Skip to main content
ACS AuthorChoice logoLink to ACS AuthorChoice
. 2022 Oct 20;35(11):1962–1973. doi: 10.1021/acs.chemrestox.2c00128

Toward Realistic Dosimetry In Vitro: Determining Effective Concentrations of Test Substances in Cell Culture and Their Prediction by an In Silico Mass Balance Model

Dunja Dimitrijevic , Eric Fabian , Beate Nicol §, Dorothee Funk-Weyer , Robert Landsiedel †,‡,*
PMCID: PMC9682521  PMID: 36264934

Abstract

graphic file with name tx2c00128_0007.jpg

Nominal concentrations (CNom) in cell culture media are routinely used to define concentration–effect relationships in the in vitro toxicology. The actual concentration in the medium (CMedium) can be affected by adsorption processes, evaporation, or degradation of chemicals. Therefore, we measured the total and free concentration of 12 chemicals, covering a wide range of lipophilicity (log KOW −0.07–6.84), in the culture medium (CMedium) and cells (CCell) after incubation with Balb/c 3T3 cells for up to 48 h. Measured values were compared to predictions using an as yet unpublished in silico mass balance model that combined relevant equations from similar models published by others. The total CMedium for all chemicals except tamoxifen (TAM) were similar to the CNom. This was attributed to the cellular uptake of TAM and accumulation into lysosomes. The free (i.e., unbound) CMedium for the low/no protein binding chemicals were similar to the CNom, whereas values of all moderately to highly protein-bound chemicals were less than 30% of the CNom. Of the 12 chemicals, the two most hydrophilic chemicals, acetaminophen (APAP) and caffeine (CAF), were the only ones for which the CCell was the same as the CNom. The CCell for all other chemicals tended to increase over time and were all 2- to 274-fold higher than CNom. Measurements of CCytosol, using a digitonin method to release cytosol, compared well with CCell (using a freeze–thaw method) for four chemicals (CAF, APAP, FLU, and KET), indicating that both methods could be used. The mass balance model predicted the total CMedium within 30% of the measured values for 11 chemicals. The free CMedium of all 12 chemicals were predicted within 3-fold of the measured values. There was a poorer prediction of CCell values, with a median overprediction of 3- to 4-fold. In conclusion, while the number of chemicals in the study is limited, it demonstrates the large differences between CNom and total and free CMedium and CCell, which were also relatively well predicted by the mass balance model.

Introduction

Modern toxicological methods aim at the reduction, refinement, and replacement of animal tests while providing reliable data for risk and hazard characterization of chemicals.13 Key events observed in vitro are linked to in vivo adverse outcomes, and the corresponding concentrations in vitro and doses in vivo can be linked by “quantitative in vitro to in vivo extrapolation” (QIVIVE). Information on in vitro biokinetics and dosimetry of test chemicals in cell-based test systems is helpful to define toxicological effects and no-effect levels based from in vitro studies.46In vitro-derived toxicological endpoints generally relate to the nominal concentration (CNom), defined as the amount of a chemical added to the test system divided by the volume of the culture medium.710 However, CNom might deviate considerably from the actual concentrations in the medium and, importantly, the cellular concentrations at the target that exerts toxic effects.11,12 Therefore, the biologically effective concentration of a chemical should more accurately correlate to plasma and tissue concentrations in vivo to enable more accurate QIVIVE.1315

There are multiple factors that can alter the distribution and concentration of free concentrations of chemicals in the in vitro assays. These include adsorption of test chemicals, e.g., binding to vessels of culture flasks16 and/or serum proteins and lipids,5,17,18 evaporation, or spontaneous and enzymatic degradation of the test chemical. Other phenomena govern the uptake of chemicals into cells, including their ionization state and affinity to cellular targets such as binding to receptors and cell membranes, as well as accumulation into lysosomes.10,1921 The extent of these processes depends on the test system (e.g., the constituents of the culture medium, material of the vessels, coatings), as well as incubation conditions such as gas atmosphere and temperature,12,22,23 the metabolic competence of the cells, and the physicochemical properties of the test chemical.12,2426

Numerous studies recommend the total (“CMedium”) and unbound freely dissolved (“free CMedium”) concentrations in the culture medium to describe in vitro concentrations.5,12,23 Several methods are available to separate free CMedium and the fraction bound to proteins: equilibrium dialysis, ultracentrifugation, ultrafiltration, and solid phase microextraction (SPME), with the latter being the most prominent and established method.1,5,12,23,27 Only a few in vitro studies have estimated the intracellular concentrations of test chemicals.12,13,24 Obtaining cellular concentrations (CCell) presents analytical challenges, while the measurement of CMedium is well implemented.20 Measuring the intracellular distribution of chemicals in other compartments, such as cytosol, membranes, or receptors are even more difficult to assess. Estimating cellular concentrations by more simple concentration concepts is applicable when interactions between the chemical and intracellular targets are noncovalent, reversible, and where the in vitro system reaches steady state. By contrast, irreversible reactions,28 transporter-mediated uptake,29 accumulation in cells,30 and instability of the test chemicals in the in vitro system31,32 require more refined methods to estimate CCell. Due to the various technical difficulties in measuring chemical concentrations in multiple cell compartments, the work here focused on overall cell concentrations (CCell), as well as free and total CMedium.

In addition to the experimental methods, in silico models have been established and used to predict in vitro-derived concentrations.33,34 Commonly, these models assume steady state and an equilibrated partitioning between the compartment culture medium, cells, headspace, and plastics. Different elements such as spontaneous and enzymatic degradation, ionization of test chemicals, or the pH of different compartments were implemented in these models.21,33,3538 More comprehensive models for predicting a test chemical’s fate in the in vitro test systems are recommended but not yet sufficiently established, mainly due to the lack of experimental data to validate them.12,14 We have developed an as yet unpublished refined mass balance model using equations from versions developed by Armitage et al.,36 Fischer et al.,33 and Kramer et al.12 While the equations used within the current model are not new, the combination of all of them is. The model assumes instantaneous equilibrium and is based on mass balance equations describing the partitioning between five compartments of an in vitro test system: headspace, serum components (proteins and lipids), cells, water phase (free), and plastic (Figure 1). The model also removes the chemical that is added to the system above the solubility limit to a “precipitate” fraction.

Figure 1.

Figure 1

Partitioning within the test system used to describe the mass balance model. Schematic representation of an in vitro system and an example cell type, including the processes influencing the concentration of a substance and partitioning within the test system (Adapted with permission from Kramer et al. “Quantifying processes determining the free concentration of phenanthrene in basal cytotoxicity assays.” Chemical Research in Toxicology, 25(2), 436–445. Copyright 2012, American Chemical Society12).

This manuscript describes a comprehensive experimental method to characterize the cell test system and to quantify the total and free CMedium and CCell of 12 test chemicals (acetaminophen (APAP), bisphenol A (BPA), caffeine (CAF), colchicine (COL), fenarimol (FEN), flutamide (FLU), genistein (GEN), ketoconazole (KET), 17α-methyltestosterone (MT), tamoxifen (TAM), trenbolone (TRE), and warfarin (WAR)) over time in culture. The structures of the chemicals are shown in Figure S1. These chemicals were suitable for HPLC-MS analysis and represented a wide range of lipophilicities, i.e., log Pow of −0.07 to 6.84, which is considered to be a key parameter that drives the cellular uptake of chemicals. Balb/c 3T3 cells were used since they are routinely used in incubations of up to 48 h in several in vitro toxicity assays, e.g., the in vitro neutral red uptake phototoxicity test (OECD guideline no. 432) and the embryonic stem cell test. This study therefore provides a robust evaluation of the comparison of predictions using the refined in silico mass balance model with a set of measured data generated under the same conditions.

Materials and Methods

Chemicals and Materials

All chemicals were of the highest purity. The suppliers of the main chemicals and materials used in the experiments are listed in Supporting Information S1.

Chemicals and Cell Culture

Embryonic murine fibroblasts, clone A31 (Balb/c 3T3 cells) were obtained from the European Collection of Authenticated Cell Cultures. Cells were cultured in 150 cm2 flasks containing Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 1% l-glutamine, 1% penicillin/streptomycin, and 10% newborn calf serum, described as “culture medium”, and incubated at 37 °C, 90% humidity, and 5% CO2. Experiments with Balb/c 3T3 cells were performed with cells at passages 5–14.

