Home Analysis of cell cycle stage, replicated DNA, and chromatin-associated proteins using high-throughput flow cytometry
Article Publicly Available

Analysis of cell cycle stage, replicated DNA, and chromatin-associated proteins using high-throughput flow cytometry

  • Marina Bejarano Franco , Safia Boujataoui ORCID logo , Majd Hadji ORCID logo , Louis Hammer ORCID logo , Helle D. Ulrich ORCID logo and L. Maximilian Reuter ORCID logo EMAIL logo
Published/Copyright: September 3, 2024

Abstract

Flow cytometry is a versatile tool used for cell sorting, DNA content imaging, and determining various cellular characteristics. With the possibility of high-throughput analyses, it combines convenient labelling techniques to serve rapid, quantitative, and qualitative workflows. The ease of sample preparation and the broad range of applications render flow cytometry a preferred approach for many scientific questions. Yet, we lack practical adaptations to fully harness the quantitative and high-throughput capabilities of most cytometers for many organisms. Here, we present simple and advanced protocols for the analysis of total DNA content, de novo DNA synthesis, and protein association to chromatin in budding yeast and human cells. Upon optimization of experimental conditions and choice of fluorescent dyes, up to four parameters can be measured simultaneously and quantitatively for each cell of a population in a multi-well plate format. Reducing sample numbers, plastic waste, costs per well, and hands-on time without compromising signal quality or single-cell accuracy are the main advantages of the presented protocols. In proof-of-principle experiments, we show that DNA content increase in S-phase correlates with de novo DNA synthesis and can be predicted by the presence of the replicative helicase MCM2-7 on genomic DNA.

1 Introduction

Efficient, error-free duplication of the genetic information and its accurate distribution in mitosis are essential prerequisites for cell proliferation. Therefore, cells have evolved sophisticated ways and molecular machines to preserve genome integrity over generations. The first step of DNA replication, also known as origin licensing, involves the successive loading of two minichromosome maintenance 2–7 protein complexes (MCM2-7) to double-stranded DNA (dsDNA) through its loader, the origin recognition complex (ORC) (Costa and Diffley 2022; Yadav and Polasek-Sedlackova 2024). In particular, the loading of MCM2-7 is tightly regulated and depends on the association of Cdc6 to ORC in late M-phase and early G1-phase. Then, with the help of Cdt1, two copies of MCM2-7 are sequentially loaded onto origin DNA in an ATPase-dependent manner (Miller et al. 2019; Remus et al. 2009). The product of this reaction is the MCM2-7 double-hexamer, which remains inactive until the cell commits to S-phase (Evrin et al. 2009; Reuter et al. 2024). Subsequently, upon origin firing, the double-hexamer splits, remodels into the CMG (Cdc45-MCM2-7-GINS) complex and unwinds the parental DNA for duplication at the replication fork (Georgescu et al. 2017; Yuan et al. 2020). Having finished DNA synthesis, converging CMGs become unloaded and cells progress to G2-phase and finally mitosis (Moreno and Gambus 2020).

Classically, DNA replication progression is monitored by DNA content analysis, while EdU incorporation is mostly visualised through either dot blots, immunofluorescence microscopy, or DNA fiber assays (Bianco et al. 2012; Cavanagh et al. 2011; Gallo et al. 2016). Protein association to chromatin is typically assessed through chromatin binding assays, chromatin immunoprecipitations, or Western Blotting (Miyazawa-Onami et al. 2017; Weinreich et al. 2001). Another standard method to investigate the cell cycle stage of a given cell population and to evaluate the DNA content of cells is flow cytometry (Rieger 2022). Additionally, cytometric analysis can be employed to assess DNA synthesis in a given cell population through the incorporation of thymidine analogues, such as 5-ethynyl-2′-deoxyuridine (EdU) or 5-bromo-2′-deoxyuridine (BrdU) (Cavanagh et al. 2011). In combination with DNA content, such two-dimensional cytometric analysis provides valuable information about the cell cycle distribution and proliferative activity of a target cell population. Yet, despite the wide usage of flow cytometry in budding yeast research, protocols have hardly evolved from single-dimension cell cycle analysis (Rieger 2022). Furthermore, protocols often require one sample per analysed dimension, e.g., DNA content or EdU incorporation, and sample processing is usually performed in single, large reaction tubes (Bianco et al. 2012). To streamline these processes and at the same time monitor chromatin-bound proteins, we have developed a series of flow cytometry protocols to address the need of researchers for quick and comfortable analyses of single or multiple parameters relevant to DNA replication. Our protocols allow fast, high-throughput analyses of key replication parameters for both budding yeast and human cells in a 96-well plate format (Figure 1). We also present the first successful detection of yeast nuclear proteins by flow cytometry. A proof-of-principle application of antibody-based FLAG-tag detection allows qualitative and quantitative comparison of the association of various proteins to chromatin. Utilising our optimised protocols, we not only show that the central protein complexes of origin licensing, ORC and MCM2-7, are recruited in the same manner to yeast and human chromatin. We also provide measures to reduce hands-on time, reagent consumption, overall cost per well, and plastic waste. In addition, signal-to-noise ratios, throughput, and uniformity of protocols were optimised. Taken together, we provide a comprehensive tool kit for efficient and fast cytometric analysis of DNA replication parameters in cellula.

Figure 1: 
Graphical representation of the experimental design for high-throughput analyses of DNA replication patterns. After harvesting, yeast and human cell samples are collected for batch workup of multiple samples in 96-well plates. Optimised protocols allow individual or combined analyses of DNA replication parameters. Using multiplexing for staining of total DNA, newly replicated DNA, and chromatin association of proteins, up to four parameters can be measured simultaneously. The 96-well design reduces both time and resources spent to store, process, and analyse samples by flow cytometry. Created with BioRender.com.
Figure 1:

Graphical representation of the experimental design for high-throughput analyses of DNA replication patterns. After harvesting, yeast and human cell samples are collected for batch workup of multiple samples in 96-well plates. Optimised protocols allow individual or combined analyses of DNA replication parameters. Using multiplexing for staining of total DNA, newly replicated DNA, and chromatin association of proteins, up to four parameters can be measured simultaneously. The 96-well design reduces both time and resources spent to store, process, and analyse samples by flow cytometry. Created with BioRender.com.

2 Results

2.1 High-throughput DNA content and DNA synthesis analysis S. cerevisiae

Budding yeast is an ideal organism to monitor cell cycle progression in synchronised populations due to the ease of arresting cells quickly and specifically at defined cell cycle stages, e.g., in G1-phase through alpha factor treatment. To test our miniaturised 96-well format procedure, we grew yeast cultures to the exponential phase, arrested them in G1-phase with alpha factor, released them into fresh media, and took samples at the indicated time points (Figure 2A). As expected, analysis by flow cytometry and appropriate gating (Table 1 and Supplementary Figure 1A) revealed that asynchronous (cycling) cultures produced two main peaks representing G1- and G2-phase, respectively, and a smaller population in S-phase. Upon alpha factor treatment, cells uniformly arrested in G1-phase, from which the culture was synchronously released as a single population (Breeden 1997). After 45–60 min, most cells had reached G2-phase and started dividing from 75 min and onwards. Our results confirm previous observations (Bianco et al. 2012; Haase and Lew 1997; Haase and Reed 2002), highlighting the feasibility of our high-throughput protocol.

Figure 2: 
High-throughput analysis of DNA content and DNA synthesis by flow cytometry in yeast cells. (A) Cells were arrested in G1-phase with alpha factor. Upon release into fresh media, samples were withdrawn every 15 min for 90 min at 25 °C and DNA content was visualised. Cells synchronously enter S-phase within 30 min and finish replication within 75 min. Cell division and another round of DNA synthesis are observable at 90 min post G1-phase release. (B) Cells were grown and sampled as in panel (A) and DNA content was visualised (left panels). Additionally, cells were treated with EdU to monitor de novo DNA synthesis (right panels). Gating strategy was used as described (Supplementary Figure 1A). a.u. = arbitrary units.
Figure 2:

High-throughput analysis of DNA content and DNA synthesis by flow cytometry in yeast cells. (A) Cells were arrested in G1-phase with alpha factor. Upon release into fresh media, samples were withdrawn every 15 min for 90 min at 25 °C and DNA content was visualised. Cells synchronously enter S-phase within 30 min and finish replication within 75 min. Cell division and another round of DNA synthesis are observable at 90 min post G1-phase release. (B) Cells were grown and sampled as in panel (A) and DNA content was visualised (left panels). Additionally, cells were treated with EdU to monitor de novo DNA synthesis (right panels). Gating strategy was used as described (Supplementary Figure 1A). a.u. = arbitrary units.

Table 1:

Flow cytometry acquisition parameters.

