APAP concentrations in female reproductive tract
All materials were sampled at the Copenhagen University Hospital, Denmark, and TFP Stork Fertility clinic, Denmark, in accordance with relevant guidelines and regulations and after consent from the regional scientific ethical committee of the Capital Region of Denmark (protocol nr.: 17003845). All material was anonymised and collected as part of standard clinical procedures.
Follicular fluid: Follicular fluid was collected from a total of 26 women during transvaginal ultrasound guided aspiration of the pre-ovulatory follicles. The procedure is part of standard fertility treatment with the aim of collecting oocytes, termed oocyte pickup (OPU). As a by-product of OPU, follicular fluid is recovered but is routinely discarded. Each patient scheduled for OPU receive, as part of pain-management 1 gram of APAP 1 hr before OPU. Collected follicular fluids were centrifuged and stored at -20°C until analysis.
Endometrial tissue and uterine fluid: Endometrial tissue and uterine fluid were collected from 7 women as part of an endometrial scratching (ES) procedure. ES is performed as part of fertility treatment to patients with recurrent implantation failure and has been suggested to improve chances of implantation41,42. Endometrial tissue and uterine fluid are recovered during ES but is routinely discarded. Each patient scheduled for ES received as part of pain-management 1 gram of APAP 1 hr before the procedure. Following intrauterine placement of an inner and outer biopsy catheter a small amount of suction was applied. The inner catheter was removed and brought to the laboratory for collection of uterine fluid. A new inner catheter was then positioned, and endometrial biopsy/scratching was performed by moving the catheter while applying strong suction with a syringe. In the laboratory the initial inner catheter was rinsed with 0.4 ml of sterile saline to recover uterine fluid. Fluid from the first inner catheter and endometrial tissue from the second catheter was stored at -20°C until analysis.
Liquid chromatography–mass spectrometry (LC-MS/MS) measurements for APAP: For measurements of APAP in follicular fluid, 200 uL aliquots of follicular fluid, calibration standards (10 solutions of native APAP standard diluted in water in the concentration range from 0.5-2000 ng/mL) and control materials (native APAP spiked in urine pool in three different concentrations) were added 20 µL of internal standard solution followed by 276 µL NH4Ac-buffer. Immediately before enzymatic de-conjugation, all sample extracts calibration and control materials were added 10 µL freshly prepared enzyme mixture (ß-glucuronidase from Escherichia coli K12), sulfatase from Aerobacter aerogenes, and NH4Ac-buffer; 1:1:3), mixed and incubated at 37ºC for 3 h, stored overnight at -20ºC and then centrifugated at 4ºC for 10 min. Supernatants were transferred to HPLC vials and were then ready for analysis.
For uterine fluid and endometrial tissues, APAP were extracted from approximately 100 mg of uterine fluid (98.9-120 mg) and endometrial tissue (35.2-141 mg) samples following a validated method for extraction of chemicals43. Briefly, samples were added 20 µL of internal standard solution (containing 200 ng/mL of APAP-d4 and 100 ng/mL 13C4-methylumbelliferone including 4-methylumbelliferyl b-D-glucuronide and 4-methylumbelliferyl-b-D-sulfate dissolved in 50% methanol), then centrifuged and stored at RT for 30 min. Samples were submerged by addition of 1 mL acetone and mechanically homogenized with a mixer. The mixer was then washed with 2 mL methanol per extract, which was collected and added to the homogenized extract. Then the extracts were sonicated in an ultrasound bath for 10 min. Subsequently, total extract volumes were reduced to < 2 mL each by evaporation under a gentle nitrogen stream at RT. Thereafter tissue residuals were removed by transferring the remaining extracts to a 2 mL Eppendorf tubes followed by storing at -20ºC for 15 min and then centrifugation at 4ºC for 10 min. Supernatants were then transferred to new glass tubes, evaporated to dryness under a stream of nitrogen and re-suspended in 496 µL 0.5 M ammonium acetate (NH4Ac) buffer (pH5.5).
