May 31, 2024

Public workspaceAn end-to-end workflow to study newly synthesized mRNA following rapid protein depletion in Saccharomyces cerevisiae V.4

CheckPeer-reviewed method
  • 1Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
Open access
Protocol CitationJohn B. Ridenour, Rafal Donczew 2024. An end-to-end workflow to study newly synthesized mRNA following rapid protein depletion in Saccharomyces cerevisiae. protocols.io https://dx.doi.org/10.17504/protocols.io.n2bvj3dj5lk5/v4Version created by John B. Ridenour
Manuscript citation:
Ridenour, J.B., Donczew, R. An end-to-end workflow to study newly synthesized mRNA following rapid protein depletion in Saccharomyces cerevisiaeBMC Methods 1, 8 (2024). https://doi.org/10.1186/s44330-024-00008-1
License: This is an open access protocol distributed under the terms of the Creative Commons Attribution License,  which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited
Protocol status: Working
We use this protocol and it's working
Created: March 04, 2024
Last Modified: May 31, 2024
Protocol Integer ID: 100972
Keywords: Saccharomyces cerevisiae, Yeast, Transcription, Targeted protein degradation, Auxin-inducible degron system, Newly synthesized RNA, 4-thiouracil, SLAM-seq
Abstract
In this protocol, we describe an end-to-end workflow for rapidly degrading a target protein using the AID system and quantifying newly synthesized mRNA using SLAM-seq in Saccharomyces cerevisiae. We describe methods for targeted protein degradation, 4-thiouracil (4tU) incorporation, rapid methanol fixation, RNA purification, RNA alkylation, 3´ mRNA-seq library construction, and data analysis. Although the individual methods described in this protocol are not novel per se, this workflow provides a complete resource for turnkey implementation of these methods, which will benefit others working with S. cerevisiae. In addition, this workflow is modular and readily adaptable to other systems, including industrial, pathogenic, or other model fungi, which will benefit the larger research community.

REFERENCES

Alalam H, Zepeda-Martínez JA, Sunnerhagen P. (2022). Global SLAM-Seq for accurate mRNA decay determination and identification of NMD targets. RNA, 28(6), 905-915. https://doi.org/10.1261/rna.079077.121

Alexander RD, Barrass JD, Dichtl B, Kos M, Obtulowicz T, Robert MC, Koper M, Karkusiewicz I, Mariconti L, Tollervey D, Dichtl B, Kufel J, Bertrand E, Beggs JD. (2010). RiboSys, a high-resolution, quantitative approach to measure the in vivo kinetics of pre-mRNA splicing and 3′-end processing in Saccharomyces cerevisiaeRNA16(12), 2570-2580. https://doi.org/10.1261/rna.2162610

Aslanzadeh V, Huang Y, Sanguinetti G, Beggs JD. (2018). Transcription rate strongly affects splicing fidelity and cotranscriptionality in budding yeast. Genome Research, 28(2), 203-213. https://doi.org/10.1101/gr.225615.117

Barrass JD, Beggs JD (2019). Extremely rapid and specific metabolic labelling of RNA in vivo with 4-thiouracil (Ers4tu). JoVE (Journal of Visualized Experiments), 150, e59952. https://doi.org/10.3791/59952

Barrass JD, Reid JE, Huang Y, Hector RD, Sanguinetti G, Beggs JD, Granneman S. (2015). Transcriptome-wide RNA processing kinetics revealed using extremely short 4tU labeling. Genome Biology, 16(1), 1-17. https://doi.org/10.1186/s13059-015-0848-1

Chappleboim,A, Joseph-Strauss D, Gershon O, Friedman N. (2022). Transcription feedback dynamics in the wake of cytoplasmic mRNA degradation shutdown. Nucleic Acids Research50(10), 5864-5880. https://doi.org/10.1093/nar/gkac411

Donczew R, Hahn S. (2021). BET family members Bdf1/2 modulate global transcription initiation and elongation in Saccharomyces cerevisiaeeLife10, e69619. https://doi.org/10.7554/eLife.69619

Donczew R, Warfield L, Pacheco D, Erijman A, Hahn S. (2020). Two roles for the yeast transcription coactivator SAGA and a set of genes redundantly regulated by TFIID and SAGA. eLife9, e50109. https://doi.org/10.7554/eLife.50109

Foley JW, Zhu C, Jolivet P, Zhu SX, Lu P, Meaney MJ, West RB. (2019). Gene expression profiling of single cells from archival tissue with laser-capture microdissection and Smart-3SEQ. Genome Research, 29(11), 1816-1825. https://doi.org/10.1101/gr.234807.118

Glenn TC, Nilsen RA, Kieran TJ, Sanders JG, Bayona-Vásquez NJ, Finger JW, Pierson TW, Bentley KE, Hoffberg SL, Louha S, Garcia-De Leon FJ, Del Rio Portilla MA, Reed KD, Anderson JL, Meece JK, Aggrey SE, Rekaya R, Alabady M, Belanger M, Winker K, Faircloth BC. (2019). Adapterama I: universal stubs and primers for 384 unique dual-indexed or 147,456 combinatorially-indexed Illumina libraries (iTru & iNext). PeerJ7. https://doi.org/10.7717/peerj.7755

Herzog VA, Reichholf B, Neumann T, Rescheneder P, Bhat P, Burkard TR, Wlotzka W, von Haeseler A, Zuber J, Ameres SL. (2017). Thiol-linked alkylation of RNA to assess expression dynamics. Nature Methods, 14(12), 1198-1204. https://doi.org/10.1038/nmeth.4435

Neumann T, Herzog VA, Muhar M, von Haeseler A, Zuber J, Ameres SL, Rescheneder P (2019). Quantification of experimentally induced nucleotide conversions in high-throughput sequencing datasets. BMC Bioinformatics20, 1-16. https://doi.org/10.1186/s12859-019-2849-7

