Elsevier

Journal of Biotechnology

Volume 164, Issue 4, 15 April 2013, Pages 469-478
Journal of Biotechnology

Probing the metabolism of an inducible mammalian expression system using extracellular isotopomer analysis

https://doi.org/10.1016/j.jbiotec.2013.01.025Get rights and content

Abstract

In an effort to quantitatively assess the impact of recombinant protein expression on the primary metabolism of mammalian cells in culture, we have employed an efficient inducible expression system and conducted a comparative study of the intracellular flux map distribution with and without the induction of recombinant protein synthesis. Cells were grown in parallel semi-continuous cultures with various singly and uniformly labeled substrates and the resulting mass isotopomer distributions of lactate and extracellular amino acids were measured by mass spectrometry. These distributions were used to quantify the main intracellular fluxes. The analysis revealed that, under mild hypothermic conditions, the onset of protein expression is correlated with small but significant changes in several key pathways related to ATP and NADPH formation. More specifically, we observed that induced cells exhibit a more efficient utilization of glucose, characterized by an increased flux of pyruvate into the TCA cycle. In contrast, the catabolic rates of most amino acids remained relatively unaffected. Such analysis is instrumental to guide the identification of robust biomarkers of productivity, as well as the development of medium formulations optimized for recombinant protein production.

Highlights

► Metabolic characterization of an inducible mammalian expression system. ► CHO cells grown in parallel semi-continuous cultures with various labeled substrates. ► Metabolic flux analysis using 13C-tracers and extracellular isotopomer analysis. ► Induced cells exhibited a more efficient nutrient utilization and greater TCA fluxes.

Introduction

Mammalian cells continue to be the preferred expression system for the production of valuable therapeutic proteins and vaccines, which require specific post-translational modifications that cannot be performed in other microorganisms. Monoclonal antibodies (MAb) produced by mammalian cell culture form the most important group of new biopharmaceuticals approved or under development and their market is expected to grow significantly in the next few years (Walsh, 2010). In order to meet the growing demand for MAb, there is a continuing need to develop robust, cost-effective and efficient cell culture processes. Conventional process development for mammalian cell culture MAb platforms has been conducted mainly by experimental and especially trial-and-error methods (Baughman et al., 2010). Despite significant advances in the field of cell culture technology, the establishment of an optimal set of process conditions remains mostly cell line-specific. Typically, improvements in product titers have been essentially achieved through an increase of the cumulative integrated viable cell density, but factors limiting the cell specific productivity of mammalian cells are still poorly understood (Meleady et al., 2011). Efforts to further enhance processes have thus rapidly underscored the need for better characterization and control of the complex mammalian cell metabolism in culture. In particular, compared to parental cells, recombinant cells typically exhibit reduced specific growth rates and increased nutrient utilization (Yallop et al., 2003). Assessing and analysing the impact of recombinant protein expression on the cells’ central metabolism is thus of utmost importance for the establishment and optimization of a productive mammalian cell expression platform.

To this end, quantification of metabolic fluxes can provide critical insights into the fundamental processes of biological systems and allow the identification of possible metabolic bottlenecks (Boghigian et al., 2010, Metallo et al., 2009, Park et al., 2008). In turn, such information has proven to be invaluable to enable the targeted optimization of cell culture processes. Many comprehensive metabolic models have now been developed for the most relevant industrial cell lines such as CHO, BHK, HEK-293 and hybridoma cells (Niklas and Heinzle, 2011). However, a complete and accurate determination of the flux distribution in mammalian cell networks is generally impossible to obtain from routine measurements and simple metabolite balancing, due to the typically large number of biochemical reactions involved on the one hand, and the existence of reversible, cyclic and parallel pathways on the other hand. To cope with this problem and alleviate the need to resort to questionable assumptions or constraints, additional independent information can be derived with the use of isotopic tracers. 13C-metabolic flux analysis (13C-MFA) has recently been increasingly applied to mammalian systems in order to obtain a more detailed and accurate description of cellular physiology. The technique is based on culturing cells with specific 13C-labeled substrates and subsequently measuring the label distribution in the network's metabolites using either mass spectroscopy (Ahn and Antoniewicz, 2011, Bonarius et al., 2001, Hofmann et al., 2008, Metallo et al., 2009, Niklas et al., 2011, Sengupta et al., 2011) or NMR analysis (Bonarius et al., 2001, Goudar et al., 2010, Mancuso et al., 1998). However, these approaches are experimentally challenging; they typically require the reliable and accurate determination of the mass distributions of several free intracellular metabolites, which necessitates careful and labor-intensive extraction/analysis procedures. Moreover, to ensure the achievement of proper metabolic and isotopic steady-states, such experiments are preferably carried out in a chemostat, which requires the setup of a complex culture system. Recent studies emphasized that adequate (pseudo-)steady conditions may be difficult to achieve in typical batch/fed-batch cultures (Deshpande et al., 2009). While transient isotopic studies can be performed (Ahn and Antoniewicz, 2011, Maier et al., 2008, Young et al., 2007), they are even more experimentally/computationally demanding since, in addition to mass isotopomer distributions, the intracellular pool concentrations must either be accurately measured or estimated along with the unknown fluxes.

