Skip to main content
HHS Author Manuscripts logoLink to HHS Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 6.
Published in final edited form as: Toxicology. 2021 Sep 9;462:152936. doi: 10.1016/j.tox.2021.152936

Genotoxicity evaluation using primary hepatocytes isolated from rhesus macaque (Macaca mulatta)

Ji-Eun Seo a, Kelly Davis b, Pritpal Malhi b, Xiaobo He c, Matthew Bryant c, John Talpos d, Susan Burks d, Nan Mei a, Xiaoqing Guo a,*
PMCID: PMC12412453  NIHMSID: NIHMS2108147  PMID: 34509578

Abstract

Non-human primates (NHPs) have played a vital role in fundamental, pre-clinical, and translational studies because of their high physiological and genetic similarity to humans. Here, we report a method to isolate primary hepatocytes from the livers of rhesus macaques (Macaca mulatta) after in situ whole liver perfusion. Isolated primary macaque hepatocytes (PMHs) were treated with various compounds known to have different pathways of genotoxicity/carcinogenicity and the resulting DNA damage was evaluated using the high-throughput CometChip assay. The comet data were quantified using benchmark dose (BMD) modeling and the BMD50 values for treatments of PMHs were compared with those generated from primary human hepatocytes (PHHs) in our previous study (Seo et al. Arch Toxicol 2020, 2207–2224). The results showed that despite varying CYP450 enzyme activities, PMHs had the same sensitivity and specificity as PHHs in detecting four indirect-acting (i.e., requiring metabolic activation) and seven direct-acting genotoxicants/carcinogens, as well as five non-carcinogens that are negative or equivocal for genotoxicity in vivo. The BMD50 estimates and their confidence intervals revealed species differences for DNA damage potency, especially for direct-acting compounds. The present study provides a practical method for maximizing the use of animal tissues by isolating primary hepatocytes from NHPs. Our data support the use of PMHs as a reliable surrogate of PHHs for evaluating the genotoxic hazards of chemical substances for humans.

Keywords: Non-human primate, Genotoxicity, Comet assay, Primary hepatocyte, Benchmark dose

1. Introduction

Human-derived metabolically competent cells are generally considered to be superior to cells from laboratory animals as in vitro models for evaluating chemical substances for their potential toxicity in humans (NAS, 2017). Human liver-derived cell lines contain various levels of phase I and II enzymes that catalyze the activation and detoxification of toxicants, and thus have been used as alternative models for genetic toxicity testing (Guo et al., 2020a). Primary human hepatocytes (PHHs) are recognized as the “gold standard” since they closely reflect human in vivo liver metabolism and functionality. In addition, several human hepatoma cell models, i.e., HepG2, HCC1.2, HuH6, and Hep3B, have phase I and II enzyme activities ranging between 5–10 % and <50 % of those found in PHHs, respectively (Misik et al., 2019). Our previous studies compared the performance of three human hepatic cell models, HepG2, HepaRG, and PHHs, using the high-throughput high-content (HTHC) CometChip assay; DNA damage dose–responses from these studies were analyzed using benchmark dose (BMD) potency ranking analysis (Seo et al., 2019, 2020). The results showed that PHHs had a higher sensitivity (90 %) for detecting the DNA damage induced by ten known genotoxicants/carcinogens than either HepG2 (60 %) or HepaRG (70 %) cells. Additionally, most of the compounds had comparable BMD values for DNA damage induction in PHHs and HepaRG cells. Despite their superior predictive ability, it is not practical to use PHHs in routine toxicity studies due to their limited availability and high costs (Zeilinger et al., 2016).

Non-human primates (NHPs) are physiologically and genetically similar to humans, and are susceptible to many human diseases (Mudry et al., 2011). NHPs are uniquely capable of reducing uncertainty/ambiguity in toxicological assessments and are often used for evaluating the preclinical toxicity of drugs intended for human use (Grow et al., 2016).

Both in vivo and in vitro NHP models have been used in a limited number of studies for genotoxicity assessment. A single dose of 50 mg/kg of N-ethyl-N-nitrosourea (ENU) significantly increased phosphatidylinositol glycan class A (Pig-a) and hypoxanthine-guanine phosphoribosyl transferase (Hprt) mutant frequencies in peripheral blood lymphocytes of male rhesus macaques, and the spectrum of Hprt mutation was similar to ENU-induced mutation determined for other species (Dobrovolsky et al., 2009). Further, both single and repeated administration of cyclophosphamide (CPA) significantly increased micronucleated reticulocytes in peripheral blood and bone marrow of rhesus macaques (Hotchkiss et al., 2008). Administration of chemotherapeutic agents (thiotepa, etoposide, or paclitaxel) also induced chromosomal aberrations in bone marrow of rhesus macaques (Rao et al., 2005). Quantitation of γ-H2AX foci was used as a biodosimeter for analyzing total-body exposure to ionizing radiation in a rhesus macaque model (Redon et al., 2010). Additionally, high levels of DNA adducts have been detected in several organs, particularly liver, kidney, and heart, in cynomolgus monkeys dosed with three heterocyclic aromatic amines, 2-amino-3-methylimidazo-[4,5-f]-quinoline (IQ), 2-amino-3, 8-dimethylimidazo[4,5-f]-quinoxaline (8-MeIQx), and 2-amino-1-me-thyl-6-phenylimidazo-[4,5-b]-pyridine (PhIP) (Thorgeirsson et al., 1995).

With regards to in vitro NHP studies, to the best of our knowledge, genotoxicity studies have only been conducted with an African green monkey kidney cell line (Vero). Treatment of Vero cells with 1.3 mM of the antifungal drug fluconazole induced DNA damage and micronucleus formation, likely due to the drug’s oxidative properties (Correa et al., 2018). An analgesic drug, dipyrone, also caused significant increases in DNA damage in Vero cells as evaluated by the comet assay (Gomes et al., 2021). These studies demonstrated the potential for NHP-based models in genotoxicity assessment.

Here, we evaluated whether primary macaque hepatocytes (PMHs) can be used as a surrogate for PHHs in genotoxicity assessments by testing 16 compounds known to have different genotoxic/carcinogenic modes of action (MoAs) using the HTHC CometChip assay (Table 1). The PMHs were isolated from liver after in situ whole liver perfusion following scheduled euthanasia of adult male rhesus macaques. The resulting DNA damage responses were quantified using BMD modeling and the BMD50s and their confidence intervals (CIs) compared with those generated using PHHs in our previous study (Seo et al., 2020).

Table 1.

Reported genotoxicity and carcinogenicity of test chemicals.

