1
|
Nakayama E, Tang B, Stewart R, Cox AL, Yan K, Bishop CR, Dumenil T, Nguyen W, Slonchak A, Sng J, Khromykh AA, Lutzky VP, Rawle DJ, Suhrbier A. Evolution of Zika virus in Rag1-deficient mice selects for unique envelope glycosylation motif mutants that show enhanced replication fitness. Virus Evol 2025; 11:veaf021. [PMID: 40291117 PMCID: PMC12024116 DOI: 10.1093/ve/veaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
N-linked glycosylation of flavivirus envelope proteins is widely viewed as being required for optimal folding, processing and/or transit of envelope proteins, and the assembling virons, through the endoplasmic reticulum (ER) and the Golgi. Zika virus (ZIKV) has a single N-linked envelope glycan located adjacent to the fusion loop. Herein we show that independent serial passage of ZIKVNatal in Rag1 -/- mice for 223 or 386 days generated two unique envelope glycan-deficient mutants, ZIKV-V153D and ZIKV-N154D, respectively. Surprisingly, these mutants grew to titres ∼1 to 2.6 logs higher than the glycosylated parental ZIKVNatal in Vero E6 cells and human brain organoids. RNA-Seq of infected organoids suggested that this increased replication fitness was associated with upregulation of the unfolded protein response (UPR). Cell death, cellular viral RNA, and viral protein levels were not significantly affected, arguing that these glycan mutants enjoyed faster ER/Golgi folding, processing, assembly, transit, and virion egress, assisted by an upregulated UPR. Thus, ZIKV envelope N-linked glycosylation is not essential for promoting envelope folding, assembly, and transit through the ER/Golgi, since aspartic acid (D) substitutions in the glycosylation motif can achieve this with significantly greater efficiency. Instead, the evolution of glycan mutants in Rag1 -/- mice indicates that such envelope glycosylation can have a fitness cost in an environment devoid of virus-specific antibody responses. The V153D and N154D mutations, generated by natural selection in Rag1 -/- mice, have to date not been employed in orthoflavivirus envelope glycosylation studies. Instead, genetic engineering has been used to generate mutant viruses that, for instance, contain a N154A substitution. The latter may impart confounding unfavourable properties, such as envelope protein insolubility, that have a detrimental impact on virus replication. The V153D and N154D substitutions may avoid imparting unfavourable properties by preserving the surface negative charge provided by the glycan moiety in the parental ZIKVNatal envelope protein. In Ifnar1 -/- mice ZIKV-V153D and -N154D showed faster viremia onsets, but reduced viremic periods, than the parental ZIKVNatal, consistent with an established contention that such glycans have evolved to delay neutralizing antibody activity.
Collapse
Affiliation(s)
- Eri Nakayama
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Department of Virology I, National Institute of Infectious Diseases, Shinjuku City, Tokyo 162-0052 Japan
| | - Bing Tang
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Romal Stewart
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Abigail L Cox
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Kexin Yan
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Cameron R Bishop
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Troy Dumenil
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Wilson Nguyen
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| | - Julian Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| | - Viviana P Lutzky
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Daniel J Rawle
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Andreas Suhrbier
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| |
Collapse
|
2
|
Regett N, Dieterle M, Peters F, Deuring M, Stegmaier K, Teleki A, Takors R. Subcellular Fractionation Enables Assessment of Nucleotide Sugar Donors Inside the Golgi Apparatus as a Prerequisite for Unraveling Culture Impacts on Glycoforms of Antibodies. Biotechnol J 2025; 20:e202400678. [PMID: 40123410 PMCID: PMC11931351 DOI: 10.1002/biot.202400678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/30/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
Glycosylation is a critical quality attribute in biopharmaceuticals that influences crucial properties, such as biological activity and blood clearance. Current methods for modeling glycosylation typically rely on imprecise or limited data on nucleotide sugar donor (NSD) dynamics. These methods use in vitro transporter kinetics or flux balance analysis, which overlook the key aspects of metabolic regulation. We devised an integrative workflow for absolute subcellular NSD quantification in both cytoplasm and secretory organelles. Using subcellular fractionation, exhaustive sample extraction, and liquid chromatography triple-quadrupole tandem mass spectrometry, we accurately measured NSD concentrations ranging from 1.6 amol/cell to 3 fmol/cell. As expected, NSD concentration profiles aligned closely with the glycan distributions on antibodies, particularly after nutrient pulsing to stimulate NSD production, showcasing method validity. This method enables empirical observation of compartment-specific NSD dynamics. Thus, this study provides novel insights indicating that N-glycosylation, which governs NSD supply, is primarily regulated within the Golgi apparatus (GA). This method offers a novel tool to obtain sophisticated data for a more efficient optimization of glycosylation processes in production cell lines.
