Skip to main content
Springer logoLink to Springer
. 2023 Apr 12;31(2):14. doi: 10.1007/s10577-023-09723-x

An essential role for the Ino80 chromatin remodeling complex in regulation of gene expression during cellular quiescence

Yasaman Zahedi 1, Shengyuan Zeng 1, Karl Ekwall 1,
PMCID: PMC10097750  PMID: 37043046

Abstract

Cellular quiescence is an important physiological state both in unicellular and multicellular eukaryotes. Quiescent cells are halted for proliferation and stop the cell cycle at the G0 stage. Using fission yeast as a model organism, we have previously found that several subunits of a conserved chromatin remodeling complex, Ino80C (INOsitol requiring nucleosome remodeling factor), are required for survival in quiescence. Here, we demonstrate that Ino80C has a key function in the regulation of gene expression in G0 cells. We show that null mutants for two Ino80C subunits, Iec1 and Ies2, a putative subunit Arp42, a null mutant for the histone variant H2A.Z, and a null mutant for the Inositol kinase Asp1 have very similar phenotypes in quiescence. These mutants show reduced transcription genome-wide and specifically fail to activate 149 quiescence genes, of which many are localized to the subtelomeric regions. Using spike in normalized ChIP-seq experiments, we show that there is a global reduction of H2A.Z levels in quiescent wild-type cells but not in iec1∆ cells and that a subtelomeric chromosome boundary element is strongly affected by Ino80C. Based on these observations, we propose a model in which Ino80C is evicting H2A.Z from chromatin in quiescent cells, thereby inactivating the subtelomeric boundary element, leading to a reorganization of the chromosome structure and activation of genes required to survive in quiescence.

Supplementary information

The online version contains supplementary material available at 10.1007/s10577-023-09723-x.

Keywords: Cellular quiescence, fission yeast, chromatin remodelling, histone variant, Ino80, H2A.Z, eviction, telomere, boundary element

Introduction

Cellular quiescence is a reversible dormant state in which cells are changing their metabolism and cytology to adapt to an environment that does not permit proliferation. The ability to exit the cell cycle and enter quiescence is essential for tissue development and homeostasis in multicellular organisms. It is also an important survival strategy for unicellular organisms in harsh conditions. When fission yeast, Schizosacharomyces pombe, cells are starved for nitrogen in the absence of cells of the opposite mating type, they stop dividing and enter a quiescent state. In this state, the cell cycle is halted at the G0 stage before DNA replication. In G0, cells adapt the metabolism to survive until a nitrogen source becomes available so that the cells can re-enter the cell cycle and start proliferating. The survival in quiescence depends on a global change in gene expression (Marguerat et al. 2012). RNA transcription is generally reduced; however, some genes need to be activated to cope with the new physiological situation, for example, authophagy genes, proteasome-encoding genes, and genes for hexose and amino acid transporters (Takeda et al. 2010) (Oya et al. 2019). The nuclei of quiescent fission yeast cells are dramatically reorganized presumably to accommodate these profound changes in gene expression (Sajiki et al. 2009). To gain more insight into the role of chromatin structure changes in this process, we conducted a genetic screen in fission yeast for genes required to maintain viability during cellular quiescence (Zahedi et al. 2020). The screen identified the Ino80 complex (Ino80C). Ino80 is the catalytic subunit of this large chromatin remodeling complex consisting of approximately 10–14 protein subunits depending on the species. In fission yeast Ino80C consists of the following core subunits: Ino80, Arp5, Arp8, Rvb1, Rvb2, Alp5 (Arp4), Act1, Ies2, Ies4, Ies6, and Taf14 (Tfg3), and the accessory subunits: Iec1, Hap2, Iec3, Iec5, and Nht1 (Hogan et al. 2010) (Shevchenko et al. 2008) (Shan et al. 2020). The Ino80C subunit Iec1 (Ino Eighty Complex subunit 1) in fission yeast is similar to the Ying-Yang 1 (YY1) subunit of the human Ino80C and the function of Iec1 is to recruit Ino80C to target genes (Hogan et al. 2010). We have previously shown that mutations in nine tested subunits of Ino80C: Ies6, Nht1, Iec1, Iec3, Tfg3, Arp8, Ies2, Ies4, and the putative Ino80C subunit Arp42, lead to quiescence mortality phenotypes (Zahedi et al. 2020). Mutations in Hap2, Iec1, Arp8, Iec3, Nht1, Arp5, Ies4, and Ies2 were recently shown to be short-lived in stationary phase in fission yeast, implicating Ino80C in chronological ageing (Romila et al. 2021). Thus, at least six Ino80C subunits: Iec1, Arp8, Iec3, Nht1, Ies4, and Ies2 are implicated both in survival in quiescence and chronological ageing.

The molecular function of Ino80C is to remove the histone variant H2A.Z from nucleosomes by a histone exchange mechanism with H2.A in a process driven by ATP hydrolysis (Papamichos-Chronakis et al. 2011). This function is important for the repair of double-strand breaks, DNA replication, and the regulation of transcription (Poli et al. 2017). The H2A.Z exchange mechanism may also involve RNA polymerase II (Pol II) activity (Ranjan et al. 2020). Ino80 has recently been reported to be involved in gene regulation in several different species. Ino80 is required to activate the transcription of genes involved in thermomorphogenesis in plants (Xue et al. 2021). This mechanism of gene activation by Ino80 in plants involves H2A.Z eviction in response to elevated temperature. In Candida albicans, Ino80 is required for hyphal development by H2A.Z eviction at hyphal genes (Zhao et al. 2022). In mouse embryonic stem cells, Ino80 is required for regulation of cell cycle transitions by activating cell cycle genes (Yoo et al. 2022). Hence, it is likely that the requirement for Ino80 during quiescence involves some aspect of transcription regulation.

In fission yeast, Ino80C was shown to be important also for histone H3 exchange (Singh et al. 2020). We found that the genes for H2A.Z (pht1) and histone H3 (hht2) are both essential for surviving quiescence suggesting that H2A.Z deposition or removal and new histone H3 expression are required to maintain viability in G0 (Zahedi et al. 2020). H2A.Z is deposited by the Swr1C complex (Swi2/Snf2-related ATPase) in fission yeast (Buchanan et al. 2009). However, mutations affecting Swr1C do not affect survival in quiescence, indicating that H2A.Z deposition in G0 is less important than its removal by Ino80C. The activity of Ino80C in budding yeast, Saccharomyces cerevisiae, is modulated by Inositol polyphosphates (Shen et al. 2003). Curiously, the inositol kinase, Asp1, was recently reported to be important for the survival of quiescence in fission yeast (Sajiki et al. 2018). To advance the understanding of the role of Inositol polyphosphates and histone exchange by Ino80C in fission yeast quiescence, here, we investigate the functions of Asp1, Iec1, Arp42, Ies2, H2A.Z, and histone H3 in the regulation of gene expression in G0 cells.

Results

Time course analysis of viability and RNA expression in G0

We performed RNA sequencing (RNA-seq) analysis in wild-type cells and in three Ino80C-related mutants: arp42∆, iec1∆, and ies2∆, in pht1∆ cells carrying a gene deletion for H2A.Z, in asp1∆ cells carrying a gene deletion for an inositol kinase, and in hht2∆ cells with a gene deletion for a histone H3 encoding gene. Cells were grown to the logarithmic phase in minimal medium and shifted to nitrogen-free minimal medium. Samples were taken for RNA extractions at time zero (T0, before shift) and after 24 h (T1D), 1 week (T1W), and/or 2 weeks (T2W) of nitrogen starvation. We measured the viability of the cultures at each time point using flow cytometry (Fig. 1, Table 1). All the cultures entered efficiently into G0, as judged from the percentage of cells 1C DNA content, within 24 h (T1D) from the shift to nitrogen-free medium (Table 2). Consistent with our previous observations the Ino80C mutants iec1∆∆ arp42∆, ies2∆ and pht1∆ showed a reduced viability after 1 week in G0 (Table 1). We also found that asp1∆ cells lost their viability after 1 week in quiescence. This is also in agreement with a previous report (Sajiki et al. 2018). However, hht2∆ cells displayed a milder mortality phenotype in quiescence, only showing reduced viability after 2 weeks.

Fig. 1.

Fig. 1

Measurements of viability and DNA content using flow cytometry. The gating strategy for measurement of viability and the proportion of G0 arrested cells during quiescence is illustrated with examples of FACS profiles for wild type (smt0) and the different mutant cells (as indicated)

Table 1.