Characterization of the Transporter Expression in Balb/c 3T3 Cells

Balb/c 3T3 cells were characterized according to the doubling time (cell number) and cell size (see Supporting Information (SI) Table S1). The expression levels of membrane transporters in Balb/c 3T3 cells were measured using mRNA sequencing. To generate cell samples, cells were washed twice with 10 mL of phosphate buffer saline (PBS) and harvested using 0.05% trypsin and 0.02% ethylenediaminetetraacetic acid. The cell number was determined before the suspension was centrifuged at 300g for 5 min at room temperature (RT) for mRNA extraction. For the purification and isolation of mRNA, cell samples were prepared as described in the user manual.39 Raw reads were checked for quality using FastQC. Transcript sequences were mapped to the genome of mouse (GRCm38) accessed from the National Center for Biotechnology Information to derive Transcript abundance values (Program: kallisto 0.44.0). Reads were normalized for sequencing depth and gene length by dividing the read counts with the length of each gene in kilobases to give reads per kilobase (RPK). All RPK values were normalized to cell number (“per million cells”) to give transcripts per million (TPM).

Cytotoxicity

The cell viability after incubation of a range of test chemical concentrations was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay40 (SI Table S2). The assay was performed as described by Kramer et al.12 with slight modifications: 3.2 × 104 Balb/c 3T3 cells/well were seeded in 24-well plates. After 24 h, the cells were exposed to five test concentrations per test chemical (in 0.2% dimethyl sulfoxide, DMSO) for 48 h. After exposure, the cells were incubated with 0.5 mL/well culture medium containing 1 mg/mL MTT for 40 min at 37 °C. Formazan was extracted with 0.5 mL/well 100% DMSO for 5 min. The absorbance was measured at 570 nm and normalized against the control.

Exposure of Balb/c 3T3 Cells with the Test Chemicals

CNom (SI Table S3) were based on the viability in Balb/c 3T3 cells (concentrations of test chemicals resulting in ≥80% cell viability according to the MTT assay or the maximum solubility in the solvent (DMSO)). This criterion was not valid for COL, for which a cell viability of 80% was only observed at 0.2 μmol/L (data not shown). Due to analytical limitations, a higher test concentration was selected for COL. Stock solutions of the test chemicals in DMSO were diluted in culture medium (500× the final concentration) and stirred on a magnetic stirrer for 24 h at 840 rpm, 43 °C to ensure homogeneity. One million Balb/c 3T3 cells were seeded in Petri dishes (60 cm2) with 15 mL of the culture medium. Test chemicals were added 24 h after seeding for 6, 24, and 48 h. After incubation, the culture medium was transferred to 15 mL tubes. The cell layer was washed twice with 10 mL of PBS and harvested using trypsin. Culture medium and cell lysate samples were stored at −20 °C until analysis. Cell lysate samples underwent three thaw and freeze cycles to destroy the cellular membrane and release the cytosolic fraction from the intercellular space.41,42

Determination of the Unbound Fraction of Test Chemicals in the Culture Medium

RED was performed as described by the manufacturer43 to determine the fraction unbound (fu) in culture medium. Briefly, the culture medium was spiked with the test chemicals at a final CNom of 5 μmol/L medium, 1% DMSO. A volume of 300 μL of spiked culture medium and 500 μL of PBS were transferred to the sample chamber of the inserts. The RED base plate with the samples was incubated for 6 h, at 37 °C, 5% CO2, on an orbital shaker at 250 rpm. After dialysis, 200 μL of each chamber and an equal volume of PBS were added. The samples were frozen at −20 °C until analysis. The assay was performed in triplicates. The fu was calculated using eq 1, where CPBS is the concentration in PBS (buffer chamber) and CMedium is the concentration of the test chemical in the culture medium (sample chamber)

graphic file with name tx2c00128_m001.jpg 1

Recovery was determined with CMedium(initial/end), VMedium(initial/end), CPBS(end), and VPBS(end). The terms “initial” and “end” indicate the concentrations before (0 h) and after the experiment (6 h). Acceptable thresholds for recovery tend from 70 to 130%

graphic file with name tx2c00128_m002.jpg 2

The recoveries of all test chemicals were all within the acceptance criterion (see SI Table S4).

The total concentration in culture medium, CMedium, was corrected by the fu determined via RED to obtain the free concentration of each test chemical in the culture medium, free CMedium (eq 3)

graphic file with name tx2c00128_m003.jpg 3

Determination of the Cell Number According to the Protein Content

The protein content of the treated Balb/c 3T3 cells was determined using the bicinchoninic acid (BCA) assay44 as a marker for the number of cells. The culture medium was removed and the cell layer was washed twice with 10 mL of PBS before the addition of 3 mL of Triton-X (0.5% in PBS). After 45 min incubation at 37 °C, the cell lysate was collected and centrifuged at 1000 rpm, RT for 5 min. The supernatant of the lysate was stored at −80 °C until analysis according to the user manual.45

Calculation of Intracellular Concentrations

A generic diameter (“d” in μm) of Balb/c 3T3 cells was determined at each incubation time point to calculate the cellular volume of treated and untreated Balb/c 3T3 cells (VCell in μL) using the Casy Cell Counter (Roche, Germany). Assuming a spherical shape, together with the diameter and cell number (nCell) using the BCA assay, the VCell was calculated using eq 4

graphic file with name tx2c00128_m004.jpg 4

The concentration of the test chemicals in the cell lysate, CLysate, was measured with the appropriate analytical method and corrected by the added volume of water and trypsin (VWater, 0.004 L). The intracellular concentration (CCell) was calculated using eq 5 and VCell

graphic file with name tx2c00128_m005.jpg 5

Determination of the Concentration of Test Chemicals in Cytosol

For potential differentiation between the intracellular and membrane-bound test chemical, an additional experiment was performed with APAP, CAF, FLU, and KET as model compounds adapted from Deusser et al.46 and Kaiser et al.47 Balb/c 3T3 cells were treated with the same concentrations of APAP, CAF, FLU, and KET for 48 h as described in the previous section. After 48 h of incubation, the culture medium was removed, and the cell layer washed twice with 10 mL of PBS. Then, 5 mL of digitonin solution (20 mg/L in PBS) was incubated with the cells for 5 min at RT and then on ice for 30 min to release the cytosol. The supernatants were collected and stored at −20 °C until analysis. The volume of the cytosol was based on generic calculations and assumptions. The volume of Balb/c 3T3 cells was measured (see Results section). It was assumed that cells consist of 70% water and the distribution between medium and cells occurs in the water phase. Although organelles in cells contribute to the total volume of the cell and also contain water, we applied a simplified assumption in which the volume of the cytosol in Balb/c 3T3 cells was set to be 30% lower than the total cell volume.

Determination of the Effect of Washing on Chemical Distribution

APAP, CAF, COL, and FLU were incubated for 6, 24, and 48 h, after which the cell monolayer was washed twice with 10 mL of PBS, as described above. In this experiment, the PBS wash samples were also collected after both steps. The test chemicals were measured in the culture medium, the two PBS wash samples, and in Balb/c 3T3 cells.

Sample Preparation and HPLC-MS/MS Analysis

The concentrations of the test chemicals in the culture medium, Balb/c 3T3 cell lysate, and RED samples were quantified with a high-performance liquid chromatography coupled with tandem mass spectrometry (HPLC-MS/MS). Details of the HPLC method, the generic tune files of the mass spectrometer, and the transitions monitored in parallel reaction monitoring are summarized in Supporting Information S2, Tables S5–S12. The samples were prepared by adding 10 μL of the respective deuterated or 13C-labeled internal standard (ISTD) and 4 mL of acetonitrile to 1 mL of the samples. After centrifugation at 4000g for 20 min, the supernatant was analyzed. The culture medium (50 μL) and buffer samples from the RED assay were mixed with 10 μL of ISTD and 200 μL of cold acetonitrile. Samples were centrifuged at 4000g, RT for 20 min and the supernatant analyzed. The concentrations were calculated using calibration standards containing the same matrix as the samples. Detailed parameters, e.g., concentration of ISTD, linearity range, limit of detection, and quantification can be found in Supporting Information S2, Tables S13–S18.

In Silico Mass Balance Model

The refined mass balance model used several equations developed by Armitage et al.,36 Fischer et al.,33 and Kramer et al.12 The free fraction of the initial amount of chemical in the aqueous phase of the medium was calculated as follows

graphic file with name tx2c00128_m006.jpg 6

or

graphic file with name tx2c00128_m007.jpg 7

where

Ffree is the fraction of chemical free in the aqueous media phase;

Kserum is the distribution coefficient between the serum matrix (lipid, protein) and water expressed as [L/L serum albumin];

Vserum/VWater is the volume ratio of the serum matrix (proteins + lipids) to media water;

Kprotein is the distribution coefficient between proteins and water [expressed as L/L];

Vserum proteins/VWater is the volume ratio of serum proteins to media water;

Klipid is the distribution coefficient between lipid and water [expressed as L/L];

Vserum lipids/VWater is the volume ratio of serum lipids to media water;

Kcell is the distribution coefficient between cells and water expressed as [L/L cells];

VCell/VWater is the volume ratio of cells to media water;

Kplastic is the distribution coefficient between plastic and water [expressed as m3/m2];

Aplastic/VWater is the ratio between exposed area of plastic [m2] and media water volume [m3];

Kair is the distribution coefficient between air and media water [L/L]; and

Vair/VWater is the volume ratio between the headspace in well and media water.