Collapse
Affiliation(s)
- Niklas Regett
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Marcel Dieterle
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Fleur Peters
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Max Deuring
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Kaja Stegmaier
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Attila Teleki
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| | - Ralf Takors
- Institute of Biochemical EngineeringUniversity of StuttgartStuttgartBaden‐WürttembergGermany
| |
Collapse
|
3
|
Flevaris K, Kotidis P, Kontoravdi C. GlyCompute: towards the automated analysis of protein N-linked glycosylation kinetics via an open-source computational framework. Anal Bioanal Chem 2025; 417:957-972. [PMID: 39322800 PMCID: PMC11782420 DOI: 10.1007/s00216-024-05522-3] [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: 05/30/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024]
Abstract
Understanding the complex biosynthetic pathways of glycosylation is crucial for the expanding field of glycosciences. Computer-aided glycosylation analysis has greatly benefited in recent years from the development of tools found in web-based portals and open-source libraries. However, the in silico analysis of cellular glycosylation kinetics is underrepresented in current glycoscience-related tools and databases. This could be partly attributed to the limited accessibility of kinetic models developed using proprietary software and the difficulty in reliably parameterising such models. This work aims to address these challenges by proposing GlyCompute, an open-source framework demonstrating a novel, streamlined approach for the assembly, simulation, and parameterisation of kinetic models of protein N-linked glycosylation. Specifically, given one or more sets of experimentally observed N-glycan structures and their relative abundances, minimum representations of a glycosylation reaction network are generated. The topology of the resulting networks is then used to automatically assemble the material balances and kinetic mechanisms underpinning the mathematical model. To match the experimentally observed relative abundances, a sequential parameter estimation strategy using Bayesian inference is proposed, with stages determined automatically based on the underlying network topology. The proposed framework was tested on a case study involving the simultaneous fitting of the kinetic model to two protein N-linked glycoprofiles produced by the same CHO cell culture, showing good agreement with experimental observations. We envision that GlyCompute could help glycoscientists gain quantitative insights into the effect of enzyme kinetics and their perturbations on experimentally observed glycoprofiles in biomanufacturing and clinical settings.
Collapse
Affiliation(s)
| | - Pavlos Kotidis
- Department of Chemical Engineering, Imperial, London, SW7 2AZ, UK
- Biopharm Process Research, GSK, Stevenage, UK
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial, London, SW7 2AZ, UK.
| |
Collapse
|
4
|
Lane AN, Higashi RM, Fan TWM. Challenges of Spatially Resolved Metabolism in Cancer Research. Metabolites 2024; 14:383. [PMID: 39057706 PMCID: PMC11278851 DOI: 10.3390/metabo14070383] [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: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Stable isotope-resolved metabolomics comprises a critical set of technologies that can be applied to a wide variety of systems, from isolated cells to whole organisms, to define metabolic pathway usage and responses to perturbations such as drugs or mutations, as well as providing the basis for flux analysis. As the diversity of stable isotope-enriched compounds is very high, and with newer approaches to multiplexing, the coverage of metabolism is now very extensive. However, as the complexity of the model increases, including more kinds of interacting cell types and interorgan communication, the analytical complexity also increases. Further, as studies move further into spatially resolved biology, new technical problems have to be overcome owing to the small number of analytes present in the confines of a single cell or cell compartment. Here, we review the overall goals and solutions made possible by stable isotope tracing and their applications to models of increasing complexity. Finally, we discuss progress and outstanding difficulties in high-resolution spatially resolved tracer-based metabolic studies.
Collapse
Affiliation(s)
- Andrew N. Lane
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, USA; (R.M.H.); (T.W.-M.F.)
| | | | | |
Collapse
|
5
|
Lee HM, Park JH, Kim TH, Kim HS, Kim DE, Lee MK, You J, Lee GM, Kim YG. Effects of autophagy-inhibiting chemicals on sialylation of Fc-fusion glycoprotein in recombinant CHO cells. Appl Microbiol Biotechnol 2024; 108:224. [PMID: 38376550 PMCID: PMC10879319 DOI: 10.1007/s00253-024-13059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/27/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
The occurrence of autophagy in recombinant Chinese hamster ovary (rCHO) cell culture has attracted attention due to its effects on therapeutic protein production. Given the significance of glycosylation in therapeutic proteins, this study examined the effects of autophagy-inhibiting chemicals on sialylation of Fc-fusion glycoproteins in rCHO cells. Three chemical autophagy inhibitors known to inhibit different stages were separately treated with two rCHO cell lines that produce the same Fc-fusion glycoprotein derived from DUKX-B11 and DG44. All autophagy inhibitors significantly decreased the sialylation of Fc-fusion glycoprotein in both cell lines. The decrease in sialylation of Fc-fusion glycoprotein is unlikely to be attributed to the release of intracellular enzymes, given the high cell viability and low activity of extracellular sialidases. Interestingly, the five intracellular nucleotide sugars remained abundant in cells treated with autophagy inhibitors. In the mRNA expression profiles of 27 N-glycosylation-related genes using the NanoString nCounter system, no significant differences in gene expression were noted. With the positive effect of supplementing nucleotide sugar precursors on sialylation, attempts were made to enhance the levels of intracellular nucleotide sugars by supplying these precursors. The addition of nucleotide sugar precursors to cultures treated with inhibitors successfully enhanced the sialylation of Fc-fusion glycoproteins compared to the control culture. This was particularly evident under mild stress conditions and not under relatively severe stress conditions, which were characterized by a high decrease in sialylation. These results suggest that inhibiting autophagy in rCHO cell culture decreases sialylation of Fc-fusion glycoprotein by constraining the availability of intracellular nucleotide sugars. KEY POINTS: • The autophagy inhibition in rCHO cell culture leads to a significant reduction in the sialylation of Fc-fusion glycoprotein. • The pool of five intracellular nucleotide sugars remained highly abundant in cells treated with autophagy inhibitors. • Supplementation of nucleotide sugar precursors effectively restores decreased sialylation, particularly under mild stress conditions but not in relatively severe stress conditions.