Parameter Scale Usage
Side scatter (SSC) Linear/logarithmic Relative cell complexity
Forward scatter (FSC) Linear/logarithmic Relative cell size
SYTOX Green (area) Linear DNA content/single cells
SYTOX Green (height) Linear Single cells
AlexaFluor Plus 405 Logarithmic Protein staining
AlexaFluor546 Logarithmic Protein staining/EdU
PE Logarithmic Protein staining
AlexaFluor 568 Logarithmic Protein staining
AlexaFluor 647 Logarithmic Protein staining/EdU

As a second dimension, we wanted to include EdU incorporation into our protocol as a readout for de novo DNA synthesis. To do so, we employed a budding yeast strain expressing HSV1-TK (thymidine kinase) and hEnt1 (equilibrative nucleoside transporter 1) (Bianco et al. 2012; Lengronne et al. 2001). As part of the overall optimisation process, we also considered different azides coupled to AlexaFluor 647 for the use in the click reaction to detect EdU incorporation. Comparing non-clicked, azide-clicked, and picolyl-azide-clicked samples from a culture synchronised in G1-phase and released into S-phase (45 min and 75 min), we observed that the picoly-azide reaction yielded stronger and more defined signals compared to regular azide-coupled dyes (Supplementary Figure 1B). Next, we repeated the cell cycle synchronisation experiment (Figure 2A) and complemented the media with 10 μM EdU to label replicating DNA in S-phase. Here, DNA content increase correlated well with EdU detection and was observed from 30 min onwards (Figure 2B). These analyses are in agreement with previously published results (Bianco et al. 2012; Hua and Kearsey 2011) and demonstrate the feasibility of the two-dimensional cytometric analysis in yeast and its performance in the 96-well plate high-throughput format.

2.2 High-throughput analysis of DNA content, DNA synthesis, and protein association to chromatin in S. cerevisiae

While DNA content and DNA synthesis are two major and important readouts in DNA replication research, they are only an indirect measure of the complex process unfolding at the replication fork. Therefore, we sought to include replication protein association to chromatin into our cytometric approach as a third dimension. To incorporate this dimension into our single well, multi-parameter approach, several changes had to be incorporated into the protocol. As most yeast flow cytometry protocols rely on ethanol fixation and permeabilisation, which denatures proteins, a different procedure was needed. Furthermore, to enable antibodies to recognise their antigens inside the budding yeast cell and, more importantly, the nuclear compartment, enzymatic digestion of the outer cell wall followed by permeabilisation of cellular membranes is essential. To define a qualitative measure for efficient cell wall digestion in our 96-well format, cells were grown in a flask, distributed into a 96-well plate, and digested with varying concentrations of zymolyase in a hypotonic buffer (Supplementary Figure 2A). Cell lysis as a readout for efficient cell wall removal was followed over time as a decrease in turbidity. Removal of the cell wall was deemed complete when the OD600 reached <10 % of the initial value. Due to the fragility of spheroplasts, we fixed cells with 1 % formaldehyde before zymolyase treatment. Next, we assessed the effect of formaldehyde fixation, cell wall digestion, and detergent-based permeabilisation of the cellular membranes on the flow cytometric detection of individual cells (Supplementary Figure 2B). Compared to spheroplasts that were lysed through osmotic shock, cells progressing through the steps of this protocol suffered some shrinking but maintained a distinct population in the forward versus side scatter (Supplementary Figure 2B, panel osmotic shock vs. fixed and permeabilised spheroplasts). Thresholds for detection in the forward and side scatter were therefore adjusted to exclude cell debris and a gating strategy was developed for a final region to capture the relevant cell population (Supplementary Figure 2B, panel adjusted thresholds, and Supplementary Figure 3).

Successful detection of proteins on chromatin in this workflow further depends on the antibody used and the conjugated dye. Therefore, we chose to utilise an established anti-FLAG antibody (clone M2) to detect FLAG-tagged proteins while avoiding differences in antibody properties and availabilities. We used a strain harbouring a genomically 5✕FLAG-tagged subunit of the MCM2-7 complex (Mcm4) to investigate its binding to chromatin before, during, and after S-phase.

Finally, the successful implementation of a multi-dimensional flow cytometry approach requires bright dyes with minimal spectral overlap. To accomplish this, we made use of a wide spectrum of available lasers found in many flow cytometers and chose the combination of the 405 nm laser with AlexaFluor Plus 405 coupled antibody (protein), the 488 nm laser with SYTOX Green (DNA), and the 637 nm laser with AlexaFluor647 (EdU) as a standard configuration (Table 2 and Supplementary Figure 5A). A fourth dimension, e.g., another protein could in principle be detected using the 561 nm laser with a corresponding dye-coupled antibody (AlexaFluor 546, phycoerythrin (PE), or AlexaFluor 568). Alternatively, if EdU is detected with a combination of the 561 nm laser and AlexaFluor 546-coupled picoly-azide, detection of a second protein would be possible with the 637 nm laser and an AlexaFluor 647-coupled antibody. With AlexaFluor 647 having the highest quantum yield, this latter option is particularly helpful whenever visualising a low-abundance protein.

Table 2:

Lasers and filter settings.

Fluorophore Excitation laser (nm) Emission filter (Band-Pass, nm) Output
AlexaFluor Plus 405 405 nm 100 mW VL 445/45 Protein stain
SYTOX Green 488 nm 100 mW BL 525/45 DNA staining
AlexaFluor546*/PE* 561 nm 100  mW YG 586/20 Protein stain/EdU
AlexaFluor 568* 561 nm 100  mW YG 615/20 Protein stain
AlexaFluor 647 637 nm 100 mW Red 667/30 Protein stain/EdU
  1. *Only one of these fluorophores can be measured in the same well/tube. A visual representation of fluorophore distribution is presented (see Supplementary Figure 5A).

2.2.1 High-throughput cytometric analysis of DNA replication proteins on chromatin

Next, we employed the newly developed protocol to investigate the replicative helicase MCM2-7 on chromatin (Figure 3 and Supplementary Figures 4 and 5). Cells from asynchronous and G1-phase synchronised and released cultures (up to 120 min post-release) were withdrawn and analysed on all dimensions for their replication patterns. As before (Figure 2), DNA content and DNA synthesis recapitulated progress and completion of S-phase within 60 min of G1-phase release, cell division, and a second S-phase. When probing for MCM2-7, asynchronous cells presented four major populations: A low- and high-signal population corresponding to G1-phase, a low-signal population corresponding to G2-phase, and a population connecting the high G1- and low G2-phase populations (Figure 3A, top panel). This pattern matches the life cycle of the replicative helicase on chromatin along the cell cycle and is in line with previous studies in human cells (see below and (Frisa and Jacobberger 2010; Håland et al. 2015)). Within the G1-phase cell populations, the high-fluorescence population represents loaded MCM2-7 double hexamers on licensed origins (Evrin et al. 2009). Upon release into S-phase (>15 min), the high-fluorescence population shifts to higher DNA content levels (increased SYTOX Green intensity) while progressively losing signal intensity. This reflects the gradual unloading of MCM2-7 until DNA replication is completed (Figure 3A, panel 30–60 min). As MCM2-7 is completely unloaded after fully replicating the genomic DNA, the low-fluorescent populations in G1- and G2-phase most likely present cells harbouring only soluble, nuclear MCM2-7 complexes or subunits. When entering mitosis (Figure 3A, panel 75 min), the signal intensity of the MCM2-7 complex shifts to the G1-phase level (low, pre-licensing), followed by another round of MCM2-7 double-hexamer loading, and entry into the next S-phase (high G1-phase population; Figure 3A, panels 75–120 min). Controls without antibodies and secondary antibody only confirmed that the protein signal is specific for MCM2-7 (Supplementary Figure 5B). We also validated the feasibility and effectiveness of different dye combinations by testing different secondary antibodies coupled to various dyes (Supplementary Figure 5C). Hereby, only minor signal intensity differences could be observed, illustrating the feasibility and effectiveness of dye selection and the overall protocol.

Figure 3: 
Analysis of DNA content, DNA synthesis, and MCM2-7 complex association with chromatin by flow cytometry in yeast cells. Cells were arrested in G1-phase with alpha factor. Upon release into fresh media, samples were withdrawn every 15 min for 120 min at 30 °C. DNA content (left panel), DNA synthesis (EdU incorporation, middle panel) and MCM2-7 complex association with chromatin (right panel) were visualised. Cells synchronously enter S-phase within 30 min and finish replication within 60 min. Cell division and another round of DNA synthesis are observable from 75 min post G1-phase release and onwards. A strong signal for MCM2-7 is observed in G1-phase, representing the inactive double-hexamer (G1 panel). Upon initiation of replication MCM2-7 complexes mature into CMGs, travel along the DNA, and are gradually unloaded until DNA replication has finished in G2-phase (60 min panel). A low signal is retained and cells divide before MCM2-7 is again loaded onto chromatin (75 min panel). A second replicate is shown in Supplementary Figure 4. (B) Bar chart representing the cell cycle phase distribution of cells in (A). G1-, S-, and G2-phase populations were calculated using the DNA content panel employing the Jean-Dean-Fox modelling algorithm (Fox 1980). (C) The appearance of EdU-positive cells coincides with S-phase progression. (D) MCM2-7 association with chromatin allows cell cycle status prediction. Dot plots from (A, right panels) were analysed for MCM2-7 binding and classified into high, intermediate, and low signal. This correlates with the cell cycle phase distribution depicted in panel (B) and reproduces the wave-like pattern of synchronised cell cycle progression. DNA synthesis (C, indicated by EdU incorporation) correlates with the moving helicase (CMG) in S-phase and allows prediction of DNA replication initiation timing. Gating strategy for MCM2-7 classification was used as described (Supplementary Figure 3). a.u. = arbitrary units. Presented are the averages and standard deviations derived from four biological replicates.
Figure 3:

