DOI: 10.17344/acsi.2018.4591 Acta Chim. Slov. 2018, 65, 769-786 /^creative ^commons Feature article Metabolic Network Modelling of Chinese Hamster Ovary (CHO) Culture Bioreactors Operated as Microbial Cell Factories Jernej Gašperšič,1,2,* Miha Kastelic,2 Uroš Novak2 and Blaž Likozar2 1 Institute of Biochemistry, Medical faculty, University of Ljubljana, Vrazov trg2, 1000 Ljubljana 2 National Institute of Chemistry, Department of Catalysis and Chemical Reaction Engineering, Hajdrihova 19, 1000 Ljubljana, Slovenia * Corresponding author: E-mail: jernej.gaspersic@mf.uni-lj.si Received: 06-07-2018 Abstract Chinese hamster ovary (CHO) epithelial cells are one of the most used therapeutic medical lines for the production of different biopharmaceutical drugs. They have a high consumption rate with a fast duplication cycle that makes them an ideal biological clone. The higher accumulated amounts of toxic intracellular intermediates may lead to lower organism viability, protein productivity and manufactured biosimilar, so a careful optimal balance of medium, bioreactor operational parameters and bioprocess is needed. A precise phenomenological knowledge of metabolism's chemical transformations can predict problems that may arise during batch, semi-continuous fed batch and continuous reactor operation. For a better detailed understanding (and relations), future performance optimization and scaling, mechanistic model systems have been built. In this specific work, the main metabolic pathways in mammalian structured CHO cultures are reviewed. It starts with organic biochemical background, controlling associated phenomena and kinetics, which govern the sustaining conversion routes of biology. Then, individual turnover paths are described, overviewing standard mathematical formulations that are commonly applied in engineering. These are the core of black box modeling, which relates the substrates/products in a simplified relationship manner. Moreover, metabolic flux analysis (MFA)/flux balance analysis (FBA), that are traditionally characterizing mechanisms, are presented to a larger portion extent. Finally, similarities are discussed, illustrating the approaches for their structural design. Stated variables' equations, employed for the description of the growth in the controllable environmental conditions of a vessel, the researched reaction series of proliferating dividing CHO population, joint with the values of maximal enzymatic activity, and solutions are outlined. Processes are listed in a way so that a reader can integrate the state-of-the-art. Our particular contribution is also denoted. Keywords: Microbial cell factory; Chinese hamster ovary (CHO) cell metabolism; Bioreactor operation modeling; Biochemical reaction kinetics; Metabolic flux analysis; Biopharmaceutical and biosimilar 1. Introduction Chinese hamster ovary (CHO) cells are one of the most important cell lines for production of therapeutic bi-opharmaceuticals.1,2 CHO cells are characterized by high consumption rates of nutrients and large amount of toxic intermediates, which may lead to lower cell viability and protein production.1,2,3 Production of recombinant proteins is promoted in growth medium with high content of glucose and glutamine.4,5 Further optimization of the medium is needed to reduce lactate and ammonia accumulation and to increase antibody yields.3,6-9 In recent years technology has made a remarkable impact in bioreactor fermentation yields using kinetic models that are commonly used to describe fermentation processes in indus- trial fermentors. These models turn out to be useful at process monitoring, acquiring and storing the data, and troubleshooting. In the last three decades databases of CHO cell metabolism have been extended, likewise the tracing of fluxes into biomass and byproducts, which led to major improvements of bioreactor models.2,5,10-17 This knowledge has guided the researchers to develop several mathematical models, which are able to describe the fluxes within metabolic pathways.4,18-23 Analysis of fluxes usually focuses on measuring concentrations of extracellular metabolites.13 Biocatalysis of substrates into commercially attractive products as well as byproducts is connected through pathways of cell metabolism.13,24-26 Pathway analysis usually relies on measuring concentration behavior of extracellular metabolites,13 nev- Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 770 Acta Chim. Slov. 2018, 65, 769-786 ertheless estimates of intracellular fluxes are readily available by using flux balance analysis (FBA) and metabolic flux analysis (MFA).27 In recent years genotype data have been also included into in silico methodologies.28 The following pathways of the cell metabolism are usually assumed: glycolysis, pentose phosphate (PPP) pathway, tricarboxylic acid (TCA) cycle, amino acid metabolism, protein synthesis, urea cycle, nucleic acid synthesis, membrane lipid synthesis, and biomass production.1-5,14-19,29-36 Glycogen5,16 synthesis and glycosylation pathways are part of detailed models.29,37-41 Numerous models were developed for the purpose of data analysis and growth optimization in cell cultures. A hybrid simulation framework was proposed to predict the dynamics in bioreactors.42 A simplified model of central carbon metabolism provides the framework for analyzing measurements of external metabolites.29 Simulation as- suming pseudo steady state assumption and extracellular metabolite concentrations accurately predicted the effects of process variables, temperature shift, seeding density, specific productivity and nutrient concentrations.43 A mathematical model was developed for optimization of batch and fed batch bioreactor.20 The kinetic model that jointed several phases of cell culture was capable to describe the time evolution of experimental data.16 Similar model was build and its correctness validated on experimental data from CHO cells grown in spinner flasks.19 The model was assembled from submodels, where each of them described a separate phase of CHO cell (growth, stationary, and decline phase).33 The model was then used to explain the experimental data from batch cultivated CHO cells. In another example, the phenotype of mammalian cells was studied by the aims of metabolic flux analysis.5 Fluxes were measured using 13C MFA variant and stoichi- Figure 1: Metabolic engineering starts with the metabolic network, which comprises biochemical transformations within living cells (bottom left: unstructured network, top left: structured network). Then we use available experimental data together with theoretical methodologies (paradigm of Metabolic Flux Analysis - MFA) to estimate flux rates of biochemical transformations (top right). These insights help us to perform bioprocess optimization. Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster Acta Chim. Slov. 2018, 65, 769-786 771 ometric modeling.2,44 MFA was also used to estimate total energy production of growing CHO-320 cells.24 New metabolic information was mined from the models and associated simulations. Analysis of experiments showed the existence of multiple steady states.17 In silico modeling of CHO cells allowed the identification of major growth-limiting factors including oxidative stress and depletion of lipid metabolites.45 These factors may lead to a better development of strategies to enhance CHO culture performance.45 Continuous cell lines can down-regulate their oxidative metabolism when nutrients get depleted or growth rate slows.15 Flux analysis shows significant rewiring of intracellular metabolic fluxes in the transition from growth to stationary phase. Changes were noticed in energy metabolism, redox metabolism, oxidative pentose phosphate pathway and anaplerosis.16 In stationary phase glycolysis is rerouted through PPP pathway - no lactate production is observed.2 Unusual lipogenic pathway was discovered with modeling. Carbon from glucose supplies mitochondrial production of alpha-ketoglutarate (aKG), which is trafficked to the cytosol and used to supply reductive carboxylation by isocitrate dehydrogenase.46 It was suggested that endogenous lactate is not being used for ATP production through TCA cycle when the medium is supplemented with galactose. It was observed that lac-tate starts to get depleted at the same time as glucose is used up and cell switches to a galactose source.1 With kinetic model of CHO cell growth Lopez-Meza estimated substrate threshold below which growth is not observed and obtained the a and ^ factor from Luedeking-Piret equation.18 Analysis showed that CTP deficient cells use different central carbon metabolism, suppress pyruvate dehydrogenase, and induce glucose dependent anaplerosis through pyruvate carboxylase.46 Composition optimization of limiting amino acids in growth medium increased maximum cell density by 55% and protein titer production by 27%.3 The medium did not have any influence on sialic acid content. Mannose carbon source also improved recombinant protein productivity.47 Glucose consumption was 5 times higher than that of glu-tamine (13C labeled glucose, 2D-NMR spectroscopy). Error propagation of Goudar metabolic flux analysis can be routinely used in bioprocess development.14 Modeled effect of specific rate error on the flux error is a function of both the sensitivity of the flux with respect to the specific rate and relative magnitudes of the flux and the specific rate.14 41% of glucose was channeled through PPP, while flux of pyruvate to lactate and to TCA cycle was evenly distributed 55%-45%; anaplerotic conversion to oxaloacetate accounted to 10% of whole pyruvate conversion.14 We describe the research done in the area of bioreac-tor modeling and contribution of our department to the field. Complete literature search and data collection of known metabolic fluxes was made to ease the design of future models. The improvement of models is ongoing with a goal to make a good theoretical framework and to foresee possible errors in the fermentation process. Schematic illustration of the interplay between metabolic engineering and bioprocess is shown in Figure 1. 2. Contribution of the Authors Modeling brings new insights into existing problems. Great advantage of modeling is to track the problem at molecular, cellular level. Good hypothesis and theories cannot be developed without sufficiently developed model. Development of biotechnology processes is thus easier and cost effective. Models have many disadvantages, because they may be developed on unproven hypothesis or theories that yet need to be confirmed. Theories may have in this case erroneous core. Complete literature search of published models and metabolism of CHO cells leads to a successful build of model of bioreactor. Review of preferred articles is included with the emphasis on bioreactor operation equations and metabolic pathways of CHO cell lines. Contribution is divided into three parts: • Overview of black box models: Evolution of mathematical equations used in CHO mechanistic models • Overview of CHO cell metabolic pathways and the unique properties of CHO cell lines • Overview of mechanistic models and our proposed approach toward solution of the problem 2. 1. Review of Black Box Models: Formulation of Mathematical Equations Protein production in a bioreactor requires perfect agitation and aeration to maintain a homogeneous distribution of cells, substrate and oxygen throughout the biore-actor. Extracellular medium composition analysis, coupled with intracellular metabolic pathway analysis give rise to CHO cell bioreactor models. 2. 1. 1. Growth of Biomass Growth of biomass is described in different ways. Growth is influenced by the specific growth factor (y) and concentration of cells (X) as seen in Eq. (1).16,19,20 Standard biomass growth equation: dx -—= [iX d t r (1) Specific growth factor (y) is described in different ways. Commonly used descriptions are Verhulst and Monod equations, which can be adequately modified to account for bioreactor conditions (Eqs. 2 and 6). One limit substrate Monod36 kinetics is: (2) Gaspersic et al.: Metabolic Network Modelling of Chinese Hamster 772 Acta Chim. Slov. 2018, 65, 769-786 while two limit substrates Monod24 kinetics is expressed as: [gi] [Sz] — ¡Imax [S1]+KSl [S2]+A-x2 and finally «-limit substrates Monod24 kinetics as: [Si] [Szl v v [Snl max [S1]+KSl [S2]+Ks2 ■ X ... X • [Snl+^Sn (3) (4) By Monod, biomass growth is influenced by limiting substrate (Eq. 2). Here X represents cell concentration -number of cells per mL; S represent substrate, ^max is the maximum value of growth rate, KS is the substrate saturation constant. Note that specific growth rate may be influenced by one