Group Chemical CAS# Ames Genotoxicity assay findings Carcinogenicity* References
in vitro in vivo
1. Indirect-acting genotoxicants/carcinogens 2,4-DAT 95-80-7 (+) (+) for MLA, CA, MN, comet, and UDS; (−) for comet in HepaRG and Hprt (+) for UDS and comet; (+/−) for MN in BM (+) for liver tumor; Group 2B (Guo et al., 2020b; Kirkland et al., 2016; Le Hegarat et al., 2014; Seo et al., 2019, 2020; Severin et al., 2005)
B[a]P 50-32-8 (+) (+) for MLA, CA, MN, Pig-a, comet, Hprt, and UDS (+) for comet, MN, Pig-a and gene mutations (+) for liver, lung tumor, etc.; Group 1 (Dertinger et al., 2019; Graupner et al., 2014; Guo et al., 2020b; Kirkland et al., 2016; Le Hegarat et al., 2010; Shah et al., 2018, 2016; Wang et al., 2018)
CPA 6055-19-2 (+) (+) for MLA, CA, and MN (+) for CA, MN, Pig-a, TG, and comet (+) for liver, lung, skin tumor, etc.; Group 1 (Bhalli et al., 2013; Dertinger et al., 2019; Dertinger et al., 2012; Kirkland et al., 2016; Le Hegarat et al., 2010, 2014; Seo et al., 2019, 2020; Yusuf et al., 2000)
DMBA 57-97-6 (+) (+) for MLA, CA, MN, comet, and UDS (+) for MN and Pig-a in blood, comet in liver, CA in BM, gene mutation (+) for mammary liver, skin tumor, etc. (Kirkland et al., 2016; Le Hegarat et al., 2014; Shi et al., 2011)
2. Direct-acting genotoxicants/carcinogens 4-NQO 56-57-5 (+) (+) for CA, MLA, Pig-a, comet (+) for MN, Pig-a, TG (+) for oral tumor (David et al., 2018; Dertinger et al., 2012; Kanojia and Vaidya, 2006; Le Hegarat et al., 2014)
CdCl2 10108-64-2 (+/−) (+) for MN, CA, MLA, Hprt, comet (+) for CA and MN, comet in liver (+) for lung, prostate, etc.; Group 1 (Guo et al., 2016; Kirkland et al. 2016; Seo et al., 2019)
Cisplatin 15663-27-1 (+) (+) for MN, Pig-a, comet (+) for MN, Pig-a, TG (+) for lung tumor, leukemia; Group 2A (Bemis et al., 2018; Bhalli et al., 2013; Dertinger et al., 2019)
Colchicine 64-86-8 (−) (+) for MN, CA, MLA; (−) for comet (+) for MN in BM and liver No adequate studies (FDA and CDER, 2013; Kirkland et al., 2016; Seo et al., 2020)
ENU 759-73-9 (+) (+) for MLA, CA, MN, Pig-a, Hprt, comet, and UDS (+) for CA, MN, Pig-a, TG, comet (+) for kidney, mammary tumor, etc.; Group 2A (Bemis et al., 2018; David et al., 2018; Dertinger et al., 2012; Habas et al., 2017; Kirkland et al., 2016; Le Hegarat et al., 2014; Seo et al., 2019, 2020)
HQ 123-31-9 (−) (+) for MLA, MN, CA, comet (+) for CA, MN; (−) for comet and TG in liver and stomach (+) for liver and kidney adenoma; Group 3 (IARC, 1999; Kirkland et al., 2016; Peng et al., 2013)
MMS 66-27-3 (+) (+) for MLA, CA, MN, Pig-a, Hprt, comet, and UDS (+) for CA, MN, UDS, Pig-a, TG, comet (+) for nasal, nervous system tumor, etc.; Group 2A (Bemis et al., 2018; Dertinger et al., 2012; Habas et al., 2017; Kirkland et al., 2016; Liu et al., 2019; Seo et al., 2019, 2020)
3. Non-carcinogens that are negative or equivocal for genotoxicity in vivo 2-Ethyl-1,3-hexanediol 94-96-2 (−) (+) for CA + S9; (−) for MLA, Hprt, MN, and SCE (−) for MN, CA (−) in dermal studies (Ballantyne, 2005; Kirkland et al., 2016; Slesinski et al., 1988)
o-Anthranilic acid 118-92-3 (−) (+) for CA, MLA, SCE; (+/−) for MN (−) for Pig-a, MN, CA, comet (−) in rats and mice; Group 3 (Dertinger et al., 2012; Kirkland et al., 2016; (NTP, 1978a)
Curcumin 458-37-7 (−) (+) for oxidative DNA damage, CA; (+/−) for MN, SCE (−) for MN (−) in male rats; E in mice and female rats (Damarla et al., 2018; Fowler et al., 2012; Li et al., 2008; NTP 1993; Sebastia et al., 2012)
Ethionamide 536-33-4 (−) (+) for MLA; (+/−) for CA; (−) for SCE, MN (−) for comet (−) in rats and mice; Group 3 (Kirkland et al., 2016; (NTP, 1978b)
Resorcinol 108-46-3 (−) (+) for MLA, SCE, CA; (+/−) for MN (+/−) for MN (−) in rats and mice; Group 3 (Kirkland et al., 2016; NTP, 1992; Soeteman-Hernandez et al., 2016)

2,4-DAT: 2,4-diaminotoluene; B[a]P: benzo[a]pyrene; CPA: cyclophosphamide; DMBA: 7,12-dimethylbenzanthracene; 4-NQO: 4-nitroquinoline1-oxide; CdCl2: cadmium chloride; ENU: N-ethyl-N-nitrosourea; HQ: hydroquinone; MMS: methyl methanesulfonate.

E, equivocal; BM, bone marrow; CA, chromosome aberration; Hprt, hypoxanthine-guanine phosphoribosyltransferase; MLA, mouse lymphoma assay; MN, micronucleus; Pig-a, phosphatidylinositol glycan class A; SCE, sister chromatid exchange; TG, transgenic mutation; UDS, unscheduled DNA synthesis.

*

Groups were classified by IARC Monographs on the identification of carcinogenic hazards to humans, Volumes 1–125 (https://monographs.iarc.who.int/list-of-classifications). +/–, both positive and negative results were reported.

2. Materials and methods

2.1. Chemicals and materials

Chemicals purchased from Sigma-Aldrich (St. Louis, MO) include: 16 test compounds (Table 1), chemicals and reagents used for hepatocyte isolation [bovine serum albumin (BSA), collagenase type IV, ethylene glycol-bis(2-aminoethylether)-N,N,N’,N’-tetraacetic acid (EGTA), D-(−)-fructose, d-(+)-glucose, heparin, 4-(2-hydroxyethyl)-1-piper-azineethanesulfonic acid (HEPES), hydrocortisone 21-hemisuccinate, insulin, L-15 medium (Leibovitz), percoll, trypsin inhibitor], six chemicals used for CYP450 enzyme substrates (phenacetin, bupropion, chlorzoxazone, diclofenac, dextromethorphan, and midazolam) and their metabolites (4-acetamidophenol, hydroxybupropion, 6-hydroxychlorzoxazone, 4-hydroxydiclofenac, dextrorphan, and 1-hydroxymidazolam, respectively). A CellTiter-Glo® 2.0 Cell Viability Assay kit (ATP assay) was obtained from Promega (Madison, WI). The CometChip and other comet supplies were purchased from R&D Systems (Minneapolis, MN).

2.2. In situ whole liver perfusion

All animal procedures were approved by the National Center for Toxicological Research (NCTR) Institutional Animal Care and Use Committee. Eight 15-year-old male rhesus macaques (body weight ranging from 11.4 to 14.7 kg) were humanely euthanized for brain tissue collection as part of a long-term neurotoxicological assessment (Rodriguez et al., 2010; Wilkinson et al., 2019; Zhang et al., 2016, 2021). Other tissues, including livers, were collected from these animals at the time of sacrifice in support of the 3Rs of animal testing. All animals had been treatment free for at least one year before their euthanasia and 7 of the 8 animals had livers with no signs of gross pathology at the time of sacrifice. No animals were sacrificed for the sole purpose of harvesting tissue to support this study. Under the guidance of a veterinarian, animals were sedated with ketamine (10 mg/kg, I.M.). Once appropriately sedated, the animals were administered Euthasol® (60–90 mg/kg, I.V., Virbac Corporation, Westlake, TX). After sedation, the brain and upper body of the animals were perfused with approximately 2 L of heparinized phosphate buffered saline (PBS, Thermo Fisher Scientific, Waltham, MA) for other tissue collection requirements. Then, the liver was flushed clean of residual blood by perfusion with washout solution (9.2 mM HEPES-NaCl buffer containing 0.5 mM EGTA, 16.65 mM fructose, and 10 U/mL heparin) at approximately 40 °C. After 20 min, perfusion was switched to the digestion solution (9.2 mM HEPES-NaCl buffer containing 6.7 mM KCl, 5 mM CaCl2, 16.65 mM fructose, 0.3 % BSA, 0.06 % collagenase type IV, 100 mg/L hyaluronidase, and 100 mg/L trypsin inhibitor) at approximately 40 °C by use of a 3-way stopcock, without stopping the peristaltic pump to avoid the incorporation of air bubbles into the system. The perfusion was stopped after 20–30 min of perfusion with digestion solution. The endpoint of digestion was determined when the liver possessed an overall soft consistency (Ulrich et al., 1990).

2.3. Primary macaque hepatocytes isolation

After perfusion, a portion of liver was removed and placed in a sterile beaker containing 200 mL suspension medium (L-15 medium supplemented with 0.2 % BSA, 1 M HEPES, 5 μg/mL insulin, and 5 % FBS) on ice and transported within 5 min into a biosafety cabinet. The Glisson’s capsule of the digested liver was cut with sterile scissors and the parenchyma was mechanically dissociated and then gently pushed through two sieves of decreasing sizes (200 and 80 μm). The first cell suspension was centrifuged at 60 ×g for 2 min. After two washes with suspension medium, the cells were purified with 90 % isotonic percoll and resuspended in plating medium (L-15 medium supplemented with 1× nonessential amino acids, 100 U/mL penicillin, 100 μg/mL streptomycin, 1× GlutaMax, 1.4 μM hydrocortisone, 100 nM insulin, and 17 % FBS). Cell number and viability were measured using the trypan blue exclusion assay, and 5 × 104 cells in 100 μL plating medium were plated in each well of collagen I coated 96-well plates and cultured at 37 °C in a humidified incubator containing 5 % CO2. PMHs were allowed to attach for 4 h, and the medium was replaced with fresh plating medium. PMHs were incubated overnight at 37 °C in a humidified incubator containing 5 % CO2.

