1
|
Reddy JV, Leibiger T, Singh SK, Lee KH, Papoutsakis E, Ierapetritou M. A Novel, Site-Specific N-Linked Glycosylation Model Provides Mechanistic Insights Into the Process-Condition Dependent Distinct Fab and Fc Glycosylation of an IgG1 Monoclonal Antibody Produced by CHO VRC01 Cells. Biotechnol Bioeng 2025; 122:761-778. [PMID: 39740206 DOI: 10.1002/bit.28916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
The CHO VRC01 cell line produces an anti-HIV IgG1 monoclonal antibody containing N-linked glycans on both the Fab (variable) and Fc (constant) regions. Site-specific glycan analysis was used to measure the complex effects of cell culture process conditions on Fab and Fc glycosylation. Experimental data revealed major differences in glycan fractions across the two sites. Bioreactor pH was found to influence fucosylation, galactosylation, and sialylation in the Fab region and galactosylation in the Fc region. To understand the complex effects of process conditions on site-specific N-linked glycosylation, a kinetic model of site-specific N-linked glycosylation was developed. The model parameters provided mechanistic insights into the differences in glycan fractions observed in the Fc and Fab regions. Enzyme activities calculated from the model provided insights into the effect of bioreactor pH on site-specific N-linked glycosylation. Model predictions were experimentally tested by measuring glycosyltransferase-enzyme mRNA-levels and intracellular nucleotide sugar concentrations. The model was used to demonstrate the effect of increasing galactosyltransferase activity on site-specific N-linked glycan fractions. Experiments involving galactose and MnCl2 supplementation were used to test model predictions. The model is capable of providing insights into experimentally measured data and also of making predictions that can be used to design media supplementation strategies.
Collapse
Affiliation(s)
| | - Thomas Leibiger
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Sumit Kumar Singh
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Eleftherios Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
- Delaware Biotechnology Institute, & Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
2
|
Akune-Taylor Y. Glyco you should know. Glycobiology 2025; 35:cwaf016. [PMID: 40111002 DOI: 10.1093/glycob/cwaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Yukie Akune-Taylor
- Glycan and Life Systems Integration Center, Soka University, 1-236, Tangimachi, Hachioji City, Tokyo, 192-8577, Japan
- Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London
| |
Collapse
|
3
|
Chen X, Li F, Li X, Otto M, Chen Y, Siewers V. Model-assisted CRISPRi/a library screening reveals central carbon metabolic targets for enhanced recombinant protein production in yeast. Metab Eng 2025; 88:1-13. [PMID: 39615667 DOI: 10.1016/j.ymben.2024.11.010] [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] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Production of recombinant proteins is regarded as an important breakthrough in the field of biomedicine and industrial biotechnology. Due to the complexity of the protein secretory pathway and its tight interaction with cellular metabolism, the application of traditional metabolic engineering tools to improve recombinant protein production faces major challenges. A systematic approach is required to generate novel design principles for superior protein secretion cell factories. Here, we applied a proteome-constrained genome-scale protein secretory model of the yeast Saccharomyces cerevisiae (pcSecYeast) to simulate α-amylase production under limited secretory capacity and predict gene targets for downregulation and upregulation to improve α-amylase production. The predicted targets were evaluated using high-throughput screening of specifically designed CRISPR interference/activation (CRISPRi/a) libraries and droplet microfluidics screening. From each library, 200 and 190 sorted clones, respectively, were manually verified. Out of them, 50% of predicted downregulation targets and 34.6% predicted upregulation targets were confirmed to improve α-amylase production. By simultaneously fine-tuning the expression of three genes in central carbon metabolism, i.e. LPD1, MDH1, and ACS1, we were able to increase the carbon flux in the fermentative pathway and α-amylase production. This study exemplifies how model-based predictions can be rapidly validated via a high-throughput screening approach. Our findings highlight novel engineering targets for cell factories and furthermore shed light on the connectivity between recombinant protein production and central carbon metabolism.
Collapse
Affiliation(s)
- Xin Chen
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark.
| | - Feiran Li
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xiaowei Li
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Maximilian Otto
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Yu Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Verena Siewers
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
4
|
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
|
5
|
Kong WZ, Fujita M. GlycoMaple: recent updates and applications in visualization and analysis of glycosylation pathways. Anal Bioanal Chem 2025; 417:885-894. [PMID: 39414644 PMCID: PMC11782371 DOI: 10.1007/s00216-024-05594-1] [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: 08/05/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Post-translational modifications including glycosylation, phosphorylation, and lipidation expand the functionality and diversity of proteins. Protein glycosylation is one of the most abundant post-translational modifications in mammalian cells. The glycosylation process is regulated at multiple steps, including transcription, translation, protein folding, intracellular transport, and localization, and activity of glycosyltransferases and glycoside hydrolases. The glycosylation process is also affected by the concentration of sugar nucleotides in the lumen of the Golgi apparatus. Unlike the synthesis of other biological macromolecules, such as nucleic acids and proteins, glycan biosynthesis is not a template-driven process. In addition, the chemical complexity of glycan structures makes the glycosylation network extraordinarily intricate. We previously developed a web-based tool specially focused on glycan metabolic pathways known as GlycoMaple, which is able to easily visualize and estimate glycosylation pathways based on gene expression data. We recently updated GlycoMaple to incorporate the new genes and glycosylation pathways. Here, we introduce and discuss the uses and upgrades of GlycoMaple.
Collapse
Affiliation(s)
- Wei-Ze Kong
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Morihisa Fujita
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
6
|
Akune-Taylor Y, Kon A, Aoki-Kinoshita KF. In silico simulation of glycosylation and related pathways. Anal Bioanal Chem 2024; 416:3687-3696. [PMID: 38748247 PMCID: PMC11180631 DOI: 10.1007/s00216-024-05331-8] [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: 10/10/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024]
Abstract
Glycans participate in a vast number of recognition systems in diverse organisms in health and in disease. However, glycans cannot be sequenced because there is no sequencer technology that can fully characterize them. There is no "template" for replicating glycans as there are for amino acids and nucleic acids. Instead, glycans are synthesized by a complicated orchestration of multitudes of glycosyltransferases and glycosidases. Thus glycans can vary greatly in structure, but they are not genetically reproducible and are usually isolated in minute amounts. To characterize (sequence) the glycome (defined as the glycans in a particular organism, tissue, cell, or protein), glycosylation pathway prediction using in silico methods based on glycogene expression data, and glycosylation simulations have been attempted. Since many of the mammalian glycogenes have been identified and cloned, it has become possible to predict the glycan biosynthesis pathway in these systems. By then incorporating systems biology and bioprocessing technologies to these pathway models, given the right enzymatic parameters including enzyme and substrate concentrations and kinetic reaction parameters, it is possible to predict the potentially synthesized glycans in the pathway. This review presents information on the data resources that are currently available to enable in silico simulations of glycosylation and related pathways. Then some of the software tools that have been developed in the past to simulate and analyze glycosylation pathways will be described, followed by a summary and vision for the future developments and research directions in this area.
Collapse
Affiliation(s)
- Yukie Akune-Taylor
- Glycan and Life Systems Integration Center, Soka University, Tokyo, Japan
| | - Akane Kon
- Graduate School of Science and Engineering, Soka University, Tokyo, Japan
| | - Kiyoko F Aoki-Kinoshita
- Glycan and Life Systems Integration Center, Soka University, Tokyo, Japan.
- Graduate School of Science and Engineering, Soka University, Tokyo, Japan.