Viability measurements by FACS

Genotype T0 T1D T1w T2w
smt-0 (wt) 99,9 ± 0,6 99,7 ± 0,2 98,8 ± 1,3 98,30 ± 0,3
hht2∆ 98,0 ± 5,8 96,5 ± 2,5 95,1 ± 1,0 92,7 ± 2,8
pht1∆ 96,0 ± 3,7 93,7 ± 0,3 85,0 ± 1,7 70,9 ± 11,5
iec1∆ 96,8 ± 2,3 92,6 ± 1,4 85,1 ± 1,7 68,1 ± 0,8
asp1∆ 99,5 ± 0,6 98,7 ± 0,1 95,1 ± 0,1 62,8 ± 3,7
arp42∆ 99,4 ± 0,5 98,7 ± 0,1 87,2 ± 1,1 75,4 ± 2,7
ies2∆ *99,0; 99,6 *98,0; 98,2 *78,0; 82,5 *68,2; 65,1

The percentage of viable cells is indicated

Mean value ± standard deviation (n = 3)

*shows two measurements

Table 2.

DNA content measurements by FACS

Genotype T1D T1w T2w
smt-0 (wt) 81,9 ± 1,91 83,3 ± 3,23 85,0 ± 0,36
hht2∆ 83,8 ± 1,07 86,4 ± 2,36 87,1 ± 2,51
pht1∆ 61,4 ± 13,1 71,6 ± 13,4 74,5 ± 11,1
iec1∆ 40,8 ± 11,5 40,8 ± 13,6 15,2 ± 0,42
asp1∆ 67,3 ± 0,80 71,4 ± 0,57 74,9 ± 0,66
arp42∆ 67,3 ± 0,85 71,9 ± 4,13 80,4 ± 1,51
ies2∆ *56,4; 56,9 *64,5; 64,8 *72,2; 70,6

The percentage of G0 cells is indicated (cells with a 1c DNA content)

Mean value ± standard deviation (n = 3)

*shows two measurements

A global repression of the transcriptome in G0 and an induction of subtelomeric genes

For RNA extractions, we used biological triplicates for wild type and each mutant. Because we expected a global change of transcription in quiescence, we could not normalize the RNA-seq data to the total number of reads, since it would give false negative and false positive results. Instead, we normalized the data to external RNA control consortium (ERCC) spike in controls that were added in proportion to the number of cells in each sample (Risso et al. 2014). It was previously shown that the fission yeast transcriptome is strongly downregulated in quiescence (Marguerat et al. 2012). Consistent with the earlier study, the ERCC normalization clearly showed that gene expression was globally repressed in wild-type cells after 1 and 2 weeks in G0 (Fig. 2). The hht2∆ cells showed a similar tendency as wild type. However, in pht1∆, iec1∆∆, arp42∆, ies2∆, and asp1∆ cells, the global repression had occurred already after 24 h (Fig. 2).

Fig. 2.

Fig. 2

A representation of the ERCC spike in normalized RNA-seq data. The box plot shows triplicate RNA-seq samples as log2 values of numbers of normalized sequence reads (Y-axis) in wild-type cells (smt0) and the different mutants (as indicated in the X-axis)

Next, a statistical threshold (FDR adjusted P < 0.05) was used to define up- and down-regulated genes. First, we defined genes affected by the shift to –nitrogen in wild type at T1D, T1W, and T2W compared to T0 (Table 3). These results are in nice agreement with earlier studies showing that most genes are downregulated and only a few genes are upregulated in G0. We compared the relatively few upregulated genes at the three time points in wild type (smt0) cells. There were 149 genes upregulated at T1D, 17 genes at T1W, and 21 genes at T2W. However, only 16 genes which were upregulated at all time points. Thus, we defined a set of 16 “core quiescence genes” that were found to be upregulated throughout the quiescence time course (Fig. 3A). Interestingly, 9 of these 16 genes (56.3%; CHI2 = 64; P < 0.001) reside in subtelomeric regions near tel1R and tel2L (Table 4). To validate the data, we compared with a previous quantitation of absolute numbers of mRNA molecules per cell (Marguerat et al. 2012). In all 16 cases, these measurements of mRNA molecules confirmed an induction of transcription in quiescent cells. Thus, our RNA-seq approach with ERCC normalization and a statistical cut off for affected genes was justified.

Table 3.

Number of genes differentially expressed in quiescent wild-type cells compared to vegetative cells

Comparison Up Down NS
T1D_T0_SMT0 149 1208 5255
T1W_T0_SMT0 17 6436 159
T2W_T0_SMT0 21 6360 231

The indicated gene lists were compared and the number of differentially expressed genes (up or down) are shown for each comparison (FDR adjusted P < 0.05)

RNA levels were normalized across all groups with ERCC spike-in controls

NS, not significant

Fig. 3.

Fig. 3

Analysis of gene expression changes during quiescence in wild type and Ino80C mutants. A Venn diagram comparing lists of genes upregulated in wild-type cells (smt0) at 24 h (T1D) 1 week (T1W) and 2 weeks (T2W) after removal of nitrogen. B Venn diagram comparing lists of genes downregulated in Ino80C mutants iec1∆, ies2∆ and arp42∆ with a list of genes upregulated in wild-type cells (smt0) at 24 h (T1D) after removal of nitrogen. C Venn diagram paring lists of genes downregulated in the Inositol kinase null mutant asp1∆ with iec1∆ and a null mutant for the histone variant H2A.Z (pht1∆) as well as the list of genes upregulated in wild-type cells (smt0) at 24 h (T1D) after removal of nitrogen

Table 4.

Features of 16 core quiescence genes

Geneid Gene name Quant. veg Quant. G0 Gene localization
SPNCRNA.821 0.038 0.17
SPAC22H10.13 zym1 2.7 17
SPAC3G9.11c pdc201 1.7 6.4
SPAC1F7.06 hsp3105 0.023 1.2
SPAC869.09 0.077 0.62 subtel1R
SPAC869.07c mel1 0.069 0.66 subtel1R
SPAC869.06c hry1 0 0.19 subtel1R
SPAC869.04 0.033 28 subtel1R
SPAC869.03c 0.025 10 subtel1R
SPBPB21E7.01c eno102 0.11 0.4 subtel2L
SPBPB21E7.02c 0 0.12 subtel2L
SPBPB21E7.10 0 0.11 subtel2L
SPBPB21E7.11 0.13 0.49 subtel2L
SPNCRNA.1364 0.079 0.94
SPNCRNA.1573 0.078 0.68
SPBC2G2.17c 0.52 3.6
SPNCRNA.577 0.016 4.6

Quantitative data (number of molecules per cell) from Marguerat, S.; Schmidt, A.; Codlin, S.; Chen, W.; Aebersold, R.; Bähler, J., Quantitative analysis of fission yeast transcriptomes and proteomes in proliferating and quiescent cells. Cell 2012, 151, 671–683

Next, we examined the 149 genes that were upregulated only at T1D, and a significant fraction of these genes are also localized close to telomeres. A total of 25 of the 149 genes (16.8%; CHI2 = 28,4; P < 0.001) are in 200 kb subtelomeric regions of chromosomes 1 and 2 (Table 5). Regarding the downregulated genes in G0, we found that as many as 1208 genes were significantly down after 24 h (T1D) and 6436 genes (including non-coding RNA genes) were down at T1W (Table 1). This represents 97.3% of the genome indicating that nearly the entire transcriptome is downregulated after 1 week in quiescence.

Table 5.

Genomic features of 149 quiescence genes. Subtelomeric genes on chromosomes I and II (< 200 kb from ends) are in bold letters