Details of the mass balance model can be found in Supporting Information S3.

Data Evaluation

For the quantification and qualification of the analytes, data were handled with Xcalibur and Chromeleon 7.2. Data were analyzed with Microsoft Excel and GraphPad Prism version 9.4.1.

Results

Characterization of the Applied Cells

The diameters of harvested untreated cells were 17.6 ± 0.5, 16.3 ± 0.2, and 16.0 ± 0.5 μm after 6, 24, and 48 h (≥9 biological replicates). The respective VCell were 2.9 ± 0.2, 2.3 ± 0.1, and 2.0 ± 0.1 μL/106 cells, assuming a spherical shape of the cells. Balb/c 3T3 cells contain 0.5 ± 0.2 mg protein/106 cells. The mRNA expression of membrane transporters in Balb/c 3T3 cells is presented in SI Figure S2. None of the expression levels exceeded 300 TPM that is assessed to represent a low expression. Membrane transporters of the solute carrier family (Slc) showed the highest expression, e.g., solute carrier transporters Slc7a5 (255.42 TPM), Slc3a2 (203.20 TPM), and Slc39a7 (137.60). The expression of other SLC transporters ranged from 30 to 90 TPM. Two transporters of the ATP binding cassette (ABC) family were prominent Abcf1 (106.18) and Abcf2 (98.69), and the other transporters of the ABC family were expressed at <40 TPM.

Measured Concentrations in the Culture Medium (Total and Free CMedium)

The initial measured concentrations of test chemicals in the culture medium at t = 0, i.e., before adding to the cells, were comparable to the CNom (with only up to 26% deviation) (Figure 2 and SI Table S19).

Figure 2.

Figure 2

Measured initial total CMedium of test chemicals before addition to Balb/c 3T3 cells (t = 0) compared to CNom. Each icon denotes one test chemical where circles represent hydrophilic (log Pow −0.07–1.30), rhombus and squares represent moderate lipophilic (log Pow 2.59–3.36), and triangles represent lipophilic (log Pow > 3.60) test chemicals. Data are represented as mean in μmol/L (standard deviation, SD, if n = 3 or mean difference between individual values, if n = 2*).

Figure 3 shows the values of fu, CCell, and total and free CMedium for all test chemicals and compares them with their CNom (concentrations are also listed in SI Table S20). The highest fu values were observed for the most hydrophilic test chemicals APAP, CAF, and COL (88.3–108.6%). MT, TRE, and WAR were moderately bound to proteins (fu was 35–52%), and BPA, FEN, GEN, KET, and TAM were more highly bound to medium proteins (fu values were ≤22%), especially TAM, which exhibited the lowest fu of 1% and the highest lipophilicity.

Figure 3.

Figure 3

Measured values of fu, CCell, and total and free CMedium for all chemicals. The CNom is denoted by the dotted line, total CMedium by black circles, free CMedium by white circles, and the CCell by red squares. Data are represented as mean in μmol/L (SD if n = 3 or mean difference between individual values if n = 2; Welch t test where * indicates p < 0.01 and **p < 0.005). The concentration of BPA could not be detected after 6 h of incubation (#).

The total CMedium for all chemicals except TAM were similar to the CNom and remained constant over the 48 h incubation. The free CMedium for the low (APAP) or no (CAF and COL) protein binding chemicals were similar to the CNom and remained constant over the 48 h incubation period (Figure 3A–C). The free CMedium values of all other chemicals remained stable but all were less than 30% of the CNom. This was especially noticeable for TAM (Figure 3L), the total and free CMedium of which decreased to 50% of the initial test concentration after 48 h of exposure.

Test chemicals could be measured in all samples, except for BPA in cell lysates after 6 h of incubation, in which CCell was below the LOQ. Of the 12 chemicals, the two most hydrophilic chemicals, APAP and CAF, were the only ones for which the CCell was the same as the CNom at t = 6 h and then decreased over the remaining time (down to 38 and 28% of the 6 h concentration, respectively). The CCell for all other chemicals tended to increase over time and were all higher than the CNom, with values 2- to 13-fold higher than CNom for six chemicals (COL, TRE, WAR, MT, FLU, and GEN (Figure 3C–H)) and 11- to 274-fold higher than CNom for four chemicals (BPA, FEN, KET, and TAM (Figure 3I–L)).

Table 1 shows the ratios of CCell/CMedium for each chemical, along with their molecular weights, Log Pow, and ionization state at pH 7.4 and measured values for fu in the medium. Chemicals that were neutral at pH 7.4 with a low log Pow and a high fu tended not to accumulate in the cells, e.g., CAF and APAP (CCell/CMedium ratios were close to 1). The CCell/CMedium tended to increase as the log Pow increased and the fu decreased. The highest cellular accumulation was observed for KET and TAM, which were lipophilic, highly protein-bound, as well as partly ionized (positively charged).

Table 1. Physicochemical Properties of Test Chemicals and Measured fu and CCell/CMedium Ratios after 6, 24, and 48 h Incubationa.

          CCell/CMedium ratio
test chemical MW [g/mol] speciation at pH 7.4 Log Pow fu 6 h 24 h 48 h
CAF 194.19 4.91 × 10–7% [neutral] – 0.07 105.6 1.1 0.5 0.3
APAP 151.16 0.86% [neutral] 0.46 88.3 1.8 0.9 0.7
COL 399.44 2.20 × 10–6% [neutral] 1.30 108.6 3.8 2.9 8.0
TRE 270.37 2.96 × 10–8% [neutral] 2.59 51.9 3.0 3.5 4.5
WAR 308.33 78.1% [acidic] 2.70 46.2 2.6 1.9 3.5
GEN 270.24 58.4% [neutral, acidic] 2.85 11.1 3.4 3.6 9.1
BPA 228.29 0.42% [neutral] 3.32 22.3 NA 13.3 25.2
FLU 276.21 1.69 × 10–4% [neutral] 3.35 20.3 5.3 3.6 11.6
MT 302.45 1.86 × 10–8% [neutral] 3.36 34.5 6.9 5.3 9.5
FEN 331.20 1.88 × 10–3% [neutral] 3.60 17.9 23.5 32.5 43.7
KET 531.43 18.2% [neutral, basic] 4.35 16.5 20.0 33.0 37.6
TAM 371.51 95.9% [neutral, basic] 6.84 1.1 93.8 1.9 597.5
a

Information about the molecular weight (MW) and log Pow were obtained from the U.S. Environmental Protection Agency CompTox Chemicals Dashboard,38 and speciation at pH 7.4 was calculated with Chemaxon. The CCell/CMedium ratio was calculated by dividing the CCell value by the total CMedium measured at each time point. The value for BPA after 6 h is not applicable (NA) due to the concentration in cell lysates being below the LOQ.

Comparison of CCell and CCytosol

Figure 4 shows the comparison of CCytosol with CCell at 48 h for four test chemicals (CAF, APAP, FLU, and KET) covering a range of lipophilicities (Log Pow of −0.07–4.35). The concentrations were the same in cell lysates and cytosol from incubations with CAF and KET. CCytosol values were statistically significantly higher than CCell after incubation with APAP (3.8-fold higher) and FLU (3.2-fold higher), although they were of the same order of magnitude.

Figure 4.

Figure 4

Comparison of CCell and CCytosol for APAP, CAF, FLU, and KET. The bars show the total CCell (gray bars) and the CCytosol (white bars) after 48 h of incubation. Data are represented as mean in μmol/L (SD of n = 3 experiments with triplicates; Welch t test, p < 0.05).

Effect of Washing on Chemical Distribution

The total CMedium and CCell after 6, 24, and 48 h measured in the repeat experiment (Table 2) were in accordance with those of the first experiment (SI Table S20). Approximately 50–90% of the chemicals were recovered in culture medium compared to only 0.04–6.4% in the cells, depending on the lipophilicity of the test chemical. Test chemicals were detected in the PBS after the first washing step and this amount represented 0.5–6.7% of the total CMedium at t = 0. The concentrations of test chemicals in PBS after the second washing step for all timepoints were almost all below the LOQ for APAP, CAF, COL, and FLU, accounting for <1.1, <0.5, <2.3, and <0.3% of the total CMedium at t = 0, respectively. Exceptions of these findings are the results in the second PBS wash for CAF and FLU after 6 h of incubation, representing 1.0 and 1.5% of the total CMedium, respectively.