- iGCORE, Nagoya University, Nagoya, Japan.
| |
Collapse
|
7
|
Reddy JV, Raudenbush K, Papoutsakis ET, Ierapetritou M. Cell-culture process optimization via model-based predictions of metabolism and protein glycosylation. Biotechnol Adv 2023; 67:108179. [PMID: 37257729 DOI: 10.1016/j.biotechadv.2023.108179] [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: 11/27/2022] [Revised: 05/18/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
In order to meet the rising demand for biologics and become competitive on the developing biosimilar market, there is a need for process intensification of biomanufacturing processes. Process development of biologics has historically relied on extensive experimentation to develop and optimize biopharmaceutical manufacturing. Experimentation to optimize media formulations, feeding schedules, bioreactor operations and bioreactor scale up is expensive, labor intensive and time consuming. Mathematical modeling frameworks have the potential to enable process intensification while reducing the experimental burden. This review focuses on mathematical modeling of cellular metabolism and N-linked glycosylation as applied to upstream manufacturing of biologics. We review developments in the field of modeling cellular metabolism of mammalian cells using kinetic and stoichiometric modeling frameworks along with their applications to simulate, optimize and improve mechanistic understanding of the process. Interest in modeling N-linked glycosylation has led to the creation of various types of parametric and non-parametric models. Most published studies on mammalian cell metabolism have performed experiments in shake flasks where the pH and dissolved oxygen cannot be controlled. Efforts to understand and model the effect of bioreactor-specific parameters such as pH, dissolved oxygen, temperature, and bioreactor heterogeneity are critically reviewed. Most modeling efforts have focused on the Chinese Hamster Ovary (CHO) cells, which are most commonly used to produce monoclonal antibodies (mAbs). However, these modeling approaches can be generalized and applied to any mammalian cell-based manufacturing platform. Current and potential future applications of these models for Vero cell-based vaccine manufacturing, CAR-T cell therapies, and viral vector manufacturing are also discussed. We offer specific recommendations for improving the applicability of these models to industrially relevant processes.
Collapse
Affiliation(s)
- Jayanth Venkatarama Reddy
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Katherine Raudenbush
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA; Delaware Biotechnology Institute, Department of Biological Sciences, University of Delaware, USA.
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA.
| |
Collapse
|
8
|
Liang C, Chiang AWT, Lewis NE. GlycoMME, a Markov modeling platform for studying N-glycosylation biosynthesis from glycomics data. STAR Protoc 2023; 4:102244. [PMID: 37086409 PMCID: PMC10160804 DOI: 10.1016/j.xpro.2023.102244] [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: 01/26/2023] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023] Open
Abstract
Variations in N-glycosylation, which is crucial to glycoprotein functions, impact many diseases and the safety and efficacy of biotherapeutic drugs. Here, we present a protocol for using GlycoMME (Glycosylation Markov Model Evaluator) to study N-glycosylation biosynthesis from glycomics data. We describe steps for annotating glycomics data and quantifying perturbations to N-glycan biosynthesis with interpretable models. We then detail procedures to predict the impact of mutations in disease or potential glycoengineering strategies in drug development. For complete details on the use and execution of this protocol, please refer to Liang et al. (2020).1.
Collapse
Affiliation(s)
- Chenguang Liang
- Department of Pediatrics, University of California, San Diego, La Jolla, San Diego, CA 92130, USA; Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA 92130, USA
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, La Jolla, San Diego, CA 92130, USA.
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, San Diego, CA 92130, USA; Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA 92130, USA.
| |
Collapse
|
9
|
Modeling N-Glycosylation: A Systems Biology Approach for Evaluating Changes in the Steady-State Organization of Golgi-Resident Proteins. Methods Mol Biol 2022; 2557:663-690. [PMID: 36512244 DOI: 10.1007/978-1-0716-2639-9_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The organization of Golgi-resident proteins is crucial for sorting molecules within the secretory pathway and regulating posttranslational modifications. However, evaluating changes to Golgi organization can be challenging, often requiring extensive experimental investigations. Here, we propose a systems biology approach in which changes to Golgi-resident protein sorting and localization can be deduced using cellular N-glycan profiles as the only experimental input.The approach detailed here utilizes the influence of Golgi organization on N-glycan biosynthesis to investigate the mechanisms involved in establishing and maintaining Golgi organization. While N-glycosylation is carried out in a non-template-driven manner, the distribution of N-glycan biosynthetic enzymes within the Golgi ensures this process is not completely random. Therefore, changes to N-glycan profiles provide clues into how altered cell phenotypes affect the sorting and localization of Golgi-resident proteins. Here, we generate a stochastic simulation of N-glycan biosynthesis to produce a simulated glycan profile similar to that obtained experimentally and then combine this with Bayesian fitting to enable inference of changes in enzyme amounts and localizations. Alterations to Golgi organization are evaluated by calculating how the fitted enzyme parameters shift when moving from simulating the glycan profile of one cellular state (e.g., a wild type) to an altered cellular state (e.g., a mutant). Our approach illustrates how an iterative combination of mathematical systems biology and minimal experimental cell biology can be utilized to maximally integrate biological knowledge to gain insightful knowledge of the underlying mechanisms in a manner inaccessible to either alone.
Collapse
|
10
|
Dworkin LA, Clausen H, Joshi HJ. Applying transcriptomics to studyglycosylation at the cell type level. iScience 2022; 25:104419. [PMID: 35663018 PMCID: PMC9156939 DOI: 10.1016/j.isci.2022.104419] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/30/2022] [Accepted: 05/12/2022] [Indexed: 11/22/2022] Open
Abstract
The complex multi-step process of glycosylation occurs in a single cell, yet current analytics generally cannot measure the output (the glycome) of a single cell. Here, we addressed this discordance by investigating how single cell RNA-seq data can be used to characterize the state of the glycosylation machinery and metabolic network in a single cell. The metabolic network involves 214 glycosylation and modification enzymes outlined in our previously built atlas of cellular glycosylation pathways. We studied differential mRNA regulation of enzymes at the organ and single cell level, finding that most of the general protein and lipid oligosaccharide scaffolds are produced by enzymes exhibiting limited transcriptional regulation among cells. We predict key enzymes within different glycosylation pathways to be highly transcriptionally regulated as regulatable hotspots of the cellular glycome. We designed the Glycopacity software that enables investigators to extract and interpret glycosylation information from transcriptome data and define hotspots of regulation. RNA-seq can provide information on the glycosylation metabolic network state It is possible to readout glycosylation capacity from single cell RNA-seq data Genes regulating the biosynthesis of common glycan scaffolds show little regulation Key enzymes in the glycosylation network are predicted to be regulatable hotspots
Collapse
Affiliation(s)
- Leo Alexander Dworkin
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Hiren Jitendra Joshi
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Corresponding author
| |
Collapse
|
11
|
Improving recombinant protein production by yeast through genome-scale modeling using proteome constraints. Nat Commun 2022; 13:2969. [PMID: 35624178 PMCID: PMC9142503 DOI: 10.1038/s41467-022-30689-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 05/12/2022] [Indexed: 01/20/2023] Open
Abstract
Eukaryotic cells are used as cell factories to produce and secrete multitudes of recombinant pharmaceutical proteins, including several of the current top-selling drugs. Due to the essential role and complexity of the secretory pathway, improvement for recombinant protein production through metabolic engineering has traditionally been relatively ad-hoc; and a more systematic approach is required to generate novel design principles. Here, we present the proteome-constrained genome-scale protein secretory model of yeast Saccharomyces cerevisiae (pcSecYeast), which enables us to simulate and explain phenotypes caused by limited secretory capacity. We further apply the pcSecYeast model to predict overexpression targets for the production of several recombinant proteins. We experimentally validate many of the predicted targets for α-amylase production to demonstrate pcSecYeast application as a computational tool in guiding yeast engineering and improving recombinant protein production. Due to the complexity of the protein secretory pathway, strategy suitable for the production of a certain recombination protein cannot be generalized. Here, the authors construct a proteome-constrained genome-scale protein secretory model for yeast and show its application in the production of different misfolded or recombinant proteins.
Collapse
|
12
|
Flevaris K, Kontoravdi C. Immunoglobulin G N-glycan Biomarkers for Autoimmune Diseases: Current State and a Glycoinformatics Perspective. Int J Mol Sci 2022; 23:5180. [PMID: 35563570 PMCID: PMC9100869 DOI: 10.3390/ijms23095180] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
The effective treatment of autoimmune disorders can greatly benefit from disease-specific biomarkers that are functionally involved in immune system regulation and can be collected through minimally invasive procedures. In this regard, human serum IgG N-glycans are promising for uncovering disease predisposition and monitoring progression, and for the identification of specific molecular targets for advanced therapies. In particular, the IgG N-glycome in diseased tissues is considered to be disease-dependent; thus, specific glycan structures may be involved in the pathophysiology of autoimmune diseases. This study provides a critical overview of the literature on human IgG N-glycomics, with a focus on the identification of disease-specific glycan alterations. In order to expedite the establishment of clinically-relevant N-glycan biomarkers, the employment of advanced computational tools for the interpretation of clinical data and their relationship with the underlying molecular mechanisms may be critical. Glycoinformatics tools, including artificial intelligence and systems glycobiology approaches, are reviewed for their potential to provide insight into patient stratification and disease etiology. Challenges in the integration of such glycoinformatics approaches in N-glycan biomarker research are critically discussed.