Veeranagouda Y, Remaury A, Guillemot JC, Didier M. (2019). RNA Fragmentation and Sequencing (RF‐Seq): Cost‐Effective, Time‐Efficient, and High‐Throughput 3′ mRNA Sequencing Library Construction in a Single Tube. Current Protocols in Molecular Biology, 129(1). https://doi.org/10.1002/cpmb.109
Guidelines
Standard laboratory guidelines and practices should be followed when performing this protocol. To ensure accurate size selection, researchers should use careful and consistent pipetting when performing this protocol.
Materials
Reagents:
  • Dimethyl sulfoxide, RNase-free (DMSO, ThermoFisher Scientific 327182500)
  • 3-indoleacetic acid (IAA, Sigma I3750)
  • 4-thiouracil (4tU, Sigma 440736)
Note
4tU is light sensitive and can crosslink upon exposure to light. Avoid exposure to light.
  • Dithiothreitol (DTT)
  • DNA/RNA Shield (Zymo Research )
  • DNase I, RNase-free, and DNA digestion buffer (Zymo Research E1010)
  • Iodoacetamide (Sigma I1149)
  • Glycogen (20 mg/ml, Roche 10901393001)
  • Sera-Mag SpeedBeads, magnetic carboxylate modified particles (Cytiva 65152105050250, ThermoFisher Scientific 09-981-123)
Note
Preparation of speedbeads and related buffers (DNA buffer and DNA binding buffer) is described in:
Protocol
A method to prepare Sera-Mag SpeedBeads for purification and size selection of nucleic acids
NAME
A method to prepare Sera-Mag SpeedBeads for purification and size selection of nucleic acids
CREATED BY
John B. Ridenour

  • Maxima H Minus Reverse Transcriptase (200 u/ul) and 5X first-strand buffer (ThermoFisher Scientific EP0752)
  • 2X Ex Premier (DNA polymerase) master mix (Takara RR370A)
  • dNTP solution set, dATP, dCTP, dGTP, and dDTP (NEB N0446S)
  • RNase H (NEB M0297L)
  • IRA_UMI_24dTVN oligo (PAGE purification, IDT)
  • IFA-isoTSO oligo (standard purification, IDT)
  • Working stocks (5 μM ) of i5 (i5-D501 through i5-D508) and i7 (i7-D701 through i7-D512) oligo mixes (standard purification, IDT)
Note
Oligo sequences and methods for oligo preparation are described in (see also Foley et al., 2019, Glenn et al., 2019, and Veeranagouda et al., 2019):

Protocol
High-throughput sequencing (HTS)  oligos and methods to prepare oligos for HTS applications
NAME
High-throughput sequencing (HTS) oligos and methods to prepare oligos for HTS applications
CREATED BY
John B. Ridenour


  • 100% methanol
  • 100% ethanol
  • Nuclease-free or DEPC-treated water
  • MilliQ water

Solutions:
  • 10 mM Tris-HCl (pH 8.0)
  • 0.5 M sodium phosphate buffer (pH 8.0, prepared with nuclease-free or DEPC-treated water)
  • 3 M sodium acetate (pH 5.2, prepared with nuclease-free or DEPC-treated water)
  • 1 M DTT
  • 100 mM DTT
  • 20 mM DTT
  • 80% ethanol (freshly prepared with nuclease-free or DEPC-treated water)

Buffers:
  • Speedbead DNA buffer
  • Speedbead DNA binding buffer

Consumables:
  • 50 ml conical tubes (sterile)
  • 1.5 ml microcentrifuge tubes (nuclease-free)
  • 0.2 ml PCR tubes or strips (low-bind, nuclease-free)
  • Quick-RNA Fungal/Bacterial Miniprep kit (Zymo Research R2014)
Note
Other RNA extraction/purification methods can be substituted. Alternative methods should be modified to maintain reducing conditions.

Equipment:
  • Mini-Beadbeater-24 (Biospec Products)
Note
Other bead beaters or cell homogenizers can be substituted. Processing conditions should be adjusted accordingly.
  • Thermomixer (e.g., ThermoMixer F1.5, Eppendorf) or dryblock
  • Vortex mixer (e.g., Vortex Genie 2, Scientific Industries)
  • Magnetic separation rack for 0.2 ml PCR tubes (e.g., NEBNext Magnetic Separation Rack, NEB)
Note
See Sergi Lab Supplies (https://sergilabsupplies.com) for economical and effective magnetic separation racks.

Safety warnings
Attention
Standard safe laboratory practices and procedures and institution-specific waste management programs should be followed when performing this protocol.
Saccharomyces cerevisiae growth, IAA treatment, 4tU labeling, and rapid fixation
Saccharomyces cerevisiae growth, IAA treatment, 4tU labeling, and rapid fixation
Streak appropriate S. cerevisiae strain on YPD agar. Incubate culture at 30°C for 2-3 days.

Note
YPD agar and YPD are prepared and supplemented with adenine following standard procedures. Other media can be used. IAA treatment and 4tU treatment are based on Donczew et al., 2020 and Donczew and Hahn, 2021. Sampling and rapid fixation are based on Alexander et al., 2010, Barrass et al., 2015, Aslanzadeh et al., 2018, and Barrass and Beggs, 2019. Samples should be protected from light during Steps 13-21 when possible (e.g., work under red light or low light and keep samples covered with foil).

Transfer an individual colony to 5 ml of YPD. Incubate culture at 30°C with shaking at 220 rpm overnight.
Dilute overnight culture to an OD600 of 0.2 in 40 ml of YPD. Incubate culture at 30°C with shaking at 220 rpm.
When the 40 ml culture reaches an OD600 of 0.3-0.4, cool centrifuge to 4°C and prepare 50 ml conical tubes, 2.0 ml microcentrifuge tubes, and 1.5 ml microcentrifuge tubes.
Add 5 ml of 100% methanol to a 50 ml conical tube for each sample to be collected. Keep tubes on dry ice. Three tubes are needed for each 40 ml culture.
When the 40 ml culture reaches an OD600 of 0.4-0.5, split the culture into three 10 ml aliquots in 50 ml conical tubes and return them to the incubator.

Each 10 ml culture represents one of three treatments:
ABC
TreatmentIAA treatment4tU treatment
ADMSO4tU
BIAA4tU
CDMSODMSO

Note
DMSO is the carrier for both IAA and 4tU. Treatment C is a control treatment and may not be required for all experiments. For example, once background T>C conversion rates have been established for an experimental system Treatment C may not be required.