The total (free and conjugated) content of APAP in the sample extracts were measured by isotope diluted online-TurboFlow-liquid chromatography mass spectrometry (LC-MS/MS) using a Thermo Scientific Aria TLX-1 LC system coupled to a TSQ Ultra triple quadrupole mass spectrometer equipped with a heated electrospray ionization source (HESI) running in positive mode. The instrument was used in combination with Aria operating software 1.6.3 and Xcalibur 2.1.0.1139 system software (ThermoFinnigan, Bellefonte, PA, USA). The TurboFlow-LC systems were equipped with a loading column; TurboFlow Cyclone P column, 0.5 x 50mm (Thermo Scientific) followed by an analytical Gemini-C18 column, 3 µm particle size, 3 x 50 mm (Phenomenex). Prepared batches were kept on the auto-samplers at 10°C. The injection volume was 100 µL. Flow rate and loading and eluting gradients were specified for this specific method and the mobile phases used were loading solvents; A: 10 nM NH4AC, B: 0.1% formic acid in methanol, C: acetone/isopropanol/acetonitrile 10:45:45 and eluting solvents; A: 3 nM ammonium hydrogen carbonate, B: acetonitrile. The method was validated, and limit of detection determined (LOD = 0.48 ng/mL) for urinary APAP as previously described44 according to the ICH guidelines45. The sample extracts were analysed in one batch, also including standards for calibration curves, three blanks and three times three spiked urine pool controls followed by one batch more with samples diluted to fit the method calibration range. The relative standard deviation (RSD) in the three control levels ranged from 2.7-7.6%. For the LC-MS/MS analyses, native APAP (N-acetyl-4-aminophenol, CAS No. 103-90-2 procured from Sigma-Aldrich), labelled APAP-d4 (Paracetamol-D4, CAS No. 64315-36-2 procured from LoGiCal®) and all other reagents and solvents were of analytical, HPLC or MS grade, and all chemicals and laboratory equipment were tested for contamination before utilisation.
Human embryogenesis experiments
All experiments were performed in accordance with relevant guidelines and regulations and conducted at the Copenhagen University Hospital – Hvidovre and Copenhagen University Hospital - Rigshospitalet, Denmark. Surplus human embryos from fertility treatment were donated at Copenhagen University Hospital - Rigshospitalet, Copenhagen University Hospital - Hvidovre, TFP Stork Fertility Clinic, and Copenhagen Fertility Center between the years 2020-2023. The study protocol was approved by the Research Ethics Committee of the Capital Region of Denmark (H-19050437) with signed informed patient consent given prior to donation. A total of 22 cleavage stage embryos and 68 blastocyst stage embryos were used. All experiments were performed with APAP dissolved directly in culture media without any changes in osmolarity (data not shown). APAP was purchased from Sigma cat. no A5000 (Sigma/Merck, Darmstadt, Germany) for these experiments and all subsequent experiments in the study.
Cleavage stage embryos: Cleavage stage embryos were frozen at D2 or D3 after fertilisation and thawed using a slow freeze protocol (Freezekit and ThawKit Cleave Vitrolife, Göteborg, Sweden) as described by the manufacturer (https://www.vitrolife.com/globalassets/support-documents/short-protocols/sp_slow_freeze_cryopreservation_Cleavage.pdf). Following thawing, embryos were placed in equilibrated SAGE 1-Step media (CooperSurgical Fertility Solutions, Ballerup, Denmark) drops covered with mineral oil (Origio, CooperSurgical Fertility Solutions, Ballerup, Denmark) at 37 ºC under 6 % CO2 and 5 % O2. Within 2 h after thawing embryos were matched in pairs and allocated to exposure groups (100 or 200 µM APAP) or control group according to a prioritized order of the following parameters: (i) female origin (sibling embryos), (ii) female age at time of embryo cryopreservation, (iii) time of cryopreservation (D2 or D3), (iv) and number of blastomeres and morphology after thawing. This was done to minimize differences between control and APAP exposed blastocysts. Experiments were performed using a timelapse incubator (EmbryoScope ESD Vitrolife, Göteborg, Sweden). As the specific cryopreservation time of the individual D2 embryos were unknown, the start point depicted in time-lapse videos (Supplemental video 1 and 2) were estimates of embryo age after fertilisation.