Collapse
Affiliation(s)
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
13
|
Bakshi T, Pham D, Kaur R, Sun B. Hidden Relationships between N-Glycosylation and Disulfide Bonds in Individual Proteins. Int J Mol Sci 2022; 23:ijms23073742. [PMID: 35409101 PMCID: PMC8998389 DOI: 10.3390/ijms23073742] [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: 03/08/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
N-Glycosylation (NG) and disulfide bonds (DBs) are two prevalent co/post-translational modifications (PTMs) that are often conserved and coexist in membrane and secreted proteins involved in a large number of diseases. Both in the past and in recent times, the enzymes and chaperones regulating these PTMs have been constantly discovered to directly interact with each other or colocalize in the ER. However, beyond a few model proteins, how such cooperation affects N-glycan modification and disulfide bonding at selective sites in individual proteins is largely unknown. Here, we reviewed the literature to discover the current status in understanding the relationships between NG and DBs in individual proteins. Our results showed that more than 2700 human proteins carry both PTMs, and fewer than 2% of them have been investigated in the associations between NG and DBs. We summarized both these proteins with the reported relationships in the two PTMs and the tools used to discover the relationships. We hope that, by exposing this largely understudied field, more investigations can be encouraged to unveil the hidden relationships of NG and DBs in the majority of membranes and secreted proteins for pathophysiological understanding and biotherapeutic development.
Collapse
Affiliation(s)
- Tania Bakshi
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada;
| | - David Pham
- Department of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada;
| | - Raminderjeet Kaur
- Faculty of Health Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada;
| | - Bingyun Sun
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada;
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Correspondence:
| |
Collapse
|
14
|
Kouka T, Akase S, Sogabe I, Jin C, Karlsson NG, Aoki-Kinoshita KF. Computational Modeling of O-Linked Glycan Biosynthesis in CHO Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061766. [PMID: 35335136 PMCID: PMC8950484 DOI: 10.3390/molecules27061766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 12/03/2022]
Abstract
Glycan biosynthesis simulation research has progressed remarkably since 1997, when the first mathematical model for N-glycan biosynthesis was proposed. An O-glycan model has also been developed to predict O-glycan biosynthesis pathways in both forward and reverse directions. In this work, we started with a set of O-glycan profiles of CHO cells transiently transfected with various combinations of glycosyltransferases. The aim was to develop a model that encapsulated all the enzymes in the CHO transfected cell lines. Due to computational power restrictions, we were forced to focus on a smaller set of glycan profiles, where we were able to propose an optimized set of kinetics parameters for each enzyme in the model. Using this optimized model we showed that the abundance of more processed glycans could be simulated compared to observed abundance, while predicting the abundance of glycans earlier in the pathway was less accurate. The data generated show that for the accurate prediction of O-linked glycosylation, additional factors need to be incorporated into the model to better reflect the experimental conditions.
Collapse
Affiliation(s)
- Thukaa Kouka
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo 192-8577, Japan; (S.A.); (I.S.)
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Correspondence: (T.K.); (K.F.A.-K.)
| | - Sachiko Akase
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo 192-8577, Japan; (S.A.); (I.S.)
| | - Isami Sogabe
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo 192-8577, Japan; (S.A.); (I.S.)
| | - Chunsheng Jin
- Proteomics Core Facility at Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden;
| | - Niclas G. Karlsson
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, 0167 Oslo, Norway;
| | - Kiyoko F. Aoki-Kinoshita
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo 192-8577, Japan; (S.A.); (I.S.)
- Glycan & Life Systems Integration Center (GaLSIC), Soka University, Tokyo 192-8577, Japan
- Correspondence: (T.K.); (K.F.A.-K.)
| |
Collapse
|
15
|
Szkodny AC, Lee KH. Biopharmaceutical Manufacturing: Historical Perspectives and Future Directions. Annu Rev Chem Biomol Eng 2022; 13:141-165. [PMID: 35300518 DOI: 10.1146/annurev-chembioeng-092220-125832] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review describes key milestones related to the production of biopharmaceuticals-therapies manufactured using recombinant DNA technology. The market for biopharmaceuticals has grown significantly since the first biopharmaceutical approval in 1982, and the scientific maturity of the technologies used in their manufacturing processes has grown concomitantly. Early processes relied on established unit operations, with research focused on process scale-up and improved culture productivity. In the early 2000s, changes in regulatory frameworks and the introduction of Quality by Design emphasized the importance of developing manufacturing processes to deliver a desired product quality profile. As a result, companies adopted platform processes and focused on understanding the dynamic interplay between product quality and processing conditions. The consistent and reproducible manufacturing processes of today's biopharmaceutical industry have set high standards for product efficacy, quality, and safety, and as the industry continues to evolve in the coming decade, intensified processing capabilities for an expanded range of therapeutic modalities will likely become routine. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering, Volume 13 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Alana C Szkodny
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA; ;
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA; ;
| |
Collapse
|
16
|
Yadav A, Vagne Q, Sens P, Iyengar G, Rao M. Glycan processing in the Golgi: optimal information coding and constraints on cisternal number and enzyme specificity. eLife 2022; 11:76757. [PMID: 35175197 PMCID: PMC9154746 DOI: 10.7554/elife.76757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Many proteins that undergo sequential enzymatic modification in the Golgi cisternae are displayed at the plasma membrane as cell identity markers. The modified proteins, called glycans, represent a molecular code. The fidelity of this glycan code is measured by how accurately the glycan synthesis machinery realises the desired target glycan distribution for a particular cell type and niche. In this paper, we construct a simplified chemical synthesis model to quantitatively analyse the tradeoffs between the number of cisternae, and the number and specificity of enzymes, required to synthesize a prescribed target glycan distribution of a certain complexity to within a given fidelity. We find that to synthesize complex distributions, such as those observed in real cells, one needs to have multiple cisternae and precise enzyme partitioning in the Golgi. Additionally, for fixed number of enzymes and cisternae, there is an optimal level of specificity (promiscuity) of enzymes that achieves the target distribution with high fidelity. The geometry of the fidelity landscape in the multidimensional space of the number and specificity of enzymes, inter-cisternal transfer rates, and number of cisternae, provides a measure for robustness and identifies stiff and sloppy directions. Our results show how the complexity of the target glycan distribution and number of glycosylation enzymes places functional constraints on the Golgi cisternal number and enzyme specificity.
Collapse
Affiliation(s)
| | - Quentin Vagne
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, Paris, France
| | - Pierre Sens
- Laboratoire Physico Chimie Curie, Institut Curie, CNRS UMR168, Paris, France
| | - Garud Iyengar
- Industrial Engineering and Operations Research, Columbia University, New York, United States
| | - Madan Rao
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Bangalore, India
| |
Collapse
|
17
|
West B, Wood AJ, Ungar D. Computational Modeling of Glycan Processing in the Golgi for Investigating Changes in the Arrangements of Biosynthetic Enzymes. Methods Mol Biol 2022; 2370:209-222. [PMID: 34611871 DOI: 10.1007/978-1-0716-1685-7_10] [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: 06/13/2023]
Abstract
Modeling glycan biosynthesis is becoming increasingly important due to the far-reaching implications that glycosylation can exhibit, from pathologies to biopharmaceutical manufacturing. Here we describe a stochastic simulation approach, to overcome the deterministic nature of previous models, that aims to simulate the action of glycan modifying enzymes to produce a glycan profile. This is then coupled with an approximate Bayesian computation methodology to systematically fit to empirical data in order to determine which set of parameters adequately describes the organization of enzymes within the Golgi. The model is described in detail along with a proof of concept and therapeutic applications.