Analysis of DNA content, DNA synthesis, and MCM2-7 complex association with chromatin by flow cytometry in yeast cells. Cells were arrested in G1-phase with alpha factor. Upon release into fresh media, samples were withdrawn every 15 min for 120 min at 30 °C. DNA content (left panel), DNA synthesis (EdU incorporation, middle panel) and MCM2-7 complex association with chromatin (right panel) were visualised. Cells synchronously enter S-phase within 30 min and finish replication within 60 min. Cell division and another round of DNA synthesis are observable from 75 min post G1-phase release and onwards. A strong signal for MCM2-7 is observed in G1-phase, representing the inactive double-hexamer (G1 panel). Upon initiation of replication MCM2-7 complexes mature into CMGs, travel along the DNA, and are gradually unloaded until DNA replication has finished in G2-phase (60 min panel). A low signal is retained and cells divide before MCM2-7 is again loaded onto chromatin (75 min panel). A second replicate is shown in Supplementary Figure 4. (B) Bar chart representing the cell cycle phase distribution of cells in (A). G1-, S-, and G2-phase populations were calculated using the DNA content panel employing the Jean-Dean-Fox modelling algorithm (Fox 1980). (C) The appearance of EdU-positive cells coincides with S-phase progression. (D) MCM2-7 association with chromatin allows cell cycle status prediction. Dot plots from (A, right panels) were analysed for MCM2-7 binding and classified into high, intermediate, and low signal. This correlates with the cell cycle phase distribution depicted in panel (B) and reproduces the wave-like pattern of synchronised cell cycle progression. DNA synthesis (C, indicated by EdU incorporation) correlates with the moving helicase (CMG) in S-phase and allows prediction of DNA replication initiation timing. Gating strategy for MCM2-7 classification was used as described (Supplementary Figure 3). a.u. = arbitrary units. Presented are the averages and standard deviations derived from four biological replicates.

To gain qualitative information on the DNA replication status, we quantified DNA content, EdU incorporation, and MCM2-7 chromatin binding (Supplementary Figures 3, 4 and Figure 3B–D). Relative cell cycle distributions were calculated from the DNA content using the Dean-Jett-Fox modelling algorithm and showed a typical progression of synchronised cells through the cell cycle (Figure 3B). Similarly, a continuous increase of EdU-positive cells was observed upon entering S-phase (Figure 3C). Finally, when quantifying the different populations of MCM2-7 binding, we found that the relative MCM2-7 population distribution recapitulated the cell cycle distribution (Figure 3D). Furthermore, the increase in EdU incorporation was highest at the point, when MCM2-7 complex signals started to decrease (compare Figure 3A, panels 30, 90, and 105 min with Figure 3C and D).

Furthermore, to test our protocol with other chromatin-associated proteins we choose Hta2 (Histone H2A) and Orc2 (ORC complex) as targets. Both proteins showed a bimodal distribution matching the DNA content of the samples (Supplementary Figure 6A and B). Hta2 showed a gradual increase with DNA synthesis and Orc2 produced a similar but less pronounced phenotype. These phenotypes were expected, as histones are continuously incorporated into newly replicated DNA within nucleosomes while ORC binds to free origin DNA. These data show that various proteins binding permanently or transiently to genomic DNA can be studied with our approach.

2.2.2 High-throughput cytometric analysis of DNA replication mutants in vivo

To further benchmark the robustness of the presented method, we sought to analyse mutants of DNA replication and their effects on the distribution of MCM2-7 on chromatin. To this end, we used two well-characterised, temperature-sensitive (ts) mutants cdc6-1 and cdc46-1 (mcm5) that, when shifted to the non-permissive temperature, exhibit a strong growth defect and cell cycle arrest in G1-phase (Aparicio et al. 1997; Hennessy et al. 1991; Tanaka et al. 1997, and Supplementary Figure 7A). Indeed, when shifting cycling cells to the non-permissive temperature, both cdc6-1 (Figure 4A) and cdc46-1 (Supplementary Figure 7B) cells produced a gradual arrest in G1-phase within 2 h. The typical MCM2-7 chromatin distribution (Figure 3A and 4B and Supplementary Figure 7C) was abolished, coinciding with an accumulation of low protein signal fraction in G1-phase (Figure 4C and Supplementary Figure 7D). As both mutants are known for failing to produce mature MCM2-7 DHs on DNA, the accumulating and low MCM2-7 signal observed in G1- and G2-phase (Figure 3A, 4 and Supplementary Figures 3–7) presents soluble, nuclear MCM2-7 complexes or subunits in a pre-licensing (G1-phase) or post-replicative (G2-phase) state.

Figure 4: 

Cdc6-1 cells arrest in a pre-licensing state in G1-phase of the cell cycle under non-permissive growth conditions. Cells were grown to exponential phase, split into two flasks, and continuously shifted to 37 °C, the non-permissive temperature of cdc6-1, (A) or kept at 25 °C (B) for indicated times. DNA content (left panels) and MCM2-7 complex association with chromatin (right panels) were visualised. a.u. = arbitrary units. (C) Quantification of the relative population distribution (A and B) of MCM2-7 under permissive and non-permissive growth conditions. Presented are the averages and standard deviations derived from three biological replicates.
Figure 4:

Cdc6-1 cells arrest in a pre-licensing state in G1-phase of the cell cycle under non-permissive growth conditions. Cells were grown to exponential phase, split into two flasks, and continuously shifted to 37 °C, the non-permissive temperature of cdc6-1, (A) or kept at 25 °C (B) for indicated times. DNA content (left panels) and MCM2-7 complex association with chromatin (right panels) were visualised. a.u. = arbitrary units. (C) Quantification of the relative population distribution (A and B) of MCM2-7 under permissive and non-permissive growth conditions. Presented are the averages and standard deviations derived from three biological replicates.

Taken together, the optimised conditions and the development of new protocols for detection of DNA content, DNA synthesis, and protein chromatin binding in a 96-well format allow for a fast, cost-effective, and accurate description of DNA replication parameters in budding yeast.

2.3 High-throughput analysis of DNA content, DNA synthesis, and protein association to chromatin in HCT116 cells

Similar to the protocols presented above, we also set out to streamline 96-well plate-based protocols for multi-dimensional analyses of DNA replication parameters in human cells. A typical experiment requires 1–1.5✕106 cells per well (one sub-confluent 6-well dish) regardless of the applied protocol. Ethanol fixation was used as a standard method for fixation, permeabilisation, and long-term storage of cells. In order to evaluate DNA content alone or DNA content and DNA synthesis (EdU), cells were pretreated accordingly, collected in a 96-well plate, ethanol-fixed and stored at 4 °C until use. To test the collection and workup protocols in 96-well plates, as well as the gating strategy (Supplementary Figure 8A), we collected asynchronous, Nocodazole-arrested, and Nocodazole-released (9 and 12 h) cells from CRISPR-engineered HCT116 cell lines harbouring MCM4-FLAG (Figure 5A) or ORC2-FLAG (Figure 5B) alleles. We also included serum-starved and released HCT116 cells (2 and 6h, Figure 5C) and EdU-treated HCT116 cells (Figure 5D). Cell cycle arrest and synchronous progression was observed for arrested cells as expected. Pulse-labelling asynchronous and sub-confluent HCT116 cells with EdU for 1 h resulted in efficient labelling of DNA when visualised by AlexaFluor 647 picolyl-azide mediated click reaction (Figure 5D and Pereira et al. (2017)). When incorporating protein binding to chromatin as a parameter, ethanol fixation was replaced by chromatin extraction followed by formaldehyde fixation and permeabilisation (Figure 5E and F). In this manner, soluble nuclear and cytoplasmic proteins were removed during the workup. However, this procedure required an adapted gating strategy (Supplementary Figure 8B). Focusing on asynchronous samples of HCT116 cell lines harbouring MCM4-FLAG (Figure 5E) or ORC2-FLAG (Figure 5F), we found signals for DNA content and EdU incorporation comparable to ethanol-fixed samples (Figure 5D). MCM4 protein detection in asynchronous cells yielded similar results as shown for yeast samples above and as previously reported (Frisa and Jacobberger 2010; Håland et al. 2015) with two specific populations, a highly fluorescent population (licensed origins) in G1-phase and a low-fluorescent population in G2-phase. ORC2 in turn produced a bimodal, lateral spread of fluorescence, suggesting constant DNA binding throughout the cell cycle. Controls without antibody and with secondary antibody only, as well as single stains and click-only reactions showcase the feasibility of the protocol.