Equilibrate cultures at 30°C for 5 min with shaking at 220 rpm.
During equilibration (or just prior to splitting the culture), prepare a 0.2 M IAA solution in DMSO. IAA is freshly prepared. To prepare a 0.2 M IAA solution, add 220 mg of IAA to 6 ml of DMSO and vortex until dissolved. Keep the IAA solution at room temperature covered with foil.

CRITICAL STEP: Add 50 μl of 0.2 M IAA solution or DMSO to the appropriate 10 ml culture. Vigorously mix the culture and return it to the incubator. The final concentration of IAA is 1 mM.
Critical
Incubate cultures at 30°C for 25 min with shaking at 220 rpm.

Note
Cultures are generally incubated for 25 min for protein degradation prior to adding 4tU in Step 12. The total time for protein degradation is ~30 min. The time can be adjusted based on the experiment.

During IAA treatment, prepare a 2 M 4tU solution in DMSO. 4tU is freshly prepared. To prepare a 2 M 4tU solution, add 260 mg of 4tU to 1 ml of DMSO and vortex until dissolved. Keep the 4tU solution at room temperature covered with foil.

CRITICAL STEP: Immediately after IAA treatment, add 25 μl of 2 M 4tU solution or DMSO to the appropriate 10 ml culture. 4tU will precipitate when added to the culture; immediately and vigorously mix the culture and return it to the incubator. It is essential that cultures are uniformly mixed to ensure 4tU is dissolved and homogeneously distributed. The final concentration of 4tU is 5 mM.

Critical
Incubate cultures at 30°C for 4 min with shaking at 220 rpm.

Note
Cultures are generally incubated for 4 min for 4tU incorporation. The time can be adjusted based on the experiment.

CRITICAL STEP: Immediately after 4tU treatment, decant each 10 ml culture directly into 5 ml of 100% methanol in a 50 ml conical tube on dry ice (prepared in Step 5). Loosely cap the slurry and gently mix by swirling to ensure homogenization. Keep the fixed sample on dry ice. The final concentration of methanol is approximately 33%.

Note
PAUSE POINT: Samples in methanol can be stored at -80°C. Thaw samples stored at -80°C at room temperature until frozen media has melted (approximately 5 min) before proceeding.

Critical
Pause
Gently mix the fixed sample and transfer 250 μl to a 1.5 ml microcentrifuge tube. Store the 250 μl aliquot at 4°C for cell counting. Estimate cell number using OD600 or count cells using a Neubauer chamber.

Note
The estimated cell number will be used to normalize samples for western blotting. It is generally sufficient to measure the OD600 for one treatment (e.g., Treatment A) for a given strain or replicate; the results for all treatments should be identical.
Gently mix the fixed sample and transfer 1.5 ml to a 2.0 ml microcentrifuge tube for western blotting. Centrifuge the sample at max speed for 1 min to pellet cells, discard the supernatant, and freeze cell pellets at -20°C.

Note
Store cell pellets at -20°C (up to 2 weeks) or -80°C (long-term storage). The volume of culture collected is 1 ml. It is helpful to record the OD600 value (Step 16) on the 2.0 ml microcentrifuge tube for future reference. Although specific methods are not described in this protocol, the depletion of target proteins must be validated by western blotting in all experiments.
Centrifuge the remaining sample at 3,000 g and 4°C for 10 min to pellet cells.
Carefully decant the supernatant and briefly invert the 50 ml conical tube on a paper towel to remove residual supernatant.
Adjust samples to a uniform cell number in DNA/RNA Shield. Use the following formula to calculate the appropriate volume of buffer for each sample: volume of buffer = (final OD600 of sample/0.70)*400. Thoroughly resuspend cells by pipetting.
Ensure cells are thoroughly resuspended and transfer 400 μl of the cell suspension to a 1.5 ml microcentrifuge tube.
PAUSE POINT: Flash freeze samples on dry ice or in liquid nitrogen and store at -80°C until proceeding.
Pause
Schizosaccharomyces pombe growth, 4tU labeling, and rapid fixation
Schizosaccharomyces pombe growth, 4tU labeling, and rapid fixation
OPTIONAL STEP: If experiments require spike-in, prepare 4tU-treated S. pombe cells as described in Steps 22-35. Streak appropriate S. pombe strain on YE agar. Incubate culture at 30°C for 3-4 days.

Note
YE agar and YE are prepared following standard procedures. Other media can be used.

Optional
Transfer an individual colony to 5 ml of YE. Incubate culture at 30°C with shaking at 220 rpm overnight.
Dilute overnight culture to an OD600 of 0.2 in 250 ml of YE. Incubate culture at 30°C with shaking at 220 rpm.
When the 250 ml culture reaches an OD600 of 0.6, cool centrifuge to 4°C and prepare 50 ml conical tubes and 1.5 ml microcentrifuge tubes.
Add 125 ml of 100% methanol to a 1 l flask for each culture to be collected. Keep flask on dry ice.
When the culture reaches an OD600 of 0.8, prepare a 2 M 4tU solution in DMSO as described in Step 11.
CRITICAL STEP: When the culture reaches an OD600 of 1.0, add 625 μl of 2 M 4tU solution. 4tU will precipitate when added to the culture; immediately and vigorously mix the culture and return it to the incubator.
Critical
Incubate the culture at 30°C for 4 min.

Note
Cultures are generally incubated for 4 min for 4tU incorporation. The time can be adjusted based on the experiment.