2.4. Determination of CYP450 activities

Activities of six major cytochrome P450 (CYP450) enzymes were measured using established methods (Seo et al., 2019). Briefly, after being cultured overnight, the PMH plating medium was changed to 100 μL/well treatment medium (L-15 medium supplemented with 1× nonessential amino acids, 100 U/mL penicillin, 100 μg/mL streptomycin, 1× GlutaMax, 17 μM hydrocortisone, 100 nM insulin, and 10 % FBS) containing 250 μM chlorzoxazone (CYP2E1) or a cocktail of five substrates, i.e., 100 μM phenacetin (CYP1A2), 100 μM bupropion (CYP2B6), 20 μM diclofenac (CYP2C9), 20 μM dextromethorphan (CYP2D6), and 50 μM midazolam (CYP3A4), in quadruplicate. Following a 2-h incubation, 100 μL medium were collected and the metabolites were quantified by high performance liquid chromatography tandem mass spectrometry (HPLC-MS/MS) (Dierks et al., 2001; Seo et al., 2019). The remaining cells were immediately washed with cold PBS and lysed in 30 μl RIPA buffer (Thermo Fisher Scientific, Waltham, MA) on ice for 30 min. The protein concentrations were measured using a Pierce BCA protein assay kit (Thermo Fisher Scientific, Waltham, MA) as described previously (Guo et al., 2015; Seo et al., 2021). Final CYP450 activities were averaged and expressed as pmol metabolite/min/mg protein.

2.5. Cell treatments

Following a 21-h incubation after plating, PMHs were treated for 24 h with 16 compounds that have been tested in PHHs in our previous study (Seo et al., 2020). The highest concentrations were selected based on the cytotoxicity of compounds in PHHs. Specifically, the 16 test compounds included four indirect-acting (requiring metabolic activation) genotoxicants/carcinogens [2,4-diaminotoluene (2,4-DAT), benzo [a]pyrene (B[a]P), cyclophosphamide (CPA), 7,12-dimethylbenz[a] anthracene (DMBA)], seven direct-acting genotoxicants/carcinogens [4-nitroquinoline 1-oxide (4-NQO), cadmium chloride (CdCl2), cisplatin, colchicine, ENU, hydroquinone (HQ), and methyl methanesulfonate (MMS)], and 5 non-carcinogens that are negative or equivocal for genotoxicity in vivo (2-ethyl-1,3-hexanediol, curcumin, ethionamide, o-anthranilic acid, and resorcinol). All chemical stock solutions were prepared as described previously (Seo et al., 2020). Two wells received 1× and 0.75× of the desired top concentrations of test article in treatment medium. A serial dilution was performed from these top two concentrations to generate 11 closely spaced concentrations with 100 μl per well on the 96-well plates. Dimethyl sulfoxide (DMSO) was used as the vehicle control with a final concentration of ≤ 0.1 %.

2.6. Cytotoxicity and CometChip assays

The cytotoxicity and CometChip assays were performed following a 24-h treatment. Cytotoxicity was measured using the ATP assay as recommended by the manufacturer (Promega). Briefly, the treatment medium was removed following treatment and the ATP reagent was added into each well at a ratio of 1:10, followed by an incubation of 10 min at room temperature. Luminescence was measured using a Cytation 5 Cell Imaging Multi-Mode Reader (BioTek, Winooski, VT). The relative viability (%) was calculated by the ratio of intensity levels of treated cells to those of vehicle controls.

The CometChip assay was performed as described previously with minor modifications (Seo et al., 2020). Briefly, the treated cells were detached by adding 30 μL Accutase® solution into each well of the 96-well plates, followed by the addition of 120 μL treatment medium to stop the reaction. Then all treated cells were transferred into each well of a 96-well CometChip for conducting the comet assay according to the manufacture’s protocol. Comet images were acquired automatically using the Cytation 5 (BioTek, Winooski, VT) and analyzed using Trevigen Comet Analysis Software. DNA damage effects were expressed as the percentage of tail DNA.

2.7. Quantification of DNA damage concentration-responses

The PMH comet data were quantified using PROASTweb (version 70.1), which was developed by the Dutch National Institute for Public Health and the Environment (RIVM). The BMD value, along with its lower and upper bounds of the 90 % CIs, BMDU and BMDL, were calculated simultaneously at the defined critical effect size (CES) or benchmark response (BMR). The present study calculated BMD50 values, with the rationale that 0.6 (60 % increase in response over vehicle control, BMD60) has been proposed as a suitable CES for evaluating in vivo rat liver comet data (Zeller et al., 2016) and that the BMD50 is commonly used in BMD quantification studies. The BMD50 values were calculated using exponential and Hill models and values from the exponential model were used for potency ranking in the present study. The PHH comet data from our previous study (Seo et al., 2020) were reanalyzed using PROAST and resulting BMD50 values were compared with those from the PMH comet data.

2.8. Statistical analysis

Comet data were averaged from at least three independent experiments using hepatocytes isolated from different animals and presented as the mean ± standard deviation (SD). The lowest effective concentration (LEC) and pairwise comparisons between treatment groups and the vehicle control were determined using one-way analysis of variance (ANOVA) followed by Dunnett’s test. The LEC was defined as the lowest concentration giving a positive response in the comet assay. Statistical significance was determined when p < 0.05 using SigmaPlot 13.0 (Systat Software, San Jose, CA). Correlation of BMD50 values between PMHs and PHHs was evaluated by Spearman’s rank correlation coefficient using GraphPad Prism version 6.04 (GraphPad Software, La Jolla, CA, USA).

3. Results

3.1. Cell viability and morphology

High quality PMHs were isolated from seven out of eight animals in the present study. After percoll purification, the viability of freshly isolated hepatocytes from the seven animals ranged from 75 to 95 %, with an average viability of 87.4 ± 6.8 %. Hepatocytes isolated from one primate (#4), which had an enlarged liver and scars in the left lateral lobe of the liver, displayed low cell viability (38 %) and were excluded from toxicity assays.

Cells were attached to the collagen-I-coated 96-well plates approximately 3 h after plating (Fig. 1). Live and attached primary hepatocytes exhibited a polygonal or typical hexagonal shape after a 4-h incubation, with nonviable cells observed on the top of the monolayer. After a 24-h incubation, cells appeared to have flattened morphology, with clearly outlined membranous boundaries. Most cells were single or binucleated and contained dense cytoplasmic granules, but multi-nucleated cells were also occasionally seen in the plate.

Fig. 1. Cell morphology of primary macaque hepatocytes (PMHs) after plating.

Fig. 1.

Light micrograph illustrates PMHs after 0 h (A), 4 h (B), and 24 h (C) in culture (200 × magnification).

3.2. CYP activities in PMHs derived from macaques

The basal activities of six major CYPs (CYP1A2, 2B6, 2C9, 2D6, 2E1, and 3A4) were examined (Fig. 2). Although variations were seen between animals, all NHPs consistently showed high levels of activities of CYP1A2, 2B6, 2D6, and 3A4, with averages between 64–93 pmol/min/mg protein. The activities of CYP2C9 and 2E1 in the 8 animals were less than 1 pmol/min/mg protein, which were significantly lower than those of the other four CYPs tested. No CYP3A4 activity could be detected in the PMHs of primate #2 likely due to technical issues.

Fig. 2. CYP450 activity in PMHs isolated from eight rhesus macaques.

Fig. 2.

Six major CYP450 activities (CYP1A2, 2B6, 2C9, 2D6, 2E1, and 3A4) were measured 24 h after plating. CYP450 activities are expressed as pmol/min/mg protein and the data are presented as the mean ± SD (n ≥ 3). PHHs and HepaRG cells data are from our previous study (Seo et al., 2020). < LOD, below the limit of detection; NT, not tested.