Figure 5: 
High-throughput analysis of DNA content, DNA synthesis, and protein association with chromatin by flow cytometry in human cells. One-dimensional, high-throughput analyses of DNA content from MCM4-FLAG (A) and ORC2-FLAG-tagged (B) HCT116 cells arrested in G2/M-phase (Nocodazole, G2/M) and released for up to 12 h. (C) Arrest and release experiment using serum-starved HCT116 cells. Synchronous release from G0/G1-phase was visualised for up to 6 h. (D) Two-dimensional analyses of DNA content and DNA synthesis in asynchronous HCT116 cells. Three-dimensional, high-throughput analyses of DNA content, DNA synthesis, and protein association with chromatin in HCT116 cells. Plots for antibody controls, single stainings, and the full protocol are shown for MCM4-FLAG (E) and ORC2-FLAG-tagged (F) cells. While ORC shows steady binding to chromatin throughout the cell cycle, MCM2-7 is loaded and unloaded from chromatin (represented through different degrees of binding) during the cell cycle. The underlying gating strategy was used as described (Supplementary Figure 8A and B). a.u. = arbitrary units.
Figure 5:

High-throughput analysis of DNA content, DNA synthesis, and protein association with chromatin by flow cytometry in human cells. One-dimensional, high-throughput analyses of DNA content from MCM4-FLAG (A) and ORC2-FLAG-tagged (B) HCT116 cells arrested in G2/M-phase (Nocodazole, G2/M) and released for up to 12 h. (C) Arrest and release experiment using serum-starved HCT116 cells. Synchronous release from G0/G1-phase was visualised for up to 6 h. (D) Two-dimensional analyses of DNA content and DNA synthesis in asynchronous HCT116 cells. Three-dimensional, high-throughput analyses of DNA content, DNA synthesis, and protein association with chromatin in HCT116 cells. Plots for antibody controls, single stainings, and the full protocol are shown for MCM4-FLAG (E) and ORC2-FLAG-tagged (F) cells. While ORC shows steady binding to chromatin throughout the cell cycle, MCM2-7 is loaded and unloaded from chromatin (represented through different degrees of binding) during the cell cycle. The underlying gating strategy was used as described (Supplementary Figure 8A and B). a.u. = arbitrary units.

3 Discussion

In this study, we present a comprehensive set of protocols for the high-throughput cytometric analysis of DNA replication parameters in budding yeast and human cells. To our knowledge, this is also the first demonstration of antibody-based flow cytometric detection of chromatin-associated proteins in yeast. Serving the unmet need for high-throughput cytometric methods, our protocols facilitate rapid multi-dimensional analysis of single or multiple cellular parameters in a single well, such as DNA content (Figures 2A and 5A-C), DNA synthesis (Figures 2B, 5D and Supplementary Figures 1B and 6A), and protein binding to chromatin (Figures 3, 4, 5E and F and Supplementary Figures 4–7). Unifying various methods (flow cytometry, BrdU/EdU dot blots, Western blot) in a single assay to screen parameters easily from large cohorts of samples is one of the main advantages. Combining bright stains and dyes, as well as picolyl-based click chemistry, we were able to reduce the cell material required. We incorporated the use of picolyl azide, which is an advanced fluorescent probe containing a copper-chelating moiety. This raises the effective concentration of copper at the reaction site boosting the copper-catalyzed azide–alkyne cycloaddition while lowering its toxicity (Uttamapinant et al. (2012) and Supplementary Figure 1B). Other advantages include a reduction of sample numbers, plastic waste (as plates and reservoirs can be reused many times), costs per well (<50 %), and hands-on time while maintaining signal quality and single-cell accuracy.

Applying our new, three-dimensional protocol to analyse yeast and human MCM2-7 and ORC, we found that the two protein complexes show unique binding behaviours tailored to their individual functions (Figures 3-5 and Supplementary Figures 4 and 6B). ORC binds to replication origins throughout the cell cycle and recruits MCM2-7 as a double-hexamer to the DNA in late M- and G1-phase (Figure 6) (Evrin et al. 2009; Li et al. 2018; Reuter et al. 2024; Sun et al. 2014; Weinreich et al. 2001; Yuan et al. 2017). This is recapitulated by the bimodal signal of ORC detected in G1- and G2-phase cells (Figures 5F, and Supplementary Figure 6B) (Reuter et al. 2024). In contrast, MCM2-7 presented several populations with varying fluorescent signals (Figures 3-5). We suggest that these populations represent distinct stages of the helicase life (Figure 6). In G1-phase, the maximum signal was detected due to the maximum loading of MCM2-7 double-hexamers to chromatin. After activation upon S-phase commitment, MCM2-7 gradually unwinds DNA at the tip of the replication fork, potentially unloading inactive double-hexamers at dormant origins (Figure 6). This process in combination with replication termination leads to the progressive elimination of MCM2-7 from chromatin until cells complete replication in G2-phase (defined population with low fluorescent signal, Figure 6), arguing that no parental MCM2-7 double-hexamer remains on replicated DNA. Once cells have undergone mitosis, this low-signal population (pre-licensing state) is again shifted to the high-signal population (licensed state) by ORC-Cdc6-dependent loading of MCM2-7 double hexamers onto dsDNA (origin licensing, Figures 3-6 and Supplementary Figures 4, 6, and 7).

Figure 6: 
Model for MCM2-7 and ORC dynamics during the cell cycle. Origins of replication serve as binding sites for the helicase loader, ORC. ORC recruits two copies of the replicative helicase MCM2-7 to double-stranded DNA in a consecutive manner during late M-phase and throughout G1-phase, thereby licensing the genome for DNA replication. Upon commitment to S-phase, the inactive MCM2-7 double hexamer (DH) is transformed into two CMGs that unwind DNA as part of the replication fork. Inactive double-hexamers and converging CMGs are gradually unloaded from the chromatin during and at the end of S-phase. Segregation of DNA during mitosis and progression into the next cell cycle stimulates helicase loading in the divided cells. Together, this highlights a life cycle of the replicative helicase MCM2-7 on DNA during the cell cycle, while ORC binds to origins similarly in every stage of the cell cycle. Created with BioRender.com.
Figure 6:

Model for MCM2-7 and ORC dynamics during the cell cycle. Origins of replication serve as binding sites for the helicase loader, ORC. ORC recruits two copies of the replicative helicase MCM2-7 to double-stranded DNA in a consecutive manner during late M-phase and throughout G1-phase, thereby licensing the genome for DNA replication. Upon commitment to S-phase, the inactive MCM2-7 double hexamer (DH) is transformed into two CMGs that unwind DNA as part of the replication fork. Inactive double-hexamers and converging CMGs are gradually unloaded from the chromatin during and at the end of S-phase. Segregation of DNA during mitosis and progression into the next cell cycle stimulates helicase loading in the divided cells. Together, this highlights a life cycle of the replicative helicase MCM2-7 on DNA during the cell cycle, while ORC binds to origins similarly in every stage of the cell cycle. Created with BioRender.com.

Future developments of our method could include the direct detection of proteins of interest with dye-coupled primary antibodies or the implementation of SNAP-tag technology to couple a fluorescent dye directly to a tagged protein (Cole 2013). Alternatively, using fluorescent protein tags such as GFP or RFP could replace the antibody-based detection of proteins of interest, as good antibody pairs are essential for efficient protein detection. Although levels of chromatin-bound proteins can be assessed globally for individual cells in a population, one major limitation of this method is that localisation and dynamic reorganisation on chromatin, e,g., foci formation, cannot be evaluated using standard flow cytometers. Similarly, any post-translational modifications that interfere with detection by the antibody would lead to an underestimation of the chromatin-bound fraction of a protein or serious skewing of the evaluation.

4 Materials and methods

4.1 Materials

Material and instruments Source Identifier - Cat#
Deepwell plates, Protein LoBind, 96-wells Eppendorf EP0030504305
Adhesive PCR foil seal VWR 732-3218
Falcon™ 96-well, non-treated, U-Shaped-Bottom Microplate Fisher Scientific 08-772-54
Tabletop refrigerated centrifuge with 96-well plate adaptors, e.g. Heraeus Multifuge X3R Thermo Scienctific 75371761
8-Channel Finnpipette Multichannel Pipettes (various volumes) Thermo Fisher n/a
Reagent reservoirs ROTILABO® Volume 5 ml Carl Roth KLE5.1
DM1000 LED Microscope Leica n/a
Q125 sonicator with 8-horn tip QSONICA n/a
NovoCyte Quanteon flow cytometer Agilent n/a
BD LSR Fortessa SORP flow cytometer BD Bioscience n/a
Flow cytometry analysis software: FlowJo v.10 BD Bioscience n/a

4.2 Antibodies

Antibody Source Identifier - Cat#
Mouse monoclonal anti-FLAG M2 Antibody Sigma-Aldrich F1804
Donkey anti-Mouse IgG, Secondary Antibody, AlexaFluor™ Plus 405 Invitrogen 17131701
Goat anti-Mouse IgG, Secondary Antibody PE Invitrogen P-852
Goat anti-Mouse IgG, Secondary Antibody, AlexaFluor™ 546 Invitrogen A-21123
Goat anti-Mouse IgG, Secondary Antibody, AlexaFluor™ 568 Invitrogen A-11031
Goat anti-Mouse IgG, Secondary Antibody, AlexaFluor™ 647 Invitrogen A-21236