CRITICAL STEP: Immediately after 4tU treatment, decant the 250 ml culture directly into 125 ml of 100% methanol in a 1 l flask on dry ice (prepared in Step 26). Gently mix the slurry by swirling to ensure homogenization. Keep the fixed sample on dry ice.
Critical
Distribute 30 ml aliquots of the culture slurry to 50 ml conical tubes and centrifuge at 3,000 g and 4°C for 10 min to pellet cells.
Carefully decant supernatant and briefly invert 50 ml conical tubes on a paper towel to remove residual supernatant.
Thoroughly resuspend the cells in each 50 ml conical tube in 540 μl of DNA/RNA Shield by pipetting and pool cell suspensions in a fresh 50 ml conical tube on ice. Samples are adjusted to the equivalent of an OD600 of 7.0.
Ensure cells are thoroughly resuspended and distribute 500 μl aliquots of the cell suspension to 1.5 ml microcentrifuge tubes on ice.
PAUSE POINT: Flash freeze the samples on dry ice or in liquid nitrogen and store at -80°C until proceeding.
Pause
RNA purification and DNase I treatment
RNA purification and DNase I treatment
Supplement the following buffers and reagents with DTT (100 mM DTT working stock):

ABCD
TreatmentReagent vol. per sampleDTT vol. per sampleFinal DTT conc.
RNA Lysis Buffer400 μl0.8 μl0.2 mM
RNA Wash Buffer1.5 ml1.5 μl0.1 mM
RNA Prep Buffer400 μl0.4 μl0.1 mM
100% ethanol 550 μl0.55 μl0.1 mM
Nuclease-free water50 μl0.5 μl1 mM

Note
RNA purification and DNase I treatment is performed using a Quick-RNA Fungal/Bacterial Miniprep kit (see Materials). Methods are based on manufacturer’s recommendations modified to maintain reducing conditions using DTT essentially as described by Alalam et al., 2022. Prewarm the required volume of nuclease-free water (supplemented with DTT) (see Step 54). Perform all centrifugation steps at 16,000 g and room temperature for 30 s (unless specified). Samples should be protected from light during Steps 36-55 when possible (e.g., work under red light or low light and keep samples covered with foil).



Thaw samples stored in DNA/RNA Shield at -80°C at room temperature.
Combine cells suspended in DNA/RNA Shield with RNA Lysis Buffer using one of the following options:
OPTION A (without spike-in): Combine 400 μl of RNA Lysis Buffer and 400 μl of cells suspended in DNA/RNA Shield.
Optional
OPTION B (with spike-in): Combine 360 μl of RNA Lysis Buffer and 400 μl of cells suspended in DNA/RNA Shield, and combine 500 μl of RNA Lysis Buffer and 500 μl of S. pombe cells suspended in DNA/RNA Shield.

Note
See Steps 22-35 for preparation of S. pombe cells. Schizosaccharomyces pombe cells are combined with S. cerevisiae cells in mass-to-mass ratio of 1:19 based on OD600. One aliquot of S. pombe cells provides material to spike 24 S. cerevisiae samples. If multiple S. pombe samples are needed for an experiment, they should be prepared individually, pooled, and thoroughly mixed to ensure the spike-in is consistent across the experiment.

Optional
Transfer cells suspended in DNA/RNA Shield:RNA Lysis Buffer to a ZR Bashingbead Lysis Tube using one of the following options:
OPTION A (without spike-in): Transfer 800 μl of cells suspended in 1:1 DNA/RNA Shield:RNA Lysis Buffer to a ZR Bashingbead Lysis Tube.
Optional
OPTION B (with spike-in): Transfer 760 μl of S. cerevisiae cells suspended in DNA/RNA Shield:RNA Lysis Buffer and 40 μl of S. pombe cells suspended in DNA/RNA Shield:RNA Lysis Buffer to a ZR Bashingbead Lysis Tube.
Optional
Process samples in a Mini-Beadbeater-24 at 3800 rpm for 45 s and immediately incubate on ice for 2 min.
Repeat Step 40 once.
While samples are incubating on ice, prepare the following reaction mixture in a 1.5 ml microcentrifuge tube on ice:

DNase I reaction mixture
ABC
Reagent1X reaction vol. (μl)Master mix vol. (μl)
DNA Digestion Buffer75
DNase I, reconstituted (1 u/μl)5
Total80

Note
Do not supplement DNA Digestion Buffer with DTT. DNase I is easily denatured. Do not vortex reaction mixture.

Centrifuge samples at 16,000 g for 1 min to pellet debris.
CRITICAL STEP: Transfer 550 μl of the cleared supernatant to a Zymo-spin IIICG Column in a collection tube and centrifuge at 16,000 g for 30 s. Save the flow through.
Critical
Add an equal volume (550 μl) of 100% ethanol to the flow through. Thoroughly mix samples by pipetting.
Transfer 700 μl of the mixture to a Zymo-spin IICR Column in a collection tube and centrifuge at 16,000 g for 30 s. Discard flow through.
Repeat Step 46 with the remaining mixture volume.

Add 400 μl of RNA Wash Buffer to the column and centrifuge at 16,000 g for 30 s. Discard flow through.
Gently mix DNase I reaction mixture (prepared in Step 42) by inversion and add 80 μl directly to the column matrix. Incubate the reaction at room temperature for 15 min.
Add 400 μl of RNA Prep Buffer to the column and centrifuge at 16,000 g for 30 s. Discard flow through.
Add 700 μl of RNA Wash Buffer to the column and centrifuge at 16,000 g for 30 s. Discard flow through.
Add 400 μl of RNA Wash Buffer to the column and centrifuge at 16,000 g for 1 min to ensure complete removal of buffer.
Carefully transfer the column to a labeled nuclease-free 1.5 ml microcentrifuge tube.
Add 50 μl of nuclease-free water prewarmed to 50°C directly to the column matrix, incubate at room temperature for 2 min, and centrifuge at 16,000 g for 30 s.
PAUSE POINT: Store purified RNA at -20°C (up to four weeks) or -80°C until proceeding.
Pause
RNA alkylation and ethanol precipitation
RNA alkylation and ethanol precipitation
Thaw RNA samples on ice. Quantify RNA using a Nanodrop.

Note
RNA alkylation (iodoacetamide treatment) method is based on Herzog et al., 2017 (see also Alalam et al., 2022 and Chappleboim et al., 2022). Alternatively, the SLAMseq Kinetics kit – Anabolic Kinetics module (Lexogen) can be used for alkylation following the manufacturer’s recommendations. Iodoacetamide is referred to as IAA in Herzog et al., 2017; IAA refers to 3-indoleacetic acid in this protocol. Samples should be protected from light during Steps 56-62 when possible (e.g., work under red light or low light and keep samples covered with foil).