limiting substrate (Eq. 2),18 two (Eq. 3)37 or more limiting substrates from the growth medium (Eq. 4).48 del Val38 proposed modified expression: Ii [s] PU (5) By del Val, an upgraded Monod equation for continuous production is utilized, where Xv is the density of viable cells and ax is the specific factor (cellular carrying capacity). Viable cells represent live cell population, while total cells represent a sum of viable and dead cell population. The ratio is changing through cell phases. Each equation is optimized for specific value, so it is necessary to take into account certain value. Verhulst49 established the following closure: M- Mm X (i- ) (6) By Verhulst, maximum cell density of the culture (Xmax) is a limiting factor to cell growth. In this case substrate is available in large quantities or may be fed additionally to the bioreactor. The equation is commonly used to describe the growth of bacterial cultures. Equations for biomass growth that are commonly used in bioreactor systems are described below (continuous, fed batch). Altamirano17 and Xing3 suggested to use the following formulation: Xy (7) By Altamirano, the specific growth rate in the continuous bioreactor is described as dilution rate (D) multiplied by the ratio of the total cell number (Xt) and the viable cell number (XV). Goudar4 wrote the growth factor as: ' V V + ■ 1 ■V dt (8) By Goudar, the specific growth rate is designed for perfusion systems, where V is the reactor volume, Fd is the discard rate, F^ is the harvest flow rate, XH is the harvest viable cell density, and XB is the density of viable cells in bioreactor. Hagrot21 used the solution of first-order kinetics: (9) By Hagrot, the specific growth rate depends upon viable cell concentration before renewal (Xv) and after renewal (Xv0). Here t and t0 are corresponding times of sampling. It is essential for the model of bioreactor to select the mathematical formulation that suits type of the biore-actor and the cell culture. To get the best formulation we need to compare numerical simulations with experimental data and choose the best fitting curve. For that we need a set of sensor concentration data of products, substrates, and biomass in bioreactor throughout fermentation process. 2. 1. 2. Final Product Formation Final product formation is described in connection with the cell concentration (X) or viable cell concentration (Xv) multiplied by product growth factors (r or a) (Eq. 10 and 11).20,33 Standard equation to describe product formation: dp —~ = rX dt (10) Naderi20 and Provost19 wrote down the following closure: dp _ dt _ a9 while Ludeking-Piret35 suggested additional term: dp dx ov — = a — + ßX dt dt r (11) (12) By Ludeking-Piret, two coefficients a and p describe the product formation, both are product specific. Further, P is the concentration of glycosylated product, and dX/dt is the slope of biomass increase. Xing3 and Berrios47 suggested two formulations: (13) (14) By Xing, product growth is described by the ratio of concentration difference of product (Q) between two time points. Xv is the viable cell density, and D is the dilution factor (Eq. 13). By Berrios concentration difference of the product (ci) was divided by the corresponding difference of integral viable cell density (IVCD) (Eq. 14). Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 773 Acta Chim. Slov. 2018, 65, 769-786 Zamorano24 proposed linear relationship: (15) By Zamorano, the r factor (Eq. 10) was substituted by multiplication of stoichiometric matrix for final product (Np) and metabolic flux (v(t)). Goudar4 utilized the extended closure: (16) By Goudar, the product growth consists of two terms. Here XB is viable cell density, and Fm is the flow rate. The equation is used to describe perfusion systems. del Val38 used the following expression: f = (YP/s)qsX (17) By del Val, the expression describes product growth as a yield of product per substrate (YPIS) multiplied by the substrate consumption (qS). The formula for product growth must be developed for each individual product and is based on the observable physical and biochemical strain characteristics and the type of bioreactor. Reviewed models are optimized to describe formation of secondary metabolites - end products 1-5,15,17,19,21, 24,29,30,31,37,38,40,43,45,46,50 por production of antibodies post translational glycosilation should be taken into account as well.37,38,40 Formation of primary metabolites - important for growth and maintenance of cell were most precisely described in Quek's model with thorough description of nucleotide and lipid synthesis.5 2. 1. 3. Mathematical Description of Substrate Consumption Substrate consumption is dependent upon cell density (X) and specific uptake rate (vS) as follows:17,24,33 (18) Jedrzejewski37 proposed the formulation with two terms: ds Tt = (vt7+maic)x ■flic (19) By Jedrzejewski substrate consumption is influenced by number of cells, steady state consumption of substrate (mglc), and biomass growth coefficient (Ygk). Goudar4 and Xu6 suggested another equation: dS dt = -(■ xS V Fm(Sm-S) dS\ Tt)x (20) By Goudar and Xu specific substrate consumption rate depends upon perfusion rate Fm, difference of sub- strate concentration (starting minus current) (Sm-S), bioreactor volume (V), and viable cell density (XV). del Val38 extended the Michaelis-Menten kinetics as: ds dt X (21) By del Val, YX/S is the yield coefficient from substrate, S represents concentration of substrate, Km is constant specific for each substrate. Ahn16 introduced another expression: dt ^ (22) By Ahn, decomposition of substrate k is independent of biomass. S represents substrate concentration, X represents cell concentration, and qs is a substrate consumption per cell concentration unit. 2. 1. 