Collapse
Affiliation(s)
- Ben West
- Department of Biology, University of York, York, UK
| | - A Jamie Wood
- Departments of Biology and Mathematics, University of York, York, UK
| | - Daniel Ungar
- Department of Biology, University of York, York, UK.
| |
Collapse
|
18
|
Mariethoz J, Alocci D, Karlsson NG, Packer NH, Lisacek F. An Interactive View of Glycosylation. Methods Mol Biol 2022; 2370:41-65. [PMID: 34611864 DOI: 10.1007/978-1-0716-1685-7_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] [Indexed: 06/13/2023]
Abstract
The present chapter focuses on the interactive and explorative aspects of bioinformatics resources that have been recently released in glycobiology. The comparative analysis of data in a field where knowledge is scattered, incomplete, and disconnected from main biology requires efficient visualization, integration, and interactive tools that are currently only partially implemented. This overview highlights converging efforts toward building a consistent picture of protein glycosylation.
Collapse
Affiliation(s)
- Julien Mariethoz
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, Geneva, Switzerland
- Computer Science Department, University of Geneva, Geneva, Switzerland
| | - Davide Alocci
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nicolle H Packer
- Department of Molecular Sciences and ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW, Australia
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Frédérique Lisacek
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
19
|
Aoki-Kinoshita KF. Functions of Glycosylation and Related Web Resources for Its Prediction. Methods Mol Biol 2022; 2499:135-144. [PMID: 35696078 DOI: 10.1007/978-1-0716-2317-6_6] [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: 06/15/2023]
Abstract
Glycosylation involves the attachment of carbohydrate sugar chains, or glycans, onto an amino acid residue of a protein. These glycans are often branched structures and serve to modulate the function of proteins. Glycans are synthesized through a complex process of enzymatic reactions that occur in the Golgi apparatus in mammalian systems. Because there is currently no sequencer for glycans, technologies such as mass spectrometry is used to characterize glycans in a biological sample to ascertain its glycome. This is a tedious process that requires high levels of expertise and equipment. Thus, the enzymes that work on glycans, called glycogenes or glycoenzymes, have been studied to better understand glycan function. With the development of glycan-related databases and a glycan repository, bioinformatics approaches have attempted to predict the glycosylation pathway and the glycosylation sites on proteins. This chapter introduces these methods and related Web resources for understanding glycan function.
Collapse
|
20
|
Luo Y, Kurian V, Ogunnaike BA. Bioprocess systems analysis, modeling, estimation, and control. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Aoki-Kinoshita KF. Glycome informatics: using systems biology to gain mechanistic insights into glycan biosynthesis. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Fung Shek C, Kotidis P, Betenbaugh M. Mechanistic and data-driven modeling of protein glycosylation. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
23
|
Štor J, Ruckerbauer DE, Széliová D, Zanghellini J, Borth N. Towards rational glyco-engineering in CHO: from data to predictive models. Curr Opin Biotechnol 2021; 71:9-17. [PMID: 34048995 DOI: 10.1016/j.copbio.2021.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
Metabolic modelling strives to develop modelling approaches that are robust and highly predictive. To achieve this, various modelling designs, including hybrid models, and parameter estimation methods that define the type and number of parameters used in the model, are adapted. Accurate input data play an important role so that the selection of experimental methods that provide input data of the required precision with low measurement errors is crucial. For the biopharmaceutically relevant protein glycosylation, the most prominent available models are kinetic models which are able to capture the dynamic nature of protein N-glycosylation. In this review we focus on how to choose the most suitable model for a specific research question, as well as on parameters and considerations to take into account before planning relevant experiments.
Collapse
Affiliation(s)
- Jerneja Štor
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, A-1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria
| | - David E Ruckerbauer
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria
| | - Diana Széliová
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria
| | - Jürgen Zanghellini
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria.
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, A-1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria.
| |
Collapse
|
24
|
Zhang L, Wang M, Castan A, Hjalmarsson H, Chotteau V. Probabilistic model by Bayesian network for the prediction of antibody glycosylation in perfusion and fed-batch cell cultures. Biotechnol Bioeng 2021; 118:3447-3459. [PMID: 33788254 DOI: 10.1002/bit.27769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 01/01/2023]
Abstract
Glycosylation is a critical quality attribute of therapeutic monoclonal antibodies (mAbs). The glycan pattern can have a large impact on the immunological functions, serum half-life and stability. The medium components and cultivation parameters are known to potentially influence the glycosylation profile. Mathematical modelling provides a strategy for rational design and control of the upstream bioprocess. However, the kinetic models usually contain a very large number of unknown parameters, which limit their practical applications. In this article, we consider the metabolic network of N-linked glycosylation as a Bayesian network (BN) and calculate the fluxes of the glycosylation process as joint probability using the culture parameters as inputs. The modelling approach is validated with data of different Chinese hamster ovary cell cultures in pseudo perfusion, perfusion, and fed batch cultures, all showing very good predictive capacities. In cases where a large number of cultivation parameters is available, it is shown here that principal components analysis can efficiently be employed for a dimension reduction of the inputs compared to Pearson correlation analysis and feature importance by decision tree. The present study demonstrates that BN model can be a powerful tool in upstream process and medium development for glycoprotein productions.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden.,AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH Royal Institute of Technology, Stockholm, Sweden
| | - MingLiang Wang
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH Royal Institute of Technology, Stockholm, Sweden.,Division of Decision and Control System, School of Electrical Engineering and Computer Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | | | - Håkan Hjalmarsson
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH Royal Institute of Technology, Stockholm, Sweden.,Division of Decision and Control System, School of Electrical Engineering and Computer Science, KTH-Royal Institute of Technology, Stockholm, Sweden.,Digital Futures - KTH Royal Institute of Technology, Stockholm, Sweden
| | - Veronique Chotteau
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden.,AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH Royal Institute of Technology, Stockholm, Sweden.,Digital Futures - KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
25
|
Huang YF, Aoki K, Akase S, Ishihara M, Liu YS, Yang G, Kizuka Y, Mizumoto S, Tiemeyer M, Gao XD, Aoki-Kinoshita KF, Fujita M. Global mapping of glycosylation pathways in human-derived cells. Dev Cell 2021; 56:1195-1209.e7. [PMID: 33730547 PMCID: PMC8086148 DOI: 10.1016/j.devcel.2021.02.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/15/2020] [Accepted: 02/12/2021] [Indexed: 01/02/2023]
Abstract
Glycans are one of the fundamental classes of macromolecules and are involved in a broad range of biological phenomena. A large variety of glycan structures can be synthesized depending on tissue or cell types and environmental changes. Here, we developed a comprehensive glycosylation mapping tool, termed GlycoMaple, to visualize and estimate glycan structures based on gene expression. We informatically selected 950 genes involved in glycosylation and its regulation. Expression profiles of these genes were mapped onto global glycan metabolic pathways to predict glycan structures, which were confirmed using glycomic analyses. Based on the predictions of N-glycan processing, we constructed 40 knockout HEK293 cell lines and analyzed the effects of gene knockout on glycan structures. Finally, the glycan structures of 64 cell lines, 37 tissues, and primary colon tumor tissues were estimated and compared using publicly available databases. Our systematic approach can accelerate glycan analyses and engineering in mammalian cells.
Collapse
Affiliation(s)
- Yi-Fan Huang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Sachiko Akase
- Graduate School of Engineering, Soka University, Hachioji, Tokyo 192-8577, Japan
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Yi-Shi Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ganglong Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yasuhiko Kizuka
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu 501-1193, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi 468-8503, Japan
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Kiyoko F Aoki-Kinoshita
- Graduate School of Engineering, Soka University, Hachioji, Tokyo 192-8577, Japan; Glycan & Life System Integration Center (GaLSIC), Faculty of Science and Engineering, Soka University, Hachioji, Tokyo 192-8577, Japan.
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
26
|
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
|
27
|
Abstract
Glycobiology is a glycan-based field of study that focuses on the structure, function, and biology of carbohydrates, and glycomics is a sub-study of the field of glycobiology that aims to define structure/function of glycans in living organisms. With the popularity of the glycobiology and glycomics, application of computational modeling expanded in the scientific area of glycobiology over the last decades. The recent availability of progressive Wet-Lab methods in the field of glycobiology and glycomics is promising for the impact of systems biology on the research area of the glycome, an emerging field that is termed “systems glycobiology.” This chapter will summarize the up-to-date leading edge in the use of bioinformatics tools in the field of glycobiology. The chapter provides basic knowledge both for glycobiologists interested in the application of bioinformatics tools and scientists of computational biology interested in studying the glycome.