The CYP activities in PMHs also were compared with those in human hepatic cells, i.e., PHHs and differentiated HepaRG cells (Fig. 2). CYP3A4 activity was comparable among the three types of liver cells. Interestingly, PMHs had much higher activities of CYP1A2, 2B6, and 2D6 than those in PHHs and HepaRG cells (with an average of 64–85 vs. 3–14 pmol/min/mg protein). Conversely, the CYP2C9 activity of PMHs was markedly lower than those in PHHs and HepaRG cells (0.2 vs. 7–31 pmol/min/mg protein). PMHs had low CYP2E1 activities, with values of 0.2–1.4 pmol/min/mg protein, while the CYP2E1 activity in HepaRG cells was below the detection limits. PHHs from three donors showed 11–126 pmol/min/million cells CYP2E1 activities as reported by the supplier (Seo et al., 2020). Regrettably, CYP2E1 activity was not tested in PHHs in our previous study, and we were unable to make a direct comparison between PMHs and PHHs in the present study.

3.3. Cytotoxicity profiles of the 16 compounds

Following a 24-h exposure, all 16 compounds induced cytotoxic effects in PMHs as measured by the ATP assay (Table 2). The maximum concentration of each chemical used in the CometChip assay was defined as the highest concentration producing cell viability of ≥ 70 % or 10 mM when no cytotoxicity was observed (OECD, 2015). Nine out of the 16 compounds induced the same cytotoxicity in PMHs and PHHs, as evaluated by the maximum concentration and cell viability. CPA displayed the most distinct cytotoxic effects between the two types of primary hepatocytes, with cell viability of 50 % at 2500 μM in PMHs and 88 % at 10,000 μM in PHHs. Three compounds, DMBA, ENU, and o-anthranilic acid, had cytotoxic effects over the same range of concentrations, but only in PMHs. Cisplatin (50 μM) and HQ (200 μM) were slightly more cytotoxic in PHHs, with a cell viability of 78–81 % in PMHs vs. 55–63 % in PHHs. CdCl2 at 4 μM induced slightly higher cytotoxicity (43 %) in PMHs than in PHHs (25 %).

Table 2.

Comparison of cytotoxicity and DNA damage responses between primary macaque (PMH) and human (PHH) hepatocytes.

Group Chemical Max. con. (μM) LECa (μM) Cytotoxicityb DNA damagec
PMH PHH PMH PHH PMH PHH PMH PHH
Indirect-acting genotoxicants/carcinogens 2,4-DAT 8000 8000 8000 1414 + + ++ ++
B[a]P 100 100 37.5 6.3 + + +++ ++
CPA 5000 10,000 937.5 1768 +++ + +++ ++
DMBA 500 250 31.25 62.5 + +++ ++
Direct-acting genotoxicants/carcinogens 4-NQO 7.5 5 7.5 2.5 + + ++ ++
CdCl2 4 4 2 1 ++ + +++ +
Cisplatin 50 50 9.4 6.3 + ++ ++ ++
Colchicine 4 4 + +
ENU 3200 3200 2400 800 + +++ +++
HQ 200 200 + ++
MMS 500 500 125 62.5 + + +++ +++
Non-carcinogens that are negative or equivocal for genotoxicity in vivo 2-Ethyl-1,3-hexanediol 10,000 10,000 + +
o-Anthranilic acid 5000 5000 ++
Curcumin 40 40 + +
Ethionamide 2000 2000 + +
Resorcinol 1000 2000 ++ ++

PHH data are from our previous study (Seo et al., 2020).

a

LEC, the lowest effective concentration, determined by one-way ANOVA followed by Dunnett’s test, is the lowest concentration that gives a positive response in the comet assay.

b

Cytotoxicity was determined by the ATP assay following a 24-h treatment. −, cytotoxicity of 0–10 %; +, 11–30 %; ++, 31–50 %; +++, > 50 %.

c

DNA damage was expressed as the relative ratio calculated by comparing the %DNA in tail at the highest concentration to those of the vehicle control. −, The ratio ≤ 1.5-fold and p ≥ 0.05; +, 1.5 < ratio ≤ 2, and p < 0.05; ++, 2 < ratio ≤ 5; +++, the ratio > 5.

3.4. DNA damage profiles of the 16 compounds

Although different levels of CYP activities and cytotoxicity were seen in PMHs and PHHs, both cell types demonstrated the same positive/negative calls for all 16 compounds tested, whether or not the compounds required metabolic activation (Table 2, Fig. 3). Overall, PMHs induced higher percentages of tail DNA in the comet assay than PHHs for four out of the nine positive compounds. At the highest acceptable concentration, B[a]P, CPA, DMBA, and CdCl2 induced 6.1–8.1-fold and 1.8–4.4-fold increases in % tail DNA (DNA damage) in PMHs and PHHs, respectively, compared to the vehicle control. Interestingly, out of the nine positive compounds, the LECs of seven compounds were 1.5–6-fold higher in PMHs than those in PHHs, while the LEC values for two compounds (CPA and DMBA) were 2-fold higher in PHHs than in PMHs. Both PHHs and PMHs also showed a 100 % specificity for detecting the five non-carcinogens that are negative or equivocal for genotoxicity in vivo (Fig. 3C).

Fig. 3. DNA damage and cytotoxicity of 16 compounds in PMHs.

Fig. 3.

PMHs were exposed to four indirect-acting (A), seven direct-acting genotoxicants or carcinogens (B), and five non-carcinogens that are negative or equivocal for genotoxicity in vivo (C) for 24 h. The relative cell viability (% of control, indicating cytotoxicity) was measured by the ATP assay (right y-axis and red line) and DNA damage (% tail DNA; left y-axis and black bar) was detected using the CometChip assay. The data are expressed as the mean ± SD (n ≥ 3). *p < 0.05, **p < 0.01, and ***p < 0.001 vs. control. See Table 1 for abbreviations of the compounds tested.

3.5. BMD modeling on DNA damage responses from nine positive compounds in PMHs

By using a PROAST covariate approach, the nine concentration-responses were roughly divided into six groups having non-overlapping BMDLs and BMDUs (Fig. 4A). CdCl2, showed the lowest BMD50 value of 0.37 μM (Table 3), indicating it was the most potent DNA damage inducer in PMH among the nine compounds. Three compounds (4-NQO, B[a]P, and DMBA) had BMD50 values of 3.5–5.5 μM; MMS and cisplatin had similar BMD50 values of 21–22 μM; while CPA, ENU, and 2,4-DAT had the highest BMD50 values of 253, 683, and 2831 μM in PMHs, respectively. The width of the CI, i.e., the BMDU/BMDL ratio, represents the uncertainty in the BMD estimate and reflects the quality of the concentration-response data (Slob, 2014; Wills et al., 2017). The ratios of the BMDU to the BMDL in the present study were between 2.3–3.2 (Table 3), indicating relatively high precision for the BMD estimates.

Fig. 4.

Fig. 4.

Comparison of BMD50 and its upper and lower 90 % confidence intervals (BMDU and BMDL) of chemical-induced DNA damages between PMHs and PHHs. The DNA damage concentration-response data in PHHs were obtained from our previous study (Seo et al., 2020). BMD and its BMDU and BMDL were calculated from PMH (A) and PHH (B) comet data using exponential (upper line) and Hill (lower line) models of PROAST. (C) The bars represent the range between BMDUs and BMDLs and are used to differentiate between responses based on non-overlapping confidence intervals (CIs). Red, PHHs; Blue, PMHs. (D) Correlation of BMD50 values between PMHs and PHHs is illustrated by BMD and its CIs for PMH comet data against those of PHHs. See Table 1 for abbreviations of the compounds tested.

Table 3.

Comparison of the benchmark dose (BMD50) and potency ranking between primary macaque and human hepatocytes.a

Chemical Primary macque hepatocytes Primary human hepatocytes
BMD50 (μM) BMDL50–BMDU50 (μM) BMDU/BMDL BMD50 (μM) BMDL50–BMDU50 (μM) BMDU/BMDL
2,4-DAT 2831 (9)b 1790–4660 2.6 880.6 (9)b 489–1580 3.2
4-NQO 3.5 (2) 2.2–6.3 2.9 0.3 (1) 0.1–0.6 4.4
B[a]P 5.5 (4) 3.4–8.5 2.5 9.2 (5) 5.2–16.3 3.2
CdCl2 0.4 (1) 0.2–0.5 2.3 0.9 (2) 0.5–1.6 3.2
Cisplatin 21.3 (5) 12.4–39.2 3.2 2.5 (4) 1.3–5.3 4.2
CPA 253.8 (7) 165–372 2.3 248.2 (8) 135–437 3.2
DMBA 4.0 (3) 2.6–6.1 2.4 15.1 (6) 8.5–25.8 3.0
ENU 683 (8) 439–1030 2.3 47.3 (7) 25.3–83.5 3.3
MMS 22.3 (6) 14.2–33.7 2.4 1.0 (3) 0.5–1.9 3.5

Primary human hepatocyte data are from our previous study (Seo et al., 2020).

a

BMDs for the CometChip data were calculated using PROAST.

b

Potency ranking.