Collapse
Affiliation(s)
- Hoon-Min Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Jong-Ho Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, Korea
| | - Tae-Ho Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, Korea
| | - Hyun-Seung Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Dae Eung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Korea
| | - Mi Kyeong Lee
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Korea
| | - Jungmok You
- Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea.
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
6
|
Jiménez del Val I, Kyriakopoulos S, Albrecht S, Stockmann H, Rudd PM, Polizzi KM, Kontoravdi C. CHOmpact: A reduced metabolic model of Chinese hamster ovary cells with enhanced interpretability. Biotechnol Bioeng 2023; 120:2479-2493. [PMID: 37272445 PMCID: PMC10952303 DOI: 10.1002/bit.28459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023]
Abstract
Metabolic modeling has emerged as a key tool for the characterization of biopharmaceutical cell culture processes. Metabolic models have also been instrumental in identifying genetic engineering targets and developing feeding strategies that optimize the growth and productivity of Chinese hamster ovary (CHO) cells. Despite their success, metabolic models of CHO cells still present considerable challenges. Genome-scale metabolic models (GeMs) of CHO cells are very large (>6000 reactions) and are difficult to constrain to yield physiologically consistent flux distributions. The large scale of GeMs also makes the interpretation of their outputs difficult. To address these challenges, we have developed CHOmpact, a reduced metabolic network that encompasses 101 metabolites linked through 144 reactions. Our compact reaction network allows us to deploy robust, nonlinear optimization and ensure that the computed flux distributions are physiologically consistent. Furthermore, our CHOmpact model delivers enhanced interpretability of simulation results and has allowed us to identify the mechanisms governing shifts in the anaplerotic consumption of asparagine and glutamate as well as an important mechanism of ammonia detoxification within mitochondria. CHOmpact, thus, addresses key challenges of large-scale metabolic models and will serve as a platform to develop dynamic metabolic models for the control and optimization of biopharmaceutical cell culture processes.
Collapse
Affiliation(s)
| | - Sarantos Kyriakopoulos
- Manufacturing Science and TechnologyBioMarin PharmaceuticalCorkIrelandIreland
- Present address:
Drug Product DevelopmentJanssen PharmaceuticalsSchaffhausenSwitzerland
| | - Simone Albrecht
- GlycoScience GroupNational Institute for Bioprocessing Research and TrainingDublinIreland
| | - Henning Stockmann
- GlycoScience GroupNational Institute for Bioprocessing Research and TrainingDublinIreland
| | - Pauline M. Rudd
- GlycoScience GroupNational Institute for Bioprocessing Research and TrainingDublinIreland
- Present address:
Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Karen M. Polizzi
- Department of Chemical EngineeringImperial College LondonLondonUK
| | - Cleo Kontoravdi
- Department of Chemical EngineeringImperial College LondonLondonUK
| |
Collapse
|
7
|
Enzyme cascades for the synthesis of nucleotide sugars: Updates to recent production strategies. Carbohydr Res 2023; 523:108727. [PMID: 36521208 DOI: 10.1016/j.carres.2022.108727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Nucleotide sugars play an elementary role in nature as building blocks of glycans, polysaccharides, and glycoconjugates used in the pharmaceutical, cosmetics, and food industries. As substrates of Leloir-glycosyltransferases, nucleotide sugars are essential for chemoenzymatic in vitro syntheses. However, high costs and the limited availability of nucleotide sugars prevent applications of biocatalytic cascades on a large industrial scale. Therefore, the focus is increasingly on nucleotide sugar synthesis strategies to make significant application processes feasible. The chemical synthesis of nucleotide sugars and their derivatives is well established, but the yields of these processes are usually low. Enzyme catalysis offers a suitable alternative here, and in the last 30 years, many synthesis routes for nucleotide sugars have been discovered and used for production. However, many of the published procedures shy away from assessing the practicability of their processes. With this review, we give an insight into the development of the (chemo)enzymatic nucleotide sugar synthesis pathways of the last years and present an assessment of critical process parameters such as total turnover number (TTN), space-time yield (STY), and enzyme loading.
Collapse
|
8
|
Losfeld ME, Scibona E, Lin CW, Aebi M. Glycosylation network mapping and site-specific glycan maturation in vivo. iScience 2022; 25:105417. [DOI: 10.1016/j.isci.2022.105417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/12/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
|
9
|
Saghaleyni R, Malm M, Moruzzi N, Zrimec J, Razavi R, Wistbacka N, Thorell H, Pintar A, Hober A, Edfors F, Chotteau V, Berggren PO, Grassi L, Zelezniak A, Svensson T, Hatton D, Nielsen J, Robinson JL, Rockberg J. Enhanced metabolism and negative regulation of ER stress support higher erythropoietin production in HEK293 cells. Cell Rep 2022; 39:110936. [PMID: 35705050 DOI: 10.1016/j.celrep.2022.110936] [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: 09/28/2020] [Revised: 01/05/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Recombinant protein production can cause severe stress on cellular metabolism, resulting in limited titer and product quality. To investigate cellular and metabolic characteristics associated with these limitations, we compare HEK293 clones producing either erythropoietin (EPO) (secretory) or GFP (non-secretory) protein at different rates. Transcriptomic and functional analyses indicate significantly higher metabolism and oxidative phosphorylation in EPO producers compared with parental and GFP cells. In addition, ribosomal genes exhibit specific expression patterns depending on the recombinant protein and the production rate. In a clone displaying a dramatically increased EPO secretion, we detect higher gene expression related to negative regulation of endoplasmic reticulum (ER) stress, including upregulation of ATF6B, which aids EPO production in a subset of clones by overexpression or small interfering RNA (siRNA) knockdown. Our results offer potential target pathways and genes for further development of the secretory power in mammalian cell factories.