4.3 Reagents

Reagent Source Identifier - Cat#
Citric acid Sigma-Aldrich C0759-1KG
Sodium citrate tribasic Sigma-Aldrich 25114-1kg
Ethanol Carl Roth 9,065.4
RNaseA Sigma-Aldrich 10109169001
Proteinase K Sigma-Aldrich P4850-5ML
Propidium iodide Sigma-Aldrich 81845
SYTOX™ Green Nucleic Acid Stain Invitrogen S7020
5-Ethynyl-2′-deoxyuridine (EdU) Life Technologies E10187
Alpha factor (WHWLQLKPGQPMY) ProteoGenix Custom peptide synthesis
Bovine serum albumin, BSA Sigma-Aldrich A7906
AlexaFluor™ 647 Azide, Triethylammonium Salt Thermo Fisher A10277
AlexaFluor™ 647 picolyl-azide Jena Bioscience CLK-1284-1
AlexaFluor™ 546 picolyl-azide Jena Bioscience CLK-1300A-5
Copper(II) sulfate pentahydrate Sigma-Aldrich 469130-50G
(+)-Sodium l-ascorbate Sigma-Aldrich A7631-25G
5x phosphate-buffered saline IMB media lab n/a
Sodium azide Sigma-Aldrich S2002-100G
Formaldehyde solution, 36.5–38 % in H2O Sigma-Aldrich F8775-4x25mL
DL-Dithiothreitol (DTT) Sigma-Aldrich D0632-25G
Zymolyase 20T (Arthrobacter luteus), (20KU/g) AMS Biotechnology 120491-1
Potassium phosphate monobasic Sigma-Aldrich P5655-500G
Potassium phosphate dibasic Sigma-Aldrich P3786-500g
D-Sorbitol Sigma-Aldrich S1876-1 KG
Magnesium chloride hexahydrate Sigma-Aldrich M9272-500G-D
Sodium dodecyl sulfate, 20 % Sigma-Aldrich 05030-1L-F
Saponin Sigma-Aldrich 47036-50G-F
Triton X-100 Sigma-Aldrich T9284-500ML
Dulbecco’s modified Eagle´s medium (DMEM) Biozym 880005
Dulbecco’s Phosphate-Buffered Saline (DPBS) Gibco 14190144
Fetal bovine serum (FBS) PAN Biotech P30-3601
0.05 % Trypsin-EDTA-Phenol red Gibco 25300054
l-Glutamine Thermo Fisher 25030081
Penicillin-Streptomycin Pan Biotech P06-07100
Permeabilisation Buffer (10✕) eBioscience™ 00-8333-56
HEPES Sigma-Aldrich H3375-500G
PIPES Carl Roth 9,156.4
Sodium chloride Sigma-Aldrich S3014-5KG
EDTA Sigma-Aldrich E9884-100G
Sucrose Sigma-Aldrich S0389-1KG
Protease Inhibitor Cocktail Tablets Roche 5056489001
Igepal CA-630 (NP-40) Sigma-Aldrich I8896-50ML
Milk powder, skim milk Sigma-Aldrich 70166-500G

4.4 Buffers

Citrate buffer: 20✕ stock: 258.07 g of sodium citrate tribasic, dissolved in 1 L H2O, and filter-sterilised. Working solution (50 mM sodium citrate): 50 mL of the 20✕ stock in 900 mL ultrapure H2O, adjusted to pH 7.0 with 0.1 M citric acid and filled up to 1 L with ultrapure H2O. 5✕ phosphate-buffered saline (PBS): 685 mM NaCl, 13.5 mM KCl, 50 mM Na2HPO4, and 9 mM KH2PO4, adjusted to pH 7.4. 0.1 M potassium phosphate buffer (pH 7.4): 8.02 mL of 1 M KH2PO4 and 1.98 mL of 1 M K2HPO4, filled up to 100 mL with ultrapure H2O. Spheroplasting buffer: 100 mM potassium phosphate buffer, pH 7.4, 1.2 M sorbitol, 0.5 mM MgCl2, 10 mM DTT and 10 U/mL Zymolyase 20T. Permeabilisation buffer (yeast): 1✕ PBS, 0.1 % Saponin (w/v), 0.1 % Triton X-100 (v/v), and 0.1 % sodium azide (w/v). Extraction buffer: 25 mM HEPES pH 7.4, 50 mM NaCl, 1 mM EDTA, 3 mM MgCl2, 300 mM sucrose, 0.5 % Triton X-100 (v/v), and 1✕ protease inhibitor cocktail. PIPES buffer: 10 mM PIPES pH 7.0, 100 mM NaCl, 3 mM MgCl2, 300 mM sucrose, 0.7 % Triton X-100 (v/v), and 1✕ protease inhibitor cocktail. PBS-B buffer: 1✕ PBS and 1 mg/mL BSA. Fixation buffer: 1✕ PBS and 2 % formaldehyde (v/v). Flow buffer: 1✕ PBS, 0.5 mM EDTA, and 0.1 % NP-40 (v/v).

4.5 Cultivation of human cell lines

Human cells (HCT116) were cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10 % fetal bovine serum (FBS), 1 % (2 mM) l-glutamine and 100 μg/mL penicillin-streptomycin at 37 °C with 5 % CO2. Cells were arrested in G2/M-phase by treatment with Nocodazole (50 ng/mL) for 16 h before cells were washed and released in fresh medium supplemented with 10 μM EdU for 9 and 12 h before harvesting. To synchronise cells in G0/G1-phase, 1–2✕106 cells were seeded in 10 cm plates. On the next day, regular medium was exchanged for serum free media (DMEM and 1 % penicillin/streptomycin) and cells were incubated for 72 h before harvesting or release with regular medium.

4.6 Yeast strains and plasmids

Yeast strains, plasmids, and DNA sequences used in this study are listed in the Supplementary information. Saccharomyces cerevisiae strains used in this study are derived of W303 (Bianco et al. 2012) and listed in Supplementary Table 1. Cells were grown at 30 °C in full medium (YPD, 2 % peptone, 1 % yeast extract, and 2 % glucose) if not stated otherwise. For arrest and release experiments, overnight cultures were diluted to OD600 =0.2 and grown to OD600= 0.5. Alpha factor (final [c]= 5 μg/mL) was added every hour for 2 h. If labelling of newly synthesised DNA was desired, 5-ethynyl-2′-deoxyuridine (EdU, final [c]= 10 µM) was added for the last 15 min of the alpha factor treatment. Subsequently, cells were washed with pre-warmed media, released into fresh YPD supplemented with EdU (final [c]= 10 µM) and samples were withdrawn at indicated time points. For experiments with cdc6-1 and cdc46-1 temperature-sensitive mutants, cells were grown overnight at 25 °C, diluted, and grown to OD600= 0.5. Then, the cultures were split and incubated either at 25 °C (permissive temperature) or 37 °C (non-permissive temperature) in a shaking water bath (180 rpm) for indicated times.

4.7 Procedures for flow cytometry analyses

All protocols presented were optimised for the use in 96-well deepwell plates but experiments can also be performed in single 1.5 or 2 mL reaction tubes without adjustments. Cells were analysed on an Agilent Novocyte Quanteon or a BD LSR Fortessa II SORP with similar results. All data were analysed with FlowJo (v10.10). For analyses of cell cycle distribution the Dean-Jett-Fox modelling algorithm assuming synchronised peaks was used (Fox 1980).

4.8 Protocols for analysing S. cerevisiae

Typically, 0.4  mL of a culture (OD600 > 0.5, 4.5✕106 cells/mL) was sufficient for each of the following protocols to yield 50,000 gated events per well. If detection of newly synthesised DNA was desired, an appropriate concentration of EdU was added to the medium at least 30–60 min before harvesting. Unless otherwise stated, all steps were performed at room temperature (RT).

4.8.1 DNA content

400 µL of culture (OD600 >0.5) were fixed with 1 mL 100 % ethanol in a 96-well deepwell plate and kept at 4 °C sealed with a foil overnight or stored at 4 °C for up to a few months. Cells were washed twice with 500 µL citrate buffer pH 7.0 (2,500 g, 2 min), the plate was decanted and hit gently onto a piece of paper to dry. Cells were resuspended in 500 µL citrate buffer pH 7.0 supplemented with RNaseA (10 mg/mL, 1/400 dilution) and incubated at 50 °C for 1 h. Then, 4 µL Proteinase K was added and the plate was incubated at 50 °C for 1 h. Subsequently, samples were either stored at 4 °C if needed or 100 µL of the cell suspension was transferred into a new U-shaped-bottom 96-well plate. After sonication to separate mother and daughter cells (70 % output 2 s on, 2 s off for 20 s for 8-tip horn), SYTOX™ Green (final [c]= 2 µM) or propidium iodide (final [c]= 25 ug/mL) was added to each well of the plate and mixed. Samples were measured on a flow cytometer equipped to measure 96-well plates (see Note 1).

