1. Introduction
The skin is the largest organ and the first to come into contact with environmental carcinogens, either incidentally, such as through exposure to household chemicals, or via the application of topical agents such as cosmetics and sunscreens. In spite of the best efforts of regulatory agencies to ensure consumer safety, the sheer volume of chemicals in everyday products makes it impractical to screen them effectively by using low-throughput models. Roughly 12,500 cosmetics are on the US market at any given time [
1]. Furthermore, at a replacement rate of roughly 25–30%, 3000 to 4000 cosmetics need to be tested each year. Most of the 85,000 chemicals commonly used in the US, outside of drugs and pesticides, and including cosmetics, personal products, household cleaners, food, fabric, and children’s toys, have not been tested for their effects on human health. More broadly, 10 million chemicals are currently being produced each year with little or no analysis of health or environmental impact [
2]. Where there has been any testing at all, it has been historically performed on animals, in conjunction with other assays. These have been used to generate databases, including those of the National Toxicology Program (NTP) (available online:
https://ntp.niehs.nih.gov/data/index.html?utm_source=direct&utm_medium=prod&utm_campaign=ntpgolinks&utm_term=datasearch (accessed on 27 February 2023)) and the International Agency for Research on Cancer (IARC; available online:
https://publications.iarc.fr/Databases/Iarc-Cancerbases (accessed on 27 February 2023)). However, animal testing has been phased out in Europe and has recently been prohibited in some US states. Even assuming that most of these chemicals are safe, overlooking certain chemicals because of lack of testing or political will has had major public health and ecological consequences, as in the cases of arsenic, asbestos, benzene, bisphenol A (BPA), chromium hexavalent compounds, dioxins, formaldehyde, polybrominated diphenyl ethers (PBDEs), polycyclic aromatic hydrocarbons (PAH), and vinyl chloride. A number of these chemical agents are both genotoxic and carcinogenic [
3]. Accumulated DNA damage and misrepair results in mutations, most of which are inconsequential. However, damage can also result in the accrual of deleterious mutations and ultimately tumorigenesis when oncogenes or tumor suppressor genes are altered [
4]. Thus, the rationale for the well-known Ames mutation test is precisely due to the fact that a large proportion of genotoxic agents are also carcinogens. Another screening challenge is that once a single compound has been approved, it no longer needs to be tested in combination with other compounds, which may act synergistically with each other to generate genotoxic combinations.
There are several established methods for measuring DNA damage and resulting mutations. Since rodent models are not practical, in vitro genotoxicity can be measured using several different techniques, including the aforementioned Ames test, mouse lymphoma assay, and in vitro micronucleus and chromosomal aberrations test. Higher sensitivities could be achieved using combinations of these assays [
5]. The prokaryote-based Ames test employs a Salmonella typhimurium reverse mutation assay (
his− to
his+) [
6]. The mouse lymphoma assay similarly measures gene mutation, usually TK, in eukaryotes [
7]. Some of these assays can be time-consuming and can yield a high proportion of false-positive results [
5]. In contrast, single-cell gel electrophoresis (SCGE), or the Comet, is now internationally recognized as a simple and inexpensive method for the detection of different types of DNA damage, including single- and double-stranded breaks, DNA adducts, crosslinks, and alkaline-labile sites, and has been used in a low-throughput format until recently [
8,
9,
10,
11,
12,
13,
14,
15,
16,
17,
18].