4. Oxygen Consumption Oxygen consumption is traditionally described as follows:5,17,50 dDO dt = OTR- OCR OTR = Kxa (DO*-DO) (23) (24) Oxygen concentration is dependent upon oxygen transfer rate (OTR) between gas and liquid phase and oxygen consumption rate (OCR) of biomass.29,51 OTR is dependent upon KLa (bioreactor specific) and dissolved oxygen concentration (DO). Note that saturated oxygen (DO*) is temperature dependent.29 Jorjani52 used the following expression: OCR = q02 Xv; qo2 qo2° e"E/RT (25) By Jorjani, OCR is dependent upon qO20 and temperature,52 while qO20 is cell culture and clone specific. It depends mostly on the number and condition of mitochondria (origin of respiratory chain). It is also dependent upon availability of substrate with which cell and later mitochondria are fed.53 Nyberg29 proposed two terms: OUR = Kxa (C*-Cr) + D (Cf-Cr) (26) By Nyberg, the formula accounts for the continuous bioreactor, where oxygen uptake rate (OUR) is dependent upon OTR as (KLa (C*-CR)), dilution rate (D), and the difference between oxygen concentrations (feed minus reactor) as (CF-CR). C represents saturated oxygen concentration at operating temperature. Oxygen is consumed in the respiratory chain. H2O is the byproduct of the respiratory chain reactions.54 Oxygen that is incorporated in waste CO2 comes from glucose or Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 774 Acta Chim. Slov. 2018, 65, 769-786 other substrates. Since metabolism is affected by pH value, O2 consumption (metabolism) is pH dependent.55 2. 1. 5. CO2 and NH3 Waste Production CO2 production is integrated into models to address reactions in TCA cycle.5,45,50 CO2 is mostly produced in Krebs cycle, while NH3 is produced during metabolism of glutamine and other amino acids.5,17,33,56 (27) CO2 concentration is dependent upon CO2 transfer rate (CO2TR), and CO2 production from biomass (CO2PR). CO2 importantly influences cell growth and productivity.57 Free CO2 can be recycled together with amino group during fixation into carbamoyl phosphate and later integrated into arginine (urea cycle). Dissolved CO2 (DCO2) partially transforms into HCO3- ions, which serve as a regulator of pH in the cell. CO2 is generated during pyruvate, isocitrate and oxoglutarate degradation (TCA cycle). It is also generated in lysis of lysine and glycine as well as in formation of ribose-6-phosphate from glucose-6-phos-phate. RCo2 represents rate of CO2 production. Nyberg29 suggested another expression: CER = % (jco2 - yFC02)+ D (C* - C/) (28) By Nyberg, expression accounts for carbon dioxide evolution rate. Here ng is molar gas flow rate to the reactor, is liquid volume of the reactor, Vr is mole fraction of CO2 in reactor headspace, yRC02 is mole fraction of CO2 in the feed gas. CRA is concentration of CO2 and bicarbonate in the liquid phase, CFA is concentration of CO2 and bicarbonate in the liquid feed phase. (29) In Eq. (29), concentration of NH3 has similar mathematical dependence as CO2 in Eq. (27). Note that KLa is different for each gas and varies with the reactor and temperature. Ammonia production is highly dependent upon lysis of amino acids that can be biomass dependent and/or independent (catalytic degradation in water medium dependent upon temperature). Ammonia is produced from the amino acids and transferred into gas phase. Then, it is recycled during formation of carbamoyl phosphate. 2. 1. 6. Temperature, pH Optimum Temperature and pH are important process parameters that influence optimal growth of CHO cell lines.58-64 Normal operating temperature for the growth of mammalian cells is 37 °C.65 At 33 °C a remarkable decrease in specific growth rate is observed. At 30 °C, growth of cells starts to stagnate. Mammalian cells grow in the range from 35 to 38 °C.55,66 On the other hand, production rate of product (unspecified recombinant protein) is increased at 33 and 30 °C. Lower temperatures (below 37 °C) inhibit cell growth, but enhance cellular productivity of the recombinant protein, maintain high cell viability, suppress consumption of nutrients from medium, and suppress release of waste products from the cells.67,68 CHO cells have been reported to grow best at pH 7.1.55 Maximum product concentration (recombinant protein) was achieved at pH 6.8; 1.8-fold higher than at pH 7.1.55 Regardless of the culture temperature, the highest specific growth rate was observed in the range of pH from 7.0 to 7.4.34 2. 1. 7. Cell Phases of CHO Cell Lines in Bioreactor When cells are transferred to a new bioreactor batch, they need time to reach stable operation (lag phase). During lag phase cells adapt to new environment and multiply enzymes that are needed to catalyze biochemical reactions. Initially, concentrations of glucose and other amino acids are falling, while lactate and glutamate (byproducts) concentrations are rising in the medium. After glucose is depleted, lactate starts to get consumed. When substrates are depleted, biomass stops to grow and uses internal reserves to maintain cell functions. Reserve glycogen and li-pids are used to supply the cell with the energy. The nutrients from dead cells can be recycled and reused as the energy source. Most commonly used substrate is glucose. If substrate is switched for example to galactose, specific enzymes must be multiplied to reach optimal concentrations, before cells can re-adapt. In the presence of large concentrations of substrates inside cells, mitochondria divide rapidly and supply the cells with large amounts of energy (ATP). Until mitochondria sufficiently multiply, the excess flux of glucose is diverted to lactate or other metabolites, such as alanine. In the growth phase cells multiply with ease. The lac-tate does not accumulate anymore and is used up by cells as a substrate. Cells stop to grow when they reach maximum cell density or when they have used up all substrates. Then, cells start to use nutrients from energy storage: gly-cogen and lipid molecules. When cells run out of the substrates and nutrients from energy storage they enter an atypical cell death due to starvation. The viable CHO cells are in G1, S, G2/M (part of the interphase) or apoptotic phase.69,70 The production of protein is usually phase specific.69,66 In apoptosis cell enter programmed cell death. After certain number of duplications, cells die off. At that time significant amount of biomass in the bioreactor belongs to dead cells. Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 775 Acta Chim. Slov. 2018, 65, 769-786 3 * o 3 (5 T3 » s a s o ^ g a v s ja *g f S3 2 J3 Ü T3 U ,û ja ft m (5 £ a s v Q & N •S j J « Js T3 £ s « s T3 o .iï -! >1 c c ^ CO CO CO CO CO W W W o w w W o ^ >1 >1 c ¡H ^ G CO CO CO CO CO W o W o o w W W ^ G >1 c G ^ ^ CO CO CO CO W o W o o W o W ^ G >1 c G ^ G CO CO CO CO CO W W W o o W o W >-! >1 c G G S3 g >H C CO CO W W >H >H W ° o o G G O o G G CO CO W W >-i ^ CO CO W W >H >H CO CO W W >H >H CO CO W W >H >H « -S "H ^ ^ OJ O O £1= O ¿S w w CO co w w >H >H CO CO CO WWW >H >H >H o o o p^ p^ o ^ ^ ^ ^ ^ ^ CO CO CO WW W >H >H >H CO CO W W >H >H o o o o pq o G G G G ^ G CO W o o o o o o G G G G G G G G G G G G O O O O o O G G G G G G W >1 W >H >H >H W CO W -, CO CO 2 W W C >H >H CO CO CO WW W >H >H >H CO CO CO WWW >H >H >H -G Sp o g CA CA 'S £ ¡1 £ 10 1 o rS o C £ c o o G G W G G " G M k^j w w ° S <3 « on « S tion g o CA 'o •is u o -G n « « •Û ci n &C « o js 'Ï3 o -C Jc O „ -H O C 2. 