Mice
C57BL/6 (Ly5.2) mice (Sankyo Labo Service Corporation, Tokyo, Japan) were used for bone marrow transplantation assays. Trp53−/− mice, in which 5′ part of exon 2 including translation initiation site of Trp53 gene was replaced with Neomycin resistance gene, were provided from the RIKEN BioResource Center (Ibaragi, Japan)15. Rosa26-LSL-Cas9 knockin mice were purchased from Jackson Laboratory (#024857)16. The Trp53−/− mice were crossed with the Cas9 knockin mice to obtain Trp53−/−/Cas9 mice. All animal experiments were approved by the Animal Care Committee of the Institute of Medical Science at the University of Tokyo (PA21-67), and were conducted following the Regulation on Animal Experimentation at University of Tokyo based on International Guiding Principles for Biomedical Research Involving Animals.
Cell culture
Human cord blood (CB) cells were obtained from the Japanese Red Cross Kanto-Koshinetsu Cord Blood Bank (Tokyo, Japan). Mono nuclear cells (MNCs) were isolated from CB by density gradient centrifugation using LymphoprepTM (density 1.077; Alere Technologies AS, Oslo, Norway). The CD34+ cell fraction was then isolated from the MNCs using the MidiMACS system (CD34+ Microbead Kit; Miltenyi Biotec; Bergisch Gladbach, Germany) according to the manufacturer’s protocols. CB CD34+ cells were incubated in StemSpanTM SFEMII (STEMCELL Technologies) supplemented with 100 ng/ml rhSCF (#255-SC, R&D Systems), 10 ng/ml rhIL-6 (#206-IL, R&D Systems), 1 ng/ml rhIL-3 (#203-IL, R&D Systems) and 1% penicillin–streptomycin (PS, #09367-34, Nacalai). CB cells were then transduced with vector or p53DD and were cultured in StemSpan SFEM II medium (#ST-09655, STEMCELL Technologies) with 2 U/ml erythropoietin (EPO, #3999412G7020, Kyowa Kirin). THP1 and Kasumi-1 cells were cultured in Roswell Park Memorial Institute (RMPI)-1640 medium (#189–02025, FUJIFILM Wako) with 10% fetal bovine serum (FBS; #FB-1365/500, Biosera) and 1% PS. F36P and TF-1 cells were cultured in RPMI-1640 medium supplemented with 10% FBS, 1% PS and 2 ng/ml GM-CSF (#215-GMP, R&D Systems). cSAM cells were cultured in RPMI-1640 medium supplemented with 10% FBS containing 1 ng/ml mouse IL-3 (#203-IL, R&D Systems). CEP53 cells were cultured in RPMI-1640 medium supplemented with 10% FBS containing 1 ng/ml EPO (#959-ME, R&D Systems). Plat-E and 293 T cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM) medium (#044–29765, Wako) with 10% FBS and 1% PS.
Plasmids
p53DD was obtained from Addgene (#25989)17, and we cloned it into pMYs-IRES-NGFR vector18. MSCV-PIG, MSCV-PIG-ERG was abstained from Addgene (#66984)19. We added the AM-tag sequence (5’-TGCCAAGATCCTCAACGCAAAGGCAACGTGATACTCTCTCAGGC
TTACGGGTGCCAAGATCCTCAACGCAAAGGCAACGTGATACTCTCTCAGGCTTACTAG-3’) into MSCV-PIG-ERG for the ChIP-Seq assay. pcDNA3-GATA1 (Addgene # 85693)20, pSG5-hEKLF (Addgene #67835)21 and pGL3-GATA-Luc (Addgene #85695)22 were obtained from Addgene. pGL4.10[Luc2] (#E6651) and pGL4.74[hRluc/TK] (#E692A) were obtained from Promega. For pGL4.1-KLF1-Luc, we amplified the 6x repeat promoter sequence (5’-AGGGTGTGG-3’) of KLF1 23 using PCR and inserted the PCR-amplified fragment into the restriction sites of pGL4.10[Luc2].