Gene id Chr Start End Gene_name T1_T0_SMT0_logFC T1_T0_SMT0 P-value
SPNCRNA.602 I 50851 51545 SPNCRNA.602 3,25973596 2,7274E-05
SPAC1F8.04c I 92387 93931 SPAC1F8.04c 3,53511935 3,0867E-06
SPNCRNA.607 I 99046 101140 fta5-antisense-1 2,7031025 0,00024108
SPAC11D3.19 I 106893 108361 SPAC11D3.19 3,01020676 4,5012E-05
SPAC11D3.03c I;I;I 110904;112098;112378 112046;112333;112499 SPAC11D3.03c 4,44873642 8,8882E-09
SPAC11D3.16c I 140381 141653 SPAC11D3.16c 2,48746727 0,000418
SPAC11D3.17 I;I 141199;141559 141510;144768 SPAC11D3.17 2,1184002 0,00225435
SPAC13G6.08 I 187701 190031 SPAC13G6.08 2,11089891 0,00512897
SPNCRNA.643 I 388352 389177 trm112-antisense-1 2,47596303 0,00370115
SPNCRNA.649 I 446175 446702 lsd2-antisense-1 2,79371868 0,00369267
SPAC23E2.03c I 450860 453603 ste7 3,25111192 7,4801E-06
SPNCRNA.673 I 664677 665246 rad15-antisense-1 2,42711956 0,00934543
SPNCRNA.690 I 829234 831663 prh1-antisense-1 3,67766921 0,00038004
SPNCRNA.220 I 1015475 1015830 SPNCRNA.220 3,3952792 0,0028625
SPAC56F8.15 I 1151431 1153638 SPAC56F8.15 1,96273702 0,00753558
SPNCRNA.737 I 1357621 1359568 SPNCRNA.737 1,93983373 0,00875406
SPAC1002.19 I 1835270 1837060 urg1 3,37759044 3,9743E-06
SPAC1399.03 I 1841908 1843995 fur4 1,81787099 0,00812566
SPNCRNA.178 I 1844130 1845339 SPNCRNA.178 2,90780759 0,00018621
SPAP11E10.02c I 1856833 1860727 mam3 2,58249425 0,00026631
SPAPB1A10.14 I 1890529 1892090 pof15 2,58471259 0,00031153
SPAC3C7.02c I 2064853 2066004 pil2 2,01101362 0,00384745
SPNCRNA.791 I 2066217 2067343 rad55-antisense-1 3,26641538 7,9496E-05
SPAC20H4.11c I;I 2131685;2132545 2132426;2133066 rho5 2,23485497 0,00175906
SPAC13F5.07c I;I 2184654;2185472 2185282;2185990 hpz2 2,95890939 6,5014E-05
SPAC13G7.13c I 2318492 2322664 msa1 2,01610833 0,00357637
SPNCRNA.821 I 2380416 2381234 SPNCRNA.821 5,22655651 2,8497E-07
SPAC22H10.13 I 2381690 2382200 zym1 5,19708373 9,4432E-11
SPNCRNA.194 I 2424735 2425515 SPNCRNA.194 2,33292483 0,00869259
SPNCRNA.43 I 2432340 2432921 prl43 3,36282112 0,00071322
SPAC4A8.04 I 2544815 2546981 isp6 2,06781376 0,00291872
SPNCRNA.835 I 2547093 2547449 SPNCRNA.835 2,20795116 0,0071483
SPSNRNA.06 I;I 2562276;2562374 2562323;2562427 snu6 1,87091948 0,00697876
SPNCRNA.198 I 2703965 2704030 SPNCRNA.198 2,78396522 0,00553514
SPNCRNA.853 I 2784605 2785492 SPNCRNA.853 4,11372383 3,3265E-07
SPNCRNA.860 I 2931622 2932800 SPNCRNA.860 5,01730583 3,0063E-09
SPAC2E1P3.04 I 2931763 2935115 cao1 2,22542246 0,00144898
SPAC31G5.09c I 2999546 3001687 spk1 1,84222622 0,00732841
SPNCRNA.71 I 3001761 3001930 SPNCRNA.71 2,61362541 0,00116616
SPNCRNA.877 I 3032579 3033397 SPNCRNA.877 2,8566656 0,00067403
SPAC24C9.16c I;I;I;I 3042749;3042872;3043096;3043225 3042817;3042909;3043157;3043380 cox8 1,88137663 0,00786675
SPAC3G9.11c I 3159753 3162245 pdc201 4,74996563 1,3887E-09
SPAC6G10.06 I;I;I 3226929;3227042;3227456 3226980;3227383;3228192 SPAC6G10.06 2,51110956 0,0013486
SPNCRNA.931 I 3731313 3732279 grx2-antisense-1 2,91994619 0,0014984
SPNCRNA.935 I 3745778 3748833 SPNCRNA.935 3,9191636 1,7498E-07
SPATRNAGLU.04 I 3776768 3776839 SPATRNAGLU.04 1,95115116 0,01020029
SPNCRNA.955 I 4004553 4005632 SPAC27E2.02-antisense-1 2,33505779 0,00572603
SPSNORNA.13 I 4154912 4155002 snoR69b 2,03181845 0,00326288
SPAC25B8.13c I 4179961 4183074 isp7 2,4050579 0,00062889
SPAC1F7.06 I 4230615 4232557 hsp3105 5,80756074 2,7966E-11
SPNCRNA.243 I 4231812 4232976 SPNCRNA.243 2,57917641 0,00141246
SPAC9E9.17c I;I;I;I 4437311;4437606;4437732;4437860 4437401;4437648;4437813;4437862 SPAC9E9.17c 5,21541272 8,4446E-09
SPNCRNA.987 I 4437466 4438921 SPAC9E9.17c-antisense-1 2,17485116 0,00635584
SPNCRNA.12 I 4439118 4439451 prl12 3,78490716 0,00470787
SPAC17C9.16c I;I 4472472;4473325 4473271;4474744 mfs1 1,83115795 0,00796667
SPAC27D7.03c I 4510983 4515015 mei2 2,22978588 0,00141295
SPNCRNA.993 I 4515182 4515993 SPNCRNA.993 2,45668478 0,00117438
SPAC11H11.04 I 4780224 4781922 mam2 4,13282167 4,892E-08
SPAC4F10.17 I 4868326 4869283 SPAC4F10.17 3,91344782 1,839E-06
SPAC4F10.22 I 4878241 4878453 cmc4 2,0237062 0,00648228
SPAPB8E5.04c I 4913971 4915513 npc2 1,84500451 0,00745054
SPAPB8E5.05 I;I;I 4916603;4916921;4917286 4916841;4917214;4917521 mfm1 7,30766685 1,2375E-16
SPAPJ691.02 I 5187647 5189187 SPAPJ691.02 4,53526941 4,6374E-09
SPAC19D5.07 I 5222447 5224795 uga1 1,88556666 0,00622319
SPAC2H10.01 I 5273012 5275983 SPAC2H10.01 2,6493213 0,00027561
SPNCRNA.1068 I 5309027 5311689 SPNCRNA.1068 2,30679984 0,00316561
SPNCRNA.1075 I 5353770 5354310 new12-antisense-1 3,31258143 9,0267E-05
SPAC1039.03 I 5452105 5453867 SPAC1039.03 2,12458926 0,00219715
SPAC1039.09 I 5465377 5468649 isp5 2,15393594 0,00202143
SPAC1039.10 I;I;I 5470432;5470574;5471284 5470525;5471227;5471520 mmf2 1,86772985 0,00694076
SPAC922.09 I 5482662 5482877 SPAC922.09 5,12304127 4,3744E-05
SPAC922.06 I 5485096 5486287 SPAC922.06 2,20990467 0,00153079
SPNCRNA.1092 I 5495270 5496597 SPNCRNA.1092 2,16976747 0,00364619
SPAC869.09 I 5496844 5497448 SPAC869.09 4,67744479 8,7417E-06
SPAC869.08 I 5497918 5499280 pcm2 4,23373637 4,4033E-07
SPAC869.07c I 5499768 5501236 mel1 4,70824039 6,0466E-09
SPAC869.06c I 5502560 5503291 SPAC869.06c 3,4837771 0,00042619
SPAC869.04 I 5511137 5512369 SPAC869.04 9,23276947 6,5934E-22
SPAC869.03c I 5512757 5514865 SPAC869.03c 9,16835946 9,8841E-22
SPAC869.01 I 5521275 5523181 SPAC869.01 3,00278941 0,00011382
SPBPB21E7.02c II;II 60553;61119 61107;61205 SPBPB21E7.02c 10,5415628 0,00031848
SPBPB21E7.10 II;II 61362;61632 61526;62126 SPBPB21E7.10 10,3509696 0,00048232
SPBPB21E7.11 II;II;II 61526;62598;62944 62449;62885;63086 SPBPB21E7.11 4,55305877 2,1632E-06
SPBC1683.05 II 147915 150574 SPBC1683.05 1,81584673 0,00821874
SPBC1271.09 II;II 350692;350787 350740;352369 tgp1 2,42462905 0,00053769
SPNCRNA.1346 II 359391 359866 SPNCRNA.1346 3,2706138 0,00044942
SPSNRNA.04 II 467233 467361 snu4 3,23066699 0,00022388
SPNCRNA.1364 II 503961 505097 SPNCRNA.1364 4,8789878 5,6148E-10
SPBC1685.05 II 504397 509056 SPBC1685.05 2,22522485 0,0014316
SPNCRNA.66 II 547325 547761 prl66 2,67117747 0,0054103
SPBC354.12 II;II 578063;578725 578181;580056 gpd3 2,77080717 9,979E-05
SPBPJ4664.03 II;II 700291;700544 700476;700700 mfm3 7,90991958 3,2907E-18
SPNCRNA.1405 II 1013315 1014054 prp2-antisense-1 3,05365283 0,00040845
SPNCRNA.1415 II 1150424 1150863 SPNCRNA.1415 6,49255957 7,8126E-08
SPSNORNA.21 II 1308634 1308745 snoU14 3,06732048 1,982E-05
SPNCRNA.15 II 1329046 1329760 prl15 7,65836499 9,1712E-11
SPBC83.19c II 1541452 1541903 SPBC83.19c 11,1826139 4,0742E-05
SPNCRNA.352 II 1542196 1542364 SPNCRNA.352 11,0312641 6,4796E-05
SPBC29B5.02c II 1550293 1553467 isp4 2,76293742 0,00010681
SPNCRNA.1443 II 1554843 1557638 SPNCRNA.1443 2,04831502 0,00323332
SPBC1D7.02c II 1752100 1755619 scr1 2,04685207 0,0032455
SPBC9B6.03 II 1817454 1819848 SPBC9B6.03 2,88027667 5,562E-05
SPBC23G7.11 II 2120231 2121050 mag2 2,67196421 0,00085483
SPNCRNA.1512 II 2417501 2418682 pvg3-antisense-1 3,40867882 0,00292084
SPNCRNA.1525 II 2543249 2544500 psm1-antisense-1 3,50755144 1,6382E-05
SPNCRNA.1530 II 2580400 2581182 SPNCRNA.1530 9,90488058 0,0021415
SPBC25B2.08 II 2611366 2612917 SPBC25B2.08 2,18518869 0,00197078
SPBC19C7.04c II 2825251 2826700 SPBC19C7.04c 2,71034397 0,00015799
SPNCRNA.1564 II 2934727 2935979 SPBC1703.11-antisense-1 2,52684517 0,00180399
SPNCRNA.1573 II 3020526 3022271 SPBC15D4.05-antisense-1 6,22415483 4,9082E-12
SPNCRNA.413 II 3150075 3150475 SPNCRNA.413 2,82207927 0,00847413
SPBC13A2.04c II 3405575 3408492 ptr2 2,25820836 0,00121891
SPBC2G2.17c II 3466270 3467797 SPBC2G2.17c 5,69625442 5,1176E-12
SPBC887.16 II 3574869 3575250 SPBC887.16 2,87571225 0,00131414
SPSNORNA.27 II 3654333 3654416 snoR47 2,08042095 0,0026702
SPNCRNA.1660 II 4049029 4049473 SPBC215.10-antisense-1 2,32255749 0,00367613
SPBC1347.11 II 4081869 4083198 sro1 2,14480969 0,00202154
SPNCRNA.1670 II 4109027 4109600 SPNCRNA.1670 5,26102836 1,1998E-06
SPNCRNA.577 II 4255755 4259164 SPBC1652.02-antisense-1 5,33538848 2,9985E-11
SPNCRNA.451 III 49802 50142 SPNCRNA.451 4,52899464 1,8957E-08
SPCC757.13 III 77080 79816 SPCC757.13 2,56643263 0,00028782
SPNCRNA.1126 III 289992 293300 SPCC553.08c-antisense-1 2,13247264 0,00505195
SPNCRNA.1137 III 394357 396657 ubp16-antisense-1 3,60875293 7,5933E-06
SPCC1183.12 III;III 604366;604857 604805;604961 spo13 2,01450177 0,00823742
SPNCRNA.1160 III 815167 816476 SPCC1393.09c-antisense-1 3,32393748 0,00134978
SPCPB16A4.06c III;III 956012;957195 957014;957627 SPCPB16A4.06c 1,91007479 0,00587734
SPCC550.07 III 1196729 1199999 SPCC550.07 3,43129476 2,8671E-06
SPCC550.10 III 1204315 1206695 atd3 2,60470778 0,00022774
SPCC338.18 III 1340628 1341847 SPCC338.18 2,64277685 0,00033584
SPCC1281.04 III 1386524 1387961 SPCC1281.04 2,38484333 0,00223696
SPCC188.12 III;III 1504862;1505062 1504999;1506851 spn6 4,13670216 1,5614E-07
SPNCRNA.51 III 1514062 1514343 prl51 3,34790704 0,00037929
SPCC584.13 III 1516197 1518230 SPCC584.13 2,22673511 0,00137358
SPNCRNA.1205 III 1559743 1561240 SPNCRNA.1205 5,35400218 1,6137E-05
SPCC417.02 III 1669944 1671551 dad5 4,36062922 1,3103E-08
SPNCRNA.1215 III 1671883 1674238 SPCC417.03-antisense-1 2,29927601 0,00099408
SPCC417.06c III;III;III 1677093;1678329;1678828 1678286;1678778;1679436 mug27 3,07341949 0,00197289
SPCC417.10 III 1694169 1697592 dal51 1,98405831 0,00416259
SPCC1450.07c III;III 1736500;1738213 1737778;1738318 dao1 2,1507211 0,00212075
SPCC1442.01 III 1765913 1768971 ste6 2,32561278 0,00091354
SPCC1223.09 III 1857150 1858621 SPCC1223.09 2,18680094 0,00172594
SPNCRNA.1236 III 1859963 1860577 SPNCRNA.1236 1,78076961 0,00932741
SPCC74.04 III 1935474 1938754 SPCC74.04 2,86776786 6,048E-05
SPCC576.01c III 2079421 2080662 xan1 4,08792138 6,3569E-08
SPCC830.04c III 2186723 2187154 mug128 2,84134074 0,00746144
SPCC965.13 III 2310096 2314400 SPCC965.13 2,22721546 0,00139801
SPCC70.04c III;III;III 2352568;2353746;2353974 2353701;2353913;2354572 SPCC70.04c 2,53908379 0,00031576
SPNCRNA.519 III 2362179 2362725 SPNCRNA.519 2,33386995 0,0013251
SPCC569.09 III;III 2413,905;2414158 2414089;2414790 SPCC569.09 274,408957 0,00012036