2. Overview of Important Metabolic Pathways In the past two decades much research has been done on CHO cell metabolism. At modeling, cell metabolism is described using kinetic laws, equilibrium equations and associated parameters. Table 1 shows metabolic pathways, which were included in the models published in the literature. The following pathways were reviewed: (i) substrate intake, (ii) glycolysis, (iii) glutaminolysis, (iv) pentose phosphate pathway (PPP), (v) UDP-monosaccharides production, (vi) nucleotide synthesis, (vii) amino acid metabolism, (viii) tricarboxylfic acid cycle (TCA), (ix) lipid metabolism, (x) glycogen synthesis, (xi) lipid synthesis, (xii) DNA duplication, (xiii) RNA transcription and protein translation, (xiv) glycosylation, and (xv) feedback loops. 2. 2. 1. Substrate Intake (Part of Metabolic Flux Pathway Analysis) High influxes of substrates contribute to high osmotic pressure.71 The cell regulates its pressure with fast conversion into more favorable intermediates and products. Other regulatory mechanisms are passive and active transport. One of the major metabolites is glycogen, which serves as energy storage.72 Glucogenic amino acids may transform into glucose and then into glycogen, while keto-genic amino acids transforms into ketone bodies that may be stored through acetyl CoA in fatty acids (Figure 2).73 Mathematical descriptions (Eqs. 18-22) of substrates' consumption are selected in accordance with phenotype, while associated constants are substrate specific. Majority Figure 2: Biochemical reactions associated with mitochondria: degradation pathway of keto- into glucogenic amino acids.45 Glucogenic amino acids are transformed into acetylCoenzyme A (AcCoA), while keto amino acids are transformed to glutamate that is further transformed into alpha-ketoglutarate (aKG). Three letter amino acid code is standard. From "A structured, dynamic model for animal cell culture systems," by C. S. Sanderson, J. P. Barford, and G. W. Barton, Biochem. Eng. J., 1999, 3, 203-211. Reprinted with permis- Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 776 Acta Chim. Slov. 2018, 65, 769-786 of the models include glucose intake, lactate, ammonia, CO2 and oxygen. More precise models include also transport of glutamine and glutamate. 2. 2. 2. Glycolysis and Glutaminolysis (AA Lysis) Glycolysis is one of the most important pathways in the metabolism of cells and is therefore included in majority of the models (Table 1).1-5,15,17,19,21,24,29-31,43,45,46,50 In the growth phase cells extensively use glucose and glutamine. Apart from normal cells, the phenomenon is enhanced in mutated, cancerous cells.74 Due to osmotic pressure, excess glucose and glutamine are diverted into glycogen,71 lactate, glutamate, and lipids.1,16,43,75 Glucose enters tricarboxylic acid (TCA) cycle through pyruvate (Figure 3). Lactate is a waste product of surplus pyruvate produced during glycolysis that cannot enter TCA cycle due to unavailable mitochondria machinery. Lack of oxygen produces similar effect (Figure 3). Lactate is removed from the cell into the cell medium and lat- er reused.1,18 Two molecules of ATP and two molecules of NADH are generated in the process. When pyruvate enters mitochondria it is converted into 3 molecules of CO2. In this process three molecules of NADH, one molecule of FADH2 and one molecule of GTP are produced per molecule of pyruvate. Each molecule of NADH produces three molecules of ATP through electron transport chain; each molecule of FADH2 produces two ATP molecules due to lower proton energy. If lactate is produced, one molecule of NADH is consumed, and recycled when lactate is being converted back. High concentration of lactate may lead to lower pH. Note that pH is regulated with NH3 release during glutamate metabolism. Lysis of amino acids gives huge amount of ATP due to independent enzymatic pathways to TCA cycle (Figure 4). In degradation pathways of isoleu-cine three NADH molecules, one acetyl CoA and one suc-cinyl CoA molecule are generated. ATP is further generated through electron transport chain. Metabolic models comprise metabolic steps of glycolysis pathway into lumped reactions due to less impor- Figure 3: Overview of metabolic pathways in the cytosol: Precise metabolic pathway of catabolism and anabolism in CHO mammalian cell cytosol. Pathways of glucose conversion into pyruvate, glycogen and other monosaccharide's are presented. Formation of nucleotides, lipids, nucleotide monosaccharide's is presented together with cytosolic amino acid synthesis reactions. Amino acids are colored in blue; phosphate rich molecules are colored in green; ADP, NADH in red; gases in orange.1-5, 13, 15, 17, 19, 21, 29, 30, 45, 46, 50, 76 Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 777 Acta Chim. Slov. 2018, 65, 769-786 Figure 4: Metabolic pathways within mitochondria: TCA cycle is the major metabolic pathway. Fatty acids are pre-metabolized in beta oxidation cycle (purple). Amino acids (blue) enter TCA cycle through oxoglutarate, fumarate, and succinyl CoA. Oligosaccharides enter TCA cycle through PPP and glycolysis pathway. Gases are colored in orange, while phosphate rich energy molecules are colored in green.1-5'15'17'19"21'24,29-31'43'45'46'50 tant, short half-life intermediates.1-4,17,21,29,46 In simplified models, fluxes are diverted directly to stable intermediates. Complete lysis of amino acids shows that amino acids regenerate intermediates of TCA cycle much faster compared to glycolysis due to independent enzyme machinery of each amino acid. Enormous energy comes from branched chain amino acids (BCAA), i.e. valine, isoleu-cine, and leucine (Figure 4). 2. 2. 