Add 5 μg of total RNA to a 1.5 ml microcentrifuge tube on ice. Adjust the volume of each sample to 20 μl with nuclease-free water.
Prepare a 100 mM iodoacetamide solution in DMSO. Iodoacetamide is freshly prepared. To prepare a 100 mM iodoacetamide solution, add 9.25 mg of iodoacetamide to 500 μl of DMSO (1 mg of iodoacetamide to 54 μl of DMSO). Keep the iodoacetamide solution at room temperature covered with foil. Do not store unused iodoacetamide.

Prepare the following reaction mixture in a 1.5 ml microcentrifuge tube and mix well by pipetting:

Alkylation reaction mixture
ABC
Reagent1X reaction vol. (μl)Master mix vol. (μl)
100 mM iodoacetamide5
0.5 M sodium phosphate buffer (pH 8.0)5
DMSO20
Total 30

Note
The sodium phosphate may form aggregates in the reaction mixture. The presence of aggregates will not interfere with the reaction. Prepare an additional 10% volume of master mix to avoid transferring aggregates.

Add 30 μl of reaction mixture to the 1.5 ml microcentrifuge tube containing 5 μg of total RNA in 20 μl nuclease-free water (prepared in Step 57). The final concentration of iodoacetamide is 10 mM.
Gently mix the reaction and incubate in a thermomixer at 900 rpm and 50°C for 15 min in the dark.
Add 1 μl of 1 M DTT to stop the reaction. Briefly vortex to mix. Exposure to light is acceptable following the addition of DTT.
Add 1 μl of glycogen (20 mg/ml) and 5 μl of 3 M sodium acetate (pH 5.2) to each sample. Briefly vortex to mix.
Add 125 μl of 100% ethanol to each sample. Briefly vortex to mix and incubate at -80°C for 30 min.
Centrifuge samples for 30 min at 21,000 g and 4°C to pellet precipitated RNA.
Carefully decant the supernatant. Take care not to dislodge pellet.
Add 1 ml of 80% ethanol to each sample. Centrifuge samples for 10 min at 21,000 g and 4°C to wash pellet.
Carefully decant the supernatant. Take care not to dislodge pellets. Carefully inspect samples and remove remaining ethanol with a 20 μl pipet.
Air dry samples for 5 min with lid open. Do not over dry samples.
Resuspend RNA in 30 μl of nuclease-free water.
PAUSE POINT: Store alkylated RNA at -20°C (up to four weeks) or -80°C until proceeding.
Pause
3´ mRNA sequencing library construction
3´ mRNA sequencing library construction
RNA fragmentation and cDNA synthesis.

Note
3´ mRNA sequencing library construction is based on Foley et al., 2019 and Veeranagouda et al., 2019. Prewarm required volume of 10 mM Tris-HCl (pH 8.0) (see Steps 73.4 and 74.11).

Thaw alkylated RNA samples on ice. Quantify RNA using an RNA-specific fluorometry method (i.e., Qubit RNA BR or HS assay kit).
Add 200 ng of RNA to a 0.2 ml PCR tube or strip on ice. Adjust the volume of each sample to 5 μl with nuclease-free water.
Prepare the following reaction mixture in a 0.2 ml PCR tube (or 1.5 ml microcentrifuge tube) on ice and mix well by pipetting:

Fragmentation reaction mixture
ABC
Reagent1X reaction vol. (μl)Master mix vol. (μl)
5X reverse transcription buffer2
20 mM dNTP mix0.5
20 μM IRA_UMI_24dTVN oligo0.5
Total3

Prepare the following reaction mixture in a 0.2 ml PCR tube (or 1.5 ml microcentrifuge tube) on ice and mix well by pipetting:

Reverse transcription (RT) reaction mixture
ABC
Reagent1X reaction vol. (μl)Master mix vol. (μl)
20 mM DTT1
20 μM IFA-isoTSO oligo0.5
Reverse transcriptase (200 u/μl)0.5
Total2

Bring the RNA and fragmentation reaction mixture to room temperature for 2-5 minutes and add 3 μl of the fragmentation reaction mixture to each sample. Mix well by pipetting.
CRITICAL STEP: Incubate samples at 94°C for exactly 4 min in a thermocycler with heated lid to fragment the RNA. Cool samples to 42°C in a thermocycler. Fragmentation time can be optimized to generate libraries of different insert sizes.
Critical
During RNA fragmentation, bring the RT reaction mixture (prepared in Step 72.4) to room temperature for 2-5 minutes.
Remove samples from the thermocycler, briefly centrifuge, and add 2 μl of the RT reaction mixture to each sample. Mix well by pipetting.
Briefly centrifuge and return samples to a thermocycler at 42°C.
Heat samples to 42°C for 60 min followed by 70°C for 10 min in a thermocycler with heated lid. Cool samples to 37°C in a thermocycler. Proceed immediately to RNA removal (Step 73.1).
RNA removal and single-stranded DNA purification.
Add 1 μl of RNase H to each sample and mix well by pipetting. Centrifuge briefly at room temperature to collect samples.
Heat samples to 37°C for 15 min in a thermocycler with heated lid. Proceed directly to speedbead purification.
Purify the single-stranded DNA (ssDNA). Add 39 μl of DNA buffer and 1 volume (50 μl) of speedbeads to each sample and proceed as described in the general method for speedbead purification (Step 76).

Note
Ensure speedbeads are equilibrated to room temperature and thoroughly mixed. DNA buffer and speedbeads can be mixed prior to use (here and in Step 74.5). If using commercial beads, it may be necessary to supplement beads with 0.05% Tween 20.

Elute DNA in 21 μl of 10 mM Tris-HCl (pH 8.0) prewarmed to 50°C.
Transfer 20 μl of the cleared supernatant to a fresh PCR tube.
PAUSE POINT: Store purified DNA at -20°C (up to eight weeks) or -80°C until proceeding.
Pause
Barcoding and enrichment PCR and library purification.
Transfer 10 μl of purified DNA to a 0.2 ml PCR tube on ice.
Add 2.5 μl of an appropriate 5 μM i5/i7 primer mix to each sample on ice.
Add 12.5 μl of 2X Ex Premier master mix to each sample on ice. Mix thoroughly by pipetting.
Barcode and enrich the library using PCR. Perform initial denaturation at 98°C for 45 s, followed by 12-18 cycles of amplification (98°C for 10 s, 60°C for 15 s, and 68°C for 15 s), and bring the reactions to 20°C.