Transcription changes in G0 and reduced viability of Asp1, Ino80C, and H2A.Z mutants

Next, we analyzed the changes of the G0 transcriptome in the mutants. It was clear already by looking at the total number of reads after ERCC normalization that all mutants showed overall changes in the G0 transcriptome as compared to wild type (Fig. 2). The overall tendency was that the mutants showed a further reduction of global transcription as compared to the wild type. The genes affected in the different mutants at each time point as compared to the wild type were defined (Table 6). This analysis revealed substantial changes in gene expression in all the tested mutants. Again, hht2∆ cells showed a weaker phenotype at T1D and T1W compared to the other mutants, but 1286 genes were downregulated in hht2∆ cells at T2W. The hht2+ gene is constitutively expressed in contrast to the other two histone H3-encoding genes (hht1+ and hht3+) in fission yeast which are strictly expressed during the S phase (Takayama and Takahashi 2007). Thus, hht2+ is the sole histone H3 gene expressed in G0 cells. The viability of hht2∆ cells drops significantly compared to wild type after 2 weeks in quiescence (TTEST; P = 0.013) and is correlated with reduced transcription (Table 1, Fig. 2, Table 6). It is possible that this reduction of transcription, caused by reduced histone H3 levels, is contributing to the mortality of hht2∆ cells in G0.

Table 6.

Number of genes differentially expressed in mutant cells compared to wild-type cells

Comparison Up Down NS
HHT2_SMT0_T0 2 6 6605
PHT1_SMT0_T0 5 9 6599
IEC1_SMT0_T0 20 11 6582
ASP1_SMT0_T0 26 19 6569
ARP42_SMT0_T0 9 114 6490
IES2_SMT0_T0 9 29 6575
HHT2_SMT0_T1D 91 1 (hht2) 6521
PHT1_SMT0_T1D 6 6143 464
IEC1_SMT0_T1D 8 6194 410
ASP1_SMT0_T1D 2 6368 243
ARP42_SMT0_T1D 0 6519 94
IES2_SMT0_T1D 7 6048 558
HHT2_SMT0_T1W 1 1 6611
PHT1_SMT0_T1W 29 1 6583
IEC1_SMT0_T1W 88 0 6525
ASP1_SMT0_T1W 230 3504 2879
ARP42_SMT0_T1W 5 6145 463
IES2_SMT0_T1W 24 6 6583
HHT2_SMT0_T2W 190 1129 5294
PHT1_SMT0_T2W 16 33 6564
IEC1_SMT0_T2W 43 78 6492
ASP1_SMT0_T2W 144 1898 4571
ARP42_SMT0_T2W 5 5238 1370

The indicated gene lists were compared and the number of differentially expressed genes (up or down) are shown for each comparison (FDR adjusted P < 0.05)

RNA levels were normalized across all groups with ERCC spike-in controls

NS, not significant

In contrast to hht2∆, pht1∆ cells show strong changes of the transcriptome already at 24 h, i.e., prior to the reduction in viability that occurs after 1 week (Fig. 1, Fig. 2, Table 6). This observation is consistent with a key role for the histone variant H2A.Z in gene regulation, being essential for survival in quiescence. Ino80C is required for the removal of H2A.Z by its chromatin remodeling activity driven by ATP hydrolysis (Papamichos-Chronakis et al. 2011). Our results show that null mutations in three Ino80C-related genes, arp42∆, iec1∆, and ies2∆ cause a massive reduction of the G0 transcriptome after 24 h as compared to the wild type, and arp42∆ shows a further reductions of gene expression after one and 2 weeks. Thus, a vast majority of genes are prematurely downregulated in Ino80C mutants compared to wild type (Fig. 2, Table 6). Although the observed transcription patterns are not strictly correlated to mortality, the generally reduced transcription is likely explaining the essential role of Ino80C in cellular quiescence that we previously observed (Zahedi et al. 2020).