3.6. Comparison of BMD values between PMHs and PHHs

The BMD values and upper and lower 90 % CIs for each test compound also were used for the comparative evaluation of DNA damage potency in PMHs and PHHs. For both PMHs and PHHs, CdCl2 and 4-NQO were the most potent, and CPA, ENU, and 2,4-DAT were the least potent DNA damage inducers (Table 3 and Fig. 4A and B). DMBA was more potent (ranked #3) in PMHs than in PHHs (ranked #6), while MMS was more potent in PHHs, with a BMD50 value 22-fold lower than that in PMHs.

Among the four indirect-acting compounds, the BMD50 CIs for both B [a]P and CPA tested in PMHs and PHHs overlapped with each other (Fig. 4C). The BMD50 for 2,4-DAT was 3.2-fold higher in PMHs than in PHHs, while DMBA had a 3.8-fold higher BMD50 value in PHHs than in PMHs (Table 3). Four out of the five positive direct-acting compounds, 4-NQO, cisplatin, ENU, and MMS, had significantly (8–22-fold) lower BMD50 values in PHHs than in PMHs. CdCl2 had a lower BMD50 value in PMHs than in PHHs (0.37 vs. 0.89 μM), but the 90 % CIs overlapped for the DNA damage responses in the two types of primary hepatocytes.

A correlation evaluation using the Spearman rank correlation test on comet data in PHHs and PMHs showed that BMD50 values had a correlation coefficient of 0.8 (p < 0.05), indicating a strong correlation between the PMH and PHH data.

4. Discussion

The phylogenetic similarity of NHP species to Homo sapiens makes them unique in their ability to mimic toxicity testing in humans. Due to their homology with humans in terms of physiology and drug metabolism, NHPs are widely used in preclinical studies for drug and antibody development (Uno et al., 2016). The present study used in situ whole liver perfusion, as was initially used in rats (Oldham et al., 1979), for isolating primary hepatocytes from rhesus macaques. In situ perfusion of rodent livers maintains liver function for up to 2 h and has better cell yield and viability compared to isolated organ perfusion (Aiken et al., 1990; Choi et al., 2019). Isolated PMHs in the present study had a relatively high average cell viability of 87.4 %. An optimal liver perfusion/digestion and normal liver morphology are likely critical factors for the successful isolation of high-quality primary hepatocytes. The hepatocytes with low viability (38 %) were obtained in the present study from one animal with an enlarged/scarred liver. However, the poor cell viability could also be due to procedural or technical issues.

CYP450 enzymes are essential for the bioactivation and/or detoxification of many xenobiotics and drugs. The present study compared the activities of six major CYPs in PMHs, PHHs, and HepaRG cells. CYP3A4/5 has been reported to be the most abundant (40 % of total) enzyme in the liver of both humans and NHPs (Gundert-Remy et al., 2014; Uehara et al., 2011), and CYP3A4 showed similar activities in the three types of cells (Fig. 2). However, significant differences in the activities of several CYPs were seen in human and rhesus macaque hepatocytes, despite CYP450 enzymes displaying 95 % homology between humans and NHPs (Martignoni et al., 2006). Interindividual variability in CYP450 activities also was observed in the present study even though the 8 animals were from the same species (rhesus macaque) and were all males. Regardless of the varying CYP450 activities and cytotoxic effects, PMHs in the present study displayed identical sensitivity and specificity for detecting the eleven genotoxicants/carcinogens and five non-carcinogens that are negative or equivocal for genotoxicity in vivo (Fig. 2 and Table 2). These results demonstrate the usefulness of PMHs as a surrogate to PHHs for predicting genotoxic risks for the xenobiotics humans are likely to be exposed to.

As genetic toxicology is moving from a qualitative, “screen and bin” binary approach to quantitative dose-response analysis and risk assessment (White and Johnson, 2016), we conducted a quantitative comparison of DNA damage potency for the-comet-assay-positive compounds between PMHs and PHHs. BMD50 values were estimated using PROAST software, with covariate analysis used to improve the precision for DNA damage potency ranking of the nine concentration-response relationships (Wills et al., 2016). Our data showed that seven out of nine compounds had a similar rank order of potency in the two types of primary hepatocytes, regardless of requiring metabolic activation or not. Thus, CdCl2 and 4-NQO ranked #1–2, B[a]P and cisplatin ranked #4–5, and CPA, ENU, and 2,4-DAT ranked #7–9 (Table 3). In contrast, DMBA, a polycyclic aromatic hydrocarbon mainly metabolized by CYP1B1/1A1 (Lin et al., 2012), showed a higher DNA damage potency ranking in PMHs (#3), with a BMD50 value 3.75-fold lower (i.e., more potent) compared to PHHs (ranked #6). CYP1A1, 1A2, and 1B1 are three highly conserved members of the CYP1 family. We didn’t measure the activities of CYP1A1 and CYP1B1 in the present study because both are expressed predominantly in extrahepatic tissues, with very low levels of expression in the liver of human and other species including NHPs. In contract, CYP1A2 is mainly expressed in the liver (Wang et al., 2016). Our results indicate that PMHs isolated from the eight NHPs all had significantly higher CYP1A2 activities compared to PHHs from three human donors (Fig. 2). Given that CYP1A2 appeared as a gene duplication event from CYP1A1 and ancestors of the CYP1A and CYP1B subfamily diverged from one another >400 million years ago (Jorge-Nebert et al., 2010), we speculate that PMHs may possess higher levels of CYP1B1 and 1A1 activities than PHHs. Consequently, more mutagenic metabolites of DMBA might be produced in PMHs than in PHHs.

To further evaluate the appropriateness of PMHs for predicting human genotoxicity risk, we compared BMD50 values and their CIs derived from the CometChip data for PMHs with those for PHHs. These comparisons led to several interesting findings. First, the DNA damage responses in PMHs and PHHs for two out of four (50 %) indirect-acting compounds had overlapping BMD50 upper and lower CIs, while only for one out of the five (20 %) direct-acting compounds had overlapping BMD50 CIs for DNA damage (Fig. 4C). Second, for the six compounds that had non-overlapping BMD50 CIs, the four direct-acting compounds (4-NQO, cisplatin, ENU, and MMS) had distinctly different BMD50 values in PMHs and PHHs compared to the two indirect-acting compounds (2,4-DAT and DMBA). These observations demonstrate that primary hepatocytes isolated from rhesus macaques can detect indirect-acting human genotoxic compounds both qualitatively and quantitatively, although the CYP activity levels varied between PMHs and PHHs. Third, all but one compound (DMBA) that had non-overlapping BMD50 CIs had more conservative BMD50 values in PHHs than in PMHs. In our previous study, 4-NQO showed remarkably higher DNA damage potency in PHHs from one donor compared to the other two donors (Seo et al., 2020). Even though we assume that the significant difference in BMD50 values between the two types of hepatocytes is due to inter-species differences rather than intraspecies individual variability, based on the observation that the BMD50 of 4-NQO was 12-fold higher in PMHs than in PHHs (Table 3). It is worth noting that the PMHs used in the present study were freshly isolated from the liver of euthanized animals, while PHHs were cryopreserved for several years before they were used for these studies. This freezing and thawing process may have resulted in slightly compromised cell viability and higher background DNA damage values (Gajski et al., 2020). Cryopreservation of primary hepatocytes provides constantly available resources for studying in vitro drug metabolism and evaluating the toxicity of various drugs intended for human use (Hengstler et al., 2000). The present study has optimized the conditions for cryopreserving freshly isolated PMHs and stored them in liquid nitrogen for future use. However, further studies are required to validate cryopreservation techniques by comparing metabolic functions and the sensitivity for detecting genotoxic compounds between fresh and frozen PMHs.