4.8.2 DNA content and newly synthesised DNA

400 µL of culture (OD600 >0.5) were fixed with 1 mL 100 % ethanol in a 96-well deepwell plate and kept at 4 °C sealed with a foil overnight or stored at 4 °C for up to a few months. Cells were washed twice with 500 µL citrate buffer pH 7.0 (2,500 g, 2 min), the plate was decanted and hit gently onto a piece of paper to dry. Cells were resuspended in 500 µL citrate buffer pH 7.0 supplemented with RNaseA (10 mg/mL, 1/400 dilution) and incubated at 50 °C for 1 h. Then, 4 µL Proteinase K was added and the plate was incubated at 50 °C for 1 h. Cells were spun for 2 min at 2,500 g and the cell pellet was resuspended 500 μL of 1✕ PBS+3 % BSA and incubated for 15 min (see Note 2). For the click reaction, samples were spun (2,500 g, 2 min), the supernatant was discarded, and the cell pellet was resuspended in 40 μL of freshly prepared dye azide mix. Per well, 27.8 μL of ultrapure water, 8 μL 5 ✕ PBS, 2 μL of CuSO4 (100 mM), 0.2 μL of AlexaFluor 647 picolyl-azide (500 μM), and 2 μL of sodium ascorbate (200 mg/mL) (see Note 3) were mixed. The click mix was distributed and the reaction incubated in the dark for 30–60 min. Cells were washed once with 500 μL of 1✕ PBS+3 % BSA, then with 500 μL of 1✕ PBS. Finally, cells were resuspended in 150 μL (cycling cells) to 300 μL (remaining samples) of 1✕ PBS and 100 µL of the cell suspension was transferred into a new U-shaped-bottom 96-well plate. Sample sonication separated mother and daughter cells (70 % output 2 s on, 2 s off for 20 s for 8-tip horn). SYTOX™ Green (final [c]= 2 µM) or propidium iodide (final [c]= 25 μg/mL) was added to each well of the plate and mixed. Samples were protected from light and measured on a flow cytometer equipped to measure 96-well plates (see Note 1).

4.8.3 DNA content, newly synthesised DNA, and protein association with chromatin

10 µL of 10 % sodium azide (final [c]= 0.1 % w/v) were added to 1 mL of culture (OD600 >0.5) in a 96-well deepwell plate to block mitochondrial respiration and inhibit cell growth. Cells were stored at 4 °C overnight or for up to few weeks until use. The plate was then removed from 4 °C and warmed up to RT. Cells were fixed with formaldehyde (final [c]= 1 % w/v) and incubated 30 min. The plate was centrifuged at 2,500 g for 2 min, the supernatant discarded and cells were washed with 1 mL of ultrapure water. After that, cells were resuspended in 500 µL spheroplasting buffer supplemented with DTT (final [c]= 10 mM) and Zymolyase 20T (final [c]= 10 U/mL). Next, the plate was incubated at 30 °C for 1 h without shaking and cell wall digestion was monitored (see Notes 4 and 5). The plate was spun (3,200 g, 2 min), the supernatant discarded, and cells were washed with 500 µL of spheroplasting buffer. Samples were centrifuged (3,200 g, 2 min), the supernatant discarded, and cells were resuspended in 50 µL permeabilisation buffer supplemented with the primary antibody (1/250, 4 µg; see Note 6). After incubation for at least 1 h, the plate was spun (3,200 g, 2 min), the supernatant discarded, and cells were washed with 250 µL permeabilisation buffer. Subsequently, cells were mixed with 50 μL permeabilisation buffer containing secondary antibody (1/200, 5 µg, fluorescently labelled; see Notes 6–8) and cells were incubated for at least 1 h in the dark. The plate was centrifuged (3,200 g, 2 min), the supernatant discarded, and cells were washed with 250 µL permeabilisation buffer. If EdU staining was not needed, the following steps (until SYTOX™ Green addition) were skipped, otherwise included. Having spun the plate and removed the supernatant, cells were resuspended in 250 μL permeabilisation buffer +3 % BSA and incubated for >15 min (see Note 2). For the click reaction, samples were spun (3,200 g, 2 min), the supernatant was discarded, and the cell pellet was resuspended in 40 μL of freshly prepared click mix. Per well, 27.8 μL of ultrapure water, 8 μL 5✕ PBS, 2 μL of CuSO4 (100 mM), 0.2 μL of AlexaFluor 647 picolyl-azide (500 μM), and 2 μL of sodium ascorbate (200 mg/mL) (see Note 3) were mixed. The click mix was distributed and the reaction incubated in the dark for 30 min. Cells were washed once with 250 μL of 1✕ PBS +3 % BSA, then with 250 μL of permeabilisation buffer. Finally, cells were resuspended in 100 μL of permeabilisation buffer supplemented with RNaseA (10 mg/mL, 1/400 dilution), SYTOX™ Green (final [c]= 2 µM) or propidium iodide (final [c]= 25 μg/mL), and 100 µL of the cell suspension was transferred into a new U-shaped-bottom 96-well plate. Samples were protected from light and measured on a flow cytometer equipped to measure 96-well plates (see Note 1).

4.9 Protocols for analysing human cells

Before starting, the number of cells and culture dishes needed for each experiment were calculated. Typically, 1–1.5✕106 cells (Supplementary Table 2) were sufficient for each of the following protocols to yield a minimum 25,000 of gated events per well. If detection of newly synthesised DNA was desired, an appropriate concentration of EdU was added to the medium at least 30–60 min before harvesting. Unless otherwise stated, all steps were performed at RT.

4.9.1 DNA content

The appropriate number of cells was grown in the respective medium. Confluency was avoided to ensure that all cell cycle stages were represented in the final analysis. Cells were washed with 1x DPBS, trypsinised, washed with 1x PBS, and diluted to 1–1.5✕106 cells/mL in 1x PBS. Cells were then transferred to a 96-well deepwell plate, spun at 400 g for 3 min at 4 °C and resuspended in 50 μL 1✕ PBS before fixation by adding 1 mL of ice-cold 70 % ethanol dropwise while vortexing the plate on a low setting. Cells were covered with a foil incubated at 4 °C overnight or for at least 1 h before storage at −20 °C if cells were not used within the next days. After fixation, cells were centrifuged at 1,000 g for 10 min at 4 °C, washed with 1✕ PBS and resuspended in 100 μL of 1✕ PBS supplemented with RNaseA (10 mg/mL, 1/400 dilution) and SYTOX™ Green (final [c]= 2 µM) or propidium iodide (final [c]= 25 μg/mL). The cell suspension was transferred into a new U-shaped-bottom 96-well plate. Samples were incubated for 20 min while protected from light and finally analysed on a flow cytometer, where DNA content was measured.

4.9.2 DNA content and newly synthesised DNA

The appropriate number of cells was grown in the respective medium. Confluency was avoided to ensure that all cell cycle stages were represented in the final analysis. Cells were washed with 1✕ DPBS, trypsinised, washed with 1x PBS, and diluted to 1–1.5✕106 cells/mL in 1✕ PBS. 1–1.5✕106 cells were then transferred to a 96-well deepwell plate and spun at 400 g for 3 min at 4 °C and resuspended in 50 μL 1✕ PBS before fixation by adding 1 mL of ice-cold 70 % ethanol dropwise while vortexing the plate on a low setting. Cells were covered with a foil and incubated at 4 °C overnight or for at least 1 h before storing at −20 °C if cells were not used within the next days. After fixation, cells were centrifuged at 1,000 g for 10 min at 4 °C, washed once with 1 mL 1✕ PBS and 1 mL 1 % BSA in 1✕ PBS. Permeabilisation of cells was achieved by resuspending cells in 100 μL 1x permeabilisation buffer (eBioscience) and pulse-vortexing followed by 30 min incubation. The plate was centrifuged (400 g, 3 min) cells were washed once with 200 μL 1✕ permeabilisation buffer, before adding 50 μL of the click mix. To prepare the click mix, 33.75 μL of ultrapure water, 10 μL 5✕ PBS, 1 μL of CuSO4 (100 mM), 0.25 μL of AlexaFluor 647 picolyl-azide (500 μM), and 5 μL of sodium ascorbate (200 mg/mL) (see Note 3) were mixed per well. Cells were treated in 50 μL of click mix and the reaction was incubated >30min in the dark. 500 μL 1✕ permeabilisation buffer was added and the plate was centrifuged (400 g, 3 min). The supernatant was discarded and cells were resuspended in 100 μL of 1✕ permeabilisation buffer supplemented with RNaseA (10 mg/ml, 1/400 dilution) and SYTOX™ Green (final [c]= 2 µM) or propidium iodide (final [c]= 25 μg/mL). The cell suspension was transferred into a new U-shaped-bottom 96-well plate. Samples were incubated for 20 min while protected from light and finally analysed on a flow cytometer, where. DNA content and newly synthesised DNA were measured.