In the current contribution, we demonstrate that reliable estimates can be obtained for several key intracellular fluxes using an approach that alleviates the need for a complex chemostat setup and the difficulties inherent in the extraction/analysis of intracellular metabolites. Cells were grown in parallel semi-continuous cultures containing various labeled glucose and glutamine tracers and only the resulting mass isotopomer distributions of extracellular metabolites (three excreted amino acids and lactate) were measured by LC-QTOF MS. The semi-continuous (or repeated batch) mode provides a simple and flexible operation with good approximation of (pseudo)steady-state conditions (Henry et al., 2008). This method was used to analyze the central carbon metabolism of a mammalian cell line in relation with cellular productivity. To this end, we have employed a CHO cell line that has been engineered with an inducible expression system called the “cumate gene-switch” (Gaillet et al., 2007, Mullick et al., 2006). These cells are capable of robust growth in serum-free and protein-free suspension cultures and, upon addition of a non-toxic small molecule (cumate) in the culture medium, they start to express a recombinant antibody. Comparing the metabolism of the induced and non-induced cells using 13C-MFA allowed investigating the effect of the onset of protein expression on the primary metabolism of the cells. Confidence intervals for the evaluated fluxes were calculated to properly judge the significance of the observed similarities/differences. Such analysis is invaluable for the rational development of improved cell culture processes on both the metabolic and process scales (Boghigian et al., 2010).

Section snippets

Cell lines, medium and culture conditions

The study was performed using an industrially relevant recombinant CHO cell line provided by Viropro International Inc. (Montreal, Canada) and producing a human anti-CD20 monoclonal antibody. The cell line was derived from CHO–Cum2 cells, which stably express the reverse cumate transactivator described in Mullick et al. (2006). Cells were grown in a costumed chemically-defined SFM4CHO medium (Hyclone). Prior to culturing the cells, the medium was supplemented with 4 mM glutamine (Hyclone), 25 mM

Kinetics of growth and substrate utilization

Cells were grown in semi-continuous mode to meet the requirements for metabolic and isotopic pseudo-steady states. The time profiles of viable cell concentrations for the cultures performed with and without cumate addition are shown in Fig. 1A. For each case, the data presented are the average and standard deviation from five parallel cultures. The apparent growth rates for the non-induced and the induced cultures were determined to be 0.36 d−1 and 0.33 d−1, respectively. To allow reliable

Discussion

Compared to parental cell lines, recombinant mammalian cell lines generally exhibit reduced growth rates and increased substrate utilization (Yallop et al., 2003), most likely to meet the specific energy and biosynthetic demands related to protein synthesis and related posttranslational modifications. Recent studies performed on yeasts and bacteria have characterized the metabolic response to the overproduction of recombinant protein and revealed significant redirections of intracellular carbon

Conclusion

The use of an efficient inducible mammalian expression system combined to a relatively straightforward experimental approach allowed us to characterize the primary metabolism of CHO cells in relation with their specific productivity. The main intracellular fluxes were quantified with relatively narrow confidence intervals, thereby allowing to decipher metabolic changes related to the onset of recombinant protein expression. Our study notably revealed small but significant variations in a number

Acknowledgements

The authors wish to acknowledge Dr. Alexandra Furtos from Université de Montréal for 13C QTOF–LCMS analysis, Jingkui Chen from Ecole Polytechnique for amino acids analysis, as well as Dr. Patrick Benoist and Dr. Patrick Daoust of Viropro International Inc. (Montreal, Canada) for providing the cell line employed in this study.

References (44)

  • J. Niklas et al.

    Metabolite channeling and compartmentation in the human cell line AGE1.HN determined by 13C labeling experiments and 13C metabolic flux analysis

    Journal of Bioscience and Bioengineering

    (2011)
  • R.P. Nolan et al.

    Dynamic model of CHO cell metabolism

    Metabolic Engineering

    (2011)
  • J.H. Park et al.

    Application of systems biology for bioprocess development

    Trends in Biotechnology

    (2008)
  • P.F. Suthers et al.

    Improved computational performance of MFA using elementary metabolite units and flux coupling

    Metabolic Engineering

    (2010)
  • J.L. Walther et al.