While DNA damage can occur in any organ, the skin withstands particularly relentless assaults from the environment and has evolved several mechanisms to protect its basal layer from DNA damage and mutation. The outer anucleate cornified skin layers serve as a first line of defense, protecting the proliferating basal cells that are resident in the lowest layer of the epidermis; protection is paramount, as mutations in stem and basal cells are necessary for tumorigenesis. Furthermore, UVA (320–400 nm) and some genotoxic chemicals can penetrate to the dermis, while other chemicals may leach into the bloodstream, as in the case of sunscreen active ingredients such as oxybenzone (clinical trial NCT03582215) [
19]. Additionally, the skin contains xenobiotic-metabolizing enzymes, including three type I cytochrome P450s (
CYPs), namely CYP1A1, 1A2, and 1B1, which oxidize carcinogens, forming intermediates that can react with DNA to form mutagenic adducts [
20]. Thus, it is important to have a reliable high-throughput method to determine the skin genotoxicity of chemicals that are likely to come into contact with humans. The need for data that can be extrapolated to humans has generated interest in organotypic human skin models that recapitulate biochemical and morphological properties of human skin, including those related to absorption and drug metabolism. Numerous endpoints have been validated and adopted, including corrosion, irritation, and sensitizing assays. Currently, there are no standardized assays to assess genotoxicity in the skin. Part of the difficulty lies in the nature of skin, which, like lens fiber and reticulocytes, contains anucleate terminally differentiated cells. Thus, while different assays are available for the screening of DNA breaks, these methods are complicated in mature skin, since the degradation of DNA is part of the normal process of epidermal differentiation, resulting in a well-organized stratified squamous epithelium, enabling its barrier function against environmental insult. It is in this context of differentiated epidermis that environmental carcinogens act on the skin. Thus, there has been growing interest in developing a reliable skin comet assay.
We combined the advantages of CometChip with those of organotypic culture. The ability to grow human basal-like cells in specialized low calcium medium containing growth factors and specific concentrations of retinoids allows for the investigation of both the short- and long-term effects of DNA damage on keratinocytes. However, the response of monolayer cells does not replicate the in vivo response to toxic insult. On the other hand, organotypic and/or xenograft skin cultures on immune-compromised mice have successfully recapitulated the response of the skin to toxic agents and irradiation, including signature cytokine, metabolomic, and transcriptomic profiles [
21]. Among other factors, these differences may be explained by differences in p21Cip1 cyclin-dependent kinase inhibitor levels inducing apoptosis in monolayers, as well as altered differentiation in the organotypic culture, with “bystander” cells contributing to the latter effect [
21]. However, this creates a conundrum: under normal growth conditions, monolayer cells are “basal-like” but lose their in vivo response to toxins, while organotypic cultures, such as human epidermis, lose their nuclei during differentiation, creating a heterogeneous background of DNA damage in published reports of skin comets [
22]. Secondly, it has long been known that proliferating basal cells have a more robust DNA-repair response than differentiated keratinocytes derived from the same epidermis [
23]. The third problem is the identification of the cell type in the skin that has sustained damage, as full-thickness skin cultures contain both fibroblasts and keratinocytes.
This platform thus adds three new parameters to the Comet assay: higher throughput, cell-type isolation and identification, and increased accuracy for DNA-damage measurements. The uniqueness of this model is thus the use of an organotypic human skin model that it more accurately recapitulates the response of human skin than other cell models of genotoxicity currently used for screening and the ability to isolate basal cells, the target of genotoxic and carcinogenic agents, and precisely and rapidly measure their DNA damage in a high-throughput CometChip format.
2. Materials and Methods
2.1. Cells
E6/E7-immortalized or HaCaT keratinocytes were maintained in DMEM media supplemented with 10% FBS and 1% of Penicillin–Streptomycin (ThermoFisher, Waltham, MA, USA, 10,000 U/mL). Conditionally reprogrammed keratinocytes (CRC-HFK) were maintained in EpiX medium according to the manufacturer’s protocol (Propagenix, Gaithersburg, MD, USA). Human Foreskin Keratinocytes (HFKs) were kind gifts from Richard Schlegel at Georgetown University Medical Center and were co-cultured in FY medium (3 parts of DMEM containing 10% FBS, 1% of Penicillin–Streptomycin, 1% of 100X glutamine, and 1 part of F12 nutrient mixture) plus Hydrocortisone/EGF (1:1000), insulin (4 µg/mL), Gentamicin (10 μg/mL), Fungizone (250 ng/mL), 0.1 nM cholera toxin, and 10 μM ROCK Inhibitor Y-27632. Jurkat T cells were cultured in RPMI supplemented with 10% FBS and 1% of Penicillin–Streptomycin. All cells were cultured at 37 °C in a 5% CO2 cell incubator.