Collapse
Affiliation(s)
- Rasool Saghaleyni
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Magdalena Malm
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, 17176 Stockholm, Sweden
| | - Jan Zrimec
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Ronia Razavi
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Num Wistbacka
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Hannes Thorell
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Anton Pintar
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Andreas Hober
- Science for Life Laboratory, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Veronique Chotteau
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Industrial Biotechnology, 106 91 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, 17176 Stockholm, Sweden
| | - Luigi Grassi
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Aleksej Zelezniak
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Thomas Svensson
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, 41258 Gothenburg, Sweden
| | - Diane Hatton
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jonathan L Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, 41258 Gothenburg, Sweden.
| | - Johan Rockberg
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden.
| |
Collapse
|
10
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
11
|
Ludwig SD, Bernstein ZJ, Agatemor C, Dammen-Brower K, Ruffolo J, Rosas JM, Post JD, Cole RN, Yarema KJ, Spangler JB. A versatile design platform for glycoengineering therapeutic antibodies. MAbs 2022; 14:2095704. [PMID: 35815437 PMCID: PMC9272841 DOI: 10.1080/19420862.2022.2095704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/24/2022] [Indexed: 11/21/2022] Open
Abstract
Manipulation of glycosylation patterns, i.e., glycoengineering, is incorporated in the therapeutic antibody development workflow to ensure clinical safety, and this approach has also been used to modulate the biological activities, functions, or pharmacological properties of antibody drugs. Whereas most existing glycoengineering strategies focus on the canonical glycans found in the constant domain of immunoglobulin G (IgG) antibodies, we report a new strategy to leverage the untapped potential of atypical glycosylation patterns in the variable domains, which naturally occur in 15% to 25% of IgG antibodies. Glycosylation sites were added to the antigen-binding regions of two functionally divergent, interleukin-2-binding monoclonal antibodies. We used computational tools to rationally install various N-glycosylation consensus sequences into the antibody variable domains, creating "glycovariants" of these molecules. Strikingly, almost all the glycovariants were successfully glycosylated at their newly installed N-glycan sites, without reduction of the antibody's native function. Importantly, certain glycovariants exhibited modified activities compared to the parent antibody, showing the potential of our glycoengineering strategy to modulate biological function of antibodies involved in multi-component receptor systems. Finally, when coupled with a high-flux sialic acid precursor, a glycovariant with two installed glycosylation sites demonstrated superior in vivo half-life. Collectively, these findings validate a versatile glycoengineering strategy that introduces atypical glycosylation into therapeutic antibodies in order to improve their efficacy and, in certain instances, modulate their activity early in the drug development process.
Collapse
Affiliation(s)
- Seth D. Ludwig
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zachary J. Bernstein
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christian Agatemor
- 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
| | - Kris Dammen-Brower
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Ruffolo
- Program in Molecular Biophysics, the Johns Hopkins University, Baltimore, MD, USA
| | - Jonah M. Rosas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy D. Post
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert N. Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin J. Yarema
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- 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 Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg–Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jamie B. Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- 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 Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg–Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Salim T, Chauhan G, Templeton N, Ling WLW. Using MVDA with stoichiometric balances to optimize amino acid concentrations in chemically defined CHO cell culture medium for improved culture performance. Biotechnol Bioeng 2021; 119:452-469. [PMID: 34811720 DOI: 10.1002/bit.27998] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/22/2021] [Accepted: 11/13/2021] [Indexed: 11/07/2022]
Abstract
Chemically defined (CD) media are routinely used in the production of biologics in Chinese hamster ovary (CHO) cell culture and provide enhanced raw material control. Nutrient optimized CD media is an important path to increase cell growth and monoclonal antibody (mAb) productivity in recombinant CHO cell lines. However, nutrient optimization efforts for CD media typically rely on multifactorial and experimental design of experiment approaches or complex mathematical models of cellular metabolism or gene expression systems. Moreover, the majority of these efforts are aimed at amino acids since they constitute essential nutrients in CD media as they directly contribute to biomass and protein production. In this study, we demonstrate the utilization of multivariate data analytics (MVDA) coupled with amino acid stoichiometric balances (SBs) to increased cell growth and mAb productivity in efforts to support CD media development efforts. SBs measure the difference between theoretical demand of amino acids and the empirically measured fluxes to identify various catabolic or anabolic states of the cell. When coupled with MVDA, the statistical models were not only able to highlight key amino acids toward cell growth or productivity, but also provided direction on metabolic favorability of the amino acid. Experimental validation of our approach resulted in a 55% increase in total cell growth and about an 80% increase in total mAb productivity. Increased specific consumption of stoichiometrically balanced amino acids and decreased specific consumption of glucose was also observed in optimized CD media suggesting favorable consumption of desired nutrients and a potential for energy redistribution toward increased cellular growth and mAb productivity.