In conclusion, by comparing DNA damage responses between macaque and human primary hepatocytes qualitatively and quantitatively, our results demonstrated that PMHs had the same sensitivity and specificity as PHHs in evaluating the genotoxicity of 16 compounds, indicating that PMHs may be an appropriate surrogate for PHHs in hazard identification studies. However, despite the fact that NHPs are the closest relatives to human, quantitative differences were seen between PMHs and PHHs regarding CYP450 enzyme activities as well as DNA damage responses. Differences in DNA damage responses were especially notable for direct-acting genotoxicants/carcinogens. Currently, animal genotoxicity and carcinogenicity data are used by regulatory bodies to derive point-of-departure metrics; and cross-species extrapolation is used to determine health-based guidance values that specify a maximum human exposure, e.g., acceptable daily intake or permitted daily exposure (White et al., 2020). Our results demonstrate the existence of differences in toxicity, even for closely related primate species. However, given the close phylogenetic relationship between NHPs and humans and the strong correlation of the BMD50 values between the PMH and PHH comet data, our data support the use of PMHs as a surrogate for PHHs for evaluating the potential genotoxic hazard of compounds in humans.

Acknowledgements

This work was supported by the U.S. Food and Drug Administration (FDA), National Center for Toxicological Research (NCTR, project number E0770501). J.E.S. was supported by an appointment to the Postgraduate Research Program at the NCTR administered by the Oak Ridge Institute for Science Education (ORISE) through an interagency agreement between the U.S. Department of Energy and the U.S. FDA. The authors thank the NCTR non-human primate (NHP) team, especially Ms. April Bilyeu and Tristin Wellmeyer, for their technical support, Dr. Qiang Shi for helpful suggestions for the hepatocyte isolation, and Drs. Mugimane G. Manjanatha and Fang Liu for their critical review of the manuscript.

Footnotes

Disclaimer

This manuscript reflects the views of the authors and does not necessarily reflect those of the U.S. Food and Drug Administration.

Declaration of Competing Interest

There was no conflict of interest declared.