Note
Use 12-14 amplification cycles if starting with ≥500 ng of total RNA, 16 amplification cycles if starting with 200 ng of total RNA, or 18 amplification cycles if starting with 100 ng of total RNA.

Purify the library. Add 25 μl of DNA buffer and 0.8 volumes (40 μl) of speedbeads to the 0.2 ml PCR tube containing the amplified library.

Mix well by pipetting up and down a minimum of 10 times. Alternatively, vortex samples on setting 4 for 3-5 seconds. If samples require centrifugation after mixing, stop the centrifuge before beads settle.
Incubate samples at room temperature for 5 min.
Place samples on magnetic stand for 2-5 min (or until solution is completely clear) to collect beads. Carefully remove and discard cleared supernatant without disturbing the bead pellet.
Remove samples from the magnetic stand. Add 30 µl of DNA buffer and mix well by pipetting or vortexing. Ensure beads are fully resuspend and incubate samples at room temperature for 2 min.
Add 0.8 volumes (24 μl) of DNA binding buffer to the 0.2 ml PCR tube containing the library and bead suspension. Proceed as described in the general method for speedbead purification (Step 76).
Elute the library in 21 μl of 10 mM Tris-HCl (pH 8.0) prewarmed to 50°C.
Transfer 20 μl of the cleared supernatant to a fresh PCR tube.
PAUSE POINT: Store purified libraries at -20°C (up to four weeks) or -80°C until proceeding.
Pause
Library quality control, quantification, pooling, and sequencing.
Assess library quality using an Agilent Bioanalyzer or agarose gel electrophoresis.
Quantify libraries using a DNA-specific fluorometry method (i.e., Qubit dsDNA BR or HS assay kit).
Pool equimolar concentrations (i.e., 5 nM) of each library. The recommended final concentration of the pool is 5 nM.

Note
See Glenn et al., 2019 for additional information on pooling and a template for calculating molar concentrations and volumes. Methods for library construction that use a template switching oligo (TSO), as described here, generate forward reads that start with three low-diversity bases. It is necessary to increase the diversity of the sequencing pool prior to sequencing on an Illumina instrument. Libraries should be pooled with 10-15% libraries prepared using standard approaches or control libraries (see Veeranagouda et al., 2019 for additional details).

Sequence the library pool on an Illumina instrument (e.g., NovaSeq 6000). The recommended sequencing depth is 10 million reads per sample.
General method for speedbead (SPRI) purification
General method for speedbead (SPRI) purification
Equilibrate speedbeads to room temperature and mix thoroughly.
CRITICAL STEP: Carefully add the indicated amount of speedbeads to the sample.
Critical
Mix well by pipetting up and down a minimum of 10 times. Alternatively, vortex samples on setting 4 for 3-5 seconds. If samples require centrifugation after mixing, stop the centrifuge before beads settle.
Incubate samples at room temperature for 5 min. Prolonged incubation or incubation at low temperature will increase binding of small nucleic acids (e.g., adapter dimers).
Place samples on magnetic stand for 2-5 min (or until solution is completely clear) to collect beads. Keep samples on magnetic stand for Steps 81-85.
Carefully remove and discard cleared supernatant without disturbing the bead pellet.
Add 200 μl of 80% ethanol (freshly prepared with MilliQ water) and incubate for 30 s. Carefully remove ethanol and discard without disturbing the bead pellet.
Repeat Step 82 once. Carefully inspect samples and remove remaining ethanol using a 10 μl pipet.
CRITICAL STEP: Air dry samples for 2 min with lid open. Do not over-dry samples. Speedbeads (and other SPRI products) will clump and can not be fully resuspended during elution if samples are over-dried.
Critical
Remove samples from the magnetic stand. Add the indicated volume of appropriate solution for elution and mix well by pipetting or vortexing (see Step 78).
Incubate mixture at room temperature for 2-5 min.
Place samples on a magnetic stand for 2 min (or until solution is completely clear) to collect beads.
Transfer cleared supernatant (volume of solution added for elution less 1 μl) to a fresh PCR tube.
Data analysis
Data analysis
Download and install the following software and dependencies:

  • fastp (version 0.23.2)
  • BBMAP (version 39.06)
  • Python (version 3.10.2)
  • SLAM-DUNK (version 0.4.3)
  • NextGenMap (version 0.5.5)
  • SAMtools (version 1.18)
  • VarScan 2 (version 2.4.5)
  • R (version 4.1.2-mkl)
  • Fastqc (version 0.12.1)
  • Multiqc (version 1.21)
Note
The most recent versions of the software should be used when possible. The input data for the subsequent steps are demultiplexed, paired-end 150 bp reads. Other data types can be processed without modification (e.g., paired-end 100 bp reads) or with slight modification (e.g., single-end 50 bp reads). The reverse reads (R2) generated from most 3′ mRNA-seq methods provide little information beyond barcoding. Unique molecular identifiers (UMIs) in the R2 read are extracted in Step 95. R2 reads are excluded from subsequent steps.

Download reference genome and annotation files. The reference genome assembly and annotation for S. cerevisiae strain S288C (version R64-3-1, RefSeq Assembly GCF_000146045.2) can be retrieved from the NCBI Datasets repository.

Note
Chromosome names should be modified in the reference genome assembly to reflect standard nomenclature (https://www.yeastgenome.org/) and maintain compatibility with BED file described below.

Download the reference BED file attached below. This BED file will be used to define counting windows in SLAM-DUNK.

Note
To generate the BED file below, all S. cerevisiae open reading frames (ORFs) were extracted from the reference annotation (version R64-3-1) using BEDOPS (version 2.4.3). Libraries constructed as described generate reads that align to gene bodies and 3′ untranslated regions (UTRs). All ORFs were extended 250 bp beyond their stop position using SAMtools (version 1.14) and BEDTools (version 2.30.0) to include 3′ UTRs. Chromosome names in the BED file were modified to reflect standard nomenclature (https://www.yeastgenome.org/). Reference files (i.e., genome assembly and BED file) must be checked to ensure the same chromosome names are used.