Blastocyst stage embryos: Embryos were vitrified at D5 or D6 and subsequent thawed (Vit Kit Freeze NX and Vit Kit®-Warm, Irvine Scientific, Santa Ana, CA, USA) as described by the manufacture (https://www.irvinesci.com/media/IrvineScientific/Resources/0/0/002773_warming_ooctyes_protocol.pdf). Embryos were subsequently transferred to a pre-equilibrated dish with 25 µl drops of SAGE 1-Step culture media (CooperSurgical Fertility Solutions, Ballerup, Denmark) and cultured at 37 ºC under 6 % CO2 and 5 % O2. Within 3 h after warming embryos were matched in pairs and allocated to exposure groups (100 or 200 µM APAP) or control group according to a prioritized order of the following parameters: female origin (sibling blastocysts), female age at time of vitrification, day of vitrification (D5 or D6) and morphology post warming. A maximum of 12 embryos were warmed per experiment by two experienced embryologists to reduce difference in culture time between warming and experiment initiation. Morphology was evaluated by light microscopy (LM) following warming, at start of the experiment, following 3 h of APAP exposure, and at the end of the 6 h culture period using the Gardner grading system46 (Supplemental. Table 1a and 1b).
Whole-mount immunofluorescence staining of human blastocysts stage embryos: Click-iT® Plus EdU Alexa Fluor® 555 Cell Proliferation imaging kit (Thermo Fisher Scientific, Waltham, MA, USA) was used to visualize and quantify newly synthesized DNA. APAP was diluted in equilibrated SAGE 1-Step media and dishes made with 25 µl culture media drops with or without APAP (100 or 200 µM) covered in mineral oil (Origio, CooperSurgical Fertility Solutions, Ballerup, Denmark) and maintained in the incubator for minimum 2 h before experiment start. Embryos were exposed to 100 or 200 µM APAP and control in SAGE 1-Step media for 6 h with the addition of 10µM EdU for the final 3 h. After treatment, the embryos were subjected to a modified procedure of the protocol from Wong (2021)47 combined with the manufacture protocol of the Click-iT® Plus EdU Alexa Fluor® 555 Cell Proliferation imaging kit. In brief, Embryos were fixed individually in Millicell® (Sigma/Merck, Darmstadt, Germany) cell culture inserts in 4% paraformaldehyde (PFA) (VWR chemicals, Radnor, PA, USA) for 15 minutes at room temperature (RT). Next, the embryos were washed twice in 3% BSA in PBS before placed in 0.5% PBSTr for 20 minutes at RT. Again, the embryos were washed twice in 3% BSA in PBS and placed in Click-iT® Plus reaction cocktail for 30 minutes. The embryos were protected from light for the remainder of the protocol and were washed once in 3% BSA in PBS before transferred to neutralization buffer and incubated for 15 minutes at RT. Embryos were then subjected to washes, primary- and secondary antibodies, and mounted as described under “Whole-mount immunofluorescence staining of human and mouse embryos”. Segregation between the ICM and TE fates was determined by the expression of OCT3/4 and CDX237,48. A definable ICM was identified by a clear cluster of cells exclusively expressing OCT3/4. An indefinable ICM was defined by either no OCT3/4 positive cells, OCT3/4 positive cells dispersed throughout the blastocyst stage embryos with no cluster or a cluster of cells expressing both CDX2 and OCT3/4.