Failure to induce quiescence genes in Ino80C mutants

Next, we studied the behavior of the larger set of 149 genes that are upregulated in the wild type after 24 h in nitrogen starvation (T1D). We found that none of these genes were upregulated in arp42∆, iec1∆, and ies2∆ cells (Fig. 3B). In fact, all the 149 genes were downregulated in these mutants compared to wild type (smt0) after 24 h in G0. This shows that Ino80C is essential for the activation of these genes in response to nitrogen starvation. Gene ontology analysis revealed that the list of 149 upregulated genes is significantly enriched for several GO terms including the fungal vacuole, amino acid, dipeptide, and nucleobase transmembrane transport (Table 7). The vacuole is required in quiescence for the autophagy process to recycle amino acids, and these transmembrane transport processes are crucial for survival during cellular quiescence. Thus, the upregulated expression of these genes is required for survival in G0 by adapting the cellular metabolism. For example, the SPAC11D3.16c gene located in near tel1L is annotated as being essential for viability in G0 (Harris et al. 2021). Two genes near tel1R encoding membrane transporters, isp5+ and SPAC869.03c fail to be activated in Ino80C mutants. Therefore, it is conceivable that the reason Ino80C mutants are dying in G0 is due to the inability to activate genes needed for the metabolic change and cellular uptake that normally occur during quiescence.

Table 7.

Gene ontology analysis of 149 genes induced in quiescent wild-type cells after 24 h

GO Term N Gene list T1D_T0_SMT0_up P-value
GO:0,000,750 pheromone-dependent signal transduction involved in conjugation with cellular fusion 14 5 9,23456E-06
GO:0,035,442 dipeptide transmembrane transport 3 3 1,12193E-05
GO:0,071,916 dipeptide transmembrane transporter activity 3 3 1,12193E-05
GO:0,043,864 indoleacetamide hydrolase activity 3 2 0,001,491,016
GO:0,015,205 nucleobase transmembrane transporter activity 3 2 0,001,491,016
GO:0,007,267 cell–cell signaling 4 2 0,002,937,898
GO:0,000,772 mating pheromone activity 4 2 0,002,937,898
GO:0,000,324 fungal-type vacuole 68 6 0,004,105,498
GO:0,035,673 oligopeptide transmembrane transporter activity 5 2 0,004,824,158
GO:0,003,333 amino acid transmembrane transport 17 3 0,006,047,446
GO:0,031,520 plasma membrane of cell tip 20 3 0,009,650,323
GO:0,006,878 cellular copper ion homeostasis 9 2 0,016,367,608

Gene Ontology (GO) analysis of GO terms obtained from PomBase

Analysis done on the upregulated genes of the wild type (SMT0) cells between T0 and T1D

N column, the number of genes in the GO

A role of Ino80 and H2A.Z in activation of quiescence genes

To get some insights into the role of H2A.Z in quiescence, we compared the list of downregulated genes in the pht1∆ mutant at T1D with the gene lists of Ino80C mutants, arp42∆ and iec1∆, and a list of genes induced in the wild type at T1D. This comparison revealed that all of the 149 genes that are upregulated in wild type (smt0) at T1D fail to be induced in pht1∆ cells lacking H2A.Z (Fig. 3C). Also, there is a very strong overlap genome-wide between genes downregulated in pht1∆ cells and those downregulated in arp42∆ and iec1∆ mutants. A total of 5951 genes were downregulated in all three mutants. Based on this and our observations, we concluded that H2A.Z and Ino80C are both somehow required for the activation of genes induced in G0, in particularly in subtelomeric regions.

Changes of H2A.Z localization in quiescent cells at subtelomeric LTR boundary elements

The molecular function of Ino80C is to remove H2A.Z in a nucleosome disassembly mechanism in which H2A.Z is exchanged with H2A (Papamichos-Chronakis et al. 2011). Furthermore, Ino80 has been shown to evict H2A.Z in diverse organisms leading to changes in gene expression. To test if the observed changes in gene expression in quiescent cells depend on H2A.Z, we performed ChIP-seq of epitope tagged H2A.Z (pht1-myc) in wild type and iec1∆ cells. We used the Drosophila spike in chromatin methodology to allow measurement of global changes of H2A.Z occupancy by ChIP-seq (see the “Materials and methods” section). The total number of matched reads after the spike in normalization revealed a strong and significant reduction of H2A.Z-myc occupancy in quiescent wild type (smt0) cells at T1D compared to vegetative wild-type cells (T0), but not redusction was observed in quiescent iec1∆ cells (Fig. 4A). The chromosomal browser view confirmed this observation showing very low signals of H2A.Z-myc along all three chromosomes in wild-type cells at T1D, whereas signals remained high in iec1∆ cells (Fig. 4B). Thus, we conclude that there is an eviction process of H2A.Z from chromsosomes in quiescent cells, which is dependent on Ino80C.

Fig. 4.

Fig. 4

ChIP-seq analysis of H2A.Z localisation at a subtelomeric boundary element in vegetative (T0) and quiescent cells (T1D). A Quantitation of total number oif ChIP-seq reads for H2A.Z-myc. The bar diagram shows the total number of reads (after spike-in normalization) in wild-type cells (smt0) and the iec1∆ mutant at the vegetative stage (T0) and 24 h after removal of nitrogen (T1D). The error bars represent Standard deviation (SD) values from triplicate samples. Unpaired t-test was used to determine data significance. *P < 0.05, **P < 0.01. B Browser images of chromosomes I, II and the LTR boundary element at tel2L. The IGV genome browser tracks present chromosome 1, chromosome 2 and LTR boundary at chromosome II subtelomeric region (chrII: 93,039–102,657). C Quantitation of ChIP-seq reads for H2A.Z-myc in subtelomeric regions. The bar diagram shows the reads in 4 subtelomeric region, chrI:1–200,000, chrI:5,379,134–5,579,133, chrII:1–200,000 and chrII:4,339,805–4,539,804. The error bars represent SD values from triplicate samples. Unpaired t-test was used to determine data significance. **P < 0.01. D Quantitation of ChIP-seq reads for H2A.Z-myc the LTR boundary element at tel2L. The bar diagram shown the reads in LTR boundary element at chromosome II subtelomeric region (chrII: 93,039–102,657). The error bars represent SD values from triplicate samples. Unpaired t-test was used to determine data significance. *P < 0.05, **P < 0.01. E Model for regulation of the activity of the tel2L boundary element. For details see Discussion

Next, we examined the subtelomeric regions for changes of H2A.Z localization in quiescent cells (Fig. 4C). As illustrated by the bar diagrams subtelomeric regions (0–200 kb) of chromosomes I and II had a significant reduction (TTEST; P < 0.01) of H2A.Z in wild-type cells at T1D. Again this reduction was not observed in iec1∆ cells at T1D. Furthermore, it was clear that H2A.Z is localized in four peaks to the long terminal repaet (LTR) containing subtelomeric boundary element near tel2L (Fig. 4B; bottom). The activity of this boundary element has previously been shown to be maintained via Fft3 (Fun thirty homolog 3) in vegetative cells (Strålfors et al. 2011) (Steglich et al. 2015). In vegetative cells (T0), we observed a small but significant increase of H2A.Z at the tel2L boundary in iec1∆ cells compared to wild type (Fig. 4D). However, in quiescent wild-type cells, the H2A.Z peaks were strongly reduced (TTEST; P < 0.01) whereas the peaks could still be detected in quiescent iec1∆ cells (Fig. 4D). It was not possible to investigate if Ino80C also plays a role at the he other subtelomeric boundary elements since they are not cleary defined (Steglich et al. 2015). Taken together, this suggests that Ino80 is involved in the eviction or relocalization of H2A.Z genomewide, in subtelomeric regions of chromosomes I and II, and at a subtelomeric LTR boundary element (tel2L) especially during quiescence.

Discussion

A role for Ino80 in quiescence and ageing?

We have shown that Ino80C is required for the expression of genes in quiescence, including the hsp3105+ gene, encoding a ThiJ domain protein implicated in autophagy and oxidative stress resistance (Table 4). This gene was shown to be required for survival in the stationary phase (Su et al. 2015). Assuming that Ino80C drives expression of hsp3105+ in the stationary phase, then it could explain the short chronological lifespan phenotype that was reported for Ino80C mutants (Romila et al. 2021). It would also suggest that there is some commonality between chronological ageing in stationary phase and quiescence.

A possible role for Inositol polyphosphates restricting Ino80C activity in fission yeast?