Collapse
|
28
|
Insights into Bioinformatic Applications for Glycosylation: Instigating an Awakening towards Applying Glycoinformatic Resources for Cancer Diagnosis and Therapy. Int J Mol Sci 2020; 21:ijms21249336. [PMID: 33302373 PMCID: PMC7762546 DOI: 10.3390/ijms21249336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 01/10/2023] Open
Abstract
Glycosylation plays a crucial role in various diseases and their etiology. This has led to a clear understanding on the functions of carbohydrates in cell communication, which eventually will result in novel therapeutic approaches for treatment of various disease. Glycomics has now become one among the top ten technologies that will change the future. The direct implication of glycosylation as a hallmark of cancer and for cancer therapy is well established. As in proteomics, where bioinformatics tools have led to revolutionary achievements, bioinformatics resources for glycosylation have improved its practical implication. Bioinformatics tools, algorithms and databases are a mandatory requirement to manage and successfully analyze large amount of glycobiological data generated from glycosylation studies. This review consolidates all the available tools and their applications in glycosylation research. The achievements made through the use of bioinformatics into glycosylation studies are also presented. The importance of glycosylation in cancer diagnosis and therapy is discussed and the gap in the application of widely available glyco-informatic tools for cancer research is highlighted. This review is expected to bring an awakening amongst glyco-informaticians as well as cancer biologists to bridge this gap, to exploit the available glyco-informatic tools for cancer.
Collapse
|
29
|
Zhang L, Schwarz H, Wang M, Castan A, Hjalmarsson H, Chotteau V. Control of IgG glycosylation in CHO cell perfusion cultures by GReBA mathematical model supported by a novel targeted feed, TAFE. Metab Eng 2020; 65:135-145. [PMID: 33161144 DOI: 10.1016/j.ymben.2020.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 10/23/2022]
Abstract
The N-linked glycosylation pattern is an important quality attribute of therapeutic glycoproteins. It has been reported by our group and by others that different carbon sources, such as glucose, mannose and galactose, can differently impact the glycosylation profile of glycoproteins in mammalian cell culture. Acting on the sugar feeding is thus an attractive strategy to tune the glycan pattern. However, in case of feeding of more than one carbon source simultaneously, the cells give priority to the one with the highest uptake rate, which limits the usage of this tuning, e.g. the cells favor consuming glucose in comparison to galactose. We present here a new feeding strategy (named 'TAFE' for targeted feeding) for perfusion culture to adjust the concentrations of fed sugars influencing the glycosylation. The strategy consists in setting the sugar feeding such that the cells are forced to consume these substrates at a target cell specific consumption rate decided by the operator and taking into account the cell specific perfusion rate (CSPR). This strategy is applied in perfusion cultures of Chinese hamster ovary (CHO) cells, illustrated by ten different regimes of sugar feeding, including glucose, galactose and mannose. Applying the TAFE strategy, different glycan profiles were obtained using the different feeding regimes. Furthermore, we successfully forced the cells to consume higher proportions of non-glucose sugars, which have lower transport rates than glucose in presence of this latter, in a controlled way. In previous work, a mathematical model named Glycan Residues Balance Analysis (GReBA) was developed to model the glycosylation profile based on the fed carbon sources. The present data were applied to the GReBA to design a feeding regime targeting a given glycosylation profile. The ability of the model to achieve this objective was confirmed by a multi-round of leave-one-out cross-validation (LOOCV), leading to the conclusion that the GReBA model can be used to design the feeding regime of a perfusion cell culture to obtain a desired glycosylation profile.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Sweden; AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH, Sweden
| | - Hubert Schwarz
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Sweden; AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH, Sweden
| | - Mingliang Wang
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH, Sweden; Division of Decision and Control System, School of Electrical Engineering and Computer Science, KTH-Royal Institute of Technology, Sweden
| | | | - Håkan Hjalmarsson
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH, Sweden; Division of Decision and Control System, School of Electrical Engineering and Computer Science, KTH-Royal Institute of Technology, Sweden
| | - Veronique Chotteau
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Sweden; AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, KTH, Sweden.
| |
Collapse
|
30
|
Luo Y, Lovelett RJ, Price JV, Radhakrishnan D, Barnthouse K, Hu P, Schaefer E, Cunningham J, Lee KH, Shivappa RB, Ogunnaike BA. Modeling the Effect of Amino Acids and Copper on Monoclonal Antibody Productivity and Glycosylation: A Modular Approach. Biotechnol J 2020; 16:e2000261. [PMID: 32875683 DOI: 10.1002/biot.202000261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/22/2020] [Indexed: 01/15/2023]
Abstract
In manufacturing monoclonal antibodies (mAbs), it is crucial to be able to predict how process conditions and supplements affect productivity and quality attributes, especially glycosylation. Supplemental inputs, such as amino acids and trace metals in the media, are reported to affect cell metabolism and glycosylation; quantifying their effects is essential for effective process development. We aim to present and validate, through a commercially relevant cell culture process, a technique for modeling such effects efficiently. While existing models can predict mAb production or glycosylation dynamics under specific process configurations, adapting them to new processes remains challenging, because it involves modifying the model structure and often requires some mechanistic understanding. Here, a modular modeling technique for adapting an existing model for a fed-batch Chinese hamster ovary (CHO) cell culture process without structural modifications or mechanistic insight is presented. Instead, data is used, obtained from designed experimental perturbations in media supplementation, to train and validate a supplemental input effect model, which is used to "patch" the existing model. The combined model can be used for model-based process development to improve productivity and to meet product quality targets more efficiently. The methodology and analysis are generally applicable to other CHO cell lines and cell types.
Collapse
Affiliation(s)
- Yu Luo
- University of Delaware, Chemical and Biomolecular Engineering, 150 Academy St, Newark, DE, 19716, USA
| | - Robert J Lovelett
- University of Delaware, Chemical and Biomolecular Engineering, 150 Academy St, Newark, DE, 19716, USA
| | - J Vincent Price
- Janssen Research and Development, Discovery, Product Development and Supply, 200 Great Valley Parkway, Malvern, PA, 19355, USA
| | - Devesh Radhakrishnan
- University of Delaware, Chemical and Biomolecular Engineering, 150 Academy St, Newark, DE, 19716, USA
| | - Kristopher Barnthouse
- Janssen Research and Development, Discovery, Product Development and Supply, 200 Great Valley Parkway, Malvern, PA, 19355, USA
| | - Ping Hu
- Janssen Research and Development, Discovery, Product Development and Supply, 200 Great Valley Parkway, Malvern, PA, 19355, USA
| | - Eugene Schaefer
- Janssen Research and Development, Discovery, Product Development and Supply, 200 Great Valley Parkway, Malvern, PA, 19355, USA
| | - John Cunningham
- Janssen Research and Development, Discovery, Product Development and Supply, 200 Great Valley Parkway, Malvern, PA, 19355, USA
| | - Kelvin H Lee
- University of Delaware, Chemical and Biomolecular Engineering, 150 Academy St, Newark, DE, 19716, USA
| | - Raghunath B Shivappa
- Takeda Pharmaceuticals, Biologics Process Development, 200 Shire Way, Lexington, MA, 02421, USA
| | - Babatunde A Ogunnaike
- University of Delaware, Chemical and Biomolecular Engineering, 150 Academy St, Newark, DE, 19716, USA
| |
Collapse
|
31
|
Dasgupta A, Chowdhury N, De RK. Metabolic pathway engineering: Perspectives and applications. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 192:105436. [PMID: 32199314 DOI: 10.1016/j.cmpb.2020.105436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Metabolic engineering aims at contriving microbes as biocatalysts for enhanced and cost-effective production of countless secondary metabolites. These secondary metabolites can be treated as the resources of industrial chemicals, pharmaceuticals and fuels. Plants are also crucial targets for metabolic engineers to produce necessary secondary metabolites. Metabolic engineering of both microorganism and plants also contributes towards drug discovery. In order to implement advanced metabolic engineering techniques efficiently, metabolic engineers should have detailed knowledge about cell physiology and metabolism. Principle behind methodologies: Genome-scale mathematical models of integrated metabolic, signal transduction, gene regulatory and protein-protein interaction networks along with experimental validation can provide such knowledge in this context. Incorporation of omics data into these models is crucial in the case of drug discovery. Inverse metabolic engineering and metabolic control analysis (MCA) can help in developing such models. Artificial intelligence methodology can also be applied for efficient and accurate metabolic engineering. CONCLUSION In this review, we discuss, at the beginning, the perspectives of metabolic engineering and its application on microorganism and plant leading to drug discovery. At the end, we elaborate why inverse metabolic engineering and MCA are closely related to modern metabolic engineering. In addition, some crucial steps ensuring efficient and optimal metabolic engineering strategies have been discussed. Moreover, we explore the use of genomics data for the activation of silent metabolic clusters and how it can be integrated with metabolic engineering. Finally, we exhibit a few applications of artificial intelligence to metabolic engineering.