2.2. Skin Organotypic Cultures
Full-thickness skin equivalents (EFT-400, MatTek, Ashland, MA, USA) and epidermal equivalents (EPI-201) were purchased from MatTek in 24-well formats, while EpiX-HFK skin equivalents were generated in our lab. MatTek tissue was maintained in DMEM for 3 days prior to exposure to compounds. Dissociated skin equivalents or detached monolayer cells were blocked with 6% BSA in PBS for 30 min and then hybridized with biotin-conjugated anti-human integrin β1 (1:200 Miltenyi Biotec cat# 130-101-262) for 1 h at room temperature. After 3 PBS washes, cells were hybridized with Qdot™ 655 Streptavidin Conjugate (20 nM, Invitrogen cat# Q10121MP) for 1 h. Cells were washed with PBS 3 times and resuspended in cell culture media prior to loading into the CometChip.
2.3. Immunostaining
For chamber slide staining, 30 K cells were loaded into each chamber; after attachment, cells were washed and fixed with 4% paraformaldehyde (PFA) for 15 min, permeabilized with 0.5% Triton X-100 (in PBS) for 30 min, and then hybridized with anti-human integrin β11 antibody and fluorescein-conjugated phalloidin (1:40, F432, ThermoFisher Scientific) overnight at 4 °C. The following day, cells were PBS-washed 3X, and incubated with Qdot™ 655 Streptavidin Conjugate (20 nM) for 1 h, along with DAPI (1:2000, D1306, Invitrogen) to visualize the nucleus. For the 3D skin tissue stain, after treatment with compounds, the membranes in culture inserts were removed, paraffin-embedded, and sectioned according to standard protocols by Histopathology & Tissue Shared Resource at Georgetown University Medical Center. For immunofluorescent staining, sections were deparaffinized in xylene for 5 min 2 times and then rehydrated in 100%, 90%, 70%, and finally 30% ethanol for 1 min each. Then antigen retrieval was performed using 10 cycles of microwave boiling and cooling. After a brief PBS wash, tissues were permeabilized with 0.2% glycine/PBS for 15 min and then blocked with SuperBlockTM Buffer (ThermoFisher cat #37515) for 1 h. Tissues were incubated overnight at 4 °C, using CYP1A1 (Abcam, Cambridge, UK, Ab3568, 1:1000) or rabbit Anti-CYP1B1 antibody Ab33586 1:1000). The next day, after three PBS washes, the tissue was hybridized with corresponding secondary antibody at room temperature for 1 h: Alexa-488-conjugated goat anti-mouse IgG - (Abcam Ab150113, 1:500) or Goat anti-rabbit IgG -Alexa 594 conjugated (Invitrogen # A32740, 1:500). DAPI (Invitrogen, Waltham, MA, USA) stain was used to visualize the nucleus.
2.4. Treatment of Cells with Compounds
A total of 50K cells/well were loaded into the comet chips. After 30 min of gravity loading, as previously described [
24], cells were treated with NTPs compounds or known DNA-damage agents at indicated concentration for 1 or 3 h. DMSO was used as a negative control.
2.5. Cell Viability Assays
First, 104 cells were seeded into 96-well culture plates. After 16 h to allow attachment, the cells were treated with 200 µM of each compound and exposed for 3 h at 37 °C in a 5% CO2 cell incubator. Compounds were then removed, the cells were washed with PBS, and the original culture medium was added back. Measurements were conducted 24 h after exposure, using an XTT Cell Viability Assay Kit (Biotium, Fremont, CA, USA) according to the manufacturer’s instructions, using Absorbance 450- Absorbance 650 at 0, 1, 2, and 3 h. To calculate % cell viability, the absorbance/time slopes were determined from four time points, as mentioned, and % cell viability = (the slope of samples/the slope of DMSO- or untreated).