Table 2. Effect of Washing on the Distribution of APAP, CAF, COL, and FLUa.

test chemical [CNom] incubation time [h] total CMedium[μmol/L medium] total CPBS1[μmol/L PBS] total CPBS2[μmol/L PBS] total CCell[μmol/L cell]
APAP [60 μmol/L medium] 0 67.8 ± 1.9 (100%)      
  6 50.0 ± 10.3 (74%) 2.6 ± 0.9 (3.8%) <0.8 (<1.1%) 75.1 ± 33.4 (0.04%)
  24 40.5 ± 2.7 (60%) 2.5 ± 0.8 (3.6%) <0.8 (<1.1%) 120.1 ± 40.0 (0.12%)
  48 39.1 ± 1.4 (58%) 1.7 ± 0.4 (2.6%) <0.8 (<1.1%) 38.1 ± 8.0 (0.09%)
CAF [160 μmol/L medium] 0 160.5 ± 5.6 (100%)      
  6 127.0 ± 15.1 (79%) 6.7 ± 0.3 (4.2%) 1.6 ± 0.4 (1.0%) 182.0 ± 91.0 (0.04%)
  24 133.1 ± 12.4 (83%) 4.5 ± 0.7 (2.8%) <0.9 (<0.5%) 161.1 ± 19.3 (0.03%)
  48 125.9 ± 15.6 (78%) 5.1 ± 0.5 (3.2%) <0.9 (<0.5%) 40.9 ± 7.1 (0.03%)
COL [10 μmol/L medium] 0 6.5 ± 0.4 (65%)      
  6 4.2 ± 0.5 (80%) 0.3 ± 0.1 (4.5%) <0.1 (< 2.3%) 9.1 ± 3.6 (0.05%)
  24 5.2 ± 0.7 (80%) 0.1 ± 0.1 (0.5%) <0.1 (< 2.3%) 10.1 ± 1.4 (0.10%)
  48 4.9 ± 0.2 (74%) 0.3 ± 0.1 (4.3%) <0.1 (< 2.3%) 4.3 ± 0.4 (0.11%)
FLU [10 μmol/L medium] 0 9.0 ± 0.3 (100%)      
  6 7.7 ± 1.4 (86%) 0.5 ± 0.1 (5.1%) 1.6 (0.4) (1.5%) 250.4 ± 16.7 (0.93%)
  24 6.4 ± 1.2 (72%) 0.3 ± 0.1 (3.8%) <0.1 (0.3%) 333.1 ± 12.0 (2.5%)
  48 5.3 ± 0.5 (59%) 0.3 ± 0.1 (3.8%) <0.1 (0.3%) 344.5 ± 2.0 (6.4%)
a

Total concentrations in the culture medium (Total CMedium) in PBS collected after the first and second washing steps (CPBS1 and CPBS2) and in cells (CCell) after 6, 24, and 48 h incubation with APAP, CAF, COL, and FLU. Data are represented as mean in μmol/L ± SD, n = 3. Values in brackets are the mass balance percentages given as mean ± SD in % (total CCell value is without conversion to cell volume).

Predictions by the Mass Balance Model

The comparisons of the predicted and measured values for total and free CMedium and total CCell after 6, 24, and 48 h are shown in Figure 5 and SI Table S20. Values of total CMedium at 6, 24, and 48 h were well predicted by the model (Figure 5A–C), with values for 11 of 12 chemicals predicted to be within 30% of the measured values, and a median ratio of predicted/measured values of 1.0. The exception to this was for TAM, for which the model predicted much lower concentrations (0.23–0.46 μmol/L) than were measured (5.7–9.8 μmol/L) at different timepoints. While the total CMedium of TAM was not well predicted, the predicted free CMedium of this chemical was within 2-fold of the measured values (Figure 5D–F). Indeed, the free CMedium of all 12 chemicals were relatively well predicted, with a median ratio of measured/predicted values of 1.1 at all three timepoints. The maximum overprediction was for GEN, which was overpredicted by 2.9-fold at 48 h, and the maximum underprediction was for WAR, which was underpredicted by 3.3-fold at all three timepoints. The highest difference between predicted and measured values was the total CCell, which was mainly overpredicted by up to 26-, 31.4-, and 15.2-fold at 6, 24, and 48 h (Figure 5G–I). The only two chemicals that were correctly predicted with 2-fold of the measured values at all three timepoints were COL and FEN. Most of the total CCell values were overpredicted, especially those for FLU, MT, TAM, and TRE (by up to 31.4-, 25.5-, 26.0-, and 17-fold, respectively). Despite these differences, the median fold overprediction for all 12 chemicals was still only 3.0-, 4.1-, and 4.1-fold of the measured values at 6, 24, and 48 h, respectively.

Figure 5.

Figure 5

Comparison of predicted and measured values of CCell and total and free CMedium for all test chemicals after 6, 24, and 48 h of incubation. The test chemicals are denoted by white circles, the black circles indicate the most lipophilic chemical of the set of substances, TAM. The line of identity is denoted by the dotted line. Detailed information on the data is presented in Supporting Information S1, Table S20. Data are represented as mean in μmol/L (SD if n = 3 or mean difference between individual values if n = 2).

Discussion

The use of in vitro dosimetry in the in vitro testing should be carefully considered and remains a challenge for the development of robust approaches to QIVIVE.12,14,33 Typically, CNom is used to extrapolate the blood and tissue concentrations, even though it does not reflect the actual in vitro effect concentration.9,15,48 The reason for this is that methods to experimentally measure concentrations in cells, cell membranes, or other cell compartments are limited or very technically demanding, especially for high-throughput assays.49 Total or free CMedium or the concentration in the cytosol are closer to the biologically effective concentration and therefore better values for QIVIVE purposes.10,35,50 In the current study, we measured the concentrations of test chemicals in the cells and cytosol, as well as the free and total concentrations in the medium.

Characterization of Balb/c 3T3 Cells

As with any assay, it is important to characterize the cells under the conditions of the assay since different cell sources and media can impact the phenotype of the cells.51 We selected Balb/c 3T3 cells for this work since they are routinely used in toxicity assays. The determination of the actual cell volume of Balb/c 3T3 cells was not experimentally performed. For the sake of simplicity, the volume of Balb/c 3T3 cells was derived based on the assumption that cultured cells take a spherical shape and a diameter of 16.0–17.6 μm as experimentally determined. The VCell ranged between 2.0 and 2.9 μL, which is in general agreement with data from Gülden et al.18 with a VCell of 1.8 ± 0.7 μL/106 cells. Balb/c 3T3 cells contained 0.5 ± 0.2 mg protein/106 cells, which is also in line with values of 0.5 and 0.4 mg protein/106 cells reported by Gülden et al.18 and Kramer et al.12 Genes coding for transporters were detected in Balb/c 3T3 cells; however, the highest expression of transporters was for Slc7a5, which was 255 TPM, which is not high according to Wagner et al.52,53 The uptake of test chemicals in Balb/c 3T3 cells can be concluded to be largely a diffusion-limited process, with active, transport-protein-mediated uptake of minor relevance. In addition, xenobiotic-metabolizing enzymes, e.g., CYP enzymes, are reported to be expressed in negligible levels in Balb/c 3T3 cells.12,5456 This was also reflected in the current dataset since there was negligible depletion of the parent chemicals over time. Therefore, these cells represent a suitable cell model for understanding general mechanisms concerning diffusion biokinetics and for the validation of in silico models based on this mechanism. Their obvious limitation is that the results cannot be extrapolated to other cell types with a higher transporter function or to chemicals that involve transporter-mediated uptake.

Experimental Design and Sample Preparation Considerations

Cell Disruption and Cell-Associated Versus Cytosolic Concentrations

Several methods have been described to prepare samples for the measurement of cell-associated chemical concentrations. These include using detergents,46,47 freezing and thawing cycles, ultrasonication,13,41 and liquid homogenization.42 One of the consequences of each method is the resulting sample may or may not contain plasma membranes together with chemicals that may have bound to the outside of the cells. In this case, the true intracellular concentration is not measured—just the “cell-associated” concentration. Therefore, we compared two methods to disrupt cells in the current study, namely, freeze–thaw cycles to derive CCell values (including plasma membranes and cytosol) and treatment with digitonin to derive CCytosol values. Digitonin permeabilizes the cell plasma membranes to release the cytosol into the medium without releasing plasma membranes and associated chemicals. Both methods yielded comparable results, indicating that none of chemicals tested associated with the plasma membrane and that CCytosol values were a good representation of intracellular concentrations. Although the method involving lysis with digitonin is practically less demanding compared to freeze–thaw cycles, there was more variability in the measurements of CCytosol of experiments (% CV values were 14–29% for CCell values and 13–73% for CCytosol values), indicating less robust results.