Asp1 encodes an inositol kinase that may affect the activity of Ino80C. Inositol polyphosphates are synthesized by a series of enzymes including Asp1. The Asp1 kinase generates one specific inositol polyphosphate, IP8, and in fission yeast asp1∆ mutants, IP8 levels are strongly reduced whereas IP6 and IP7 levels are increased (Pascual-Ortiz et al. 2018). In budding yeast IP6 directly inhibits Ino80C (Shen et al. 2003). Thus, our results showing a similar effect on the G0 transcriptome and G0 mortality phenotypes between asp1∆ and Ino80C mutants and the strong overlap of downregulated genes (Fig. 3B) suggest that inositol polyphosphates IP6 and IP7, which accumulate in the asp1∆ mutant, may inhibit Ino80C activity also in fission yeast.

A change in nuclear organization in quiescence mediated by Ino80C?

In fission yeast, all four telomeres of chromosomes I and II, i.e., tel1L, tel1R, tel2L, and tel2R, form a peripheral cluster near the nucleolus in quiescent cells (Maestroni et al. 2020). It was previously shown that genes induced during meiosis and sporulation are enriched in subtelomeric regions (Mata et al. 2002). It is conceivable that Ino80C is involved in the formation of a transcriptionally active nuclear compartment comprising subtelomeres in response to nitrogen starvation or cellular quiescence when a mating partner is absent. Expression of this active compartment may support for mating and sporulation if cells of the opposite mating type are present. In agreement with this notion, the iec1∆ mutant was reported to show a decreased mating efficiency (Hogan et al. 2010). In budding yeast, Ino80C and its ATPase activity are required for chromosomal movements within the nucleus (Neumann et al. 2012). Hence, it is possible that the formation of this actively transcribed subtelomeric nuclear compartment, in response to nitrogen starvation, involves chromatin movements facilitated by Ino80C within the nucleus.

Based on our new results, we propose a model in which Ino80C activity, possibly modulated by inositol kinase Asp1, is required to remove H2A.Z from chromatin by a nucleosome disassembly mechanism in quiescent cells (Fig. 4E). This includes H2A.Z eviction at a subtelomeric boundary element leading to inactivation of the boundary and gene expression of subtelomeric genes, including transmembrane transporter genes, required to survive in quiescence. We hypothesize that this process may involve a drastic reorganization of chromosome structures in quiescent cells and clustering of telomeres to maintain an active nuclear compartment. Interestingly, it is known that fft3∆ cells have a reduced efficiency of to enter and exit quiescence (Sajiki et al. 2018; Zahedi et al. 2020). This is probably due to a failure in maintaining and restoring the subtelomeric boundary elements during these cellular transitions. To speculate further, the activation of quiescence genes by Ino80C is likely linked to the observed reduction of subtelomeric heterochromatin regions that occurs in quiescent cells (Oya et al. 2019). Consistent with this notion, it is known from studies in budding yeast that H2A.Z incorporation into acetylated chromatin by the SWR1-C complex is maintains heterochromatin boundary activity at silenced HMR loci and near telomeres (Zhou et al. 2010). Hence, in two distinct yeast species, H2A.Z is involved in maintaining heterochromatin boundaries.

In Drosophila, insulator boundary elements bound by the CTCF protein play important roles during development by partitioning the genome into distinct topologically associating domains (TADs), for example, in the Antennopedia gene complex where they prevent inappropriate enhancer promoter interactions between TADs (reviewed by (Batut et al. 2022)). The fission yeast genome is also organized into cohesion dependent TAD-like structures in vegetative cells (Mizuguchi et al. 2014). It is therefore tempting to speculate that the subtelomeric TAD structures are drastically reorganized in quiescent fission yeast cells is response to H2A.Z removal by Ino80. Finally, H2A.Z was recently implicated in regulating CTCF binding to chromatin by modulating the unwrapping of nucleosomes in mouse ES cells (Wen et al. 2020). Yeast cells do not have a CTCF protein; however, it is plausible that H2A.Z has a conserved function at boundary elements related to nucleosome disassembly both in unicellular and multicellular eukaryotes.

Materials and methods

Yeast strains and media

All five null mutants, hht2∆, asp1∆, iec1∆∆, arp42∆, pht1∆, and ies2∆ were derived from the version 5 Bioneer library, i.e., a large collection of gene deletion mutants carrying the kanMX4 cassette marking the gene deletion and leu1-32 ade6-M216/M210 ura4-D18 auxotrophic markers. The Bioneer strains cannot survive under the absence of nitrogen. Therefore, to produce prototrophic null mutant strains, the Bioneer strains were crossed with the Hu2843 mat1-M smt0 wild-type strain using standard methods (Ekwall and Thon 2017). The prototrophic mutants produced from each cross were selected using Edinburgh Minimal Medium (EMM) minus leucine, adenine, and uracil and subsequently YES medium containing G418 (150 ug/mL). The resulting mutant strains were named Hu3103 smt0 hht2∆ kanMX4, Hu3101 smt0 asp1∆ kanMX4, Hu3104 smt0 iec1∆ kanMX4, Hu3100 smt0 arp42∆ kanMX4, Hu3102 smt0 pht1∆ kanMX4, and Hu3113 smt0 ies2∆ kanMX4. The epitope tagged H2A.Z (pht1-myc) strains were produced from a cross using parental strains from (Buchanan et al. 2009) and Hu2843 resulting in the Hu3110 smt0 iec1D::ura4 pht1-myc and the Hu3112 smt0 pht1-myc strains.

All strains were grown in semi-solid YES complete media for 2 days 30 ℃ for 48 h and were regrown in liquid Pombe minimal glutamate medium (PMG) + nitrogen in a 200 ml flask to reach 106 cells /ml using a shaking incubator at 200 rpm at 30 °C. Before washing the cells, take 50 ml of culture as time 0 investigation and the rest of culture washed with 200 ml pre-warmed PMG-N and incubated them in 200 ml of PMG-N media then incubated (shaking incubator at 200 rpm at 30 °C).

Flow cytometry and viability measurements

For flow cytometry analysis (FACS) four time points were considered in this study, T0 (before shift to -nitrogen), T1D (24 h after shift), T1W (1 week after shift) and T2W (2 weeks after shift). For T0 and T1D 50 ml of culture and for T1D and T2W, 100 ml of culture was used. For each time point cells were pelleted and transferred into a 96-round bottom well plate and washed with 200 l of PBS (centrifuged at 400 g, 5 min, at room temperature) and stained with 150 l of Live-or-Dye™-Fixable Viability Stain (Biotium, Fremont, CA, USA). This stain was used at 1/1000 dilution in PBS, in the dark, and incubated for 30 min on ice with mild shaking. Then, cells were washed with 200 l PBS and centrifuged (400 g, 5 min, at room temperature) followed by a fixation step using 200 l of 70% ethanol and incubated for 30 min on ice in the dark. After washing with 200 l PBS, cells were incubated for 15 min in 200 l sodium citrate buffer (50 mM sodium citrate, pH 7.0), washed once with 200 l sodium citrate buffer, pelleted and resuspended in 200 l sodium citrate buffer containing 0.2 mg/ml DNAse-free RNase A (Roche diagnostics Scandinavia, Solna, Sweden, 10,109,169,001) and incubated for 3 h at 37 °C. Then, cells were stained with 100 l of PBS containing 12.5 mg/ml propidium iodide (PI) (Invitrogen AB, Stockholm, Sweden, P4864) by incubating for 30 min at room temperature in the dark. Before FACS analysis, 100 μl of PBS was added into each well and the 96-well plate was immediately analyzed using the multiplex flow-cytometer CytoflexS (Beckman Coulter) and the CytExpert software (www.mybeckman.se). Slow mode running was used to collect and run the samples and the data was recorded based on 20,000 events of live cells in each sample. The total number of cells was selected via forward (FSC) and side (SSC) scattering, and single cells were sorted via FSC vs FSH (height) to exclude doublets. Then, the live cell population was selected via negative signal of Live-or-Dye™ Fixable Viability Staining λEx/λEm 642/662 nm through the FSC-A vs FL3A (R660) channels (FL3A::660A). The DNA content histogram analysis and cell cycle population analysis were performed on live cells population using the signals of PI staining using the gating strategy described in (Zahedi et al. 2020). At the T0 time point, the mononuclear G2 cell population was selected through the total area of DNA signal (DNA-A) vs. the width of the DNA signal (DNA-W), and in quiescence, G0 cells with 1C DNA content were selected via same gating (DNA-A negative, DNA-W negative). A minimal cut-off of 1000 single cells was considered for each sample measurement. The selected data was analyzed via FlowJo software version 9 (https://www.flowjo.com/solutions/flowjo/downloads).

RNA isolation.