2.6. CometChip Assay and Analysis
CometChips were cast using a mixture of 1% agarose and 1 mg/mL collagen (Gibco, Bovine Collagen I); all other physical characteristics remain as previously described [
24]. The collagen added to the CometChip allows for the binding of only β1-integrin-expressing basal keratinocytes. The comet chip assay system (R&D system) was used according to manufacturer’s instructions. In our previous study, microwells that were 30 µM diameter were able to trap most of the cells. Gels comprised agarose (0.8% w/v) or agarose containing 1 mg/mL of collagen I (Gibco, New York, NY, USA, A1048301), as follows: First, 1.6% NA (normal agarose (Invitrogen, #16500-100) was dissolved in 100 mL 1X PBS pH 7.4 (Invitrogen, 10010-049) in a 250 sterile flask, using a microwave, and adding sterile water until the original mass is restored. The temperature of the gel was then equilibrated to 55 °C in a water bath before mixing with 2 mg/mL collagen I. For long-term storage, 30 mL aliquots can be stored in 50 mL conical tubes at 4 °C. A 2 mg/mL collagen I solution was prepared separately in a 10 mL conical tube. Then 4 mL of sterile dH
2O was added to 1 mL 10X PBS. The mixture was heated to 90 °C, and then 4 mL of 5 mg/mL rat-tail collagen I (Gibco, cat# 1048301) was added and mixed by inversion. The solution was cooled to room temperature, and the pH was adjusted to 7.4, using fresh 7.5% Sodium Bicarbonate (60–240 µL, verified by a pH meter). The temperature was equilibrated to 55 °C before mixing with the 1.6% agarose solution. For long-term storage, the 2 mg/mL collagen I can be stored at 4 °C. In a 50 mL conical tube, 7 mL of 2 mg/mL collagen I (equilibrated to 55 °C) was added to 7 mL 1.6% agarose (equilibrated to 55 °C) and poured slowly into the CometChip mold. The gel was allowed to harden for 30 min at room temperature. Cells were counted to achieve 5 × 10
5/mL, and then 100 µL (50K cells) was loaded into each of the 96 macro-wells and trapped by gravitation and integrin–collagen interactions. After 30 min, the cells were exposed to the indicated chemical compounds or positive control DNA damaging agents. All incubations were performed at 37 °C, 5% CO
2. We analyzed comets by using Comet Analysis Software, as described below.
The CometNet software initially makes a grayscale copy of the macro-well stitched image. This allows for the measurement of pixel intensities required for downstream analysis. Next, CometNet does a primary scan of the image with a fixed threshold value to separate the foreground from the background. It then utilizes the “findContours” function from the OpenCV module (Open Source Computer Vision Library;
http://opencv.org (accessed on 27 February 2023) to find all the contours in the image; these are defined as “comet candidates” or potential comets. It further filters the comet candidates via a “for loop” with specified parameters that indicate a “true comet”; parameters include a bounding box that is either square for comets with low damage or rectangular for comets with damage above ~10% damage. Once it finds the candidate comets, the program extracts the bounding box and saves a sub-image of each comet. On the extracted bounding box sub-image, the CometNet performs a further analysis to find the comet head and calculate the key statistics for the individual comet damage profile. The CAS interrogates the selected bounding box for each comet and takes a square region on the leftmost side of the bounding box where the head will be in all comets. This method improves upon the previous CAS ability to find heads that appear detached from comet tails, as the square bounding box that CometNet draws will always include the full head. From here, the program utilizes image morphology techniques, including erosions and dilations and the findContours () function to include the corona shape of the head. It then draws a contour of this corona and fills it in to create a “mask”, which specifies the head for further calculations. The program has a cometStats () function which outputs the width, height, total comet area, and tail area of the comet into a CSV file. The area is the sum of pixel intensities in a contour. The area and intensity of the DNA in the tail is correlative with the amount of DNA damage in the cell.
2.7. qRT-PCR (Quantitative Reverse-Transcriptase-Mediated PCR)
Total RNA was isolated using the RNeasy mini Kit (74104, Qiagen, Venlo, The Netherlands). A total of 1 µg of RNA was converted to cDNA by using the Verso cDNA Synthesis Kit (AB1453A, ThermoFisher Scientific). The qPCR mixture contained the cDNA diluted 50-fold in nuclease-free water.
The primer pairs Human CYP1A1- F: CAAGAGGAGCTAGACACAGTGATT; Human CYP1A1- R: AGCCTTTCAAACTTGTGTCTCTTGT; Human CYP1B1- F: TTCGGCCACTACTCGGAGC; Human CYP1B1- R: AAGAAGTTGCGCATCATGCTG; Human CYP1A2- F: TGGCCTCTGCCATCTTCTG; and Human CYP1A2- R: GGACCCGAGGCCTCAAAC and SYBR Green Master Mix (Applied Biosystems, Waltham, MA, USA) were added, and the signal was detected by a MiniOpticon Real-Time PCR System (BioRad, Hercules, CA, USA). GAPDH served as an internal control. The fold-change was calculated based on the formula 2−△△CT, where △△Ct = [Ct (CYP)TREATMENT − Ct (GAPDH)TREATMENT] – [Ct (CYP)CONTROL− Ct (GAPDH)CONTROL]. Ct = threshold response; CONTROL = solvent alone.