3. Pentose Phosphate Pathway and UDP-Monosaccharides Stock Like glycolysis, pentose phosphate pathway (PPP) is also included in several models (Table 1).1,2,4,5,15,17,19,30,45,46,50 An alternative route for glucose is generation of different monosaccharides (Figure 3). From glucose-6-phosphate cell produces ribulose-5-phosphate, xylulose-5-phosphate, erythrose-4-phosphate, fructose-6-phosphate sedoheptu-lose-7-phosphate and ribose-5-phosphate.33 Ribose-5-phosphate enters synthesis of nucleotides.5 Monosaccha- rides from PPP can be transformed into glyceraldehyde-3-phosphate and fructose-6-phosphate, which again enters glycolysis pathway (Figure 3). Few models also include generation of UDP-mono-saccharides (Figure 3).37,38,40 In excess of UTP, different nucleotide sugars are generated, including GDP-mannose, UDP-galactose, UDP-glucose, UDP-N-acetyl glucosamine (GlcNAc), and CMP-sialic acid. Nucleotide sugars are included in the models, which describe glycosylation pattern of proteins (Figure 3).29,37-39 Nucleotide sugars are transported into the endoplasmic reticulum (ER) and the Golgi apparatus (GA), where they get concentrated and ready to be attached to proteins (Figure 3). The attachment is carried out through covalent bond between sugar residue and protein through N-phospho-glycosylation.37,77 2. 2. 4. Glycogen Synthesis Glycogen synthesis is included only in a few models (Table 1),5 despite its important role in glucose homeosta- Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 778 Acta Chim. Slov. 2018, 65, 769-786 Figure 5: Synthesis of fatty acids from acetyl-CoA: Complex enzymatic pathway leads to different lipid molecules that assemble into membrane bilayer. The saturated fatty acids and unsaturated fatty acids of the n-3, n-6, n-7 and n-9 series can be synthesized from myristic acid (C14) and palmitic acid (C16), produced from ACC and FASN. Long-chain fatty acids of n-6 and n-3 series can also be synthesized from precursors obtained from dietary precursors to elongation (ELOVL) and desaturation (FADS) steps as indicated in the pathways. Lipids marked red and green are "up" and "down" regulated in our analysis, respectively. Increase in enzyme activities is framed in red whereas a decrease is framed in green. ACC: acetyl-CoA carboxylase; ELOVL: elongase of very long chain fatty acid; FASN: fatty acid synthase; FADS: fatty acid desaturase; SCD: stea-royl-CoA desaturase.78 From "Metabolism dysregulation induces a specific lipid signature of nonalcoholic steatohepatitis in patients," by F. Chiappini, A. Coilly, H. Kadar, P. Gual, A. Tran, C. Desterke, D. Samuel, J. C. Duclos-Vallée, D. Touboul, J. Bertrand-Michel, A. Brunelle, C. Guettier, and F. Le Naour, Sci. Rep., 2017, 7, 1-17. Reprinted with permission. sis. In the excess of ATP, cell transfers extra glucose into glycogen that is used as energy source, when glucose is run out from the medium or cytosol. One molecule of glycogen can store up to 30000 molecules of glucose. This contributes to lower osmotic pressure and thus removes harmful effects of high glucose content. Glucose is readily available, if there is a demand on energy or carbon source (Figure 3). The path described above is ATP controlled. Glucose is transformed into glucose-1-phosphate; in this process one molecule of ATP is consumed. UTP molecule binds to glucose-1-phosphate, leading to production of UDP-glucose. UDP-glucose attaches itself to glycogen, leading to release of UDP. 2. 2. 5. Lipid Synthesis Lipid synthesis is included in several models (Table 1). They used simplified pathways13,15,17,50,76 or very precisely defined pathways.5 During cell division, lipid synthesis is enhanced to simulate assembly of membrane. Phosphoglycerides, triglycerides, phosphatidylserine, phosphatidyllecithine, cholesterol, sphingomyelin, and geranyl pyrophosphate are synthesized (Figure 3). Successive enzymatic pathway starts wih the acetyl-CoA and leads to complex lipid molecules (Figure 5). Different glycerides are synthesized with the addition of fatty acids and other groups to glycerol. 2. 2. 6. Amino Acid Synthesis Amino acid degradation is described in most models, but usually only partial set of amino acids is integrated in a model (Table 1).1-5,15,17,19,21,24,29-31,43,45,46,50 Altamira-no et al17 includes synthesis of alanine, aspartate and glu-tamine in highly interconnected metabolic network. Quek et al5 describes precise metabolic network with interconnected synthesis of amino acids: alanine, glutamate, aspar-agine, proline, serine, glycine, aspartate. Provost et al19 includes synthesis of alanine into the model. Essential amino acids cannot be synthesized de novo (from scratch) by the organism, therefore must be supplemented from the medium. There are nine amino acids that humans cannot synthesize: phenylalanine, valine, threo-nine, tryptophan, methionine, leucine, isoleucine, lysine, and histidine (single letter abbreviations in the order they appear: F, V, T, W, M, L, I, K, and H). Other amino acids are synthesized from essential amino acids and other cell intermediates (Figure 6). 2. 2. 7. DNA Duplication, RNA Transcription and Protein Translation Nucleotides are synthesized through PPP. In a few models nucleotide synthesis is incorporated.5,19 Provost et al19 describes highly simplified model. Quek et al5 describes precise metabolic pathway, where glucose is transformed into ribose, which is further converted to IMP (in-osine monophosphate-purine precursor) and UMP (uridine monophosphate-pyrimidine precursor). These precursors are then transformed into ATP, CTP, UTP, GTP, dATP, dCTP, dTTP, and dGTP (Figure 3). Duplication of DNA and translation of RNAs take place in the cell nucleus. mRNA is transported to the cytosol and rRNA takes position in ribosome, while mRNA is translated into protein by the aid of tRNA. Nucleotide triphosphates (NTPs) are used as an energy source during transcription. For each amino acid, codon is assembled from three NTPs. mRNA is assembled of: 5' cap, 5' and 3' untranslated region (UTR), sequence for signal peptide, coding region, and poly AAA end. mRNA exits the nucleus and enters a ribosome, where the protein is translated Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 779 Acta Chim. Slov. 2018, 65, 769-786 ketobutyrate 5-phosphoribosyl 1-pyrophosphate ATP Serine NH4 Cys Cystathione Lysine Threonine Leucine Isoleucine Valine Histidine Phe—>Tyr Phosphoribosyl-ATP Homocysteine t SAM I ADP ATP Methionine Alanine Kynurenine Pyruvate Tryptophane Phosphorlbosyl-AMP H20____Glutamine Glycerate-3P —> 3-Phosphohydroxypyruvate(3.0.CO;2-G 12. H. P. Bonarius, V. Hatzimanikatis, K. P. Meesters, C. D. de Gooijer, G. Schmid, J. Tramper, Biotechnol. Bioeng. 