Whole-mount immunofluorescence staining of human and mouse embryos: The whole-mount immunofluorescence staining of embryos was performed following a procedure modified from Wong 202147. In brief, preimplantation embryos were fixed in 4% paraformaldehyde (PFA) (VWR chemicals, Radnor, PA, USA) for 15 minutes at RT with human embryos fixed individually using Millicell® (Sigma/Merck, Darmstadt, Germany) cell culture inserts. Next, the embryos were placed in 0.5% PBSTr (0.5% Triton X-100 (Sigma/Merck, Darmstadt, Germany) in PBS) for 20 minutes at RT and then transferred to neutralization buffer (1M Glycine in 0.1% PBSTr) and incubated for 15 minutes at RT. The embryos were then washed three times in 0.01% PBSTw (0.01% Tween-20 (Sigma/Merck, Darmstadt, Germany) in PBS) for 10 minutes at RT. After the last wash, the embryos were transferred to a well containing blocking buffer (3% donkey serum and 1% BSA (Sigma/Merck, Darmstadt, Germany) in PBSTw) and incubated in a humidified box containing wet tissue paper at 4°C overnight. The embryos were then transferred to primary antibodies diluted 1:200 in blocking buffer and incubated for 2-3 days at 4°C. The embryos were then washed 3 times in 0.01% PBSTw for 10 minutes at RT. The embryos were then transferred to a well with secondary antibodies diluted 1:500 in blocking buffer (DAPI diluted 1:5,000 or phalloidin diluted 1:100) and incubated, in the dark, at RT for 3-4 h. Afterwards, the embryos were washed 3 times in 0.01% PBSTw for 10 minutes at RT before being transferred to a coated microscope slide (Dako Agilent, Glostrup, Denmark). The slides were left to dry for a few minutes at RT, in the dark, before addition of 10 μl of mounting medium (90% glycerol and 2% n-propyl gallate in PBS) and mounting with cover glasses (VWR, Radnor, PA, USA)49. The cover glass edges were sealed with transparent nail polish.
Immunofluorescence microscopy and imaging: The immunofluorescence and differential interference contrast (DIC) images were obtained using an Olympus BX63 upright microscope with an Olympus DP72 color, 12.8-megapixel, 4140 Å~3096 resolution camera (Olympus, Tokyo, Japan). Olympus CellSense Dimension software version 1.7 (Olympus, Tokyo, Japan) was used for deconvolution and 3D reconstruction of Z-stacks as previously described50. Images were later processed in ImageJ (Bethesda, MD, USA) and Adobe Photoshop CS6 (Adobe, San Jose, CA, USA). For quantification of the EdU intensity, an outline was drawn around each cell double positive for OCT3/4 and EdU. Only cells or parts of cells that did not overlap with other cells were included. Using the measurement and region of interest (ROI)-function in the CellSens dimension software (Olympus, Tokyo, Japan), the mean red fluorescence intensity was measured along with a background reading. The corrected mean fluorescence for the individual cells were subsequently calculated by subtracting the corresponding background value. The following primary antibodies were used at a 1:200 dilution: rabbit anti-CDX2, #D11D10 (Cell Signaling Technology, Danvers, MA, USA), goat anti-OCT3/4, #sc-8629 (Santa Cruz Biotechnology, Dallas, TX, USA), mouse-GATA4, #sc-25310 (Santa Cruz Biotechnology, Dallas, Tx, USA), and mouse anti-α-Tubulin, #T5168 (Merck/Sigma-Aldrich, Darmstadt, Germany). Secondary antibodies (all from Thermo Fisher Scientific, Waltham, MA, USA) were used at a 1:500 dilution: Donkey-anti-mouse IgG Alexa Fluor® 568, Donkey-anti-rabbit IgG Alexa Fluor® 568, Donkey-anti-goat IgG Alexa Fluor® 488, and Donkey-anti-rabbit IgG Alexa Fluor® 647. F-actin and nuclei were stained with Phalloidin (Alexa Fluor® 488 or 568, Thermo Fisher Scientific, Waltham, MA, USA) and DAPI (Thermo Fisher Scientific, Waltham, MA, USA), respectively.