Collapse
Affiliation(s)
- Abhijit Dasgupta
- Department of Data Science, School of Interdisciplinary Studies, University of Kalyani, Kalyani, Nadia 741235, West Bengal, India
| | - Nirmalya Chowdhury
- Department of Computer Science & Engineering, Jadavpur University, Kolkata 700032, India
| | - Rajat K De
- Machine Intelligence Unit, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India.
| |
Collapse
|
32
|
Modeling Glycan Processing Reveals Golgi-Enzyme Homeostasis upon Trafficking Defects and Cellular Differentiation. Cell Rep 2020; 27:1231-1243.e6. [PMID: 31018136 PMCID: PMC6486481 DOI: 10.1016/j.celrep.2019.03.107] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/24/2019] [Accepted: 03/27/2019] [Indexed: 01/11/2023] Open
Abstract
The decoration of proteins by carbohydrates is essential for eukaryotic life yet heterogeneous due to a lack of biosynthetic templates. This complex carbohydrate mixture—the glycan profile—is generated in the compartmentalized Golgi, in which level and localization of glycosylation enzymes are key determinants. Here, we develop and validate a computational model for glycan biosynthesis to probe how the biosynthetic machinery creates different glycan profiles. We combined stochastic modeling with Bayesian fitting that enables rigorous comparison to experimental data despite starting with uncertain initial parameters. This is an important development in the field of glycan modeling, which revealed biological insights about the glycosylation machinery in altered cellular states. We experimentally validated changes in N-linked glycan-modifying enzymes in cells with perturbed intra-Golgi-enzyme sorting and the predicted glycan-branching activity during osteogenesis. Our model can provide detailed information on altered biosynthetic paths, with potential for advancing treatments for glycosylation-related diseases and glyco-engineering of cells. Developed a stochastic model of N-glycosylation coupled with Bayesian fitting Validated predicted changes of Golgi organization in trafficking mutants Model pinpointed functionally relevant glycan alterations in osteogenesis
Collapse
|
33
|
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
|
34
|
Kotidis P, Kontoravdi C. Harnessing the potential of artificial neural networks for predicting protein glycosylation. Metab Eng Commun 2020; 10:e00131. [PMID: 32489858 PMCID: PMC7256630 DOI: 10.1016/j.mec.2020.e00131] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022] Open
Abstract
Kinetic models offer incomparable insight on cellular mechanisms controlling protein glycosylation. However, their ability to reproduce site-specific glycoform distributions depends on accurate estimation of a large number of protein-specific kinetic parameters and prior knowledge of enzyme and transport protein levels in the Golgi membrane. Herein we propose an artificial neural network (ANN) for protein glycosylation and apply this to four recombinant glycoproteins produced in Chinese hamster ovary (CHO) cells, two monoclonal antibodies and two fusion proteins. We demonstrate that the ANN model accurately predicts site-specific glycoform distributions of up to eighteen glycan species with an average absolute error of 1.1%, correctly reproducing the effect of metabolic perturbations as part of a hybrid, kinetic/ANN, glycosylation model (HyGlycoM), as well as the impact of manganese supplementation and glycosyltransferase knock out experiments as a stand-alone machine learning algorithm. These results showcase the potential of machine learning and hybrid approaches for rapidly developing performance-driven models of protein glycosylation.
Collapse
|
35
|
Saeui CT, Cho KC, Dharmarha V, Nairn AV, Galizzi M, Shah SR, Gowda P, Park M, Austin M, Clarke A, Cai E, Buettner MJ, Ariss R, Moremen KW, Zhang H, Yarema KJ. Cell Line-, Protein-, and Sialoglycosite-Specific Control of Flux-Based Sialylation in Human Breast Cells: Implications for Cancer Progression. Front Chem 2020; 8:13. [PMID: 32117864 PMCID: PMC7013041 DOI: 10.3389/fchem.2020.00013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Sialylation, a post-translational modification that impacts the structure, activity, and longevity of glycoproteins has been thought to be controlled primarily by the expression of sialyltransferases (STs). In this report we explore the complementary impact of metabolic flux on sialylation using a glycoengineering approach. Specifically, we treated three human breast cell lines (MCF10A, T-47D, and MDA-MB-231) with 1,3,4-O-Bu3ManNAc, a "high flux" metabolic precursor for the sialic acid biosynthetic pathway. We then analyzed N-glycan sialylation using solid phase extraction of glycopeptides (SPEG) mass spectrometry-based proteomics under conditions that selectively captured sialic acid-containing glycopeptides, referred to as "sialoglycosites." Gene ontology (GO) analysis showed that flux-based changes to sialylation were broadly distributed across classes of proteins in 1,3,4-O-Bu3ManNAc-treated cells. Only three categories of proteins, however, were "highly responsive" to flux (defined as two or more sialylation changes of 10-fold or greater). Two of these categories were cell signaling and cell adhesion, which reflect well-known roles of sialic acid in oncogenesis. A third category-protein folding chaperones-was unexpected because little precedent exists for the role of glycosylation in the activity of these proteins. The highly flux-responsive proteins were all linked to cancer but sometimes as tumor suppressors, other times as proto-oncogenes, or sometimes both depending on sialylation status. A notable aspect of our analysis of metabolically glycoengineered breast cells was decreased sialylation of a subset of glycosites, which was unexpected because of the increased intracellular levels of sialometabolite "building blocks" in the 1,3,4-O-Bu3ManNAc-treated cells. Sites of decreased sialylation were minor in the MCF10A (<25% of all glycosites) and T-47D (<15%) cells but dominated in the MDA-MB-231 line (~60%) suggesting that excess sialic acid could be detrimental in advanced cancer and cancer cells can evolve mechanisms to guard against hypersialylation. In summary, flux-driven changes to sialylation offer an intriguing and novel mechanism to switch between context-dependent pro- or anti-cancer activities of the several oncoproteins identified in this study. These findings illustrate how metabolic glycoengineering can uncover novel roles of sialic acid in oncogenesis.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kyung-Cho Cho
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Vrinda Dharmarha
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Alison V Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Sagar R Shah
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Prateek Gowda
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Marian Park
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Melissa Austin
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Amelia Clarke
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Edward Cai
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Matthew J Buettner
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
A Markov model of glycosylation elucidates isozyme specificity and glycosyltransferase interactions for glycoengineering. CURRENT RESEARCH IN BIOTECHNOLOGY 2020; 2:22-36. [PMID: 32285041 DOI: 10.1016/j.crbiot.2020.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glycosylated biopharmaceuticals are important in the global pharmaceutical market. Despite the importance of their glycan structures, our limited knowledge of the glycosylation machinery still hinders controllability of this critical quality attribute. To facilitate discovery of glycosyltransferase specificity and predict glycoengineering efforts, here we extend the approach to model N-linked protein glycosylation as a Markov process. Our model leverages putative glycosyltransferase (GT) specificity to define the biosynthetic pathways for all measured glycans, and the Markov chain modelling is used to learn glycosyltransferase isoform activities and predict glycosylation following glycosyltransferase knock-in/knockout. We apply our methodology to four different glycoengineered therapeutics (i.e., Rituximab, erythropoietin, Enbrel, and alpha-1 antitrypsin) produced in CHO cells. Our model accurately predicted N-linked glycosylation following glycoengineering and further quantified the impact of glycosyltransferase mutations on reactions catalyzed by other glycosyltransferases. By applying these learned GT-GT interaction rules identified from single glycosyltransferase mutants, our model further predicts the outcome of multi-gene glycosyltransferase mutations on the diverse biotherapeutics. Thus, this modeling approach enables rational glycoengineering and the elucidation of relationships between glycosyltransferases, thereby facilitating biopharmaceutical research and aiding the broader study of glycosylation to elucidate the genetic basis of complex changes in glycosylation.