Download Scerevisiae_R64-3-1_gene3utr.bedScerevisiae_R64-3-1_gene3utr.bed

CRITICAL STEP: Set required variables using the commands below. Directories or files within quotations must be changed appropriately before running.
Command
DATA="path_to_zipped_fastq_files"
SAMPLES="file_listing_fastq_files_to_process"
DIRECTORY="path_to_outfiles"
BBMAP_RESOURCES="path_to_bbmap_reference_files"
GENOME_scer="reference_S.cerevisiae_genome_file"
BED_scer="reference_S.cerevisiae_bed_file"

Note
As an example, if zipped FASTQ files are stored in a directory named RDHTSP3 and the path to the directory is /archive/HighThroughputSequencing/, the command DATA="path_to_zipped_fastq_files" would be changed to DATA="/archive/HighThroughputSequencing/RDHTSP3" before running. An example "SAMPLES" file is provided below.

Download RDHTSP3_slamworkflow_ids.txtRDHTSP3_slamworkflow_ids.txt


Critical
Make subdirectories to store outfiles created in Steps 94-101 using the following commands:
Command
mkdir -p $DIRECTORY/logs/fastqc/fastq
mkdir -p $DIRECTORY/logs/multiqc/fastq
mkdir -p $DIRECTORY/fastp
mkdir -p $DIRECTORY/logs/fastp
mkdir -p $DIRECTORY/bbduk
mkdir -p $DIRECTORY/logs/bbduk
mkdir -p $DIRECTORY/logs/fastqc/bbduk
mkdir -p $DIRECTORY/logs/multiqc/bbduk
mkdir -p $DIRECTORY/slamdunk_scer
mkdir -p $DIRECTORY/slamdunk_scer/count_twotcreadcount
mkdir -p $DIRECTORY/slamdunk_scer/alleyoop
mkdir -p $DIRECTORY/logs/multiqc/slamdunk_scer

Run fastqc to assess raw data quality and multiqc to compile the fastqc output using following the commands:
Command
echo "running fastqc on raw data for sample:"
for sample_name in $(cat $SAMPLES | sed "s/_001.fastq.gz//" | uniq)
do
	echo "  ${sample_name}..."

	(fastqc \
		$DATA/${sample_name}_001.fastq.gz \
		--outdir $DIRECTORY/logs/fastqc/fastq \
		--threads 64 \
		--quiet \
		&> /dev/null)

done


echo "running multiqc on raw data..."
multiqc	\
	$DIRECTORY/logs/fastqc/fastq \
	--outdir $DIRECTORY/logs/multiqc/fastq \
	--quiet \
	&> /dev/null

Note
Here and in subsequent steps, the number of requested threads can be changed based on system capacity.

Run fastp to extract unique molecular identifiers (UMIs) using the following commands:
Command
echo "processing UMIs for sample:"
for sample_name in $(cat $SAMPLES | sed "s/_R._001.fastq.gz//" | uniq)
do
	echo "  ${sample_name}..."

	(fastp \
		--in1 $DATA/${sample_name}_R1_001.fastq.gz \
		--out1 $DIRECTORY/fastp/${sample_name}_umi_R1_001.fastq.gz \
		--in2 $DATA/${sample_name}_R2_001.fastq.gz \
		--out2 $DIRECTORY/fastp/${sample_name}_umi_R2_001.fastq.gz \
		--disable_adapter_trimming \
		--disable_quality_filtering \
		--disable_length_filtering \
		--umi \
		--umi_loc read2 \
		--umi_len 12 \
		--json $DIRECTORY/logs/fastp/${sample_name}_umi_fastp.json \
		--html $DIRECTORY/logs/fastp/${sample_name}_umi_fastp.html \
		2> $DIRECTORY/logs/fastp/${sample_name}_umi_fastp.log)

done

Note
UMI information is stored in R1 header. UMI information is not currently used in subsequent steps. In addition to extracting UMIs, fastp produces a quality control report for raw and processed data.


Run bbduk from the BBMap package to trim adapter sequences and polyA tails using the following commands:
Command
echo "trimming adapters for sample:"
for sample_name in $(ls -1 $DIRECTORY/fastp | sed "s/_umi_R._001.fastq.gz//" | uniq)
do
	echo "  ${sample_name}..."

	(bbduk.sh \
		in=$DIRECTORY/fastp/${sample_name}_umi_R1_001.fastq.gz \
		out=$DIRECTORY/bbduk/${sample_name}_trimmed_R1_001.fastq.gz \
		ref=$BBMAP_RESOURCES/truseq.fa.gz,$BBMAP_RESOURCES/polyA.fa.gz \
		k=13 \
		ktrim=r \
		mink=5 \
		qtrim=r \
		trimq=10 \
		minlength=20 \
		threads=16 \
		2> $DIRECTORY/logs/bbduk/${sample_name}_trimmed_bbduk.log)

done

Run fastqc to assess preprocessed data quality and multiqc to compile the fastqc output using following the commands:
Command
echo "running fastqc on preprocessed data..."
fastqc \
	$DIRECTORY/bbduk/*.fastq.gz \
	--outdir $DIRECTORY/logs/fastqc/bbduk \
	--threads 64 \
	--quiet \
	&> /dev/null


echo "running multiqc on preprocessed data..."
multiqc	\
	$DIRECTORY/logs/fastqc/bbduk \
	--outdir $DIRECTORY/logs/multiqc/bbduk \
	--quiet \
	&> /dev/null

Run slamdunk all from the SLAM-DUNK package to align data and quantify total reads and reads that have ≥1 T>C conversions using the following commands:
Command
echo "running slamdunk all..."
slamdunk all \
	--reference $GENOME_scer \
	--bed $BED_scer \
	--outputDir $DIRECTORY/slamdunk_scer \
	--trim-5p 5 \
	--topn 100 \
	--multimap \
	--min-coverage 10 \
	--var-fraction 0.8 \
	--max-read-length 151 \
	--min-base-qual 26 \
	--skip-sam \
	--threads 24 \
	$DIRECTORY/bbduk/*.fastq.gz

Note
See Neumann et al., 2019 and the SLAM-DUNK documentation (https://t-neumann.github.io/slamdunk/) for detailed information on the package.