Mouse models
Mouse pregnancy model: Experiments were performed under license number 2019-15-0201-00175 from the Danish Animal Experiments Inspectorate and under EU directive 2010/63/EU on the protection of animals used for scientific purposes. C57Bl6/J BomTac mice (Taconic, Lille Skensved, Denmark) arrived at the animal facility at 7-8 weeks of age. Experiments were run in two independent cohorts of 24 females and 12 males. Upon arrival, females were housed in twelve boxes of two and males six-by-six. Otherwise, housing and environment was as described previously51. Mice were acclimatized 2 weeks prior to starting the experiments. On the day of experiment initiation, one male was introduced to two dams in one home cage and the following day (day 1) APAP administration was initiated. Female cages were allocated by draw to either 200 mg/kg/day APAP administrated as a single daily oral gavage in tap water or tap water (vehicle control) for a total of 10 days (days 1-10). Females sharing cages received the same treatment and dosing based on body weight on day 0. Males were separated from females on day 5 to avoid further breeding. After end administration at day 10, dams were transferred to clean cages. Dams were euthanized on day 18 and number of fetuses and resorptions counted blindly by two experienced veterinarians. 3 controls and 4 APAP dams were excluded due to lack of pregnancy.
Mouse embryo culture and transfer model: Experiments were performed under license number 2021-15-0201-00851 from the Danish Animal Experiments Inspectorate and under EU directive 2010/63/EU on the protection of animals used for scientific purposes. For the mouse embryo collection, culture, and embryo transfer, inbred C57BL/6JRj females were used as embryo donors and outbred CD1 (RjOrl:SWISS) were used as embryo recipients. Mice were kept in individually ventilated cages at a temperature of 22 °C (±2 °C), with a humidity of 55% (±10%), under 12/12-hr light/dark cycles. Embryos were harvested from prepubescent (4–5-week-old) C57BL/6JRj females. The donor females were subjected to a hormone treatment before mating that consisted of an intraperitoneal injection of 5 IU/female of PMSG (Pregnant Mare Serum Gonadotropin, Prospec, Rehovot, Israel,), followed 47 h later by a second intraperitoneal injection of 5 IU/female of hCG (Human Chorionic Gonadotropin, Chorulon Vet, ref. 422741). After the second injection, each female was set in cross with a C57BL/6JRj stud male. The following morning, mating was monitored by the observation of copulation plugs in the vagina of the females. The day of plug detection was considered embryonic day 0.5 (E0.5). 2-cell-stage embryos (E1.5) were harvested 1.5 and morulae 2.5 dpc. On the day of dissection, pregnant females were sacrificed by cervical dislocation and immediately after the oviducts were dissected and placed on a petri dish containing M2 medium (Sigma/Merck, Darmstadt, Germany, ref. M7167). Embryos were flushed out of the oviduct using a blunt 30G needle attached to a 1 ml syringe filled with M2 medium. Embryos were collected from the eluted medium using a 115 μm diameter glass capillary (Retransferpipette, Biomedical Instruments, Zöllnitz, Germany) attached to a mouth-pipetting system (Mouthpipette, Biomedical Instruments, Zöllnitz, Germany) and washed through three 50 μl drops of M2 medium. Finally, embryos were moved to a fresh petri dish and placed in a 50 µl drop of KSOM medium (Embyomax KSOM + AA, Merck Millipore, Burlington, MA, US, ref. MR-106-D) covered by sterile mineral oil (NidOil, NordicCell, Copenhagen, Denmark, ref. 90142). Embryos were cultured for either 24 or 48 h at 37C, 5% C02, in KSOM only (control groups) or in different concentrations of APAP diluted in KSOM. All experiments were performed with APAP dissolved directly in culture media without any changes in osmolarity (data not shown).