4.9.3 DNA content, newly synthesised DNA, and protein association with chromatin

The appropriate number of cells was grown in the respective medium. Confluency was avoided to ensure that all cell cycle stages were represented in the final analysis. Cells were washed with 1✕ DPBS, trypsinised, washed with 1✕ PBS, and diluted to 1–1.5✕106 cells/mL in 1✕ PBS. 1–1.5✕106 cells were then transferred to a 96-well deepwell plate, spun at 400 g for 3 min at 4 °C and resuspended in 100 μL cold extraction buffer to pre-extract cytoplasmic proteins. After 7 min of incubation on ice, 1 mL PBS-B buffer was added on ice and the plate was centrifuged for 3 min, 400 g, at 4 °C. The supernatant was discarded and the cell pellet fixed in 100 μL fixation buffer for 20 min. The protocol was either continued directly, or cells were spun, the supernatant removed and cells resuspended in 1✕ PBS +3 % BSA and stored at 4 °C overnight, covered with a foil. If the plate was stored overnight, cells were washed once with 250 µL flow buffer before continuing. To stain chromatin-bound proteins, cells were washed with 500 μL flow buffer and resuspended in 50 µL flow buffer + 4 % milk powder with first antibody (1/250, 4 µg, see Note 9). After incubation for at least 1h, the plate was spun (400 g, 3 min), the supernatant discarded, and cells were washed with 250 µL flow buffer. Subsequently, cells were mixed with 50 μL flow buffer + 4 % milk powder containing diluted secondary antibody (1/200, 5 μg, fluorescently labelled) and the cells were incubated for at least 1 h in the dark at RT. The plate was centrifuged (400 g, 3 min, RT), the supernatant discarded, and cells were washed with 250 µL flow buffer + 4 % milk powder. If EdU staining was not needed the following steps (until SYTOX™ Green addition) were skipped, otherwise included. To prepare the click mix, 33.75 μL of ultrapure water, 10 μL 5✕ PBS, 1 μL of CuSO4 (100 mM), 0.25 μL of AlexaFluor 647 picolyl-azide (500 μM), and 5 μL of sodium ascorbate (200 mg/mL) (see Note 3) per well were mixed. Cells were treated in 50 μL of click mix and the reaction was incubated >30 min in the dark. 500 μL flow buffer + 4 % milk powder was added thereafter and the plate was centrifuged (400 g, 3 min, RT). The supernatant was discarded and cells were washed with flow buffer (no milk powder) twice. Cells were then resuspended in 100 μL of flow buffer supplemented with RNaseA (10 mg/mL, 1/400 dilution) and SYTOX™ Green (final [c]= 2 µM) or propidium (final [c]= 25 μg/mL). The cell suspension was transferred into a new U-shaped-bottom 96-well plate. Samples were incubated for 20 min while protected from light and finally analysed on a flow cytometer, where DNA content, newly synthesised DNA, and chromatin-bound proteins were measured with appropriate settings.

4.10 Notes

  1. Use of SYTOX™ Green is highly recommended due to sharper and stronger peaks when compared to propidium iodide (Rieger 2022). The following settings apply: For Agilent Novocyte Quanteon: flow rate 14 μL/min, sample volume: 50 µL, mixing at 1,500 rpm every 6 wells for one cycle. For BD LSR Fortessa II with HTS: flow rate 1 μL/s, sample volume: 30 µL, mixing volume: 100 µL, mixing speed: 200 µL, 3 mixes, wash volume: 400 µL. This should yield 50,000 of gated events in around 30 s for samples with an initial OD600 >0.5.

  2. A minimum of 15 min is recommended to allow the BSA to act as a blocking agent. This step is necessary to reduce non-target azide reactions, boosting the signal-to-noise ratio.

  3. Reagents for the click reaction should be added in the indicated order. However, when working with multiple samples, it is convenient to prepare a master mix for all wells/tubes. The master mix should then be used within 15 min of preparation. The copper solution can be stored indefinitely at RT, preferentially in the dark. The picolyl-azide was resuspended in DMSO and stored in the fridge until use. Sodium ascorbate should be prepared freshly each time and presents as a clear, transparent solution. Brown or yellowish discoloration means that the ascorbate has been oxidised and needs to be discarded. Alternatively, aliquots can be stored at −20 °C for up to one year and thawed prior to use.

  4. A low concentration of Zymolyase and longer incubation time was chosen to minimise over-digestion and premature lysis of the cells. A higher concentration/or incubation temperature may require shorter incubation times.

  5. Plates should be placed on a pre-warmed metal block for 96-well plates for even digestion across the deepwell plate. Zymolyase digestion can be monitored by microscopy. Spheroplasts become translucent and round as they lose their typical yeast shape. Alternatively, 10 μL of treated cells can be mixed with 500 μL of 1 % SDS (w/v) for monitoring the OD600. Complete digestion of the cell wall is achieved once the OD600 has dropped to ≤ 10 % of the initial value.

  6. As an alternative buffer, the flow buffer can be used with similar staining efficiency. If cytoplasmic staining of proteins is desired, omission of Triton X-100 from the permeabilisation buffer will result in intact nuclei and no nuclear staining.

  7. When choosing secondary antibodies, we found that AlexaFluor 647 = AlexaFluor 405 plus > PE > AlexaFluor 568.

  8. If more than one protein is to be visualised, one can either use a fluorescent tag, such as GFP, or switch to AlexaFluor 546-linked picolyl azide in the click reaction. This allows protein detection with a secondary antibody coupled to AlexaFluor 647, which has an excellent quantum yield. Alternatively, antibodies coupled to PE, AlexaFluor 546, or AlexaFluor 568 have been used successfully.

  9. If the permeabilisation buffer is too harsh for the protein of interest, PIPES buffer or the 1x permeabilisation buffer (eBioscience) can be used alternatively with similar efficiency.


Corresponding author: L. Maximilian Reuter, Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, D-55128 Mainz, Germany, E-mail:
Safia Boujataoui, Majd Hadji and Louis Hammer contributed equally to this work.

Award Identifier / Grant number: 210253511

Award Identifier / Grant number: 393547839 – SFB 1361

Award Identifier / Grant number: 505087959

Acknowledgments

We thank Ronald Wong for scientific discussions and Kirill Petriukov for help with setting up the CRISPR-Cas9 tagging. We thank Bruce Stillman for the provision of the cdc6-1 and cdc46-1 yeast strains. The method protocols described here were inspired by (Bay et al. 2023; Forment and Jackson 2015; Hill et al. 2020). We thank the IMB Media Lab for their support. In addition, we thank the IMB Flow Cytometry Core Facility, especially Stefanie Möckel and Stephanie Nick, for their assistance with experiment setup and analysis.

  1. Research ethics: Not applicable.

  2. Author contributions: The authors have accepted responsibility for the entire content of this manuscript and approved its submission. Conceptualisation: L.M.R..; Investigation and Formal Analysis: M.B.F., S.B., M.H., L.H., and L.M.R..; Resources: M.B.F, L.H., and L.M.R.; Writing – Original Draft: H.D.U. and L.M.R.; Review & Editing: all authors; Supervision: H.D.U. and L.M.R. ; Funding Acquisition: H.D.U. and L.M.R.

  3. Competing interests: The authors state no conflict of interest.

  4. Research funding: This project was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation Project-ID: 505087959 (L.M.R.) and Project-ID: 393547839 – SFB 1361 (H.D.U.). Funding of the German Research Foundation supported the BD LSRFORTESSA SORP (Project-ID: 210253511, IMB Flow Cytometry Core Facility).

  5. Data availability: The raw data can be obtained on request from the corresponding author. Raw data annotated according to MIFlowCyt descriptions (Lee et al. 2008) are available through FlowRepository (Spidlen et al. 2012; https://flowrepository.org) via the following experiment ID: FR-FCM-Z82B.

References

Aparicio, O.M., Weinstein, D.M., and Bell, S.P. (1997). Components and dynamics of DNA replication complexes in S. cerevisiae: redistribution of MCM proteins and Cdc45p during S phase. Cell 91: 59–69, https://doi.org/10.1016/s0092-8674(01)80009-x.Search in Google Scholar

Bay, L., Stokke, T., Syljuåsen, R., and Landsverk, H. (2023). Analysis of RNA polymerase II chromatin binding by flow cytometry. Bio-Protoc 13, https://doi.org/10.21769/BioProtoc.4659.Search in Google Scholar

Bianco, J.N., Poli, J., Saksouk, J., Bacal, J., Silva, M.J., Yoshida, K., Lin, Y.L., Tourriere, H., Lengronne, A., and Pasero, P. (2012). Analysis of DNA replication profiles in budding yeast and mammalian cells using DNA combing. Methods 57: 149–157, https://doi.org/10.1016/j.ymeth.2012.04.007.Search in Google Scholar

Breeden, L.L. (1997). Alpha-factor synchronization of budding yeast. Methods Enzymol. 283: 332–341, https://doi.org/10.1016/s0076-6879(97)83027-3.Search in Google Scholar

Cavanagh, B.L., Walker, T., Norazit, A., and Meedeniya, A.C.B. (2011). Thymidine analogues for tracking DNA synthesis. Molecules 16: 7980–7993, https://doi.org/10.3390/molecules16097980.Search in Google Scholar

Cole, N.B. (2013). Site-specific protein labeling with SNAP-tags. Curr Protoc Protein Sci 73: 30.1.1–30.1.16, https://doi.org/10.1002/0471140864.ps3001s73.Search in Google Scholar

Costa, A. and Diffley, J.F.X. (2022). The initiation of eukaryotic DNA replication. Ann Rev Biochem 91: 107–131, https://doi.org/10.1146/annurev-biochem-072321-110228.Search in Google Scholar

Evrin, C., Clarke, P., Zech, J., Lurz, R., Sun, J., Uhle, S., Li, H., Stillman, B., and Speck, C. (2009). A double-hexameric MCM2-7 complex is loaded onto origin DNA during licensing of eukaryotic DNA replication. Proc. Natl. Acad. Sci. U.S.A. 106: 20240–20245, https://doi.org/10.1073/pnas.0911500106.Search in Google Scholar

Forment, J.V. and Jackson, S.P. (2015). A flow cytometry–based method to simplify the analysis and quantification of protein association to chromatin in mammalian cells. Nat. Protoc. 10: 1297–1307, https://doi.org/10.1038/nprot.2015.066.Search in Google Scholar