Although it is possible to measure total and free CCell, this was not conducted in this study due to the technically challenging issues with handling low volumes yielded from cell culture preparations.20,57 While others could demonstrate the measurement of free CCell in HEK293 cells and primary human hepatocytes,58,59 this may be an exceptional case. For many purposes, CCell may be a sufficient proxy and refinements by, e.g., using free CCytosol may only yield improvements within the experimental error of measuring the concentrations. The current study indicates that two methods provide comparable concentrations: (i) trypsinization and a following disruption of cells by thawing and freezing cycles12 and (ii) lysis with a digitonin solution.46 This needs to be verified by further studies, including controls with buffer, and addressing the possible wash out effect.

Impact of PBS Wash on Cell Distribution

A technical concern relating to the washing procedure is that it may contribute to the removal of chemicals from the cells, i.e., diffusing back into the wash medium, thus, resulting in artificially lower CCell values. To address this, the concentrations of four chemicals removed in the PBS washes were measured in a follow up experiment. There was no link between the percentage of chemical removed in the first wash with their lipophilicity.

The amounts of compounds in the second wash were (with only two exceptions in the wash after 6 h for CAF and FLU) below the LOQ and significantly lower than the first wash. However, the calculated amounts of the compound at the LOQ still exceed the recovered amounts of the compound in the cells for APAP, COL, and CAF and account for about 5–12% of the recovered amounts of FLU in the cells. Although these data were originally generated to prove that chemicals in the cells do not diffuse back into the PBS during washing, this statement cannot be supported based on the current data.

Concentrations in Culture Medium

CNom of the test chemicals were generally in accordance with the measured total CMedium at t0, indicating that the preparation of the solutions was in accordance with the target concentrations and that nonspecific binding to the tubes did not occur. The total CMedium remained constant over 48 h of incubation for 11 of the 12 test chemicals. The exception to this was TAM, the CMedium of which decreased over time. This was attributed to the cellular uptake of TAM and accumulation into lysosomes.61

One factor affecting the effect concentration resulting in a biological effect is protein binding, as demonstrated for 9 of the 12 chemicals tested in this study. When extrapolating to no-effect levels in the in vitro assays, chemicals exhibiting low binding to proteins would not need a correction of the total CMedium by fu since the total CMedium and free CMedium are similar.10,25 The more lipophilic test chemicals exhibiting higher binding to proteins, resulting in the free CMedium being lower than total CMedium, may require a correction factor before correlating with an in vitro effect. This reduction of free CMedium in the in vitro test systems has also been described by Henneberger et al.5 and Huchthausen et al.25 While human plasma contains 60–80 g protein/L, of which 50–60% is albumin and is similar to that in newborn calf serum (71.5 g protein/L proteins; with 39.5 g/L albumin),60,62 in this study, the medium contained only 10% serum (which is typical for many cell cultures); hence, protein concentrations were lower in cell culture media compared to the human serum in vivo. Therefore, when performing the correction for protein binding and then extrapolating to in vivo concentrations, the physiological concentrations of proteins in human plasma and the in vitro incubation should be considered.

Factors Impacting Intracellular Concentrations of Chemicals in Balb/c 3T3 Cells

The kinetics of the distribution of chemicals will depend on several properties. Lipinski et al. defined the “Rule of 5” postulating that molecules with the following criteria can pass the cell membrane by diffusion: a molecular weight of <500 g/mol, log Pow < 5, five H-bond donors, and ten H-bond acceptors, e.g., oxygen and nitrogen.62 All of the test chemicals were of a molecular weight near to or lower than 500 g/mol and most had a log Pow < 5. These data showed that hydrophilic chemicals (CAF and APAP) did enter the cells but did not accumulate, while lipophilic chemicals accumulated, with the extent correlated with the log Pow. This correlation between the log Pow and cellular uptake has also been reported by others.12,57,6365 In addition, lipophilic chemicals preferentially distributed to the cells, with CCell/CMedium ratios between 9 and 598 for chemicals with log Pow values at or greater than 2.85.

The mass balance model assumes instantaneous equilibrium of the test chemicals and, indeed, many drugs pass membranes in seconds to minutes.21,66 However, due to the technical difficulties of measuring the distribution in multiple wells, such short incubations were not possible in the current study. The timepoints chosen were relevant to the assays in which the cells are used. Most accumulation of chemicals occurred in the first 6 h (although this may have occurred in the first few minutes of incubation) but CCell/CMedium ratios continued to increase until 48 h, indicating additional slower accumulation after this time.

The passage through the lipid layer and the negatively charged cell membrane also facilitates the movement of cationic molecules.12,21,37,57 Most of the test chemicals were uncharged molecules at a pH 7.4, except GEN, KET, TAM, and WAR which were partly ionized. Due to the negatively charged nature of GEN and WAR, their diffusion through the negatively charged membrane barrier would be impeded and might result in lower CCell.10,67

The free CMedium values could be expected to be linked to a lower cellular uptake of chemicals, as binding the proteins in the medium may prevent this. McManus et al.41 reported that the use of serum-free medium resulted in higher cellular concentrations in prostate cancer cells than the serum-containing medium. However, our results do not support this hypothesis since chemicals with high CCell/CMedium ratios were moderately or highly bound (fu < 35%). The impact of protein may therefore also depend on the affinity of the interaction, with covalently bound chemicals exhibiting lower cellular uptake.

In addition to the properties described above, a chemical’s affinity to cellular targets can enhance its uptake into cells, e.g., lysosomal trapping.57,68 This was observed in this study for TAM and confirmed by other groups.17,57 TAM is a positively charged molecule at pH 7.4; it is lipophilic and of rather small molecular size. These characteristics tend to facilitate adsorption of TAM to the cell membrane of Balb/c 3T3 cells.29,37,63 COL also appeared to accumulate more than expected based on its log Pow, which may be due to it binding to tubulin, where it blocks the polymerization of microtubules and suppresses the cell division and proliferation.21,69

Prediction Capacity of the Mass Balance Model

The mass balance model predicted the total CMedium within 30% of the measured values for all but one of the test chemicals. The exception was TAM, for which total CMedium was underpredicted by ∼25-fold. The reason for this was attributed to the uptake of this positively charged molecule into the cells and accumulation into the lysosomes. Despite this, the model was able to predict the free CMedium of TAM at each time point. The free CMedium of the remaining chemicals were also relatively well predicted by the model. Notably, values for GEN, KET, and WAR were over- or underpredicted by factors of up to 2.9-, 2.5-, and 3.3-fold, respectively. These test chemicals are ionized and lipophilic molecules. In cell culture media with pH 7.4, KET is positively charged and GEN and WAR are negatively charged. Positively charged molecules are known to have a strong affinity to α-glycoproteins and negatively charged molecules to albumin.70,71 This may contribute to the difference between the predicted and measured values, since the model parameterization was calibrated with neutral molecules. The prediction of the partitioning of chemicals into cells was based on a model predicting binding to liposomes and serum albumin and the ionization of the test chemicals was not considered. This may account for the poorer prediction of CCell values by the current model. Moreover, binding to serum albumin may not be predictive of binding to other proteins, such as microfilaments, microtubules, and intermediate filaments.14 Future efforts will aim to refine the model for charged molecules, as well as chemicals that bind to microfilaments, e.g., COL. It is hoped that datasets such as the one presented here will enable such refinements to be conducted.

Although the mass balance model is relatively easy to use and predicts the biokinetics of neutral chemicals relatively well, it does, however, have significant limitations that experimental models also face, i.e., it does not reflect xenobiotic metabolism or active transport. Cell types proficient in xenobiotic metabolism and transport-mediated uptake and efflux, e.g., hepatocytes, will require appropriate, dynamic models. Likewise, concentrations of volatile, ionizing, and spontaneously degrading test chemicals will not be accurately predicted and will require additional refinements to account for these common attributes of test chemicals.

Conclusions

This study compared measured biokinetics data in Balb/c 3T3 cells with predicted values using a refined in silico mass balance model. While the number of chemicals in the study is limited, this is the first time, to our knowledge, that a study combining in vitro and in silico biokinetics techniques has been published. These data provide information on cell preparation techniques with a well-established and toxicologically relevant cell line, using accurate analytical methods. It is hoped that these experimental data can be used by others for the validation of similar mass balance models. The mass balance model combined relevant, albeit known, QSARs to result in a version that could accurately predict total CMedium and free CMedium for nonvolatile, mostly neutral chemicals with a log Pow between −1 and 6.6. Predictions were of chemicals with predominantly diffusion-based uptake into cells with low xenobiotic-metabolizing and low active transport capacity. Comparisons of CNom with free CMedium and CCytosol already demonstrated the large differences between them and that nominal concentrations may not always be the most relevant when comparing to a bioactivity in the same cells. These measured and predicted values allow the extrapolation of (a) free CMedium to an unbound concentration in human blood; (b) total CMedium to the total concentration in human blood; and (c) the total CCell as surrogate to tissue concentrations in humans in vivo. Future studies will aim to expand the set of test chemicals to increase the confidence in the experimental method and improve the accuracy of the in silico model. Likewise, the methods should be expanded to be applicable to ionized chemicals and to cells with metabolizing capacities.