References

  1. Aiken J, Cima L, Schloo B, Mooney D, Johnson L, Langer R, Vacanti JP, 1990. Studies in rat liver perfusion for optimal harvest of hepatocytes. J. Pediatr. Surg 25, 140–145. [DOI] [PubMed] [Google Scholar]
  2. Ballantyne B, 2005. 2-Ethyl-1,3-hexanediol. J. Appl. Toxicol 25, 248–259. [DOI] [PubMed] [Google Scholar]
  3. Bemis JC, Avlasevich SL, Labash C, McKinzie P, Revollo J, Dobrovolsky VN, Dertinger SD, 2018. Glycosylphosphatidylinositol (GPI) anchored protein deficiency serves as a reliable reporter of Pig-a gene Mutation: support from an in vitro assay based on L5178Y/Tk(+/−) cells and the CD90.2 antigen. Environ. Mol. Mutagen 59, 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bhalli JA, Shaddock JG, Pearce MG, Dobrovolsky VN, 2013. Sensitivity of the Pig-a assay for detecting gene mutation in rats exposed acutely to strong clastogens. Mutagenesis 28, 447–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Choi WM, Eun HS, Lee YS, Kim SJ, Kim MH, Lee JH, Shim YR, Kim HH, Kim YE, Yi HS, Jeong WI, 2019. Experimental applications of in situ liver perfusion machinery for the study of liver disease. Mol. Cells 42, 45–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Correa R, Mota TC, Guimaraes AC, Bonfim LT, Burbano RR, Bahia MO, 2018. Cytotoxic and genotoxic effects of fluconazole on African green monkey kidney (Vero) cell line. Biomed Res. Int 2018, 6271547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Damarla SR, Komma R, Bhatnagar U, Rajesh N, Mulla SMA, 2018. An evaluation of the genotoxicity and subchronic oral toxicity of synthetic curcumin. J. Toxicol 2018, 6872753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. David R, Talbot E, Allen B, Wilson A, Arshad U, Doherty A, 2018. The development of an in vitro Pig-a assay in L5178Y cells. Arch. Toxicol 92, 1609–1623. [DOI] [PubMed] [Google Scholar]
  9. Dertinger SD, Phonethepswath S, Avlasevich SL, Torous DK, Mereness J, Bryce SM, Bemis JC, Bell S, Weller P, Macgregor JT, 2012. Efficient monitoring of in vivo pig-a gene mutation and chromosomal damage: summary of 7 published studies and results from 11 new reference compounds. Toxicol. Sci 130, 328–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Dertinger SD, Avlasevich SL, Torous DK, Singh P, Khanal S, Kirby C, Drake A, MacGregor JT, Bemis JC, 2019. 3Rs friendly study designs facilitate rat liver and blood micronucleus assays and Pig-a gene mutation assessments: proof-of-concept with 13 reference chemicals. Environ. Mol. Mutagen 60, 704–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Dierks EA, Stams KR, Lim HK, Cornelius G, Zhang H, Ball SE, 2001. A method for the simultaneous evaluation of the activities of seven major human drug-metabolizing cytochrome P450s using an in vitro cocktail of probe substrates and fast gradient liquid chromatography tandem mass spectrometry. Drug Metab. Dispos 29, 23–29. [PubMed] [Google Scholar]
  12. Dobrovolsky VN, Shaddock JG, Mittelstaedt RA, Manjanatha MG, Miura D, Uchikawa M, Mattison DR, Morris SM, 2009. Evaluation of Macaca mulatta as a model for genotoxicity studies. Mutat. Res 673, 21–28. [DOI] [PubMed] [Google Scholar]
  13. FDA, CDER, 2013. Pharmacology Review(s): Secondary Pharmacology and Toxicology Review for NDA 204–820. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/204820Orig1s000PharmR.pdf.
  14. Fowler P, Smith K, Young J, Jeffrey L, Kirkland D, Pfuhler S, Carmichael P, 2012. Reduction of misleading (“false”) positive results in mammalian cell genotoxicity assays. I. Choice of cell type. Mutat. Res 742, 11–25. [DOI] [PubMed] [Google Scholar]
  15. Gajski G, Geric M, Zivkovic Semren T, Tariba Lovakovic B, Orescanin V, Pizent A, 2020. Application of the comet assay for the evaluation of DNA damage from frozen human whole blood samples: implications for human biomonitoring. Toxicol. Lett 319, 58–65. [DOI] [PubMed] [Google Scholar]
  16. Gomes LM, Moyses DA, Nascimento HFS, Mota TC, Bonfim LT, Cardoso PCS, Burbano RMR, Bahia MO, 2021. Genotoxic and cytotoxic effects of the drug dipyrone sodium in African green monkey kidney (Vero) cell line exposed in vitro. Naunyn Schmiedebergs Arch. Pharmacol 394, 1529–1535. [DOI] [PubMed] [Google Scholar]
  17. Graupner A, Instanes C, Dertinger SD, Andersen JM, Lindeman B, Rongved TD, Brunborg G, Olsen AK, 2014. Single cell gel electrophoresis (SCGE) and Pig-a mutation assay in vivo-tools for genotoxicity testing from a regulatory perspective: a study of benzo[a]pyrene in Ogg1(−/−) mice. Mutat. Res 772, 34–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Grow DA, McCarrey JR, Navara CS, 2016. Advantages of nonhuman primates as preclinical models for evaluating stem cell-based therapies for Parkinson’s disease. Stem Cell Res. 17, 352–366. [DOI] [PubMed] [Google Scholar]
  19. Gundert-Remy U, Bernauer U, Blomeke B, Doring B, Fabian E, Goebel C, Hessel S, Jackh C, Lampen A, Oesch F, Petzinger E, Volkel W, Roos PH, 2014. Extrahepatic metabolism at the body’s internal-external interfaces. Drug Metab. Rev 46, 291–324. [DOI] [PubMed] [Google Scholar]
  20. Guo X, Chen S, Zhang Z, Dobrovolsky VN, Dial SL, Guo L, Mei N, 2015. Reactive oxygen species and c-Jun N-terminal kinases contribute to TEMPO-induced apoptosis in L5178Y cells. Chem. Biol. Interact 235, 27–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Guo X, Heflich RH, Dial SL, Richter PA, Moore MM, Mei N, 2016. Quantitative analysis of the relative mutagenicity of five chemical constituents of tobacco smoke in the mouse lymphoma assay. Mutagenesis 31, 287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Guo X, Seo JE, Li X, Mei N, 2020a. Genetic toxicity assessment using liver cell models: past, present, and future. J. Toxicol. Environ. Health B Crit. Rev 23, 27–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Guo X, Seo JE, Petibone D, Tryndyak V, Lee UJ, Zhou T, Robison TW, Mei N, 2020b. Performance of HepaRG and HepG2 cells in the high-throughput micronucleus assay for in vitro genotoxicity assessment. J. Toxicol. Environ. Health A 83, 702–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Habas K, Brinkworth MH, Anderson D, 2017. In vitro responses to known in vivo genotoxic agents in mouse germ cells. Environ. Mol. Mutagen 58, 99–107. [DOI] [PubMed] [Google Scholar]
  25. Hengstler JG, Utesch D, Steinberg P, Platt KL, Diener B, Ringel M, Swales N, Fischer T, Biefang K, Gerl M, Bottger T, Oesch F, 2000. Cryopreserved primary hepatocytes as a constantly available in vitro model for the evaluation of human and animal drug metabolism and enzyme induction. Drug Metab. Rev 32, 81–118. [DOI] [PubMed] [Google Scholar]
  26. Hotchkiss CE, Bishop ME, Dertinger SD, Slikker W Jr., Moore MM, Macgregor JT, 2008. Flow cytometric analysis of micronuclei in peripheral blood reticulocytes IV: an index of chromosomal damage in the rhesus monkey (Macaca mulatta). Toxicol. Sci 102, 352–358. [DOI] [PubMed] [Google Scholar]
  27. IARC, 1999. Re-evaluation of some organic chemicals, hydrazine and hydrogen peroxide (Part 1, Part 2, Part 3). In: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, vol. 71 (Accessed 1 September 2021). https://publications.iarc.fr/Book-And-Report-Series/Iarc-Monographs-On-The-Identification-Of-Carcinogenic-Hazards-To-Humans/Re-evaluation-Of-Some-Organic-Chemicals-Hydrazine-And-Hydrogen-Peroxide-Part-1-Part-2-Part-3-1999. [PMC free article] [PubMed] [Google Scholar]
  28. Jorge-Nebert LF, Jiang Z, Chakraborty R, Watson J, Jin L, McGarvey ST, Deka R, Nebert DW, 2010. Analysis of human CYP1A1 and CYP1A2 genes and their shared bidirectional promoter in eight world populations. Hum. Mutat 31, 27–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Kanojia D, Vaidya MM, 2006. 4-nitroquinoline-1-oxide induced experimental oral carcinogenesis. Oral Oncol. 42, 655–667. [DOI] [PubMed] [Google Scholar]
  30. Kirkland D, Kasper P, Martus HJ, Muller L, van Benthem J, Madia F, Corvi R, 2016. Updated recommended lists of genotoxic and non-genotoxic chemicals for assessment of the performance of new or improved genotoxicity tests. Mutat. Res 795, 7–30. [DOI] [PubMed] [Google Scholar]
  31. Le Hegarat L, Dumont J, Josse R, Huet S, Lanceleur R, Mourot A, Poul JM, Guguen-Guillouzo C, Guillouzo A, Fessard V, 2010. Assessment of the genotoxic potential of indirect chemical mutagens in HepaRG cells by the comet and the cytokinesis-block micronucleus assays. Mutagenesis 25, 555–560. [DOI] [PubMed] [Google Scholar]
  32. Le Hegarat L, Mourot A, Huet S, Vasseur L, Camus S, Chesne C, Fessard V, 2014. Performance of comet and micronucleus assays in metabolic competent HepaRG cells to predict in vivo genotoxicity. Toxicol. Sci 138, 300–309. [DOI] [PubMed] [Google Scholar]
  33. Li H, van Berlo D, Shi T, Speit G, Knaapen AM, Borm PJ, Albrecht C, Schins RP, 2008. Curcumin protects against cytotoxic and inflammatory effects of quartz particles but causes oxidative DNA damage in a rat lung epithelial cell line. Toxicol. Appl. Pharmacol 227, 115–124. [DOI] [PubMed] [Google Scholar]
  34. Lin Y, Yao Y, Liu S, Wang L, Moorthy B, Xiong D, Cheng T, Ding X, Gu J, 2012. Role of mammary epithelial and stromal P450 enzymes in the clearance and metabolic activation of 7,12-dimethylbenz(a)anthracene in mice. Toxicol. Lett 212, 97–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Liu W, Xi J, Cao Y, You X, Chen R, Zhang X, Han L, Pan G, Luan Y, 2019. An adaption of human-induced hepatocytes to in vitro genetic toxicity tests. Mutagenesis 34, 165–171. [DOI] [PubMed] [Google Scholar]
  36. Martignoni M, Groothuis GM, de Kanter R, 2006. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin. Drug Metab. Toxicol 2, 875–894. [DOI] [PubMed] [Google Scholar]
  37. Misik M, Nersesyan A, Ropek N, Huber WW, Haslinger E, Knasmueller S, 2019. Use of human derived liver cells for the detection of genotoxins in comet assays. Mutat. Res 845, 402995. [DOI] [PubMed] [Google Scholar]
