Affiliation(s)
- Jun Yuan
- Quacell Biotechnology Co., Ltd., Guangdong, China
| | - Wayne W. Xu
- Quacell Biotechnology Co., Ltd., Guangdong, China
| | - Snake Jiang
- Quacell Biotechnology Co., Ltd., Guangdong, China
| | - Henry Yu
- Quacell Biotechnology Co., Ltd., Guangdong, China
| | - H. Fai Poon
- Quacell Biotechnology Co., Ltd., Guangdong, China
| |
Collapse
|
47
|
Sasso E, Latino D, Froechlich G, Succoio M, Passariello M, De Lorenzo C, Nicosia A, Zambrano N. A long non-coding SINEUP RNA boosts semi-stable production of fully human monoclonal antibodies in HEK293E cells. MAbs 2018; 10:730-737. [PMID: 29658818 PMCID: PMC6150626 DOI: 10.1080/19420862.2018.1463945] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Use of monoclonal antibodies is emerging as a highly promising and fast-developing scenario for innovative treatment of viral, autoimmune and tumour diseases. The search for diagnostic and therapeutic antibodies currently depends on in vitro screening approaches, such as phage and yeast display technologies. Antibody production still represents a critical step for preclinical and clinical evaluations. Accordingly, improving production of monoclonal antibodies represents an opportunity, to facilitate downstream target validations. SINEUP RNAs are long non-coding transcripts, possessing the ability to enhance translation of selected mRNAs. We applied SINEUP technology to semi-stable production of monoclonal antibodies in HEK293E cells, which allows for episomal propagation of the expression vectors encoding the heavy and light chains of IgGs. Co-expression of SINEUP RNA with mRNAs encoding heavy and light chains of IgG4s was able to increase the production of different anti-CLDN1 antibodies up to three-fold. Improved production of monoclonal antibodies was achieved both in transiently transfected HEK293E cells and in cellular clones with stable expression of the SINEUP. Compared to antibody preparations obtained under standard conditions, the anti-CLDN1 IgG4s produced in the presence of the SINEUP transcript showed unaltered post-translational modifications, and retained the ability to recognize their target. We thus propose SINEUP technology as a valuable tool to enhance semi-stable antibody production in human cell lines.
Collapse
Affiliation(s)
- Emanuele Sasso
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy.,c Associazione Culturale DiSciMuS RFC , Casoria , NA , Italy
| | - Debora Latino
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy
| | - Guendalina Froechlich
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy
| | - Mariangela Succoio
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy.,c Associazione Culturale DiSciMuS RFC , Casoria , NA , Italy
| | - Margherita Passariello
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy
| | - Claudia De Lorenzo
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy
| | - Alfredo Nicosia
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy.,d ReiThera S.R.L. Roma , Italy
| | - Nicola Zambrano
- a Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II , Napoli , Italy.,b CEINGE Biotecnologie Avanzate S.C.AR.L. , Napoli , Italy.,c Associazione Culturale DiSciMuS RFC , Casoria , NA , Italy
| |
Collapse
|
48
|
Lee JH, Park JH, Park SH, Kim SH, Kim JY, Min JK, Lee GM, Kim YG. Co-amplification of EBNA-1 and PyLT through dhfr-mediated gene amplification for improving foreign protein production in transient gene expression in CHO cells. Appl Microbiol Biotechnol 2018; 102:4729-4739. [PMID: 29654557 DOI: 10.1007/s00253-018-8977-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/23/2018] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
Despite the relatively low transfection efficiency and low specific foreign protein productivity (qp) of Chinese hamster ovary (CHO) cell-based transient gene expression (TGE) systems, TGE-based recombinant protein production technology predominantly employs CHO cells for pre-clinical research and development purposes. To improve TGE in CHO cells, Epstein-Barr virus nuclear antigen-1 (EBNA-1)/polyoma virus large T antigen (PyLT)-co-amplified recombinant CHO (rCHO) cells stably expressing EBNA-1 and PyLT were established using dihydrofolate reductase/methotrexate-mediated gene amplification. The level of transiently expressed Fc-fusion protein was significantly higher in the EBNA-1/PyLT-co-amplified pools compared to control cultures. Increased Fc-fusion protein production by EBNA-1/PyLT-co-amplification resulted from a higher qp attributable to EBNA-1 but not PyLT expression. The qp for TGE-based production with EBNA-1/PyLT-co-amplified rCHO cells (EP-amp-20) was approximately 22.9-fold that of the control culture with CHO-DG44 cells. Rather than improved transfection efficiency, this cell line demonstrated increased levels of mRNA expression and replicated DNA, contributing to an increased qp. Furthermore, there was no significant difference in N-glycan profiles in Fc-fusion proteins produced in the TGE system. Taken together, these results showed that the use of rCHO cells with co-amplified expression of the viral elements EBNA-1 and PyLT improves TGE-based therapeutic protein production dramatically. Therefore, EBNA-1/PyLT-co-amplified rCHO cells will likely be useful as host cells in CHO cell-based TGE systems.
Collapse
Affiliation(s)
- Joo-Hyoung Lee
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.,Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - Jong-Ho Park
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.,Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - Sun-Hye Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.,Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Sun-Hong Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.,Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Jee Yon Kim
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea.
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea. .,Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea.
| |
Collapse
|
49
|
Fuenmayor J, Cervera L, Rigau C, Gòdia F. Enhancement of HIV-1 VLP production using gene inhibition strategies. Appl Microbiol Biotechnol 2018; 102:4477-4487. [DOI: 10.1007/s00253-018-8930-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 10/17/2022]
|
50
|
Mansouri M, Berger P. Multigene delivery in mammalian cells: Recent advances and applications. Biotechnol Adv 2018; 36:871-879. [PMID: 29374595 DOI: 10.1016/j.biotechadv.2018.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 12/27/2022]
Abstract
Systems for multigene delivery in mammalian cells, particularly in the context of genome engineering, have gained a lot of attention in biomolecular research and medicine. Initially these methods were based on RNA polymerase II promoters and were used for the production of protein complexes and for applications in cell biology such as reprogramming of somatic cells to stem cells. Emerging technologies such as CRISPR/Cas9-based genome engineering, which enable any alteration at the genomic level of an organism, require additional elements including U6-driven expression cassettes for RNA expression and homology constructs for designed genome modifications. For these applications, systems with high DNA capacity, flexibility and transfer rates are needed. In this article, we briefly give an update on some of recent strategies that facilitate multigene assembly and delivery into mammalian cells. Also, we review applications in various fields of biology that rely on multigene delivery systems.
Collapse
Affiliation(s)
- Maysam Mansouri
- Paul Scherrer Institute, Biomolecular Research, Applied Molecular Biology, CH-5232 Villigen, Switzerland
| | - Philipp Berger
- Paul Scherrer Institute, Biomolecular Research, Applied Molecular Biology, CH-5232 Villigen, Switzerland.
| |
Collapse
|