Acknowledgments

The authors thank Dr. Anita Samuga and her team, BASF Corporation, Research Triangle Park, NC, for the analysis of the mRNA expression levels in the Balb/c 3T3 cell lysate.

Glossary

Abbreviations

APAP

acetaminophen

APlastic

area of culture vessel

BCA

bicinchoninic acid

BPA

bisphenol A

BPA-d16

bisphenol A d16

CCell

total concentration in Balb/c 3T3 cells

CCytosol

total concentration in the cytosol of Balb/c 3T3 cells

CLysate

total concentration of a test chemical in Balb/c 3T3 cell lysate

CMedium

concentration of a test chemical in the culture medium

CNom

nominal concentration of a test chemical

CPBS

concentration in PBS

CAF

caffeine

COL

colchicine

d

diameter

DMEM

Dulbecco’s modified Eagle’s medium

DMSO

dimethyl sulfoxide

DZP-d5

diazepam-d5

EDTA

ethylenediaminetetraacetic acid

FAir

fraction in air

FCells

total fraction of a chemical in cells

Ffree

fraction of chemical free in the aqueous media phase

FMedia

total fraction of a chemical in medium

FPlastic

fraction of a chemical bound to plastic

Fprecip

precipitated fraction of a chemical in the test system

FEN

fenarimol

FLU

flutamide

fu

fraction unbound of a test chemical

GEN

genistein

h hour(s) ISTD

internal standard

KAir

distribution coefficient between air and water

KCell

distribution coefficient between cells and water

KLipid

distribution coefficient between lipid and water

KProtein

distribution coefficient between proteins and water

KSerum

distribution coefficient between serum matrix (lipid, protein) and water

KET

ketoconazole

Log Kaw

water–air partition coefficient

Log Pow

octanol–water partition coefficient

MT

methyltestosterone

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

nCell

cell number

PBS

phosphate buffered saline

RED

rapid equilibrium dialysis

RT

room temperature

Slc

solute carrier

Stotal,max

maximum solubility of the test chemical in the test system

TAM

tamoxifen

TAM-13C2

tamoxifen-13C2

TES-d3

testosterone-d3

TPM

transcripts per million

TRE

trenbolone

VCell

volume of Balb/c 3T3 cells

VMedium

volume of medium

VSerum

volume of serum

VSerum lipids

volume of lipids in serum

VSerum proteins

volume of proteins in serum

VWater

volume of water

WAR

warfarin

Supporting Information Available

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.chemrestox.2c00128.

  • Detailed information on test chemicals, analytical methods, data summaries, and predicted values using the mass balance model (PDF)

Author Contributions

This work was funded by the BASF Key Technology Capability Building “Alternative Toxicological Methods”.

The authors declare no competing financial interest.

Supplementary Material

tx2c00128_si_001.pdf (332.3KB, pdf)