For embryo transfer to pseudo pregnant recipient dams, 10–12-week-old CD1 females in anestrus were stimulated to enter the cycle by setting them in cross with vasectomized CD1 males. Formation of vaginal plug was monitored every morning for three days. Dams that showed a plug on the third day of breeding were selected as pseudo pregnant females and used to perform the embryo transfer of the cultured embryos. The dam was anesthetized by an intraperitoneal injection of 20 mg/ml Avertin from a stock solution of 1g Tribromoethanol (VWR chemicals, Radnor, PA, USA, ref. ACRO421430500) diluted in 630 µl 2-methyl-2-butanol (Sigma/Merck, Darmstadt, Germany, ref. 240486). 120 µl of this stock was diluted up to 10 ml in saline water to obtain the ready-to use solution at a dose of 25 µl/gram. 10-12 embryos were transferred to each oviduct of the pseudo pregnant dam. The embryo transfer was performed according to the standard procedure described in52. Control embryos were always transferred to the right oviduct, and APAP exposed embryos to the left oviduct. 18 days after the transfer (embryonic day E18.5), the pregnant dams were euthanized and dissected to count the number of fetuses developed in each uterine horn.
Human cell culture
Cell culture: HEK293 cells were cultured in high glucose Dulbecco's modified Eagle's medium (DMEM, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS, Thermo Fisher Scientific, Waltham, MA, USA) and 1% penicillin–streptomycin. The cells were kept at 37°C in a humidified 5% CO2 incubator and cultures beyond passage 25 were discarded. The human embryonic stem cell lines H1 (WA01, WiCell; RRID: CVCL_9771) and PDX1EGFP/+ HUES4, acquired from our facility53, were cultured in DEF-CS culture media (Takara Biosciences, San Jose, CA, USA) according to the manufacturer's guidelines. The culture medium was changed daily, and cells were passaged every 2-3 days using TrypLE Express Enzyme (Thermo Fisher, Waltham, MA, USA). All cell cultures were maintained in a humidified incubator at 37°C with 5% CO2. Culturing the human embryonic stem cell lines H1 and HUES4 was done in accordance with relevant guidelines and regulations and after consent from the regional scientific ethical committee of the Capital Region of Denmark (protocol nr.: H-21043866; appendix 94634).
Cell- and viability count: HEK293 cells were plated in 60 mm petri dishes (≈ 20% confluency) and HuES4 and H1 cells were seeded in 12-well cell culture dishes with a density of 15,000 cells per well. After 24 h of settling, cells were treated with either DMSO (control) (Sigma/Merck, Darmstadt, Germany) or APAP (HEK293: 500 µM, HuES4 and H1: 200 µM), for 24, 48, or 72 h. To detach the cells, Trypsin or TrypLE Express Enzyme (Thermo Fisher, Waltham, MA, USA) were used. The cells were then centrifuged at 1000 rpm for 5 minutes, resuspended in 1 mL of culture medium, and counted. HuES4 and H1 cell counting was performed on quadruplicate technical replicate wells. The counting process was performed with Via1-Cassettes™ (ChemoMetec, Lillerød, Denmark) and a NucleoCounter® NC-200™ (ChemoMetec, Lillerød, Denmark) automated cell counter following the manufacturer's protocol and resulted in measurement of total number of cells and the percentage of viable cells. All three cell lines were analysed at a ≈ 80 % confluency to avoid restricted growth and to ensure that the analyses was performed on cells within their exponential growth phase.