Rename the count directory created by slamdunk all using the following command:
Command
mv $DIRECTORY/slamdunk_scer/count $DIRECTORY/slamdunk_scer/count_onetcreadcount

Run slamdunk count from the SLAM-DUNK package to quantify reads that have ≥2 T>C conversions using the following commands:
Command
echo "running slamdunk count..."
slamdunk count \
	--reference $GENOME_scer \
	--bed $BED_scer \
	--snp-directory $DIRECTORY/slamdunk_scer/snp \
	--outputDir $DIRECTORY/slamdunk_scer/count_twotcreadcount \
	--max-read-length 151 \
	--min-base-qual 26 \
	--conversion-threshold 2 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam

Run the following alleyoop commands from the SLAM-DUNK package to summarize and merge slamdunk output:
Command
echo "running alleyoop..."
alleyoop rates \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/rates \
	--reference $GENOME_scer \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop tccontext \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/tccontext \
	--reference $GENOME_scer \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop utrrates \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/utrrates \
	--reference $GENOME_scer \
	--bed $BED_scer \
	--multiTCStringency \
	--max-read-length 151 \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop snpeval \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/snpeval \
	--snp-directory $DIRECTORY/slamdunk_scer/snp \
	--reference $GENOME_scer \
	--bed $BED_scer \
	--min-coverage 10 \
	--var-fraction 0.8 \
	--multiTCStringency \
	--max-read-length 151 \
	--min-base-qual 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop summary \
	--output $DIRECTORY/slamdunk_scer/alleyoop/summary_onetcreadcount.tsv \
	--tcountDir $DIRECTORY/slamdunk_scer/count_onetcreadcount \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop summary \
	--output $DIRECTORY/slamdunk_scer/alleyoop/summary_twotcreadcount.tsv \
	--tcountDir $DIRECTORY/slamdunk_scer/count_twotcreadcount \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop merge \
	--output $DIRECTORY/slamdunk_scer/alleyoop/merge_totalreadcount.tsv \
	--column "ReadCount" \
	$DIRECTORY/slamdunk_scer/count_onetcreadcount/*.tsv


alleyoop merge \
	--output $DIRECTORY/slamdunk_scer/alleyoop/merge_onetcreadcount.tsv \
	--column "TcReadCount" \
	$DIRECTORY/slamdunk_scer/count_onetcreadcount/*.tsv


alleyoop merge \
	--output $DIRECTORY/slamdunk_scer/alleyoop/merge_twotcreadcount.tsv \
	--column "TcReadCount" \
	$DIRECTORY/slamdunk_scer/count_twotcreadcount/*.tsv


alleyoop tcperreadpos \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/tcperreadpos \
	--reference $GENOME_scer \
	--snp-directory $DIRECTORY/slamdunk_scer/snp \
	--max-read-length 151 \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop tcperutrpos \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/tcperutrpos \
	--reference $GENOME_scer \
	--bed $BED_scer \
	--snp-directory $DIRECTORY/slamdunk_scer/snp \
	--max-read-length 151 \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam


alleyoop dump \
	--outputDir $DIRECTORY/slamdunk_scer/alleyoop/dump \
	--reference $GENOME_scer \
	--snp-directory $DIRECTORY/slamdunk_scer/snp \
	--min-basequality 26 \
	--threads 24 \
	$DIRECTORY/slamdunk_scer/filter/*.bam

Run multiqc to compile output from the SLAM-DUNK package using the following commands:
Command
echo "running multiqc on slamdunk output..."
multiqc\
	$DIRECTORY/slamdunk_scer/alleyoop \
	--outdir $DIRECTORY/logs/multiqc/slamdunk_scer \
	--quiet \
	&> /dev/null

Note
See the SLAM-DUNK documentation (https://t-neumann.github.io/slamdunk/) for an example MultiQC report and additional information.

Key results from this analysis are the three count tables described below. These files are located in the alleyoop directory (i.e., $DIRECTORY/slamdunk_scer/alleyoop).

  • merge_totalreadcount.tsv - a table (tab-separated values) providing total read counts for all processed samples
  • merge_onetcreadcount.tsv - a table (tab-separated values) providing read counts with ≥1 T>C conversions for all processed samples
  • merge_twotcreadcount.tsv - a table (tab-separated values) providing read counts with ≥2 T>C conversions for all processed samples

Note
The filter directory (i.e., "$DIRECTORY/slamdunk_scer/filter") contains final mapping files in BAM format that can be useful for visualization and/or further processing. See the SLAM-DUNK documentation (https://t-neumann.github.io/slamdunk/) for additional information on output files.

Additional processing, analysis, and plotting can be performed in R (version 4.2.3) (https://www.R-project.org/) and RStudio (version 2023.12.0+369) (http://www.rstudio.com/) based on specific experimental goals.
OPTIONAL STEP: If experiments used S. pombe spike-in cells, set variables and make subdirectories using the commands below. Download the reference S. pombe genome assembly. Download the reference S. pombe BED file attached below. Repeat Steps 98-103 to generate spike-in count tables.
Command
GENOME_spom="reference_S.pombe_genome_file"
BED_spom="reference_S.pombe_bed_file"

mkdir -p $DIRECTORY/slamdunk_spom
mkdir -p $DIRECTORY/slamdunk_spom/count_conversionthreshold
mkdir -p $DIRECTORY/slamdunk_spom/alleyoop
mkdir -p $DIRECTORY/logs/multiqc/slamdunk_spom

Note
The reference genome assembly and annotation for S. pombe strain 972h- (version ASM294v2, RefSeq Assembly GCF_000002945.1) can be retrieved from the NCBI Datasets repository. The reference S. pombe BED file was generated as described in Step 91.

Download Spombe_ASM294v2_gene3utr.bedSpombe_ASM294v2_gene3utr.bed

Optional
Protocol references



CITATION
Foley JW, Zhu C, Jolivet P, Zhu SX, Lu P, Meaney MJ, West RB (2019). Gene expression profiling of single cells from archival tissue with laser-capture microdissection and Smart-3SEQ..