1996, 50, 299-318. DOI:10.1002/(SICI)1097-0290(19960505)50:3< 299::AID-BIT9>3.0.CO;2-B 13. F. Zamorano, A. V. Wouwer, G. Bastin, J. Biotechnol. 2010, 150, 497-508. DOI:10.1016/j.jbiotec.2010.09.944 14. C. Goudar, R. Biener, C. Boisart, R. Heidemann, J. Piret, A. de Graff, K. Konstantinov, Metab. Eng. 2010, 12, 138-149. DOI:10.1016/j.ymben.2009.10.007 15. J. D. Young, Curr. Opin. Biotech. 2013, 24, 1108-1115. DOI:10.1016/j.copbio.2013.04.016 16. W. S. Ahn, M. R. Antoniewicz, Metab. Eng. 2011, 13, 598609. DOI:10.1016/j.ymben.2011.07.002 17. C. Altamirano, A. Illanes, A. Casablancas, X. Gamez, J. J. Cairo, C. Godia, Biotechnol. Progr.. 2001, 17, 1032-1041. DOI:10.1021/bp0100981 18. J. Lopez-Meza, D. Araiz-Hernandez, L. M. Carrillo-Cocom, F. Lopez-Pacheco, M. del Refugio Rocha-Pizan, M. M. Alvarez, Cytotechnology. 2016, 68, 1287-1300. 19. A. Provost, G. Bastin, J. Process Control. 2004, 14, 717-728. DOI:10.1016/j.jprocont.2003.12.004 20. S. Naderi, M. Meshram, C. Wei, B. McConkey, B. Ingalls, H. Budman, J. Scharer, Biotechnol. Prog. 2011, 27, 1197-1205. DOI:10.1002/btpr.647 21. E. Hagrot, H. A. Oddsdottir, J. G. Hosta, E. W. Jacobsen, V. Chotteau, J. Biotechnol. 2016, 228, 37-49. DOI:10.1016/j.jbiotec.2016.03.015 22. J. Robitaille, J. K. Chen, M. Jolicoeur, PLoS ONE. 2015, 10, e0136815. DOI:10.1371/journal.pone.0136815 23. J. A. Papin, N. D. Stelling,; S. Price Klamt; S. Schuster; B. O. Palsson, Trends Biotechnol. 2004, 22, 400-405. DOI: 10.1016/j.tibtech.2004.06.010 24. F. Zamorano, A. V. Wouwer, R. M. Jungers, G. Bastin, J. Biotechnol. 2013, 164, 409-422. DOI:10.1016/j.jbiotec.2012.05.005 25. P. Farzan, B. Mistry, M. G. Ierapetritou, AIChE J. 2017, 63, 398-408. DOI:10.1002/aic.15442 26. B. Berry, J. Moretto, T. Matthews, J. Smelko, K. Wiltberger, Biotechnol. Progr. 2015, 31, 566-577. DOI:10.1002/btpr.2035 27. X. Chen, A.P. Alonso, D.K. Allen, J. L. Reed, Metab. Eng. 2011, 13, 38-48. DOI:10.1016/j.ymben.2010.11.004 28. N. E. Lewis, H. Nagarajan, B. O. Palsson, Nat. Rev. Microbiol. 2012, 10, 291-305. DOI:10.1038/nrmicro2737 29. G. B. Nyberg, R. R. Balcarel, B. D. Follstad, G. Stephanopou-los, D. I. C. Wang, Biotechnol. Bioeng. 1998, 62, 324-335. DOI:10.1002/(SICI)1097-0290(19990205)62:3<324::AID- BIT9>3.0.CO;2-C 30. J. Dean, P. Reddy, Biotechnol. Bioeng. 2013, 110, 1735-1747. DOI:10.1002/bit.24826 31. A. Ghorbaniaghdam, O. Henry, M. Jolicoeur, Bioproc. Bio-syst. Eng. 2013, 36, 469-487. DOI:10.1007/s00449-012-0804-3 32. B. Ben Yahia, B. Gourevitch, L. Malphettes, E. Heinzle, Bio-technol. Bioeng. 2017, 114, 785-797. DOI:10.1002/bit.26214 33. A. Provost, G. Bastin, S. N. Agathos, Y. J. Schneider, Biopro-cess Biosyst. Eng. 2006, 29, 349-366. DOI: 10.1007/s00449-006-0083-y 34. S. N. Galleguillos, D. Ruckerbauer, M. P. Gerstl, N. Borth, M. Hanscho, J. Zanghellini, Comput. Struct. Biotechnol. J. 2017, 15, 212-221. DOI:10.1016/j.csbj.2017.01.005 35. Meng Ji, Yelian Miao, Jie Yu Chen, Yebing You, Feilong Liu, and Lin Xu. Springerplus 2016, 5, 503. DOI:10.1186/s40064-016-2151-3 36. Monod J. Annu. Rev. Microbiol., 1949, 3(1), 371-394. DOI: 10.1146/annurev.mi.03.100149.002103 37. P. M. Jedrzejewski, I. J. del Val, A. Constantinou, A. Dell, S. M. Haslam, K. M. Polizzi, C. Kontoravdi, Int. J. Mol. Sci. 2014, 15, 4492-4522. DOI:10.3390/ijms15034492 38. I. J. del Val, K. M. Polizzi, C. Kontoravdi, Sci. Rep. 2016, 6, 1-16. DOI:10.1038/srep28547 39. P. N. Spahn, N. E. Lewis, Curr. Opin, Biotech. 2014, 30, 218224. DOI:10.1016/j.copbio.2014.08.004 40. J. Krambeck, M. J. Betenbaugh, Biotechnol. Bioeng. 2005, 92, 711-728. DOI: 10.1002/bit.20645 41. S. Hutter, T. K. Villiger, D. Bruhlmann, M. Stettler, H. Broly, M. Soos, R. Gunawan, Metab. Eng. 2017, 43, 9-20. DOI:10.1016/j.ymben.2017.07.005 42. E. S. Bayrak, T. Wang, A. Cinar, C. Undey, IFAC-PapersON-Line. 2015, 48, 1252-1257. DOI:10.1016/j.ifacol.2015.09.140 43. R. P. Nolan, K. Lee, Metab. Eng. 2011, 13, 108-124. DOI:10.1016/j.ymben.2010.09.003 44. N. Templeton, K.D. Smith, A. G. Mcatee-Prereira, H. Dorai, M. J. Betenbaugh, S. E. Lang, J. D. Young, Metab. Eng. 2016, 43, 218-225. DOI:10.1016/j.ymben.2017.01.008 45. S. Selvarasu, Y. S. Ho, W. P. K. Chong, N. S. C. Wong, F. N. K. Yusufi, Y. Y. Lee, M. G. S. Yap, D. Y. Lee, Biotechnol. Bioeng. 2012, 109, 1415-1429. DOI:10.1002/bit.24445 46. L. Jiang, A. Boufersaoui, C. Yang, B. Ko, D. Rakheja, G. Guevara, Z. Hu, R. J. Debarardinis, Metab. Eng. 2017, 43, 198-207. DOI:10.1016/j.ymben.2016.11.004 47. J. Berrios, C. Alatmirano, N. Osses, R. Gonzales, Chem. Eng. Sci. 2011, 66, 2431-2439. DOI:10.1016/j.ces.2011.03.011 48. E. E. Gonzo, S. Wuertz, V.B. Rajal, Biotechnol. Bioeng. 2014, 111, 2252-2264. DOI:10.1002/bit.25284 49. Nishikant Shirsat, Avesh Mohd, Jessica Whelan, Niall J. English, Brian Glennon, and Mohamed Al-Rubeai. Cytotechnology 2015, 67(3), 515-530. DOI:10.1007/s10616-014-9712-5 50. C. S. Sanderson, J. P. Barford, G. W. Barton, Biochem. Eng. J. 1999, 3, 203-211. DOI:10.1016/S1369-703X(99)00021-2 51. T. Goudar, R. Biener, B. K. Konstantinov, J. M. Piret, Biotechnol. Progr. 2012, 25, 986-998. DOI:10.1002/btpr.155 52. P. Jorjani, S. S. Ozturk, Biotechnol. Bioeng. 1999, 64, 349-356. DOI:10.1002/(SICI)1097-0290(19990805)64:3<349::AID- BIT11>3.0.C0;2-V 53. S. R. Chowdhury, J. Djordjevic, B. C. Albensi, P. Fernyhough, Bioscience Rep. 2016, 36, e00286. DOI: 10.1042/BSR20150244 54. L. P. Zielinski, A. C. Smith, A. G. Smith, A. J. Robinson, Mito- Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 785 Acta Chim. Slov. 2018, 65, 769-786 chondrion. 2016, 31, 45-55. D01:10.1016/j.mito.2016.09.003 55. E. Trummer, K. Fauland, S. Seidinger, K. Schriebl, C. Lattenmayer, R. Kunert, K Vorauer-Uhl, R. Weik, N. Borth, H. Katinger, D. Muller, Biotechnol. Bioeng. 2006, 94, 1033-1044. D0I:10.1002/bit.21013 56. W. S. Ahn, M. R. Antoniewicz, Biotechnol. J. 2012, 7, 61-74. D0I:10.1002/biot.201100052 57. Z. Z. Xing, A. M. Lewis, M. C. Borys, Z. J. Li, Biotechnol. Bioeng. 2017, 114, 1184-1194. D0I:10.1002/bit.26232 58. N. Jenkins and A. Hovey, Biotechnol. Bioeng. 