Collapse
|
37
|
Stach CS, McCann MG, O’Brien CM, Le TS, Somia N, Chen X, Lee K, Fu HY, Daoutidis P, Zhao L, Hu WS, Smanski M. Model-Driven Engineering of N-Linked Glycosylation in Chinese Hamster Ovary Cells. ACS Synth Biol 2019; 8:2524-2535. [PMID: 31596566 PMCID: PMC7034315 DOI: 10.1021/acssynbio.9b00215] [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: 12/31/2022]
Abstract
Chinese hamster ovary (CHO) cells are used for industrial production of protein-based therapeutics (i.e., "biologics"). Here we describe a method for combining systems-level kinetic models with a synthetic biology platform for multigene overexpression to rationally perturb N-linked glycosylation. Specifically, we sought to increase galactose incorporation on a secreted Immunoglobulin G (IgG) protein. We rationally design, build, and test a total of 23 transgenic cell pools that express single or three-gene glycoengineering cassettes comprising a total of 100 kilobases of engineered DNA sequence. Through iterative engineering and model refinement, we rationally increase the fraction of bigalactosylated glycans five-fold from 11.9% to 61.9% and simultaneously decrease the glycan heterogeneity on the secreted IgG. Our approach allows for rapid hypothesis testing and identification of synergistic behavior from genetic perturbations by bridging systems and synthetic biology.
Collapse
Affiliation(s)
- Christopher S. Stach
- Department of Biochemistry, Molecular Biology & Biophysics and Biotechnology Institute
| | | | | | - Tung S. Le
- Department of Chemical Engineering and Materials Science
| | - Nikunj Somia
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Xinning Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Kyoungho Lee
- Department of Chemical Engineering and Materials Science
| | - Hsu-Yuan Fu
- Department of Chemical Engineering and Materials Science
| | | | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, PR China
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science
| | - Michael Smanski
- Department of Biochemistry, Molecular Biology & Biophysics and Biotechnology Institute
| |
Collapse
|
38
|
Klein J, Carvalho L, Zaia J. Application of network smoothing to glycan LC-MS profiling. Bioinformatics 2019; 34:3511-3518. [PMID: 29790907 DOI: 10.1093/bioinformatics/bty397] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/10/2018] [Indexed: 11/14/2022] Open
Abstract
Motivation Glycosylation is one of the most heterogeneous and complex protein post-translational modifications. Liquid chromatography coupled mass spectrometry (LC-MS) is a common high throughput method for analyzing complex biological samples. Accurate study of glycans require high resolution mass spectrometry. Mass spectrometry data contains intricate sub-structures that encode mass and abundance, requiring several transformations before it can be used to identify biological molecules, requiring automated tools to analyze samples in a high throughput setting. Existing tools for interpreting the resulting data do not take into account related glycans when evaluating individual observations, limiting their sensitivity. Results We developed an algorithm for assigning glycan compositions from LC-MS data by exploring biosynthetic network relationships among glycans. Our algorithm optimizes a set of likelihood scoring functions based on glycan chemical properties but uses network Laplacian regularization and optionally prior information about expected glycan families to smooth the likelihood and thus achieve a consistent and more representative solution. Our method was able to identify as many, or more glycan compositions compared to previous approaches, and demonstrated greater sensitivity with regularization. Our network definition was tailored to N-glycans but the method may be applied to glycomics data from other glycan families like O-glycans or heparan sulfate where the relationships between compositions can be expressed as a graph. Availability and implementation Built Executable http://www.bumc.bu.edu/msr/glycresoft/ and Source Code: https://github.com/BostonUniversityCBMS/glycresoft. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Joshua Klein
- Program for Bioinformatics, Boston University, Boston, MA, USA
| | - Luis Carvalho
- Program for Bioinformatics, Boston University, Boston, MA, USA.,Department of Math and Statistics, Boston University, Boston, MA, USA
| | - Joseph Zaia
- Program for Bioinformatics, Boston University, Boston, MA, USA.,Department of Biochemistry, Boston University, Boston, MA, USA
| |
Collapse
|
39
|
Dhiman H, Gerstl MP, Ruckerbauer D, Hanscho M, Himmelbauer H, Clarke C, Barron N, Zanghellini J, Borth N. Genetic and Epigenetic Variation across Genes Involved in Energy Metabolism and Mitochondria of Chinese Hamster Ovary Cell Lines. Biotechnol J 2019; 14:e1800681. [DOI: 10.1002/biot.201800681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/14/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Heena Dhiman
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesVienna Austria
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
| | - Matthias P. Gerstl
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesVienna Austria
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
| | - David Ruckerbauer
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
| | - Michael Hanscho
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
| | - Heinz Himmelbauer
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesVienna Austria
| | - Colin Clarke
- National Institute for Bioprocessing Research and TrainingBlackrock, Co Dublin Ireland
| | - Niall Barron
- National Institute for Bioprocessing Research and TrainingBlackrock, Co Dublin Ireland
- School of Chemical and Bioprocess EngineeringUniversity College DublinGlasnevin Whitehall Dublin Ireland
| | - Jürgen Zanghellini
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
- Austrian Biotech University of Applied SciencesKonrad Lorenz Strasse 10 3430 Tulln Austria
| | - Nicole Borth
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesVienna Austria
- Austrian Centre of Industrial BiotechnologyMuthgasse 11 1190 Vienna Austria
| |
Collapse
|
40
|
Prediction of N-linked Glycoform Profiles of Monoclonal Antibody with Extracellular Metabolites and Two-Step Intracellular Models. Processes (Basel) 2019. [DOI: 10.3390/pr7040227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Monoclonal antibodies (mAbs) are commonly glycosylated and show varying levels of galactose attachment. A set of experiments in our work showed that the galactosylation level of mAbs was altered by the culture conditions of hybridoma cells. The uridine diphosphate galactose (UDP-Gal) is one of the substrates of galactosylation. Based on that, we proposed a two-step model to predict N-linked glycoform profiles by solely using extracellular metabolites from cell culture. At the first step, the flux level of UDP-Gal in each culture was estimated based on a computational flux balance analysis (FBA); its level was found to be linear with the galactosylation degree on mAbs. At the second step, the glycoform profiles especially for G0F (agalactosylated), G1F (monogalactosylated) and G2F (digalactosylated) were predicted by a kinetic model. The model outputs well matched with the experimental data. Our study demonstrated that the integrated mathematical approach combining FBA and kinetic model is a promising strategy to predict glycoform profiles for mAbs during cell culture processes.
Collapse
|
41
|
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
|
42
|
Sumit M, Dolatshahi S, Chu AHA, Cote K, Scarcelli JJ, Marshall JK, Cornell RJ, Weiss R, Lauffenburger DA, Mulukutla BC, Figueroa B. Dissecting N-Glycosylation Dynamics in Chinese Hamster Ovary Cells Fed-batch Cultures using Time Course Omics Analyses. iScience 2019; 12:102-120. [PMID: 30682623 PMCID: PMC6352710 DOI: 10.1016/j.isci.2019.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/19/2018] [Accepted: 01/03/2019] [Indexed: 12/24/2022] Open
Abstract
N-linked glycosylation affects the potency, safety, immunogenicity, and pharmacokinetic clearance of several therapeutic proteins including monoclonal antibodies. A robust control strategy is needed to dial in appropriate glycosylation profile during the course of cell culture processes accurately. However, N-glycosylation dynamics remains insufficiently understood owing to the lack of integrative analyses of factors that influence the dynamics, including sugar nucleotide donors, glycosyltransferases, and glycosidases. Here, an integrative approach involving multi-dimensional omics analyses was employed to dissect the temporal dynamics of glycoforms produced during fed-batch cultures of CHO cells. Several pathways including glycolysis, tricarboxylic citric acid cycle, and nucleotide biosynthesis exhibited temporal dynamics over the cell culture period. The steps involving galactose and sialic acid addition were determined as temporal bottlenecks. Our results show that galactose, and not manganese, is able to mitigate the temporal bottleneck, despite both being known effectors of galactosylation. Furthermore, sialylation is limited by the galactosylated precursors and autoregulation of cytidine monophosphate-sialic acid biosynthesis. Major glycosylated species exhibit temporal dynamics during fed-batch processes Key metabolic pathways linked to N-glycosylation exhibit significant temporal dynamics Dynamics in nucleotide sugar donors (NSDs) directly influences glycoform heterogeneity Glycoform heterogeneity can be mitigated by supplementing NSD biosynthetic precursors
Collapse
Affiliation(s)
- Madhuresh Sumit
- Culture Process Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - Sepideh Dolatshahi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - An-Hsiang Adam Chu
- Analytical Research and Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - Kaffa Cote
- Analytical Research and Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - John J Scarcelli
- Cell Line Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - Jeffrey K Marshall
- Analytical Research and Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - Richard J Cornell
- Analytical Research and Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bhanu Chandra Mulukutla
- Culture Process Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA.