Collapse
Affiliation(s)
- Taha Salim
- Merck & Co. Inc., Kenilworth, New Jersey, USA
- Taha Salim, Regeneron, Tarrytown, New York, USA
| | | | | | - Wai Lam W Ling
- Merck & Co. Inc., Kenilworth, New Jersey, USA
- Wai L. W. Ling, Rocket Pharma, Cranbury, New Jersey, USA
| |
Collapse
|
13
|
Antonakoudis A, Strain B, Barbosa R, Jimenez del Val I, Kontoravdi C. Synergising stoichiometric modelling with artificial neural networks to predict antibody glycosylation patterns in Chinese hamster ovary cells. Comput Chem Eng 2021. [DOI: 10.1016/j.compchemeng.2021.107471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Kellman BP, Lewis NE. Big-Data Glycomics: Tools to Connect Glycan Biosynthesis to Extracellular Communication. Trends Biochem Sci 2021; 46:284-300. [PMID: 33349503 PMCID: PMC7954846 DOI: 10.1016/j.tibs.2020.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022]
Abstract
Characteristically, cells must sense and respond to environmental cues. Despite the importance of cell-cell communication, our understanding remains limited and often lacks glycans. Glycans decorate proteins and cell membranes at the cell-environment interface, and modulate intercellular communication, from development to pathogenesis. Providing further challenges, glycan biosynthesis and cellular behavior are co-regulating systems. Here, we discuss how glycosylation contributes to extracellular responses and signaling. We further organize approaches for disentangling the roles of glycans in multicellular interactions using newly available datasets and tools, including glycan biosynthesis models, omics datasets, and systems-level analyses. Thus, emerging tools in big data analytics and systems biology are facilitating novel insights on glycans and their relationship with multicellular behavior.
Collapse
Affiliation(s)
- Benjamin P Kellman
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA; Department of Bioengineering, University of California San Diego School of Medicine, La Jolla, CA, USA; Bioinformatics and Systems Biology Program, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA; Department of Bioengineering, University of California San Diego School of Medicine, La Jolla, CA, USA; Bioinformatics and Systems Biology Program, University of California San Diego School of Medicine, La Jolla, CA, USA; Novo Nordisk Foundation Center for Biosustainability at the University of California San Diego School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
15
|
Jalali A, Bandehpour M, Chegeni R, Ghanbarian H, Mardani-Dashti Y, Zarkesh-Esfahani SH, Kazemi B. Expression, Purification, and Evaluation of Anti-IL-2Rα Antibody Secreted by Leishmania tarentolae. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-020-10088-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
Kellman BP, Zhang Y, Logomasini E, Meinhardt E, Godinez-Macias KP, Chiang AWT, Sorrentino JT, Liang C, Bao B, Zhou Y, Akase S, Sogabe I, Kouka T, Winzeler EA, Wilson IBH, Campbell MP, Neelamegham S, Krambeck FJ, Aoki-Kinoshita KF, Lewis NE. A consensus-based and readable extension of Linear Code for Reaction Rules (LiCoRR). Beilstein J Org Chem 2020; 16:2645-2662. [PMID: 33178355 PMCID: PMC7607430 DOI: 10.3762/bjoc.16.215] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022] Open
Abstract
Systems glycobiology aims to provide models and analysis tools that account for the biosynthesis, regulation, and interactions with glycoconjugates. To facilitate these methods, there is a need for a clear glycan representation accessible to both computers and humans. Linear Code, a linearized and readily parsable glycan structure representation, is such a language. For this reason, Linear Code was adapted to represent reaction rules, but the syntax has drifted from its original description to accommodate new and originally unforeseen challenges. Here, we delineate the consensuses and inconsistencies that have arisen through this adaptation. We recommend options for a consensus-based extension of Linear Code that can be used for reaction rule specification going forward. Through this extension and specification of Linear Code to reaction rules, we aim to minimize inconsistent symbology thereby making glycan database queries easier. With a clear guide for generating reaction rule descriptions, glycan synthesis models will be more interoperable and reproducible thereby moving glycoinformatics closer to compliance with FAIR standards. Here, we present Linear Code for Reaction Rules (LiCoRR), version 1.0, an unambiguous representation for describing glycosylation reactions in both literature and code.
Collapse
|
17
|
Majewska NI, Tejada ML, Betenbaugh MJ, Agarwal N. N-Glycosylation of IgG and IgG-Like Recombinant Therapeutic Proteins: Why Is It Important and How Can We Control It? Annu Rev Chem Biomol Eng 2020; 11:311-338. [DOI: 10.1146/annurev-chembioeng-102419-010001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Regulatory bodies worldwide consider N-glycosylation to be a critical quality attribute for immunoglobulin G (IgG) and IgG-like therapeutics. This consideration is due to the importance of posttranslational modifications in determining the efficacy, safety, and pharmacokinetic properties of biologics. Given its critical role in protein therapeutic production, we review N-glycosylation beginning with an overview of the myriad interactions of N-glycans with other biological factors. We examine the mechanism and drivers for N-glycosylation during biotherapeutic production and the several competing factors that impact glycan formation, including the abundance of precursor nucleotide sugars, transporters, glycosidases, glycosyltransferases, and process conditions. We explore the role of these factors with a focus on the analytical approaches used to characterize glycosylation and associated processes, followed by the current state of advanced glycosylation modeling techniques. This combination of disciplines allows for a deeper understanding of N-glycosylation and will lead to more rational glycan control.