Viral transduction
Retroviruses for mouse cells were generated by transient transfection of retroviral constructs into Plat-E packaging cells24 using the calcium phosphate method. Retroviruses for human cells were generated by transient transfection of retroviral constructs along with M57 and RD114 into 293T cells using the calcium phosphate method. Retrovirus transduction to the cells was performed using Retronectin (Takara Bio Inc., Otsu, Shiga, Japan). Lentiviruses were produced by transient transfection of lentiviral plasmids along with pCMV-VSV-G (Addgene, #8454)25 and psPAX2 (Addgene, #12260) into 293T cells using the calcium-phosphate method.26
Gene depletion using the CRISPR/Cas9 system
To generate short guide RNA (sgRNA) constructs, annealed oligos were inserted into pLentiguide-puro vector (#52963) or pLKO5.sgRNA.EFS.tRFP657 vector (#57824)27, which were obtained from Addgene. Cas9 expressing vector was also obtained from addgene (lentiCas9-Blast #52962)28. Lentiviruses were produced by transient transfection of 293T cells as described above. Cells were infected with the virus for 24 hours and were selected for stable expression of Cas9 using blasticidin (10 µg/ml). The sgRNA-transduced cells were selected using puromycin (1µg/ml) or FACS-based sorting of tRFP657-positive cells. The sequences of sgRNAs are provided in Supplemental Table 1.
Luciferase assay
1x105 293T cells were seeded in 12-well culture plates with cells in 500 µL medium. 18 h after seeding, the cells were transfected with pGL4.1-6X-KLF1 or pGL3-GATA-Luc (co-expressing Firefly Luciferase [FLuc]), pSG5-hEKLF (KLF1) or pcDNA3-GATA1, and pGL4.74 vector (co-expressing Renilla Luciferase [RLuc]) with MSCV-PIG-ERG or MSCV-PIG, using polyethylenimine (PEI). The cells were harvested 48 h after transfection and were assayed for the luciferase activity using the luciferase assay system (Promega) and a luminometer (BMG LABTECH, FLUOstar OPTIMA). Promoter activity was calculated as a ratio of Fluc to Rluc.
Transplantation assay
Mouse bone marrow cells were collected from the Cas9 mice and Trp53−/−/Cas9 mice. Bone marrow progenitors (c-Kit+ cells) were selected using the CD117 MicroBead Kit (Miltenyi Biotec) and were pre-cultured in RPMI-1640 containing 10% FBS, 1% penicillin–streptomycin and 50 ng/ml murine SCF, 10 ng/ml TPO, 10 ng/ml IL-3 and 10 ng/ml IL-6 for 16hr. These cells were then transduced with ERG and transplanted into sublethally (5.25 Gy) irradiated 12 weeks-old C57BL/6 mice. Each mouse received 2x105 cells. For transplantation of CEP53 cells, each mouse received 5x106 CEP53 cells without irradiation. For the in vivo transplantation assay using sgRNAs targeting Hdac7, spleen cells collected from the moribund AEL mice were transduced with NT or Hdac7-sgRNAs, and 5x105 cells were transplanted into sublethally (525 cGy) irradiated 12 weeks-old C57BL/6 mice.
Flow Cytometry
Cells were stained by fluoro-conjugated antibodies for 30min at 4°C. After staining, cells were washed with cold PBS two times, and were resuspended in PBS containing 2% FBS. Cells were analyzed with Canto II (BD Biosciences, San Jose, CA, USA) and FlowJo software (FlowJo) or sorted with FACS Aria III (BD Biosciences, San Jose, CA, USA). The antibodies and their dilution ratios are provided in Supplemental Table 2.
Western blotting
Cells were washed with PBS several times and lysed with pre-heated Laemmli sample buffer (Bio-rad, USA; #1610737). Total cell lysates were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to a polyvinylidene fluoride membrane (Bio-Rad). Bands were visualized by LAS-4000 Luminescent Image Analyzer (FUJIFILM). The antibodies and their dilution ratios are provided in Supplemental Table 3.