2.8. CYP450 Activity Assays
CYP1A1/CYP1B1 activity was measured using Luciferin-CEE as a substrate, according to the manufacturer’s instructions (P450 Glo Assay, Promega, Madison, WI, USA). Luminescence was detected using an Enspire® Alpha Plate Reader (PerkinElmer, Waltham, MA, USA) and steady-state luminescence plotted. Human S9 Fractions (Catalog number HMS9PL; ThermoFisher) were purchased at a stock concentration of 20 mg/mL and diluted as controls for the CYP1A1/1B1 assay.
2.9. Statistical Analysis
All experiments were performed in at least duplicate, with representative experiments shown. One-way ANOVA was used, with p-values < 0.05, 0.01, 0.001, and 0.0001 considered significant and designated with one, two, three, or four asterisks, respectively.
4. Discussion
To mimic human skin, a 3D EpiDerm culture was used to replace traditional animal experiments, which are incompatible with large-scale screening [
41,
42]. Here, we selected 14 compounds from the NTP library that significantly decreased cell viability and increased DNA damage in HaCaT and NHEK keratinocytes, and then we used the CometChip assays to determine cytotoxicity and genotoxicity when applied topically to the 3D full-thickness skin model. We found that nine compounds significantly reduced cell viability in comparison to the acetone control. On the other hand, for the collagen CometChip, we dissociated the full thickness skin culture and labeled it with integrin β1. By overlapping the SYBR and Qdot655 fluorescent signals, the latter of which we specifically labeled keratinocytes, we could further quantify keratinocyte DNA damage without interference from other cell types. In summary, the pipeline we established is able to identify some keratinocyte-specific genotoxins in the absence of a separate keratinocyte isolation step, which was described previously [
22,
43,
44]. Further studies validated at multiple locations and additional compounds will allow us to establish the accuracy and reproducibility of the test compared to 2D cultures.
We optimized the CometChip assay by incorporating 0.1% collagen to mimic a dermal component of human skin and to selectively capture keratinocytes. This method enabled the CometChip to trap basal and stem cells by protein–protein interactions and obviate interference from certain DNA damage in tissues containing multiple cell types. The traditional CometChip captured and aligned the cells in microwells by gravity loading, as well as the size of the cells. However, multiple cell loading within single wells is an issue that can interfere with post-analysis. In addition, cell clusters dramatically decreased the loading efficiency. We therefore developed a protocol where the cells were immunofluorescently stained with specific surface marker initially and then loaded into the corresponding ligand-coated CometChip, which was collagen in our case. The strong interaction between collagen and integrin-β1-containing heterodimers retains keratinocytes loaded into the microwell. This method can be adapted to any tissue that contains multiple cell types.
After performing the alkaline electrophoresis, the Qdot nanodye was able to maintain the signal to indicate the integrin β1 and further to stain the DNA by SYBR Green/Gold. We designed a program that can recognize double-stained cells and measure the specific DNA damage in keratinocytes. The platform could be a new tool to investigate the DNA damage in the tissue level in a high-throughput format. However, some drawbacks need to be addressed in the future. First, the cellular Qdot labeling process may cause additional DNA damage or repair. It was therefore important to have a suitable control, such as untreated cells, and a positive control, which consisted of cells treated with known DNA-damaging agents, to calibrate the %DNA in tail and tail moments. Furthermore, the nonspecific Qdot stain may lead to difficulty in the post-analysis, thus requiring threshold adjustment. Moreover, the size of microwell was critical to trap certain cells and prevent cells from washing out. We attempted to address these challenges by differential fluorophore labeling and the registration of cells in the CometChip prior to alkaline electrophoresis to assign degrees of DNA damage to specific cell types.