Collapse
Affiliation(s)
- Natalia I. Majewska
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA;,
- Cell Culture and Fermentation Sciences, AstraZeneca, Gaithersburg, Maryland 20878, USA
| | - Max L. Tejada
- Bioassay, Impurities and Quality, AstraZeneca, Gaithersburg, Maryland 20878, USA
| | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA;,
| | - Nitin Agarwal
- Cell Culture and Fermentation Sciences, AstraZeneca, Gaithersburg, Maryland 20878, USA
| |
Collapse
|
18
|
Sha S, Handelman G, Agarabi C, Yoon S. A high-resolution measurement of nucleotide sugars by using ion-pair reverse chromatography and tandem columns. Anal Bioanal Chem 2020; 412:3683-3693. [PMID: 32300845 DOI: 10.1007/s00216-020-02608-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/08/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
N-Linked glycosylation is a cellular process transferring sugars from glycosyl donors to proteins or lipids. Biopharmaceutical products widely produced by culturing mammalian cells such as Chinese hamster ovary (CHO) cells are typically glycosylated during biosynthesis. For some biologics, the N-linked glycan is a critical quality attribute of the drugs. Nucleotide sugars are the glycan donors and impact the intracellular glycosylation process. In current analytical methods, robust separation of nucleotide sugar isomers such as UDP glucose and UDP galactose remains a challenge because of their structural similarity. In this study, we developed a strategy to resolve the separation of major nucleotide sugars including challenging isomers based on the use of ion-pair reverse phase (IP-RP) chromatography. The strategy applies core-shell columns and connects multiple columns in tandem to increase separation power and ultimately enables high-resolution detection of nucleotide sugars from cell extracts. The key parameters in the IP-RP method, including temperature, mobile phase, and flow rates, have been systematically evaluated in this work and the theoretical mechanisms of the chromatographic behavior were proposed. Graphical abstract.
Collapse
Affiliation(s)
- Sha Sha
- Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Garry Handelman
- Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Cyrus Agarabi
- U.S. FDA, CDER/OBP/Division of Biotechnology Review and Research II, Silver Spring, MD, 20993, USA
| | - Seongkyu Yoon
- Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, MA, 01854, USA. .,Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, 01854, USA.
| |
Collapse
|
19
|
Gutierrez JM, Feizi A, Li S, Kallehauge TB, Hefzi H, Grav LM, Ley D, Baycin Hizal D, Betenbaugh MJ, Voldborg B, Faustrup Kildegaard H, Min Lee G, Palsson BO, Nielsen J, Lewis NE. Genome-scale reconstructions of the mammalian secretory pathway predict metabolic costs and limitations of protein secretion. Nat Commun 2020; 11:68. [PMID: 31896772 PMCID: PMC6940358 DOI: 10.1038/s41467-019-13867-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/22/2019] [Indexed: 01/08/2023] Open
Abstract
In mammalian cells, >25% of synthesized proteins are exported through the secretory pathway. The pathway complexity, however, obfuscates its impact on the secretion of different proteins. Unraveling its impact on diverse proteins is particularly important for biopharmaceutical production. Here we delineate the core secretory pathway functions and integrate them with genome-scale metabolic reconstructions of human, mouse, and Chinese hamster ovary cells. The resulting reconstructions enable the computation of energetic costs and machinery demands of each secreted protein. By integrating additional omics data, we find that highly secretory cells have adapted to reduce expression and secretion of other expensive host cell proteins. Furthermore, we predict metabolic costs and maximum productivities of biotherapeutic proteins and identify protein features that most significantly impact protein secretion. Finally, the model successfully predicts the increase in secretion of a monoclonal antibody after silencing a highly expressed selection marker. This work represents a knowledgebase of the mammalian secretory pathway that serves as a novel tool for systems biotechnology.
Collapse
Affiliation(s)
- Jahir M Gutierrez
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Amir Feizi
- Department of Biology and Biological Engineering, Kemivägen 10, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Shangzhong Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Thomas B Kallehauge
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Lise M Grav
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Daniel Ley
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Department of Systems Biology, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Michael J Betenbaugh
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218-2686, USA
| | - Bjorn Voldborg
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Helene Faustrup Kildegaard
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Gyun Min Lee
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Kemivägen 10, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Kotidis P, Demis P, Goey CH, Correa E, McIntosh C, Trepekli S, Shah N, Klymenko OV, Kontoravdi C. Constrained global sensitivity analysis for bioprocess design space identification. Comput Chem Eng 2019. [DOI: 10.1016/j.compchemeng.2019.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
21
|
Kotidis P, Jedrzejewski P, Sou SN, Sellick C, Polizzi K, Del Val IJ, Kontoravdi C. Model-based optimization of antibody galactosylation in CHO cell culture. Biotechnol Bioeng 2019; 116:1612-1626. [PMID: 30802295 DOI: 10.1002/bit.26960] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/22/2019] [Accepted: 02/21/2019] [Indexed: 01/13/2023]
Abstract
Exerting control over the glycan moieties of antibody therapeutics is highly desirable from a product safety and batch-to-batch consistency perspective. Strategies to improve antibody productivity may compromise quality, while interventions for improving glycoform distribution can adversely affect cell growth and productivity. Process design therefore needs to consider the trade-off between preserving cellular health and productivity while enhancing antibody quality. In this work, we present a modeling platform that quantifies the impact of glycosylation precursor feeding - specifically that of galactose and uridine - on cellular growth, metabolism as well as antibody productivity and glycoform distribution. The platform has been parameterized using an initial training data set yielding an accuracy of ±5% with respect to glycoform distribution. It was then used to design an optimized feeding strategy that enhances the final concentration of galactosylated antibody in the supernatant by over 90% compared with the control without compromising the integral of viable cell density or final antibody titer. This work supports the implementation of Quality by Design towards higher-performing bioprocesses.