Fox, M.H. (1980). A model for the computer analysis of synchronous DNA distributions obtained by flow cytometry. Cytometry 1: 71–77, https://doi.org/10.1002/cyto.990010114.Search in Google Scholar

Frisa, P.S. and Jacobberger, J.W. (2010). Cytometry of chromatin bound Mcm6 and PCNA identifies two states in G1 that are separated functionally by the G1 restriction point1. BMC Cell Biol. 11: 26, https://doi.org/10.1186/1471-2121-11-26.Search in Google Scholar

Gallo, D., Wang, G., Yip, C.M., and Brown, G.W. (2016). Analysis of replicating yeast chromosomes by DNA combing. Cold Spring Harb Protoc 2016, pdb prot085118 https://doi.org/10.1101/pdb.prot085118.Search in Google Scholar

Georgescu, R., Yuan, Z., Bai, L., de Luna Almeida Santos, R., Sun, J., Zhang, D., Yurieva, O., Li, H., and O’Donnell, M.E. (2017). Structure of eukaryotic CMG helicase at a replication fork and implications to replisome architecture and origin initiation. Proc. Natl. Acad. Sci. U.S.A. 114: E697–E706, https://doi.org/10.1073/pnas.1620500114.Search in Google Scholar

Haase, S.B. and Lew, D.J. (1997). Flow cytometric analysis of DNA content in budding yeast. Methods Enzymol. 283: 322–332, https://doi.org/10.1016/s0076-6879(97)83026-1.Search in Google Scholar

Haase, S.B. and Reed, S.I. (2002). Improved flow cytometric analysis of the budding yeast cell cycle. Cell Cycle 1: 132–136, https://doi.org/10.4161/cc.1.2.114.Search in Google Scholar

Håland, T.W., Boye, E., Stokke, T., Grallert, B., and Syljuåsen, R.G. (2015). Simultaneous measurement of passage through the restriction point and MCM loading in single cells. Nucleic Acids Res. 43: e150, https://doi.org/10.1093/nar/gkv744.Search in Google Scholar

Hennessy, K.M., Lee, A., Chen, E., and Botstein, D. (1991). A group of interacting yeast DNA replication genes. Genes Dev. 5: 958–969, https://doi.org/10.1101/gad.5.6.958.Search in Google Scholar

Hill, B.D., Prabhu, P., Rizvi, S.M., and Wen, F. (2020). Yeast intracellular staining (yICS): enabling high-throughput, quantitative detection of intracellular proteins via flow cytometry for pathway engineering. ACS Synth. Biol. 9: 2119–2131, https://doi.org/10.1021/acssynbio.0c00199.Search in Google Scholar

Hua, H. and Kearsey, S.E. (2011). Monitoring DNA replication in fission yeast by incorporation of 5-ethynyl-2′-deoxyuridine. Nucleic Acids Res. 39: e60, https://doi.org/10.1093/nar/gkr063.Search in Google Scholar

Lee, J.A., Spidlen, J., Boyce, K., Cai, J., Crosbie, N., Dalphin, M., Furlong, J., Gasparetto, M., Goldberg, M., Goralczyk, E.M., et al.. (2008). MIFlowCyt: the minimum information about a flow cytometry experiment. Cytometry Part A 73A: 926–930, https://doi.org/10.1002/cyto.a.20623.Search in Google Scholar

Lengronne, A., Pasero, P., Bensimon, A., and Schwob, E. (2001). Monitoring S phase progression globally and locally using BrdU incorporation in TK(+) yeast strains. Nucleic Acids Res. 29: 1433–1442, https://doi.org/10.1093/nar/29.7.1433.Search in Google Scholar

Li, N., Lam, W.H., Zhai, Y., Cheng, J., Cheng, E., Zhao, Y., Gao, N., and Tye, B.K. (2018). Structure of the origin recognition complex bound to DNA replication origin. Nature 559: 217–222, https://doi.org/10.1038/s41586-018-0293-x.Search in Google Scholar

Miller, T.C.R., Locke, J., Greiwe, J.F., Diffley, J.F.X., and Costa, A. (2019). Mechanism of head-to-head MCM double-hexamer formation revealed by cryo-EM. Nature 575: 704–710, https://doi.org/10.1038/s41586-019-1768-0.Search in Google Scholar

Miyazawa-Onami, M., Araki, H., and Tanaka, S. (2017). Pre-initiation complex assembly functions as a molecular switch that splits the Mcm2-7 double hexamer. EMBO reports 18: 1752–1761, https://doi.org/10.15252/embr.201744206.Search in Google Scholar

Moreno, S.P. and Gambus, A. (2020). Mechanisms of eukaryotic replisome disassembly. Biochem Soc Transact 48: 823–836, https://doi.org/10.1042/BST20190363.Search in Google Scholar

Pereira, P.D., Serra-Caetano, A., Cabrita, M., Bekman, E., Braga, J., Rino, J., Santus, R., Filipe, P.L., Sousa, A.E., and Ferreira, J.A. (2017). Quantification of cell cycle kinetics by EdU (5-ethynyl-2′-deoxyuridine)-coupled-fluorescence-intensity analysis. Oncotarget 8: 40514–40532, https://doi.org/10.18632/oncotarget.17121.Search in Google Scholar

Remus, D., Beuron, F., Tolun, G., Griffith, J.D., Morris, E.P., and Diffley, J.F. (2009). Concerted loading of Mcm2-7 double hexamers around DNA during DNA replication origin licensing. Cell 139: 719–730, https://doi.org/10.1016/j.cell.2009.10.015.Search in Google Scholar

Reuter, L.M., Khadayate, S.P., Mossler, A., Liebl, K., Faull, S.V., Karimi, M.M., and Speck, C. (2024). MCM2-7 loading-dependent ORC release ensures genome-wide origin licensing. Nat. Commun. 15: 7306.10.1038/s41467-024-51538-9Search in Google Scholar

Rieger, A.M. (2022) Flow cytometry and cell cycle analysis: an overview. In: Wang, Z. (Ed.). Cell-cycle synchronization: Methods and protocols. Springer US, New York, NY, pp. 47–57.10.1007/978-1-0716-2736-5_4Search in Google Scholar

Spidlen, J., Breuer, K., and Brinkman, R. (2012). Preparing a minimum information about a flow cytometry experiment (MIFlowCyt) compliant manuscript using the International Society for Advancement of cytometry (ISAC) FCS file Repository (FlowRepository.org). Curr Protoc Cytometry 61: 10.18.1–10.18.26, https://doi.org/10.1002/0471142956.cy1018s61.Search in Google Scholar

Sun, J., Fernandez-Cid, A., Riera, A., Tognetti, S., Yuan, Z., Stillman, B., Speck, C., and Li, H. (2014). Structural and mechanistic insights into Mcm2-7 double-hexamer assembly and function. Genes Dev. 28: 2291–2303, https://doi.org/10.1101/gad.242313.114.Search in Google Scholar

Tanaka, T., Knapp, D., and Nasmyth, K. (1997). Loading of an Mcm protein onto DNA replication origins is regulated by Cdc6p and CDKs. Cell 90: 649–660, https://doi.org/10.1016/S0092-8674(00)80526-7.Search in Google Scholar

Uttamapinant, C., Tangpeerachaikul, A., Grecian, S., Clarke, S., Singh, U., Slade, P., Gee, K.R., and Ting, A.Y. (2012). Fast, cell-compatible click chemistry with copper-chelating azides for biomolecular labeling. Angew Chem Int Ed 51: 5852–5856, https://doi.org/10.1002/anie.201108181.Search in Google Scholar

Weinreich, M., Liang, C., Chen, H.H., and Stillman, B. (2001). Binding of cyclin-dependent kinases to ORC and Cdc6p regulates the chromosome replication cycle. Proc. Natl. Acad. Sci. U.S.A. 98: 11211–11217, https://doi.org/10.1073/pnas.201387198.Search in Google Scholar

Yadav, A.K. and Polasek-Sedlackova, H. (2024). Quantity and quality of minichromosome maintenance protein complexes couple replication licensing to genome integrity. Commun. Biol. 7: 1–14, https://doi.org/10.1038/s42003-024-05855-w.Search in Google Scholar

Yuan, Z., Georgescu, R., Bai, L., Zhang, D., Li, H., and O’Donnell, M.E. (2020). DNA unwinding mechanism of a eukaryotic replicative CMG helicase. Nat. Commun. 11: 688, https://doi.org/10.1038/s41467-020-14577-6.Search in Google Scholar

Yuan, Z., Riera, A., Bai, L., Sun, J., Nandi, S., Spanos, C., Chen, Z.A., Barbon, M., Rappsilber, J., Stillman, B., et al.. (2017). Structural basis of Mcm2-7 replicative helicase loading by ORC-Cdc6 and Cdt1. Nature Struct Mol Biol 24: 316–324, https://doi.org/10.1038/nsmb.3372.Search in Google Scholar


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/hsz-2024-0058).


Received: 2024-04-14
Accepted: 2024-08-15
Published Online: 2024-09-03
Published in Print: 2024-10-28

© 2024 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 7.10.2025 from https://www.degruyterbrill.com/document/doi/10.1515/hsz-2024-0058/html
Scroll to top button