For the cytochrome p450 detection platform, we observed that BaP induces CYP1A1 in HaCaT and HepG2 cells but is constitutively expressed in 3D skin equivalents, while CYP1B1 is induced in the latter. Recent studies have suggested that the organotypic 3D cultures could mimic responses of tissues, allowing for a more accurate assessment of CYP expression compared to a 2D culture [
42,
43]. Moreover, the concept of receptor–ligand interaction in CometChip can be utilized to capture the specific cell types, and allow single cell analysis. The platform can be a cell-sorting tool similar to flow cytometry, with the advantage that it requires fewer cells and provides a real-time study without stabilizing and culturing the cells in order to perform subsequent experiments.
A comparison between the three CAS revealed that Opencomet identified a high percentage of the comets present but overestimated DNA damage when the levels were low. In contrast, the Trevigen CAS was more sensitive and could detect and quantify low levels of DNA damage but was more problematic when DNA-damage levels were above 50%. CometNet was able to find and quantitate low levels of DNA damage and was also able to maintain linearity over a greater range.
Several labs recently collaborated by using organotypic 3D human epidermal skin models, including EpiDerm™ (MatTek, Ashland, MA, USA) and Phenion FT (Henkel, Düsseldorf, Germany), that were exposed to test chemicals, and dissociated cells subjected to a comet assay [
22,
43,
44]. However, in an attempt to overcome the problem of multiple cell types, the recently published studies required the isolation of keratinocytes and fibroblasts prior to the comet assay, a rigorous protocol that introduces unwanted assay artifacts [
44]. Secondly, while the second study by the same group employed solvent controls for background DNA damage [
43], a later study [
44] did not. Some of these studies showed a dose-dependent increase in DNA damage with increased concentrations of genotoxic agents. However, a high background of keratinocyte-differentiation-associated damage was originally observed, with unacceptably high levels of DNA damage and resultant comet tail DNA in 25% of unexposed cells, limiting sensitivity, accuracy, and adaptability for a high-capacity format that will be necessary for screening large numbers of potentially genotoxic compounds [
22]. Third, the addition of aphidicolin, an inhibitor of DNA polymerases α, δ, and ε, was used to observe damage in the latter studies [
43,
44], thus inhibiting both replication and hence also replication-associated DNA-repair pathways [
45], and partially negating the use of 3D cultures that have separate proliferating and differentiating compartments. Finally, the microwells were used in low-throughput format comet slides. In the current study, we showed that we can overcome these limitations by (1) utilizing an Immuno-CometChip, (2) developing new software to ensure simultaneous measurement of basal cell markers and DNA damage, (3) including internal controls for DNA damage, and (4) measuring levels of skin cytochrome p450 activity that can convert procarcinogens to carcinogens. Some aspects of the previous skin comet were improved by simultaneously isolating and visualizing DNA damage in basal cells by using integrin β1 as a marker and collagen-binding receptor when complexed with the α2 integrin subunit. We also enhanced the assay by adapting a gel with 96 macro-wells, each containing roughly 400 microwells in a grid to enhance reproducibility and capacity (~40K cells at once), as described previously [
24]. We varied a number of parameters, including the microwell diameter and depth and the washing stringencies. Several controls were employed to monitor success, including the use of cryopreserved cells to measure against standardized levels of DNA damage, and monitor the purity of basal cell isolation by using biotin-labeled anti-β1 integrins and Qdot® 655 streptavidin conjugate, which consists of semi-conductive nanoparticles that fluoresce under blue light. Once deposited, they can be visualized following the completion of the assay, reducing analysis time and allowing for the visualization of results. Our modified automated software was used to quantify levels of DNA damage only in basal cells. We were able to begin to validate the final Immuno-CometChip assay in the presence of several household chemicals obtained from Tox21, which might need activation via CYP enzymes. In the presented analysis, we avoided going into detail about the potential molecular mechanisms of each of the tested compounds. Many of the compounds used in this research were also used in previous publications of ours, in particular, Sykora et al. [
24]. In this report we focused on the technology and the utility of the assay on different biological materials.
To conclude, we developed the Immuno-CometChip and protocols that can be utilized to measure DNA damage at tissue level, providing a high throughput platform to isolate epidermal basal and stem cells from complex mixtures of cells in organotypic culture, and to simultaneously quantify DNA damage.