Flow cytometric analysis of HEK293, HuES4, and H1 cell lines: HEK293, HuES4, and H1 cell lines were plated and left to settle for 24 h prior to treatment with control (DMSO) or APAP for 48 h. The cells were detached, fixed by slowly adding ice cold 70% ethanol while gently vortexing the tube, and left overnight in the fridge at 4°C. After centrifugation and removal of the supernatant, the pellet was resuspended in PBS containing 0.25% triton X-100 and incubated on ice for 15 min to permeabilize the cells. The cell pellet was then incubated for 30 min in the dark at RT with PBS containing 10 ug/mL RNase A (diluted from a stock 10 mg/mL RNase A (Sigma/Merck (R-5000), Darmstadt, Germany) in 10 mM Tris pH 7.5, 10 mM MgCl2) and 20 ug/ml propidium iodide (diluted from a stock 3.6 mg/ml propidium iodide (Sigma/Merck, Darmstadt, Germany) in DMSO). After DNA staining, the cells were loaded and analysed on a FACSverse multicolour flow cytometer using the FACSuite software (BD Bioscience, San Jose, CA, USA). Cells were gated according to forward-scatter/side-scatter (FSC/SSC) principles, followed by an FSC-A/FSC-H to ensure the analysis of single cells. Approximately 6.000-10.000 cells were analysed per condition. For an unbiased analysis of the cell cycle profile the Flowing Software 2.5 (Turku Bioscience Centre, Turku, Finland) automated cell cycle tool was used to define the distribution of cells within the different cell cycle stages, G1/G0, S, and G2/M. All three cell lines were harvested at a 70-80 % confluency to avoid restricted growth and to ensure that the analyses were perform on cells within their exponential growth phase.
DNA replication analysis: Click-iT® Plus EdU Pacific Blue® Flow Cytometry Assay Kits (Thermo Fisher Scientific,Waltham, MA, USA) were used to measure de novo DNA synthesis according to the manufacturers protocol. Briefly, HEK293, HuES4, and H1 cell lines were plated and left to settle for 24 h prior to treatment with control (DMSO) or APAP for 3 h with the addition of 10µM EdU for the final 2 h. Cells were loaded and analysed on a FACSverse multicolour flow cytometer using the FACSuite software (BD Bioscience, San Jose, CA, USA). Cells were gated according to forward-scatter/side-scatter (FSC/SSC) principles, followed by an FSC-A/FSC-H to ensure the analysis of single cells. Approximately 6.000-10.000 cells were analysed per condition. For the detection of EdU Pacific Blue® a 405 nm excitation filter with a violet emission filter (448/45) were used.
Fission yeast culture
For growth assessment on solid agar, fission yeast (S. pombe) strains of indicated genotypes were grown over night in yeast extract liquid medium. Cells were then counted, and diluted to 5000, 500, 50 and 5 cells/µl. 5 µl of each dilution were spotted onto rich medium agar plates containing 1% DMSO or 1% DMSO/APAP and incubated for 3 days at 32°C before photography. For growth assessment in liquid yeast extract, fission yeast strains of indicated genotypes were grown at 32°C in the presence of 1% DMSO or 40 mM APAP/1% DMSO and were monitored by triplicate cell counting using a NucleoCounter® NC-3000 ™ (ChemoMetec, Lillerød, Denmark) as described by the manufacturer. Doubling time was calculated assuming exponential growth. Samples for DNA content were fixed in 70% ethanol, washed in 20 mM EDTA, and treated with RNAse A overnight before staining of the DNA with CytoxGreen and analysis of cellular DNA content using the NucleoCounter® NC-3000 ™ (ChemoMetec, Lillerød, Denmark) as described by the manufacturer.
Data analysis and statistics
Data analyses were performed with GraphPad Prism 9 (San Diego, CA, USA), except for the Fisher-Freeman-Halton and Phi and Cramer's tests that were performed in SPSS 29.0.1.0 (Chicago, IL, USA). For the cell cultures the ‘n’ signifies biological replicate experiments obtained from different passages. For mouse and human embryo culture experiments individual embryo was considered as one n. For animal studies each animal was considered as one n. The specific statistical test used to analyse the given data is stated in the figure legends. Data is represented as the mean ± SD or SEM and P< 0.05 was considered statistically significant.