  38. Mudry MD, Martinez RA, Nieves M, Carballo MA, 2011. Biomarkers of genotoxicity and genomic instability in a non-human primate, Cebus libidinosus (Cebidae, Platyrrhini), exposed to nitroimidazole derivatives. Mutat. Res 721, 108–113. [DOI] [PubMed] [Google Scholar]
  39. NAS, 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington (DC). https://www.nap.edu/catalog/24635/using-21st-century-science-to-improve-risk-related-evaluations (Accessed 1 September 2021). [Google Scholar]
  40. NTP, 1978a. Bioassay of anthranilic acid for possible carcinogenicity. National Cancer Institute Carcinogenesis Technical Report Series No. 36 (Accessed 1 September 2021). https://ntp.niehs.nih.gov/ntp/htdocs/lt_rpts/tr036.pdf. [PubMed] [Google Scholar]
  41. NTP, 1978b. Bioassay of ethionamide for possible carcinogenicity. Cancer Inst. Carcinog. Tech. Rep. Ser 46, 1–107. [PubMed] [Google Scholar]
  42. NTP, 1992. NTP Technical Report on the Toxicology and Carcinogenesis Studies of Resorcinol (CAS No. 108-46-3) in F344/N Rats and B6C3F1 Mice (Gavage Studies). NTP TR403 https://ntp.niehs.nih.gov/ntp/htdocs/lt_rpts/tr403.pdf?utm_source=direct&utm_medium=prod&utm_campaign=ntpgolinks&utm_term=tr403 (Accessed 1 September 2021). [PubMed] [Google Scholar]
  43. NTP, 1993. NTP toxicology and carcinogenesis studies of turmeric oleoresin (CAS No. 8024-37-1) (major component 79%−85% curcumin, CAS No. 458-37-7) in F344/N rats and B6C3F1 mice (feed studies). Toxicol. Program Tech. Rep. Ser 427, 1–275. [PubMed] [Google Scholar]
  44. OECD, 2015. Guidance document on revisions to OECD genetic toxicology test guidelines. OECD Workgroup of National Coordinators for Test 42 Guidelines (WNT) (Accessed 1 September 2021). https://www.oecd.org/chemicalsafety/testing/Genetic%20Toxicology%20Guidance%20Document%20Aug%2031%202015.pdf. [Google Scholar]
  45. Oldham JW, D.A., C JA, F, 1979. The isolation and primary culture of viable, nonproliferating rat hepatocytes. TCA Manual 5, 1047–1050. [Google Scholar]
  46. Peng C, Arthur D, Liu F, Lee J, Xia Q, Lavin MF, Ng JC, 2013. Genotoxicity of hydroquinone in A549 cells. Cell Biol. Toxicol 29, 213–227. [DOI] [PubMed] [Google Scholar]
  47. Rao VK, Knutsen T, Ried T, Wangsa D, Flynn BM, Langham G, Egorin MJ, Cole D, Balis F, Steinberg SM, Bates S, Fojo T, 2005. The extent of chromosomal aberrations induced by chemotherapy in non-human primates depends on the schedule of administration. Mutat. Res 583, 105–119. [DOI] [PubMed] [Google Scholar]
  48. Redon CE, Nakamura AJ, Gouliaeva K, Rahman A, Blakely WF, Bonner WM, 2010. The use of gamma-H2AX as a biodosimeter for total-body radiation exposure in non-human primates. PLoS One 5, e15544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Rodriguez JS, Morris SM, Hotchkiss CE, Doerge DR, Allen RR, Mattison DR, Paule MG, 2010. The effects of chronic methylphenidate administration on operant test battery performance in juvenile rhesus monkeys. Neurotoxicol. Teratol 32, 142–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Sebastia N, Soriano JM, Barquinero JF, Villaescusa JI, Almonacid M, Cervera J, Such E, Silla MA, Montoro A, 2012. In vitro cytogenetic and genotoxic effects of curcumin on human peripheral blood lymphocytes. Food Chem. Toxicol 50, 3229–3233. [DOI] [PubMed] [Google Scholar]
  51. Seo JE, Tryndyak V, Wu Q, Dreval K, Pogribny I, Bryant M, Zhou T, Robison TW, Mei N, Guo X, 2019. Quantitative comparison of in vitro genotoxicity between metabolically competent HepaRG cells and HepG2 cells using the high-throughput high-content CometChip assay. Arch. Toxicol 93, 1433–1448. [DOI] [PubMed] [Google Scholar]
  52. Seo JE, Wu Q, Bryant M, Ren L, Shi Q, Robison TW, Mei N, Manjanatha MG, Guo X, 2020. Performance of high-throughput CometChip assay using primary human hepatocytes: a comparison of DNA damage responses with in vitro human hepatoma cell lines. Arch. Toxicol 94, 2207–2224. [DOI] [PubMed] [Google Scholar]
  53. Seo JE, Guo X, Petibone DM, Shelton SD, Chen Y, Li X, Tryndyak V, Smith-Roe SL, Witt KL, Mei N, Manjanatha MG, 2021. Mechanistic evaluation of black cohosh extract-induced genotoxicity in human cells. Toxicol. Sci 182, 96–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Severin I, Jondeau A, Dahbi L, Chagnon MC, 2005. 2,4-Diaminotoluene (2,4-DAT)-induced DNA damage, DNA repair and micronucleus formation in the human hepatoma cell line HepG2. Toxicology 213, 138–146. [DOI] [PubMed] [Google Scholar]
  55. Shah UK, Seager AL, Fowler P, Doak SH, Johnson GE, Scott SJ, Scott AD, Jenkins GJ, 2016. A comparison of the genotoxicity of benzo[a]pyrene in four cell lines with differing metabolic capacity. Mutat. Res. Genet. Toxicol. Environ. Mutagen 808, 8–19. [DOI] [PubMed] [Google Scholar]
  56. Shah UK, Mallia JO, Singh N, Chapman KE, Doak SH, Jenkins GJS, 2018. A three-dimensional in vitro HepG2 cells liver spheroid model for genotoxicity studies. Mutat. Res 825, 51–58. [DOI] [PubMed] [Google Scholar]
  57. Shi J, Krsmanovic L, Bruce S, Kelly T, Paranjpe M, Szabo K, Arevalo M, Atta-Safoh S, Debelie F, LaForce MK, Sly J, Springer S, 2011. Assessment of genotoxicity induced by 7,12-dimethylbenz(a)anthracene or diethylnitrosamine in the Pig-a, micronucleus and Comet assays integrated into 28-day repeat dose studies. Environ. Mol. Mutagen 52, 711–720. [DOI] [PubMed] [Google Scholar]
  58. Slesinski RS, Guzzie PJ, Putman DL, Ballantyne B, 1988. In vitro and in vivo evaluation of the genotoxic potential of 2-ethyl-1,3-hexanediol. Toxicology 53, 179–198. [DOI] [PubMed] [Google Scholar]
  59. Slob W, 2014. Benchmark dose and the three Rs. Part I. Getting more information from the same number of animals. Crit. Rev. Toxicol 44, 557–567. [DOI] [PubMed] [Google Scholar]
  60. Soeteman-Hernandez LG, Johnson GE, Slob W, 2016. Estimating the carcinogenic potency of chemicals from the in vivo micronucleus test. Mutagenesis 31, 347–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Thorgeirsson SS, Davis CD, Schut HA, Adamson RH, Snyderwine EG, 1995. Possible relationship between tissue distribution of DNA adducts and genotoxicity of food-derived heterocyclic amines. Princess Takamatsu Symp. 23, 85–92. [PubMed] [Google Scholar]
  62. Uehara S, Murayama N, Nakanishi Y, Zeldin DC, Yamazaki H, Uno Y, 2011. Immunochemical detection of cytochrome P450 enzymes in liver microsomes of 27 cynomolgus monkeys. J. Pharmacol. Exp. Ther 339, 654–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Ulrich RG, Aspar DG, Cramer CT, Kletzien RF, Ginsberg LC, 1990. Isolation and culture of hepatocytes from the cynomolgus monkey (Macaca fascicularis). In Vitro Cell. Dev. Biol 26, 815–823. [DOI] [PubMed] [Google Scholar]
  64. Uno Y, Uehara S, Yamazaki H, 2016. Utility of non-human primates in drug development: comparison of non-human primate and human drug-metabolizing cytochrome P450 enzymes. Biochem. Pharmacol 121, 1–7. [DOI] [PubMed] [Google Scholar]
  65. Wang Z, Chen Y, Drbohlav LM, Wu JQ, Wang MZ, 2016. Development of an in vitro model to screen CYP1B1-targeted anticancer prodrugs. J. Biomol. Screen 21, 1090–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Wang Y, Revollo J, McKinzie P, Pearce MG, Dad A, Yucesoy B, Rosenfeldt H, Heflich RH, Dobrovolsky VN, 2018. Establishing a novel Pig-a gene mutation assay in L5178YTk(+/−) mouse lymphoma cells. Environ. Mol. Mutagen 59, 4–17. [DOI] [PubMed] [Google Scholar]
  67. White PA, Johnson GE, 2016. Genetic toxicology at the crossroads-from qualitative hazard evaluation to quantitative risk assessment. Mutagenesis 31, 233–237. [DOI] [PubMed] [Google Scholar]
  68. White PA, Long AS, Johnson GE, 2020. Quantitative Interpretation of genetic toxicity dose-response data for risk assessment and regulatory decision-making: current status and emerging priorities. Environ. Mol. Mutagen 61, 66–83. [DOI] [PubMed] [Google Scholar]
  69. Wilkinson JD, Callicott R, Salminen WF, Sandhu SK, Greenhaw J, Paredes A, Davis K, Jones Y, Paule MG, Slikker W Jr., Rusconi PG, Czachor J, Bodien A, Westphal JA, Dauphin DD, Lipshultz SE, 2019. A randomized controlled laboratory study on the long-term effects of methylphenidate on cardiovascular function and structure in rhesus monkeys. Pediatr. Res 85, 398–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Wills JW, Long AS, Johnson GE, Bemis JC, Dertinger SD, Slob W, White PA, 2016. Empirical analysis of BMD metrics in genetic toxicology part II: in vivo potency comparisons to promote reductions in the use of experimental animals for genetic toxicity assessment. Mutagenesis 31, 265–275. [DOI] [PubMed] [Google Scholar]
  71. Wills JW, Johnson GE, Battaion HL, Slob W, White PA, 2017. Comparing BMD-derived genotoxic potency estimations across variants of the transgenic rodent gene mutation assay. Environ. Mol. Mutagen 58, 632–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Yusuf AT, Vian L, Sabatier R, Cano JP, 2000. In vitro detection of indirect-acting genotoxins in the comet assay using Hep G2 cells. Mutat. Res 468, 227–234. [DOI] [PubMed] [Google Scholar]
  73. Zeilinger K, Freyer N, Damm G, Seehofer D, Knospel F, 2016. Cell sources for in vitro human liver cell culture models. Exp. Biol. Med. (Maywood) 241, 1684–1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Zeller A, Tang L, Dertinger SD, Funk J, Duran-Pacheco G, Guerard M, 2016. A proposal for a novel rationale for critical effect size in dose-response analysis based on a multi-endpoint in vivo study with methyl methanesulfonate. Mutagenesis 31, 239–253. [DOI] [PubMed] [Google Scholar]
  75. Zhang X, Newport GD, Callicott R, Liu S, Thompson J, Berridge MS, Apana SM, Slikker W Jr., Wang C, Paule MG, 2016. MicroPET/CT assessment of FDG uptake in brain after long-term methylphenidate treatment in nonhuman primates. Neurotoxicol. Teratol 56, 68–74. [DOI] [PubMed] [Google Scholar]
  76. Zhang X, Talpos J, Berridge MS, Apana SM, Slikker W Jr., Wang C, Paule MG, 2021. MicroPET/CT assessment of neurochemical effects in the brain after long-term methylphenidate treatment in nonhuman primates. Neurotoxicol. Teratol 87, 107017. [DOI] [PubMed] [Google Scholar]

RESOURCES