Collapse
Affiliation(s)
- Pavlos Kotidis
- Department of Chemical Engineering, Imperial College London, United Kingdom
| | - Philip Jedrzejewski
- Department of Chemical Engineering, Imperial College London, United Kingdom
- Department of Life Sciences, Imperial College London, United Kingdom
- Centre for Synthetic Biology and Innovation, Imperial College London, United Kingdom
| | - Si Nga Sou
- Department of Chemical Engineering, Imperial College London, United Kingdom
- Department of Life Sciences, Imperial College London, United Kingdom
- Centre for Synthetic Biology and Innovation, Imperial College London, United Kingdom
| | - Christopher Sellick
- Cell Culture and Fermentation Sciences BioPharmaceutical Development, MedImmune, Granta Park, Cambridge, United Kingdom
| | - Karen Polizzi
- Department of Life Sciences, Imperial College London, United Kingdom
- Centre for Synthetic Biology and Innovation, Imperial College London, United Kingdom
| | | | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, United Kingdom
| |
Collapse
|
22
|
Kontoravdi C, Jimenez del Val I. Computational tools for predicting and controlling the glycosylation of biopharmaceuticals. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.08.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Glycosylation Flux Analysis of Immunoglobulin G in Chinese Hamster Ovary Perfusion Cell Culture. Processes (Basel) 2018. [DOI: 10.3390/pr6100176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The terminal sugar molecules of the N-linked glycan attached to the fragment crystalizable (Fc) region is a critical quality attribute of therapeutic monoclonal antibodies (mAbs) such as immunoglobulin G (IgG). There exists naturally-occurring heterogeneity in the N-linked glycan structure of mAbs, and such heterogeneity has a significant influence on the clinical safety and efficacy of mAb drugs. We previously proposed a constraint-based modeling method called glycosylation flux analysis (GFA) to characterize the rates (fluxes) of intracellular glycosylation reactions. One contribution of this work is a significant improvement in the computational efficiency of the GFA, which is beneficial for analyzing large datasets. Another contribution of our study is the analysis of IgG glycosylation in continuous perfusion Chinese Hamster Ovary (CHO) cell cultures. The GFA of the perfusion cell culture data indicated that the dynamical changes of IgG glycan heterogeneity are mostly attributed to alterations in the galactosylation flux activity. By using a random forest regression analysis of the IgG galactosylation flux activity, we were further able to link the dynamics of galactosylation with two process parameters: cell-specific productivity of IgG and extracellular ammonia concentration. The characteristics of IgG galactosylation dynamics agree well with what we previously reported for fed-batch cultivations of the same CHO cell strain.
Collapse
|
24
|
Hu D, Zhao L, Wang J, Fan L, Liu X, Wang H, Tan WS. Physiological responses of Chinese hamster ovary cells to a productivity-enhancing yeast extract. J Biosci Bioeng 2018; 126:636-643. [PMID: 29853300 DOI: 10.1016/j.jbiosc.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/24/2018] [Accepted: 05/05/2018] [Indexed: 12/23/2022]
Abstract
Hydrolysates play important roles in enhancing the productivity of recombinant proteins in mammalian cell cultures. Lacking of detailed understanding of the mechanisms, hydrolysate is commonly regarded as an unstable factor which should be used with cautions. A yeast extract (YE) was approved to improve the Fc-fusion protein productivity in a recombinant Chinese hamster ovary (CHO) cell line. To elucidate the responses of cells to hydrolysates, we further elaborate their physiological changes during the processes in the presence and absence of YE. Firstly, cell sizes and the cellular components including dry cell weight, cellular fatty acid, and total cellular protein were increased in the presence of YE. Then, by comparing the extracellular and intracellular concentrations of the main metabolites and their consumption rates, we excluded the possibility of nutrient depletion in the absence of YE and observed a distinct improvement on the net consumption rates of metabolites in the presence of YE. Furthermore, the increase on the contents of intracellular nucleotides illustrated an abundance of the nucleic acid precursors and energy charge for recombinant protein synthesis in the presence of YE. In conclusion, this study systematically elucidated YE enhanced cell mass and capacity, activated substrate and energy metabolism of cells in addition to a boost in product synthesis process. The findings provide valuable information for process optimization and cell engineering.