Wild type and mutant strains were grown in a 200 ml liquid PMG + N medium using a shaking incubator (200 rpm at 30 °C) to reach between 1.0 × 106 and 10 × 106 cells/ml. For each culture, 100 ml was removed for the T0 timepoint, and the rest of the culture was washed with pre-warmed PMG-N and incubated for 24 h in 500 ml of pre-warmed PMG-N using a shaking incubator (200 rpm at 30 °C). For RNA extraction, cells were washed with ice-cold PBS and resuspended in 500 l of ice-cold RNA extraction buffer (10 mM Tris–HCl pH 8.0, 1 mM EDTA, 2% Triton X-100, 1% SDS, 100 mM NaCl). Then we added 500 µl of Phenol (acidic phenol pH 4.5, Sigma) and 500 µl of glass beads (acid washed, Sigma). The tubes were vortexed vigorously and incubated at 65 °C for 45–60 min. Next, the tube was placed on ice for 5 min and centrifuged (1300 g, 5 min, 4 °C). The upper aqueous part was collected and transferred to a tube with 500 µl of chloroform (Sigma Aldrich), vortexed and centrifuged (1300 g, 5 min, 4 °C). The upper phase was collected and subjected to RNA precipitation at − 20 °C overnight. The precipitated RNA was washed once with 70% ethanol and dissolved in 30 l H2O.

Chromatin immunoprecipitation sequencing (ChIP-Seq)

Log phase cells grown in PMG or PMG-N media were harvested and cross-linked by 1% formaldehyde for 30 min, and then 125 mM Glycine was added to quench the crosslinking for 5 min. After three time washing with cold PBS, the cells pellet was resuspended in ChIP lysis buffer with 0.5 mm Zirconia/Silica Beads, and then lysed in FastPrep machine for 7 times at max power 6.5. Sonication was done by using Bioruptor® Pico for 10 cycles, and then chromatin concentration was measured with Qubit dsDNA HS assay kit. Immunoprecipitation was performed with 20 µg sheared chromatin, 40 ng spike-in chromatin (activemotif 53,083), 1.6 µl spike-in antibody (activemotif 61,686) and 6 µl anti-c-Myc antibody (Sigma-Aldrich, M4439). After three times washing with low salt wash buffer, high salt wash buffer and LiCl wash buffer successively, ChIP-DNA was extracted by ChIP DNA Clean & Concentrator kit (ZYMO RESEARCH, D5205) and DNA concentration measured by Qubit dsDNA HS assay kit. Sequence library prepared by ThruPLEX DNA-Seq kit (TaKaRa, R400676) with DNA HT Dual Index Kit – 96N Set A (TaKaRa, R400660). Before sequencing, we performed quality control with bioanalyzer high sensitivity DNA analysis, and then the sequencing was performed using the Illumina Nextseq 2000 platform with P3 v3 50 kit (36 + 8 + 8 + 36 cycles, single-end sequencing) at the BEA facility (Huddinge, Sweden).

Raw sequencing data from Nextseq 2000 (Bcl files) were converted and demultiplexed to fastq files using the bcl2fastq v2.20.0.422 program. The STAR 2.7.9a program (Dobin et al. 2013) was used for alignment with Schizosaccharomyces pombe reference genome (ASM294v2) and Drosophila melanogaster reference genome (dm6). We used Drosophila spike in normalization strategy for ChIP-seq data normalization described in (Egan et al. 2016). Samtools was used to count the reads in specific regions, and then we normalized the reads by following spike-in normalization strategy. Data were visualized with the Integrated Genomics Viewer (IGV). For bar diagrams, Microsoft Excel was used to create bar diagrams with unpaired T-test statistics.

RNA-seq and bioinformatics

To remove rRNA, 3 µg of purified total RNA was treated with Ribominus Eukaryote System v.2 kit (Ambion, Thermo Fisher Scientific). To generate sequencing libraries, a total of 100 ng of rRNA-depleted stocks and Illumina Stranded mRNA Prep Ligation kit (Illumina) were used. To quantify the samples, Qubit (HS dsDNA) was used, and samples were sequenced using an Illumina Nextseq 2000 platform (P3 100 cycle kit, 58 + 58 cycles, paired-end sequencing) at the BEA facility (Huddinge, Sweden) following the manufacturer’s instruction. To normalize samples, ERCC RNA Spike-In Mix 1, dilution 1:100 (Invitrogen, Thermo Fisher Scientific) was added in proportion on the number of vegetative cells in each culture that was used for RNA isolation.

Raw sequencing data from Nextseq 2000 (Bcl files) were converted and demultiplexed to fastq files using the bcl2fastq v2.20.0.422 program. The STAR 2.7.9a program (Dobin et al. 2013) was used to index the Schizosaccharomyces_pombe reference genome (ASM294v2) and the ERCC spike in sequences, and then the resulting fastq files were aligned. The mapped reads were then counted in annotated exons using featureCounts v1.5.1 (Liao et al. 2014). The genome fasta file and annotations (Schizosaccharomyces_pombe.ASM294v2.35.gff3) were obtained from ensembl. The count table from ‘featureCounts’ was imported into the R/Bioconductor program and differential gene expression analysis was performed using the EdgeR package (Robinson et al. 2010). The linear models pipeline of EdgeR was used. For the gene expression analysis, genes that had > 1 count per million in 3 or more samples were used and normalized based only on the ERCC spike in counts using the TMM normalization. To correct for batch effects the second batch with ies2 samples were normalized at T0 with the average ERCC factor from the first batch.

Supplementary information

Below is the link to the electronic supplementary material.

Acknowledgements

We thank the BEA core facility at Karolinska Institutet, particularly A. Damdimopoulos, for help in processing and analysis of gene expression data. We are grateful for services at the FACS core facility, Department of Haematology and Regenerative Medicine, Karolinska Institutet.

Author contribution

KE and YZ conceived and designed the research project. YZ, SZ, and KE performed the experiments. YZ, SZ, and KE analyzed that data. KE and YZ wrote the manuscript with help from SZ. All authors read and approved the manuscript.

Funding

Open access funding provided by Karolinska Institute. The Swedish Research Council, grant number 2021–02238, and Cancerfonden, grant number CAN 2021/1895, funded this research with grants to KE.

Data availability

Yeast strains can be requested by writing to KE. The RNA-seq data and the ChIP-seq data have been submitted to the NCBI Gene Expression Omnibus (GEO) under the accession number GSE200378. The processed ERCC normalized RNA-seq data is provided in Supplementary data excel file 1.

Declarations

Competing interests

The authors declare no competing interests.

Ethics approval

Ethics approval is not required for yeast research.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Conflict of interest

The authors declare no competing interests.