1993, 42, 10291036. D0I:10.1002/bit.260420903 59. B. Rossler, H. Lubben, G. Kretzmer, Enzyme Microb. Tech. 1996, 18, 423-427. D0I:10.1016/0141-0229(95)00121-2 60. S. Becerra, J. Berrios, N. Osses, C. Altamirano, Biochem. Eng. J. 2012, 60, 1-8. D0I:10.1016/j.bej.2011.10.003 61. Watanabe and Okada S., J. Cell Biol. 1967, 32, 309-323. D0I:10.1083/jcb.32.2.309 62. P. Ducommun, P. A. Ruffieux, A. Kadouri, U. von Stockar, I. W. Marison, Biotechnol. Bioeng. 2002, 77, 838-42. D0I:10.1002/bit.10185 63. S. K. Yoon, S. L. Choi, J. Y. Song, G. M. Lee, Biotechnol. Bioeng. 2005, 89, 345-56. D0I:10.1002/bit.20353 64. V Avello, B. Tapia, M. Vergara, C. Acevedo, J. Berrios, J. G. Reyes, C. Altamirano, Electron. J.Biotechn. 2017, 27, 55-62. D0I:10.1016/j.ejbt.2017.03.008 6.5 N. Kurano, C. Leist, F. Messi, C. Gandor, S. Kurano, A. Fiech-ter, J. Biotechnol. 1990, 6, 245-258. D01:10.1016/0168-1656(90)90040-1 66. J. S. Seo, Y. J. Kim, J. M. Cho, E. Baek, G. M. Lee, Appl. Microbiol. Biot. 2013, 97, 5283-5291. D0I:10.1007/s00253-013-4849-2 67. K. Furukawa, K. Ohsuye, Cytotechnology, 1998, 26, 153-64. D0I:10.1023/A:1007934216507 68. H. Kaufmann, X. Mazur, M. Fussenegger, J. E. Bailey, Biotech-nol. Bioeng., 1999, 63, 573-582. D0I:10.1002/(SICI)1097-0290(19990605)63:5<573::AID-BIT7>3.0.C0;2-Y 69. R. L. Dutton, J. Scharer, M. Moo-Young, Cytotechnology, 2006, 52, 55-69. D0I:10.1007/s10616-006-9041-4 70. V Hendrick, P. Winnepenninckx, C. Abdelkafi, O. Vande-putte, M. Cherlet, T. Marique, G. Renemann, A. Loa, G. Kretzmer, J. Werenne, Cytotechnology 2001, 36, 71-83. D0I:10.1023/A:1014088919546 71. M. Rasouli, Clin. Biochem. 2016, 49, 936-941. D01:10.1016/j.clinbiochem.2016.06.001 72. A. Philip, M. Hargreaves, K. Baar, Am. J. Phisiol. 2012, 302, 1343-1351. 73. L. Hildebrandt, T. Spennetta, R. Ackerman, C. Elson, E. Shra- go, R. T. Ackermann, Biochem. Bioph. Res. Co. 1996, 225, 207-212. DOI:10.1006/bbrc.1996.1171 74. Moncada, S. Higgs, E. A. Colombo, S. L., Biochem. J. 2012, 446, 1-7. DOI:10.1042/BJ20120427 75. O. Feron, Radiother. Oncol. 2009, 92, 329-333. DOI:10.1016/j.radonc.2009.06.025 76. E. J. M. Blondeel, R. Ho, S. Schulze, S. Sokolenko, S. R. Guil-lemette, I. Sliva, Y. Durocher, J. G. Guillemette, B. J. McCon-key, D. Chang, M. G. Aucoin. J. Biotechnol 2016, 234, 127138. DOI:10.1016/j.jbiotec.2016.07.027 77. E. Maverakis, K. Kim, M. Shimoda, M. E. Gershwin, F. Patel, R. Wilken, S. Raychaudhuri, L. R. Ruhaak, C. B Lebrilla, J. Autoimmun. 2015, 57, 1-13. DOI:10.1016/j.jaut.2014.12.002 78. F. Chiappini, A. Coilly, H. Kadar, P. Gual, A. Tran, C. Desterke, D. Samuel, J. C. Duclos-Vallée, D. Touboul, J. Bertrand-Michel, A. Brunelle, C. Guettier, F. Le Naour, Sci. Rep. 2017, 7, 1-17. DOI: 10.1038/srep46658 79. B. Imperiali and S.E. O'Connor, Curr. Opin. Chem. Biol. 1999, 3, 643-649. DOI:10.1016/S1367-5931(99)00021-6 80. P. J. D'Huys, I. Lule, D. Vercammen, J. Anné, J. F. Van Impe, K. Bernaerts, J. Biotechnol. 2012, 161, 1-13. DOI: 10.1016/j.jbiotec.2012.04.010 81. F. Llaneras, A. Sala, J. Picó, J. Process Contr. 2012, 22, 19461955. DOI:10.1016/j.jprocont.2012.09.001 82. E. O. Voit, WIREs Syst. Biol. Med. 2017, 9, e1391. 83. H. Hefzi, K. S. Ang, M. Hanscho, A. Bordbar, D. Ruckerbauer, M. Lakshmanan, C. A. Orellana, D. Baycin-Hizal, Y. Huang, D. Ley, V. S. Martinez, S. Kyriakopoulos, N. E. Jiménez, D.C. Zielinski, L.E. Quek, T. Wulff, J. Arnsdorf, S. Li, J. S. Lee, G. Paglia, N. Loira, P.N. Spahn, L. E. Pedersen, J. M. Gutierrez, Z. A. King, A. M. Lund, H. Nagarajan, A. Thomas, A. M. Ab-del-Haleem, J. Zanghellini, H. F. Kildegaard, B. G. Voldborg, Z. P. Gerdtzen, M. J. Betenbaugh, B. O. Palsson, M. R. Andersen, L. K. Nielsen, N. Borth, D. Y. Lee, N. E. Lewis, Cell Syst. 2016, 3, 434-443. DOI:10.1016/j.cels.2016.10.020 84. Ž. Rejc, L. Magdevska, T. Tršelič, T. Osolin, R. Vodopivec, J. Mraz, E. Pavliha, N. Zimic, T. Cvitanovic, D. Rozman, M. Moškon, M. Mraz, Comput. Biol. Med. 2017, 88, 150-160. 85. I. J. del VAL, J. M. Nagy, C. A. Kontoravdi, Biotechnol. Prog. 2011, 27, 1730-1743. 86. T. K. Villiger, E. Scibona, M. Stettler, H. Broly, M. Morbidelli, M. Soos. Biotechnol Prog. 2016, 32, 1135-1148. DOI:10.1002/btpr.2315 87. Kaučič, V. (Ed.), J. Gašperšič, U. Novak, A. Pohar, B. Likozar, Zbornik referatov in povzetkov, Slovenski kemijski dnevi 2017, Portorož, Slovenia 2017, 1-6. 88. M. Kastelic, D. Kopač, U. Novak, B. Likozar, 2018, Biochemical Engineering Journal. https://authors.elsevier.com/tracking/article/details.do?aid =7095&jid=BEJ&surname=Kastelic) Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster 786 Acta Chim. Slov. 2018, 65, 769-786 Povzetek Epitelijske celice kitajskega hrčka (CHO) so ena izmed najbolj uporabljanih terapevtskih medicinskih linij za proizvodnjo različnih biofarmacevtskih zdravil. Imajo visoko stopnjo porabe s hitrim podvajanjem, kar jih naredi idealen biološki klon. Večja količina akumuliranih toksičnih intracelularnih intermediatov lahko privede do nižje viabilnosti organizma, produktivnosti proteinov in proizvedenih bioloških zdravil, zato je potrebna optimalna izbira sestave medija ter bioreaktorskih operativnih parametrov za vodenje bioprocesa. Natančno fenomenološko znanje o biokemijskih transformacijah v celičnem metabolizmu omogoča zaznavo težav, ki se lahko pojavijo med šaržnim, polkontinuirnim in konti-nuirnim obratovanjem bioreaktorja. Za boljše razumevanje (in povezave), so bili izdelani mehanistični modeli, ki se lahko uporabijo za optimiziranje obratovanja in povečevanja skale. V tem delu je narejen pregled glavnih metabolni poti v strukturiranih sesalskih kulturah CHO. Pregled se začne z biokemijskim ozadjem znotraj celica, ki nadzirajo povezane pojave in kinetiko, ki urejajo vzpostavljene biološke poti. Nato sledi opis posamezne pretvorbene poti, preko pregleda standardnih matematičnih formulacij, ki se običajno uporabljajo v inženirstvu. Te formulacije sledijo principu modeliranja t.i. črne škatle (black box), ki povezuje substrate/produkte na poenostavljen način. Poleg tega so predstavljeni modeli, ki vključujejo analizo metabolnih fluksov (MFA) in analizo uteženih fluksov (FBA). Na koncu je pregled podobnosti med različnimi pristopi za njihovo strukturno zasnovo. Izpostavljene so enačbe s spremenljivkami, ki se uporabljajo za opis rasti kontroliranih pogojev v bioreaktorju, raziskane reakcijske serije rasti populacije CHO, povezane z vrednostmi maksimalne encimske aktivnosti. Procesi so našteti tako, da lahko bralec integrira najsodobnejši pristop. Posebej je izpostavljen tudi prispevek avtorjev. Gašperšič et al.: Metabolic Network Modelling of Chinese Hamster