| | - Bruno Figueroa
- Culture Process Development, Bio Therapeutics Pharmaceutical Sciences, Pfizer, 1 Burtt Road, Andover, MA 01810, USA
| |
Collapse
|
43
|
Zhang L, Castan A, Stevenson J, Chatzissavidou N, Vilaplana F, Chotteau V. Combined effects of glycosylation precursors and lactate on the glycoprofile of IgG produced by CHO cells. J Biotechnol 2019; 289:71-79. [DOI: 10.1016/j.jbiotec.2018.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 12/29/2022]
|
44
|
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]
|
45
|
Buettner MJ, Shah SR, Saeui CT, Ariss R, Yarema KJ. Improving Immunotherapy Through Glycodesign. Front Immunol 2018; 9:2485. [PMID: 30450094 PMCID: PMC6224361 DOI: 10.3389/fimmu.2018.02485] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is revolutionizing health care, with the majority of high impact "drugs" approved in the past decade falling into this category of therapy. Despite considerable success, glycosylation-a key design parameter that ensures safety, optimizes biological response, and influences the pharmacokinetic properties of an immunotherapeutic-has slowed the development of this class of drugs in the past and remains challenging at present. This article describes how optimizing glycosylation through a variety of glycoengineering strategies provides enticing opportunities to not only avoid past pitfalls, but also to substantially improve immunotherapies including antibodies and recombinant proteins, and cell-based therapies. We cover design principles important for early stage pre-clinical development and also discuss how various glycoengineering strategies can augment the biomanufacturing process to ensure the overall effectiveness of immunotherapeutics.
Collapse
Affiliation(s)
- Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Pharmacology/Toxicology Branch I, Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
46
|
Gupta U, Le T, Hu WS, Bhan A, Daoutidis P. Automated network generation and analysis of biochemical reaction pathways using RING. Metab Eng 2018; 49:84-93. [DOI: 10.1016/j.ymben.2018.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/20/2018] [Accepted: 07/18/2018] [Indexed: 10/28/2022]
|
47
|
Kremkow BG, Lee KH. Glyco-Mapper: A Chinese hamster ovary (CHO) genome-specific glycosylation prediction tool. Metab Eng 2018. [PMID: 29522825 DOI: 10.1016/j.ymben.2018.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Glyco-Mapper is a novel systems biology product quality prediction tool created using a new framework termed: Discretized Reaction Network Modeling using Fuzzy Parameters (DReaM-zyP). Within Glyco-Mapper, users fix the nutrient feed composition and the glycosylation reaction fluxes to fit the model glycoform to the reference experimental glycoform, enabling cell-line specific glycoform predictions as a result of cell engineering strategies. Glyco-Mapper accurately predicts glycoforms associated with genetic alterations that result in the appearance or disappearance of one or more glycans with an accuracy, sensitivity, and specificity of 96%, 85%, and 97%, respectively, for publications between 1999 and 2014. The modeled glycoforms span a large range of glycoform engineering strategies, including the altered expression of glycosylation, nucleotide sugar transport, and metabolism genes, as well as an altered nutrient feeding strategy. A glycoprotein-producing CHO cell line reference glycoform was modeled and a novel Glyco-Mapper prediction was experimentally confirmed with an accuracy and specificity of 95% and 98%, respectively. Glyco-Mapper is a product quality prediction tool that provides a streamlined way to design host cell line genomes to achieve specific product quality attributes.
Collapse
Affiliation(s)
- Benjamin G Kremkow
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA.
| |
Collapse
|
48
|
Tejwani V, Andersen MR, Nam JH, Sharfstein ST. Glycoengineering in CHO Cells: Advances in Systems Biology. Biotechnol J 2018; 13:e1700234. [PMID: 29316325 DOI: 10.1002/biot.201700234] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/28/2017] [Indexed: 12/19/2022]
Abstract
For several decades, glycoprotein biologics have been successfully produced from Chinese hamster ovary (CHO) cells. The therapeutic efficacy and potency of glycoprotein biologics are often dictated by their post-translational modifications, particularly glycosylation, which unlike protein synthesis, is a non-templated process. Consequently, both native and recombinant glycoprotein production generate heterogeneous mixtures containing variable amounts of different glycoforms. Stability, potency, plasma half-life, and immunogenicity of the glycoprotein biologic are directly influenced by the glycoforms. Recently, CHO cells have also been explored for production of therapeutic glycosaminoglycans (e.g., heparin), which presents similar challenges as producing glycoproteins biologics. Approaches to controlling heterogeneity in CHO cells and directing the biosynthetic process toward desired glycoforms are not well understood. A systems biology approach combining different technologies is needed for complete understanding of the molecular processes accounting for this variability and to open up new venues in cell line development. In this review, we describe several advances in genetic manipulation, modeling, and glycan and glycoprotein analysis that together will provide new strategies for glycoengineering of CHO cells with desired or enhanced glycosylation capabilities.
Collapse
Affiliation(s)
- Vijay Tejwani
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY, 12203, USA
| | - Mikael R Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Susan T Sharfstein
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY, 12203, USA
| |
Collapse
|
49
|
Kyriakopoulos S, Ang KS, Lakshmanan M, Huang Z, Yoon S, Gunawan R, Lee DY. Kinetic Modeling of Mammalian Cell Culture Bioprocessing: The Quest to Advance Biomanufacturing. Biotechnol J 2017; 13:e1700229. [DOI: 10.1002/biot.201700229] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Sarantos Kyriakopoulos
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Kok Siong Ang
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Zhuangrong Huang
- Department of Chemical Engineering; University of Massachusetts Lowell; Lowell MA USA
| | - Seongkyu Yoon
- Department of Chemical Engineering; University of Massachusetts Lowell; Lowell MA USA
| | - Rudiyanto Gunawan
- Institute for Chemical and Bioengineering; ETH Zurich; Zurich Switzerland
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
- Department of Chemical and Biomolecular Engineering; National University of Singapore; Singapore
| |
Collapse
|
50
|
Glycosylation flux analysis reveals dynamic changes of intracellular glycosylation flux distribution in Chinese hamster ovary fed-batch cultures. Metab Eng 2017; 43:9-20. [PMID: 28754360 DOI: 10.1016/j.ymben.2017.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/29/2017] [Accepted: 07/20/2017] [Indexed: 01/06/2023]
Abstract
N-linked glycosylation of proteins has both functional and structural significance. Importantly, the glycan structure of a therapeutic protein influences its efficacy, pharmacokinetics, pharmacodynamics and immunogenicity. In this work, we developed glycosylation flux analysis (GFA) for predicting intracellular production and consumption rates (fluxes) of glycoforms, and applied this analysis to CHO fed-batch immunoglobulin G (IgG) production using two different media compositions, with and without additional manganese feeding. The GFA is based on a constraint-based modeling of the glycosylation network, employing a pseudo steady state assumption. While the glycosylation fluxes in the network are balanced at each time point, the GFA allows the fluxes to vary with time by way of two scaling factors: (1) an enzyme-specific factor that captures the temporal changes among glycosylation reactions catalysed by the same enzyme, and (2) the cell specific productivity factor that accounts for the dynamic changes in the IgG production rate. The GFA of the CHO fed-batch cultivations showed that regardless of the media composition, galactosylation fluxes decreased with the cultivation time more significantly than the other glycosylation reactions. Furthermore, the GFA showed that the addition of Mn, a cofactor of galactosyltransferase, has the effect of increasing the galactosylation fluxes but only during the beginning of the cultivation period. The results thus demonstrated the power of the GFA in delineating the dynamic alterations of the glycosylation fluxes by local (enzyme-specific) and global (cell specific productivity) factors.
Collapse
|