Footnotes

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  1. Batut PJ, Bing XY, Sisco Z, Raimundo J, Levo M, Levine MS. Genome organization controls transcriptional dynamics during development. Science. 2022;375(6580):566–570. doi: 10.1126/science.abi7178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Buchanan L, Durand-Dubief M, Roguev A, Sakalar C, Wilhelm B, Strålfors A, Shevchenko A, Aasland R, Shevchenko A, Ekwall K, Francis Stewart A. The Schizosaccharomyces pombe JmjC-protein, Msc1, prevents H2A.Z localization in centromeric and subtelomeric chromatin domains. PLoS Genet. 2009;5(11):e1000726. doi: 10.1371/journal.pgen.1000726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. doi: 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Egan B, Yuan C-C, Craske ML, Labhart P, Guler GD, Arnott D, et al. An alternative approach to ChIP-Seq normalization enables detection of genome-wide changes in histone H3 lysine 27 trimethylation upon EZH2 inhibition. PLoS ONE. 2016;11(11):e0166438. doi: 10.1371/journal.pone.0166438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ekwall, K, Thon G (2017) Setting up Schizosaccharomyces pombe Crosses/Matings. Cold Spring Harb Protoc (7). 10.1101/pdbprot091694 [DOI] [PubMed]
  6. Harris MA, Rutherford KM, Hayles J, Lock A, Bähler J, Oliver SG, Mata J, Wood V (2022) Fission stories: using PomBase to understand Schizosaccharomyces pombe biology. Genetics 220(4). 10.1093/genetics/iyab222 [DOI] [PMC free article] [PubMed]
  7. Hogan CJ, Aligianni S, Durand-Dubief M, Persson J, Will WR, Webster J, Wheeler L, Mathews CK, Elderkin S, Oxley D, Ekwall K, Varga-Weisz PD. Fission yeast Iec1-ino80-mediated nucleosome eviction regulates nucleotide and phosphate metabolism. Mol Cell Biol. 2010;30(3):657–674. doi: 10.1128/MCB.01117-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–930. doi: 10.1093/bioinformatics/btt656. [DOI] [PubMed] [Google Scholar]
  9. Maestroni L, Reyes C, Vaurs M, Gachet Y, Tournier S, Géli V, Coulon S. Nuclear envelope attachment of telomeres limits TERRA and telomeric rearrangements in quiescent fission yeast cells. Nucleic Acids Res. 2020;48(6):3029–3041. doi: 10.1093/nar/gkaa043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Marguerat S, Schmidt A, Codlin S, Chen W, Aebersold R, Bähler J. Quantitative analysis of fission yeast transcriptomes and proteomes in proliferating and quiescent cells. Cell. 2012;151(3):671–683. doi: 10.1016/j.cell.2012.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Mata J, Lyne R, Burns G, Bähler J. The transcriptional program of meiosis and sporulation in fission yeast. Nat Genet. 2002;32(1):143–147. doi: 10.1038/ng951. [DOI] [PubMed] [Google Scholar]
  12. Mizuguchi T, Fudenberg G, Mehta S, Belton JM, Taneja N, Folco HD, FitzGerald P, Dekker J, Mirny L, Barrowman J, Grewal SIS. Cohesin-dependent globules and heterochromatin shape 3D genome architecture in S. pombe. Nature. 2014;516(7531):432–435. doi: 10.1038/nature13833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Neumann FR, Dion V, Gehlen LR, Tsai-Pflugfelder M, Schmid R, Taddei A, Gasser SM. Targeted INO80 enhances subnuclear chromatin movement and ectopic homologous recombination. Genes Dev. 2012;26(4):369–383. doi: 10.1101/gad.176156.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Oya E, Durand-Dubief M, Cohen A, Maksimov V, Schurra C, Nakayama JI, Weisman R, Arcangioli B, Ekwall K. Leo1 is essential for the dynamic regulation of heterochromatin and gene expression during cellular quiescence. Epigenetics Chromatin. 2019;12(1):45. doi: 10.1186/s13072-019-0292-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Papamichos-Chronakis M, Watanabe S, Rando OJ, Peterson CL. Global regulation of H2A.Z localization by the INO80 chromatin-remodeling enzyme is essential for genome integrity. Cell. 2011;144(2):200–213. doi: 10.1016/j.cell.2010.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Pascual-Ortiz M, Saiardi A, Walla E, Jakopec V, Künzel NA, Span I, Vangala A, Fleig U. Asp1 bifunctional activity modulates spindle function via controlling cellular Inositol pyrophosphate levels in Schizosaccharomyces pombe. Mol Cell Biol. 2018;38(9):e00047–18. doi: 10.1128/MCB.00047-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Poli J, Gasser SM, Papamichos-Chronakis M (2017) The INO80 remodeller in transcription, replication and repair. Philos Trans R Soc Lond B Biol Sci 372(1731). 10.1098/rstb.2016.0290 [DOI] [PMC free article] [PubMed]
  18. Ranjan A, Nguyen VQ, Liu S, Wisniewski J, Kim JM, Tang X, Mizuguchi G, Elalaoui E,  Nickels TJ, Jou V, English BP, Zheng Q, Luk E, Lavis LD, Lionnet T, Wu C (2020) Live-cell single particle imaging reveals the role of RNA polymerase II in histone H2A.Z eviction. Elife (9):e5567. 10.7554/eLife.55667 [DOI] [PMC free article] [PubMed]
  19. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat Biotechnol. 2014;32(9):896–902. doi: 10.1038/nbt.2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. doi: 10.1093/bioinformatics/btp616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Romila CA, Townsend S, Malecki M, Kamrad S, Rodríguez-López M, Hillson O, Cotobal C, Ralser M, Bähler J. Barcode sequencing and a high-throughput assay for chronological lifespan uncover ageing-associated genes in fission yeast. Microb Cell. 2021;8(7):146–160. doi: 10.15698/mic2021.07.754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Sajiki K, Hatanaka M, Nakamura T, Takeda K, Shimanuki M, Yoshida T, Hanyu Y, Hayashi T, Nakaseko Y, Yanagida M. Genetic control of cellular quiescence in S. pombe. J Cell Sci. 2009;122(Pt 9):1418–1429. doi: 10.1242/jcs.046466. [DOI] [PubMed] [Google Scholar]
  23. Sajiki K, Tahara Y, Uehara L, Sasaki T, Pluskal T, Yanagida M. Genetic regulation of mitotic competence in G(0) quiescent cells. Sci Adv. 2018;4(8):eaat5685. doi: 10.1126/sciadv.aat5685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Shan CM, Bao K, Diedrich J, Chen X, Lu C, Yates JR, 3rd, Jia S. The INO80 complex regulates epigenetic inheritance of heterochromatin. Cell Rep. 2020;33(13):108561. doi: 10.1016/j.celrep.2020.108561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Shen X, Xiao H, Ranallo R, Wu WH, Wu C. Modulation of ATP-dependent chromatin-remodeling complexes by inositol polyphosphates. Science. 2003;299(5603):112–114. doi: 10.1126/science.1078068. [DOI] [PubMed] [Google Scholar]
  26. Shevchenko A, Roguev A, Schaft D, Buchanan L, Habermann B, Sakalar C, Thomas H, Krogan NJ, Shevchenko A, Stewart AF. Chromatin central: towards the comparative proteome by accurate mapping of the yeast proteomic environment. Genome Biol. 2008;9(11):R167. doi: 10.1186/gb-2008-9-11-r167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Singh PP, Shukla M, White SA, Lafos M, Tong P, Auchynnikava T, Spanos C, Rappsilber J, Pidoux AL, Allshire RC. Hap2-Ino80-facilitated transcription promotes de novo establishment of CENP-A chromatin. Genes Dev. 2020;34(3–4):226–238. doi: 10.1101/gad.332536.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Steglich B, Strålfors A, Khorosjutina O, Persson J, Smialowska A, Javerzat JP, Ekwall K. The Fun30 chromatin remodeler Fft3 controls nuclear organization and chromatin structure of insulators and subtelomeres in fission yeast. PLoS Genet. 2015;11(3):e1005101. doi: 10.1371/journal.pgen.1005101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Strålfors A, Walfridsson J, Bhuiyan H, Ekwall K. The FUN30 chromatin remodeler, Fft3, protects centromeric and subtelomeric domains from euchromatin formation. PLoS Genet. 2011;7(3):e1001334. doi: 10.1371/journal.pgen.1001334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Su Y, Chen C, Huang L, Yan J, Huang Y. Schizosaccharomyces pombe homologs of human DJ-1 are stationary phase-associated proteins that are involved in autophagy and oxidative stress resistance. PLoS ONE. 2015;10(12):e0143888. doi: 10.1371/journal.pone.0143888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Takayama Y, Takahashi K. Differential regulation of repeated histone genes during the fission yeast cell cycle. Nucleic Acids Res. 2007;35(10):3223–3237. doi: 10.1093/nar/gkm213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Takeda K, Yoshida T, Kikuchi S, Nagao K, Kokubu A, Pluskal T, Villar-Briones A, Nakamura T, Yanagida M. Synergistic roles of the proteasome and autophagy for mitochondrial maintenance and chronological lifespan in fission yeast. Proc Natl Acad Sci USA. 2010;107(8):3540–3545. doi: 10.1073/pnas.0911055107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Wen Z, Zhang L, Ruan H, Li G. Histone variant H2A.Z regulates nucleosome unwrapping and CTCF binding in mouse ES cells. Nucleic Acids Res. 2020;48(11):5939–5952. doi: 10.1093/nar/gkaa360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Xue M, Zhang H, Zhao F, Zhao T, Li H, Jiang D. The INO80 chromatin remodeling complex promotes thermomorphogenesis by connecting H2A.Z eviction and active transcription in Arabidopsis. Mol Plant. 2021;14(11):1799–1813. doi: 10.1016/j.molp.2021.07.001. [DOI] [PubMed] [Google Scholar]
  35. Yoo S, Lee EJ, Thang NX, La H, Lee H, Park C, Han DW, Uhm SJ, Song H, Do JT, Choi Y, Hong K. INO80 is required for the cell cycle control, survival, and differentiation of mouse ESCs by transcriptional regulation. Int J Mol Sci. 2022;23(23):15402. doi: 10.3390/ijms232315402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Zahedi Y, Durand-Dubief M, Ekwall K. High-Throughput FlowCytometry Combined with Genetic Analysis Brings New Insights into the Understanding of Chromatin Regulation of Cellular Quiescence. Int J Mol Sci. 2020;21(23):9022. doi: 10.3390/ijms21239022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Zhao Q, Dai B, Wu H, Zhu W, Chen J. Ino80 is required for H2A.Z eviction from hypha-specific promoters and hyphal development of Candida albicans. Mol Microbiol. 2022;118(1–2):92–104. doi: 10.1111/mmi.14954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Zhou BO, Wang SS, Xu LX, Meng FL, Xuan YJ, Duan YM, Wang JY, Hu H, Dong X, Ding J, Zhou JQ. SWR1 complex poises heterochromatin boundaries for antisilencing activity propagation. Mol Cell Biol. 2010;30(10):2391–2400. doi: 10.1128/MCB.01106-09. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Availability Statement

Yeast strains can be requested by writing to KE. The RNA-seq data and the ChIP-seq data have been submitted to the NCBI Gene Expression Omnibus (GEO) under the accession number GSE200378. The processed ERCC normalized RNA-seq data is provided in Supplementary data excel file 1.


Articles from Chromosome Research are provided here courtesy of Springer

RESOURCES