RNA-Seq
For RNA-seq with mouse AEL cells and erythroid progenitors, bone marrow cells were collected from normal C57BL/6 mice and the mice transplanted with Trp53−/−ERG-expressing cells. Cells were then stained with biotinylated Ter119 and CD71 antibodies and 2x106 Ter119+CD71+ cells were sorted by AriaIII (BD Biosciences, San Jose, CA, USA). For RNA-seq with F36P cells, F36P cells were transduced with vector/ERG (coexpress GFP) or NT/HDAC7-sgRNAs. GFP+ cells were sorted by AriaIII. The sgRNA-transduced cells were selected with 1 µg/ml puromycin. Total RNA was extracted using RNeasy Mini Kit (Qiagen). After RNA fragmentation, cDNA was synthesized by random hexamer-primed reverse transcription, followed by a second-strand cDNA synthesis with dUTP. Following end repair, add A and adaptor ligation, the DNA fragments were amplified by PCR. Libraries were sequenced using Illumina NovaSeq 6000 with paired-end mode (2x100 bp). Pair-end sequencing FASTQ files were aligned to the mouse reference genome (mm10). Raw gene counts were derived from the read alignments by Rsubread29 (v2.12.3) and further transferred into count per million (CPM) by edgeR30 (v3.40.2). After filtering out low-expression genes with CPM lower than 1, all CPM values were log2 transformed for generating unsupervised clustering dendrograms and heatmaps. Differential expression was analyzed with the linear model using limma31 (v3.54.2). Genes with false discovery rate (FDR) < 0.05 adjusted by the Benjamini-Hochberg method were considered significant differentially expressing genes (DEGs). 32 Pathway analyses were performed using GO Enrichment Analysis33 and Gene Set Enrichment Analysis34,35
ChIP-Seq
Chromatin immunoprecipitation was performed using Simple chip kit (Cell signaling technology, #9002) following the manufacturer’s instructions. 1x107 F36P cells transduced with vector or ERG were fixed with 1% formaldehyde (Sigma) and then quenched with glycine. After washing and cell lysis, the chromatin was fragmented with 0.75 µl micrococcal nuclease (MNase) at 37 C for 15–20 min and nuclei were completely lysed by sonication. The 10 µg of chromatin in each reaction was incubated with 10 µl of anti-AM (#91111, Active motif), anti-H3K27ac (#8173, CST) or IgG antibody (#2729,CST) overnight at 4 C with rotation. Immunoprecipitation was performed with protein G magnetic beads. Following elution, reverse-crosslinks, and purification, DNA was used for sequencing. ChIP-seq libraries were prepared and sequenced using Illumina Novaseq 6000 with paired-end mode (2x150 bp). Pair-end sequencing FASTQ files were aligned to the human reference genome (hg38) using Bowtie236 on Galaxy platform (https://usegalaxy.org). Mapped reads were transformed by bamCoverage with the parameter “normalize using RPKM” from the deepTools37. Heatmap was generated by plotHeatmap which also from the deepTools. Peak calling was performed with MACS2 callpeak.38 Gene list of peak calling was established by using R package CHIPseeker.39
Cell viability assay
The cytotoxicity of the class IIA HDAC inhibitor (TMP269) against various cell lines was assessed using the Cell Counting Kit-8 (Dojindo, Kumamoto, Japan) according to the provided instructions. Cells were plated at a density of 1×104 cells/well in 0.1 ml of RPMI medium in 96-well plates and then treated with different concentrations of each compound. After 72-hours of incubation with the compounds at 37°C, 8 µl of Cell Counting Kit-8 solution was added to each well. Following 1-hour incubation at 37°C, absorbance at 450 nm was measured using a microplate reader (CLARIOstar Plus, BMG LABTECH, Ortenberg, GER). Relative cell viability was calculated as the ratio of the absorbance in each treatment group to that of the corresponding untreated control group. Data are presented as mean ± standard deviation (SD) of more than three independent experiments.
Histone deacetylase enzyme activity measure assay
HDAC activity was evaluated using the HDAC-Glo™ I/II Assay and Screening System (Promega #G6420). 1×104 cells/well were seeded in a 96-well plate with 100 µL HDAC-Glo™ I/II buffer. Then, 100 µL HDAC-Glo™ I/II reagent and 1 µL developer reagent were added to each well. After incubation for 30 minutes at room temperature, luminescence was measured using the FLUOstar OPTIMA. Data are presented as mean ± standard deviation (SD) of more than three independent experiments.
Statistical analyses
GraphPad Prism 9 was used for statistical analyses. Unpaired Student's t-test (two-tailed) and Ordinary one-way ANOVA were used for pairwise comparisons of significance. The log-rank (Mantel-Cox) was used for the survival curves comparison. Animal experiments were neither blinded nor randomized. The type of replication (biological or technical) is indicated in figure legends. Sample size was decided based on our previous experience in the field, not predetermined by a statistical method. All data are shown as mean ± SD.