References

  1. Heringa M. B.; Schreurs R. H.; Busser F.; Van Der Saag P. T.; Van Der Burg B.; Hermens J. L. Toward more useful in vitro toxicity data with measured free concentrations. Environ. Sci. Technol. 2004, 38, 6263–6270. 10.1021/es049285w. [DOI] [PubMed] [Google Scholar]
  2. Proença S.; Paini A.; Joossens E.; Benito J. V. S.; Berggren E.; Worth A.; Whelan M.; Prieto P. Insights into in vitro biokinetics using Virtual Cell Based Assay simulations. Altern. Anim. Exp. 2019, 36, 447–461. 10.14573/altex.1812101. [DOI] [PubMed] [Google Scholar]
  3. Yoon M.; Blaauboer B. J.; Clewell H. J. Quantitative in vitro to in vivo extrapolation (QIVIVE): An essential element for in vitro-based risk assessment. Toxicology 2015, 332, 1–3. 10.1016/j.tox.2015.02.002. [DOI] [PubMed] [Google Scholar]
  4. Basketter D.; Clewell H.; Kimber I.; Rossi A.; Blaauboer B.; Burrier R.; Hasiwa N. A roadmap for the development of alternative (Non-animal) methods for systemic toxicity testing-t4 report. Altern. Anim. Exp. 2012, 29, 3–91. 10.14573/altex.2012.1.003. [DOI] [PubMed] [Google Scholar]
  5. Henneberger L.; Mühlenbrink M.; König M.; Schlichting R.; Fischer F. C.; Escher B. I. Quantification of Freely Dissolved Effect Concentrations in in vitro cell-based bioassays. Arch. Toxicol. 2019, 93, 2295–2305. 10.1007/s00204-019-02498-3. [DOI] [PubMed] [Google Scholar]
  6. Wilk-Zasadna I.; Bernasconi C.; Pelkonen O.; Coecke S. Biotransformation in vitro: An essential consideration in the quantitative in vitro-to-in vivo extrapolation (QIVIVE) of toxicity data. Toxicology 2015, 332, 8–19. 10.1016/j.tox.2014.10.006. [DOI] [PubMed] [Google Scholar]
  7. Baltazar M. T.; Cable S.; Carmichael P. L.; Cubberley R.; Cull T.; Delagrange M.; Dent M. P.; Hatherell S.; Houghton J.; Kukic P.; et al. A next-generation risk assessment case study for coumarin in cosmetic products. Toxicol. Sci. 2020, 176, 236–252. 10.1093/toxsci/kfaa048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Blaauboer B. J. Biokinetic modeling and in vitro–in vivo extrapolations. J. Toxicol. Environ. Health, Part B 2010, 13, 242–252. 10.1080/10937404.2010.483940. [DOI] [PubMed] [Google Scholar]
  9. Fabian E.; Gomes C.; Birk B.; Williford T.; Hernandez T. R.; Haase C.; Zbranek R.; van Ravenzwaay B.; Landsiedel R. In vitro-to-in vivo extrapolation (IVIVE) by PBTK modeling for animal-free risk assessment approaches of potential endocrine-disrupting compounds. Arch. Toxicol. 2019, 93, 401–416. 10.1007/s00204-018-2372-z. [DOI] [PubMed] [Google Scholar]
  10. Groothuis F. A.; Heringa M. B.; Nicol B.; Hermens J. L.; Blaauboer B. J.; Kramer N. I. Dose metric considerations in in vitro assays to improve quantitative in vitro–in vivo dose extrapolations. Toxicology 2015, 332, 30–40. 10.1016/j.tox.2013.08.012. [DOI] [PubMed] [Google Scholar]
  11. Wambaugh J. F.; Hughes M. F.; Ring C. L.; MacMillan D. K.; Ford J.; Fennell T. R.; Black S. R.; Snyder R. W.; Sipes N. S.; Wetmore B. A.; et al. Evaluating in vitro-in vivo extrapolation of toxicokinetics. Toxicol. Sci. 2018, 163, 152–169. 10.1093/toxsci/kfy020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Kramer N. I.; Krismartina M.; Rico-Rico A. N.; Blaauboer B. J.; Hermens J. L. Quantifying processes determining the free concentration of phenanthrene in basal cytotoxicity assays. Chem. Res. Toxicol. 2012, 25, 436–445. 10.1021/tx200479k. [DOI] [PubMed] [Google Scholar]
  13. Doskey C. M.; van ‘t Erve T. J.; Wagner B. A.; Buettner G. R. Moles of a substance per cell is a highly informative dosing metric in cell culture. PLoS One 2015, 10, e0132572 10.1371/journal.pone.0132572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Henneberger L.; Huchthausen J.; Wojtysiak N.; Escher B. I. Quantitative in vitro-to-in vivo extrapolation: nominal versus freely dissolved concentration. Chem. Res. Toxicol. 2021, 34, 1175–1182. 10.1021/acs.chemrestox.1c00037. [DOI] [PubMed] [Google Scholar]
  15. Ning J.; Chen L.; Strikwold M.; Louisse J.; Wesseling S.; Rietjens I. M. C. M. Use of an in vitro–in silico testing strategy to predict inter-species and inter-ethnic human differences in liver toxicity of the pyrrolizidine alkaloids lasiocarpine and riddelliine. Arch. Toxicol. 2019, 93, 801–818. 10.1007/s00204-019-02397-7. [DOI] [PubMed] [Google Scholar]
  16. Fischer F. C.; Cirpka O. A.; Goss K. U.; Henneberger L.; Escher B. I. Application of experimental polystyrene partition constants and diffusion coefficients to predict the sorption of neutral organic chemicals to multiwell plates in in vivo and in vitro bioassays. Environ. Sci. Technol. 2018, 52, 13511–13522. 10.1021/acs.est.8b04246. [DOI] [PubMed] [Google Scholar]
  17. Stadnicka-Michalak J.; Tanneberger K.; Schirmer K.; Ashauer R. Measured and modeled toxicokinetics in cultured fish cells and application to in vitro-in vivo toxicity extrapolation. PLoS One 2014, 9, e92303 10.1371/journal.pone.0092303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Gülden M.; Mörchel S.; Tahan S.; Seibert H. Impact of protein binding on the availability and cytotoxic potency of organochlorine pesticides and chlorophenols in Vitro. Toxicology 2002, 175, 201–213. 10.1016/S0300-483X(02)00085-9. [DOI] [PubMed] [Google Scholar]
  19. Heringa M. B.; Hermens J. L. M. Measurement of free concentrations using negligible depletion-solid phase microextraction (nd-SPME). TrAC, Trends Anal. Chem. 2003, 22, 575–587. 10.1016/S0165-9936(03)01006-9. [DOI] [Google Scholar]
  20. Tolonen A.; Pelkonen O. Analytical challenges for conducting rapid metabolism characterization for QIVIVE. Toxicology 2015, 332, 20–29. 10.1016/j.tox.2013.08.010. [DOI] [PubMed] [Google Scholar]
  21. Kisitu J.; Hollert H.; Fisher C.; Leist M. Chemical concentrations in cell culture compartments (C5)—free concentrations. Altern. Anim. Exp. 2020, 37, 693–708. 10.14573/altex.2008251. [DOI] [PubMed] [Google Scholar]
  22. Henneberger L.; Mühlenbrink M.; Heinrich D. J.; Teixeira A.; Nicol B.; Escher B. I. Experimental validation of mass balance models for in vitro cell-based bioassays. Environ. Sci. Technol. 2020, 54, 1120–1127. 10.1021/acs.est.9b06144. [DOI] [PubMed] [Google Scholar]
  23. Fischer F. C.; Abele C.; Droge S. T.; Henneberger L.; König M.; Schlichting R.; Scholz S.; Escher B. I. Cellular uptake kinetics of neutral and charged chemicals in in vitro assays measured by fluorescence microscopy. Chem. Res. Toxicol. 2018, 31, 646–657. 10.1021/acs.chemrestox.8b00019. [DOI] [PubMed] [Google Scholar]
  24. Broeders J. J.; Blaauboer B. J.; Hermens J. L. In vitro biokinetics of Chlorpromazine and the influence of different dose metrics on effect concentrations for cytotoxicity in Balb/c 3T3, Caco-2 and HepaRG cell cultures. Toxicol. In Vitro 2013, 27, 1057–1064. 10.1016/j.tiv.2013.01.010. [DOI] [PubMed] [Google Scholar]
  25. Huchthausen J.; Mühlenbrink M.; König M.; Escher B. I.; Henneberger L. Experimental exposure assessment of ionizable organic chemicals in in vitro cell-based bioassays. Chem. Res. Toxicol. 2020, 33, 1845–1854. 10.1021/acs.chemrestox.0c00067. [DOI] [PubMed] [Google Scholar]
  26. Birch H.; Kramer N. I.; Mayer P. Time-resolved freely dissolved concentrations of semivolatile and hydrophobic test chemicals in in vitro assays—measuring high losses and crossover by headspace solid-phase microextraction. Chem. Res. Toxicol. 2019, 32, 1780–1790. 10.1021/acs.chemrestox.9b00133. [DOI] [PubMed] [Google Scholar]
  27. Fischer F. C.; Abele C.; Henneberger L.; Klüver N.; König M.; Mühlenbrink M.; Schlichting R.; Escher B. I. Cellular Metabolism in High-Throughput In Vitro Reporter Gene Assays and Implications for the Quantitative In Vitro–In Vivo Extrapolation. Chem. Res. Toxicol. 2020, 33, 1770–1779. 10.1021/acs.chemrestox.0c00037. [DOI] [PubMed] [Google Scholar]
  28. Bläser J.; Triebel S.; Reinke H.; Tschesche H. Formation of a covalent Hg-Cys-bond during mercurial activation of PMNL procollagenase gives evidence of a cysteine-switch mechanism. FEBS Lett. 1992, 313, 59–61. 10.1016/0014-5793(92)81184-n. [DOI] [PubMed] [Google Scholar]
  29. Schildknecht S.; Pape R.; Meiser J.; et al. Preferential extracellular generation of the active parkinsonian toxin MPP+ by transporter-independent export of the intermediate MPDP+. Antioxid. Redox Signaling 2015, 23, 1001–1016. 10.1089/ars.2015.6297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Zimmer B.; Kuegler P. B.; Baudis B.; Genewsky A.; Tanavde V.; Koh W.; Tan B.; Waldmann T.; Kadereit S.; Leist M. Coordinated waves of gene expression during neuronal differentiation of embryonic stem cells as basis for novel approaches to developmental neurotoxicity testing. Cell Death Differ. 2011, 18, 383–395. 10.1038/cdd.2010.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Gülden M.; Jess A.; Kammann J.; Maser E.; Seibert H. Cytotoxic potency of H2O2 in cell cultures: impact of cell concentration and exposure time. Free Radicals Biol. Med. 2010, 49, 1298–1305. 10.1016/j.freeradbiomed.2010.07.015. [DOI] [PubMed] [Google Scholar]
  32. Kisitu J.; Hougaard Bennekou S.; Leist M. Chemical concentrations in cell culture compartments (C5): concentration definitions. Altern. Anim. Exp. 2019, 36, 154–160. 10.14573/altex.1901031. [DOI] [PubMed] [Google Scholar]
  33. Fischer F. C.; Henneberger L.; König M.; Bittermann K.; Linden L.; Goss K.-U.; Escher B. I. Modeling exposure in the Tox21 in vitro bioassays. Chem. Res. Toxicol. 2017, 30, 1197–1208. 10.1021/acs.chemrestox.7b00023. [DOI] [PubMed] [Google Scholar]