    Optimization of (13)C isotopic tracers for metabolic flux analysis in mammalian cells

    Metabolic Engineering

    (2012)
  • J.D. Young et al.

    Mapping photoautotrophic metabolism with isotopically nonstationary (13)C flux analysis

    Metabolic Engineering

    (2011)
  • C. Altamirano et al.

    Analysis of CHO cells metabolic redistribution in a glutamate-based defined medium in continuous culture

    Biotechnology Progress

    (2001)
  • H.P. Bonarius et al.

    Metabolic-flux analysis of continuously cultured hybridoma cells using (13)CO(2) mass spectrometry in combination with (13)C-lactate nuclear magnetic resonance spectroscopy and metabolite balancing

    Biotechnology and Bioengineering

    (2001)
  • M.C.F. Dalm et al.

    Effect of feed and bleed rate on hybridoma cells in an acoustic perfusion bioreactor: metabolic analysis

    Biotechnology Progress

    (2007)
  • R. Deshpande et al.

    Towards a metabolic and isotopic steady state in CHO batch cultures for reliable isotope-based metabolic profiling

    Biotechnology Journal

    (2009)
  • D.M. Dinnis et al.

    Functional proteomic analysis of GS-NS0 murine myeloma cell lines with varying recombinant monoclonal antibody production rate

    Biotechnology and Bioengineering

    (2006)
  • B. Gaillet et al.

    High-level recombinant protein productionin CHO cells using an adenoviral vector and the cumate gene switch

    Biotechnology Progress

    (2007)
  • Cited by (30)

    • <sup>13</sup>C metabolic flux analysis in cell line and bioprocess development

      2021, Current Opinion in Chemical Engineering
      Citation Excerpt :

      Because of the increased burden of recombinant protein production, there is interest in inducible expression systems to initiate protein production after cells have reached stationary phase. Sheikholeslami et al. [28] used 13C MFA to study a CHO cell line that had been engineered with a cumate-inducible expression system, allowing the expression of the recombinant protein to be controlled in order to decouple cell growth from product formation. This study highlighted the metabolic changes that occur following induction of product expression.

    • From nutritional wealth to autophagy: In vivo metabolic dynamics in the cytosol, mitochondrion and shuttles of IgG producing CHO cells

      2019, Metabolic Engineering
      Citation Excerpt :

      As depicted in Fig. S4, fluxes via PDH mirrored d-glucose uptake levels well, whereas fluxes via pyruvate carboxylase (PC) revealed steady declines from phases I to III.1. Observations under the ample nutrient conditions of phase I agree well with the findings of Sheikholeslami et al. (2013), which indicated that the majority of Pyr is derived from glycolysis (about 69%), and the remaining from cytosolic ME 1. The mitochondrial Pyr pool fueled the TCA cycle via PDH producing AcCoA and PC producing OAA in approximately equal proportions.

    • Application of <sup>13</sup>C flux analysis to identify high-productivity CHO metabolic phenotypes

      2017, Metabolic Engineering
      Citation Excerpt :

      Several recent studies have examined how metabolic fluxes of CHO cells respond to alternative nutrient feeds (Nargund et al., 2015; Sheikholeslami et al., 2014), inducible expression of a recombinant protein (Sheikholeslami et al., 2013), expression of anti-apoptotic factors, (Templeton et al., 2014) and the different stages of fed-batch culture (Ahn and Antoniewicz, 2013; Sengupta et al., 2011; Templeton et al., 2013). These studies have consistently reported that CHO cells rely more heavily on citric acid cycle (CAC) and oxidative phosphorylation (OXPHOS) to supply energy during stationary phase when production of recombinant protein is typically highest (Sengupta et al., 2011; Sheikholeslami et al., 2013, 2014; Templeton et al., 2013). This metabolic phenotype is often associated with a shift from lactate production to lactate consumption (Ahn and Antoniewicz, 2013; Templeton et al., 2013, 2014).

    • What can mathematical modelling say about CHO metabolism and protein glycosylation?

      2017, Computational and Structural Biotechnology Journal
      Citation Excerpt :

      Similarly, Sheikholeslami et al. [51] used 13C MFA to compare two semicontinuous cultures grown on chemically defined media with 1 mM and 5 mM glutamine, respectively, and found that low glutamine uptake (in the 1 mM culture) was more metabolically efficient in terms of the proportion of pyruvate that enters the TCA cycle (and therefore is not converted to lactate). Furthermore, the CHO cell line used in this study was found to be particularly efficient, mostly under hypothermic conditions, as confirmed on their previous work [52]. In this case, the use of 13C MFA was simplified by analyzing only extracellular 13C -labelled metabolites and then performing MFA to predict the intracellular fluxes.

    View all citing articles on Scopus
    View full text