Collapse
Affiliation(s)
- Dongdong Hu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiaqi Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Li Fan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xuping Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Haibin Wang
- Zhejiang Hisun Pharmaceutical Co. Ltd., Fuyang, Hangzhou, Zhejiang 311404, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
25
|
Losfeld ME, Scibona E, Lin CW, Villiger TK, Gauss R, Morbidelli M, Aebi M. Influence of protein/glycan interaction on site-specific glycan heterogeneity. FASEB J 2017; 31:4623-4635. [PMID: 28679530 DOI: 10.1096/fj.201700403r] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/19/2017] [Indexed: 01/23/2023]
Abstract
To study how the interaction between N-linked glycans and the surrounding amino acids influences oligosaccharide processing, we used protein disulfide isomerase (PDI), a glycoprotein bearing 5 N-glycosylation sites, as a model system and expressed it transiently in a Chinese hamster ovary (CHO)-S cell line. PDI was produced as both secreted Sec-PDI and endoplasmic reticulum-retained glycoprotein (ER)-PDI, to study glycan processing by ER and Golgi resident enzymes. Quantitative site-specific glycosylation profiles were obtained, and flux analysis enabled modeling site-specific glycan processing. By altering the primary sequence of PDI, we changed the glycan/protein interaction and thus the site-specific glycoprofile because of the improved enzymatic fluxes at enzymatic bottlenecks. Our results highlight the importance of direct interactions between N-glycans and surface-exposed amino acids of glycoproteins on processing in the ER and the Golgi and the possibility of changing a site-specific N-glycan profile by modulating such interactions and thus the associated enzymatic fluxes. Altering the primary protein sequence can therefore be used to glycoengineer recombinant proteins.-Losfeld, M.-E., Scibona, E., Lin, C.-W., Villiger, T. K., Gauss, R., Morbidelli, M., Aebi, M. Influence of protein/glycan interaction on site-specific glycan heterogeneity.
Collapse
Affiliation(s)
- Marie-Estelle Losfeld
- Department of Biology, Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Ernesto Scibona
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology ETH Zürich, Zürich, Switzerland
| | - Chia-Wei Lin
- Department of Biology, Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Thomas K Villiger
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology ETH Zürich, Zürich, Switzerland
| | - Robert Gauss
- Department of Biology, Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Massimo Morbidelli
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology ETH Zürich, Zürich, Switzerland
| | - Markus Aebi
- Department of Biology, Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland;
| |
Collapse
|
26
|
Brühlmann D, Sokolov M, Butté A, Sauer M, Hemberger J, Souquet J, Broly H, Jordan M. Parallel experimental design and multivariate analysis provides efficient screening of cell culture media supplements to improve biosimilar product quality. Biotechnol Bioeng 2017; 114:1448-1458. [DOI: 10.1002/bit.26269] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/01/2017] [Accepted: 02/08/2017] [Indexed: 01/15/2023]
Affiliation(s)
- David Brühlmann
- Merck Biopharma; Biotech Process Sciences; Merck Biopharma; Route de Fenil 25; 1804; Corsier-sur-Vevey Switzerland
- Department of Biotechnology and Biophysics; Biozentrum; Julius-Maximilians-Universität Würzburg; Germany
| | - Michael Sokolov
- Department of Chemistry and Applied Biosciences; Institute of Chemical and Bioengineering; ETH Zürich Switzerland
| | - Alessandro Butté
- Department of Chemistry and Applied Biosciences; Institute of Chemical and Bioengineering; ETH Zürich Switzerland
| | - Markus Sauer
- Department of Biotechnology and Biophysics; Biozentrum; Julius-Maximilians-Universität Würzburg; Germany
| | - Jürgen Hemberger
- Institute for Biochemical Engineering and Analytics; University of Applied Sciences Giessen; Germany
| | - Jonathan Souquet
- Merck Biopharma; Biotech Process Sciences; Merck Biopharma; Route de Fenil 25; 1804; Corsier-sur-Vevey Switzerland
| | - Hervé Broly
- Merck Biopharma; Biotech Process Sciences; Merck Biopharma; Route de Fenil 25; 1804; Corsier-sur-Vevey Switzerland
| | - Martin Jordan
- Merck Biopharma; Biotech Process Sciences; Merck Biopharma; Route de Fenil 25; 1804; Corsier-sur-Vevey Switzerland
| |
Collapse
|
27
|
Galleguillos SN, Ruckerbauer D, Gerstl MP, Borth N, Hanscho M, Zanghellini J. What can mathematical modelling say about CHO metabolism and protein glycosylation? Comput Struct Biotechnol J 2017; 15:212-221. [PMID: 28228925 PMCID: PMC5310201 DOI: 10.1016/j.csbj.2017.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 11/15/2022] Open
Abstract
Chinese hamster ovary cells have been in the spotlight for process optimization in recent years, due to being the major, long established cell factory for the production of recombinant proteins. A deep, quantitative understanding of CHO metabolism and mechanisms involved in protein glycosylation has proven to be attainable through the development of high throughput technologies. Here we review the most notable accomplishments in the field of modelling CHO metabolism and protein glycosylation.
Collapse
Affiliation(s)
- Sarah N Galleguillos
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - David Ruckerbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Matthias P Gerstl
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Michael Hanscho
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Jürgen Zanghellini
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| |
Collapse
|
28
|
McDonald AG, Hayes JM, Davey GP. Metabolic flux control in glycosylation. Curr Opin Struct Biol 2016; 40:97-103. [DOI: 10.1016/j.sbi.2016.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/04/2016] [Accepted: 08/29/2016] [Indexed: 11/17/2022]
|