  34. Graepel R.; Lamon L.; Asturiol D.; Berggren E.; Joossens E.; Paini A.; Prieto P.; Whelan M.; Worth A. The virtual cell based assay: Current status and future perspectives. Toxicol. In Vitro 2017, 45, 258–267. 10.1016/j.tiv.2017.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Proença S.; Escher B. I.; Fischer F. C.; Fisher C.; Grégoire S.; Hewitt N. J.; Nicol B.; Paini A.; Kramer N. I. Effective exposure of chemicals in in vitro cell systems: A review of chemical distribution models. Toxicol. In Vitro 2021, 73, 105133. 10.1016/j.tiv.2021.105133. [DOI] [PubMed] [Google Scholar]
  36. Armitage J. M.; Wania F.; Arnot J. A. Application of mass balance models and the chemical activity concept to facilitate the use of in vitro toxicity data for risk assessment. Environ. Sci. Technol. 2014, 48, 9770–9779. 10.1021/es501955g. [DOI] [PubMed] [Google Scholar]
  37. Fisher C.; Siméon S.; Jamei M.; Gardner I.; Bois Y. VIVD: Virtual in vitro distribution model for the mechanistic prediction of intracellular concentrations of chemicals in in vitro toxicity assays. Toxicol. In Vitro 2019, 58, 42–50. 10.1016/j.tiv.2018.12.017. [DOI] [PubMed] [Google Scholar]
  38. Zaldívar J.; Mennecozzi M.; Marcelino Rodrigues R.; Bouhifd M.. A Biology-Based Dynamic Approach for the Modelling of Toxicity in Cell-Based Assays. Part I: Fate Modelling; JRC Institute for Health and Consumer Protection: Luxembourg, 2010. [Google Scholar]
  39. QIAGEN. RNeasy Mini Handbook, 2019. https://www.qiagen.com/us/products/discovery-and-translational-research/dna-rna-purification/rna-purification/total-rna/rneasy-kits/?catno=74104&clear=true (accessed March 29, 2021).
  40. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. 10.1016/0022-1759(83)90303-4. [DOI] [PubMed] [Google Scholar]
  41. McManus J. M.; Bohn K.; Alyamani M.; Chung Y. M.; Klein E. A.; Sharifi N. Rapid and structure-specific cellular uptake of selected steroids. PLoS One 2019, 14, e0224081 10.1371/journal.pone.0224081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Walker J. M.The Protein Protocols Handbook; Springer Science & Business Media, 1996; Vol. 1996. [Google Scholar]
  43. Thermo Scientific Manual. RED Device Inserts, 2012. https://www.thermofisher.com/order/catalog/product/89809 (accessed March 29, 2021).
  44. Smith P. K.; Krohn R. I.; Hermanson G. T.; Mallia A. K.; Gartner F. H.; Provenzano M. D.; Fujimoto E. K.; Goeke N. M.; Olson B. J.; Klenk D. C. Measurement of protein using bicinchoninic acid. Anal. Biochem. 1985, 150, 76–85. 10.1016/0003-2697(85)90442-7. [DOI] [PubMed] [Google Scholar]
  45. Thermo Scientific Manual. Pierce BCA Protein Assay Kit, 2020. https://www.thermofisher.com/order/catalog/product/23225 (accessed March 29, 2021).
  46. Deusser H.; Groh I.; Bakuradze T.; Simson N.; Kaiser E.; Barth H.; Richling E. Are compounds membrane-associated or Present in the cytosol? A study using polyphenols in a colon carcinoma cell line model. Curr. Pharmacol. Rep. 2020, 6, 1–6. 10.1007/s40495-020-00242-5. [DOI] [Google Scholar]
  47. Kaiser E.; Pust S.; Kroll C.; Barth H. Cyclophilin A facilitates translocation of the Clostridium botulinum C2 toxin across membranes of acidified endosomes into the cytosol of mammalian cells. Cell. Microbiol. 2009, 11, 780–795. 10.1111/j.1462-5822.2009.01291.x. [DOI] [PubMed] [Google Scholar]
  48. Zhang M.; van Ravenzwaay B.; Fabian E.; Rietjens I. M.; Louisse J. Towards a generic physiologically based kinetic model to predict in vivo uterotrophic responses in rats by reverse dosimetry of in vitro estrogenicity data. Arch. Toxicol. 2018, 92, 1075–1088. 10.1007/s00204-017-2140-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Fischer F. C.; Henneberger L.; Schlichting R.; Escher B. I. How to improve the dosing of chemicals in high-throughput in vitro mammalian cell assays. Chem. Res. Toxicol. 2019, 32, 1462–1468. 10.1021/acs.chemrestox.9b00167. [DOI] [PubMed] [Google Scholar]
  50. a Escher B. I.; Ashauer R.; Dyer S.; Hermens J. L.; Lee J. H.; Leslie H. A.; Warne M. S.; et al. Crucial role of mechanisms and modes of toxic action for understanding tissue residue toxicity and internal effect concentrations of organic chemicals. Integr. Environ. Assess. Manage. 2011, 7, 28–49. 10.1002/ieam.100. [DOI] [PubMed] [Google Scholar]; b Hewitt N. J.; Hewitt P. Phase I and II enzyme characterization of two sources of HepG2 cell lines. Xenobiotica 2004, 34, 243–256. 10.1080/00498250310001657568. [DOI] [PubMed] [Google Scholar]
  51. Bray N. L.; Pimentel H.; Melsted P.; Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 2016, 34, 525–527. 10.1038/nbt.3519. [DOI] [PubMed] [Google Scholar]
  52. Wagner G. P.; Kin K.; Lynch V. J. A model based criterion for gene expression calls using RNA-seq data. Theory Biosci. 2013, 132, 159–164. 10.1007/s12064-013-0178-3. [DOI] [PubMed] [Google Scholar]
  53. Clothier R.; Gómez-Lechón M.; Kinsner-Ovaskainen A.; Kopp-Schneider A.; O’Connor J.; Prieto P.; Stanzel S. Comparative analysis of eight cytotoxicity assays evaluated within the ACuteTox Project. Toxicol. In Vitro 2013, 27, 1347–1356. 10.1016/j.tiv.2012.08.015. [DOI] [PubMed] [Google Scholar]
  54. Mannerström M.; Toimela T.; Sarkanen J. R.; Heinonen T. Human BJ fibroblasts is an alternative to mouse BALB/c 3T3 cells in in vitro neutral red uptake assay. Basic Clin. Pharmacol. Toxicol. 2017, 121, 109–115. 10.1111/bcpt.12790. [DOI] [PubMed] [Google Scholar]
  55. Wang K.; Shindoh H.; Inoue T.; Horii I. Advantages of in vitro cytotoxicity testing by using primary rat hepatocytes in comparison with established cell lines. J. Toxicol. Sci. 2002, 27, 229–237. 10.2131/jts.27.229. [DOI] [PubMed] [Google Scholar]
  56. Gülden M.; Schreiner J.; Seibert H. In vitro toxicity testing with microplate cell cultures: Impact of cell binding. Toxicology 2015, 332, 41–51. 10.1016/j.tox.2013.11.006. [DOI] [PubMed] [Google Scholar]
  57. Mateus A.; Matsson Pr.; Artursson P. Rapid measurement of intracellular unbound drug concentrations. Mol. Pharmaceutics 2013, 10, 2467–2478. 10.1021/mp4000822. [DOI] [PubMed] [Google Scholar]
  58. Mateus A.; Treyer A.; Wegler C.; Karlgren M.; Matsson P.; Artursson P. Intracellular drug bioavailability: a new predictor of system dependent drug disposition. Sci. Rep. 2017, 7, 43047 10.1038/srep43047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Cheever M.; Master A.; Versteegen R. A method for differentiating fetal bovine serum from newborn calf serum. BioProcess. J. 2017, 16, 16. 10.12665/J16OA.Cheever. [DOI] [Google Scholar]
  60. Lenz B.; Brink A.; Mihatsch M. J.; Altmann B.; Niederhauser U.; Steinhuber B.; Wyttenbach N.; Fischer H. Multiorgan Crystal Deposition of an Amphoteric Drug in Rats Due to Lysosomal Accumulation and Conversion to a Poorly Soluble Hydrochloride Salt. Toxicol. Sci. 2021, 180, 383–394. 10.1093/toxsci/kfaa191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Leeman M.; Choi J.; Hansson S.; Storm M. U.; Nilsson L. Proteins and antibodies in serum, plasma, and whole blood-size characterization using asymmetrical flow field-flow fractionation (AF4). Anal. Bioanal. Chem. 2018, 410, 4867–4873. 10.1007/s00216-018-1127-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Lipinski C. A.; Lombardo F.; Dominy B. W.; Feeney P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 2001, 46, 3–26. 10.1016/S0169-409X(00)00129-0. [DOI] [PubMed] [Google Scholar]
  63. Gülden M.; Mörchel S.; Seibert H. Factors influencing nominal effective concentrations of chemical compounds in vitro: cell concentration. Toxicol. In Vitro 2001, 15, 233–243. 10.1016/S0887-2333(01)00008-X. [DOI] [PubMed] [Google Scholar]
  64. Nakatsugawa T.; Bradford W. L.; Usui K. Hepatic disposition of parathion: uptake by isolated hepatocytes and chromatographic translobular migration. Pestic. Biochem. Physiol. 1980, 14, 13–25. 10.1016/0048-3575(80)90019-X. [DOI] [Google Scholar]
  65. Proença S.; Escher B. I.; Fischer F. C.; Fisher C.; Grégoire S.; Hewitt N. J.; Nicol B.; Paini A.; Kramer N. I. Effective exposure of chemicals in in vitro cell systems: A review of chemical distribution models. Toxicol. In Vitro 2021, 73, 105133 10.1016/j.tiv.2021.105133. [DOI] [PubMed] [Google Scholar]
  66. Austin R. P.; Barton P.; Davis A. M.; Fessey R. E.; Wenlock M. C. The thermodynamics of the partitioning of ionizing molecules between aqueous buffers and phospholipid membranes. Pharm. Res. 2005, 22, 1649–1657. 10.1007/s11095-005-6336-7. [DOI] [PubMed] [Google Scholar]
  67. Hallifax D.; Houston J. B. Saturable uptake of lipophilic amine drugs into isolated hepatocytes: mechanisms and consequences for quantitative clearance prediction. Drug Metab. Dispos. 2007, 35, 1325–1332. 10.1124/dmd.107.015131. [DOI] [PubMed] [Google Scholar]
  68. Leung Y. Y.; Yao Hui L. L.; Kraus V. B. Colchicine—Update on mechanisms of action and therapeutic uses. Semin. Arthritis Rheum. 2015, 45, 341–350. 10.1016/j.semarthrit.2015.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kratochwil N. A.; Huber W.; Müller F.; Kansy M.; Gerber P. R. Predicting plasma protein binding of drugs: a new approach. Biochem. Pharmacol. 2002, 64, 1355–1374. 10.1016/S0006-2952(02)01074-2. [DOI] [PubMed] [Google Scholar]
  70. Zhang F.; Xue J.; Shao J.; Jia L. Compilation of 222 drugs’ plasma protein binding data and guidance for study designs. Drug Discovery Today 2012, 17, 475–485. 10.1016/j.drudis.2011.12.018. [DOI] [PubMed] [Google Scholar]
  71. Endo S.; Goss K. U. Serum albumin binding of structurally diverse neutral organic compounds: data and models. Chem. Res. Toxicol. 2011, 24, 2293–2301. 10.1021/tx200431b. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

tx2c00128_si_001.pdf (332.3KB, pdf)

Articles from Chemical Research in Toxicology are provided here courtesy of American Chemical Society

RESOURCES