Mol. Cells 2018; 41(11): 953-963
Published online November 1, 2018
https://doi.org/10.14348/molcells.2018.0213
© The Korean Society for Molecular and Cellular Biology
Correspondence to : *Correspondence: kimsy@kribb.re.kr
The stepwise development of T cells from a multipotent precursor is guided by diverse mechanisms, including interactions among lineage-specific transcription factors (TFs) and epigenetic changes, such as DNA methylation and hydroxymethylation, which play crucial roles in mammalian development and lineage commitment. To elucidate the transcriptional networks and epigenetic mechanisms underlying T-cell lineage commitment, we investigated genome-wide changes in gene expression, DNA methylation and hydroxymethylation among populations representing five successive stages of T-cell development (DN3, DN4, DP, CD4+, and CD8+) by performing RNA-seq, MBD-seq and hMeDIP-seq, respectively. The most significant changes in the transcriptomes and epigenomes occurred during the DN4 to DP transition. During the DP stage, many genes involved in chromatin modification were up-regulated and exhibited dramatic changes in DNA hydroxymethylation. We also observed 436 alternative splicing events, and approximately 57% (252) of these events occurred during the DP stage. Many stage-specific, differentially methylated regions were observed near the stage-specific, differentially expressed genes. The dynamic changes in DNA methylation and hydroxymethylation were associated with the recruitment of stage-specific TFs. We elucidated interactive networks comprising TFs, chromatin modifiers, and DNA methylation and hope that this study provides a framework for the understanding of the molecular networks underlying T-cell lineage commitment.
Keywords DNA hydroxymethylation, DNA methylation, T-cell development
The lineage-specific differentiation of immature T cells into CD4+ or CD8+ mature T cells is a stepwise process with well-defined sequential developmental stages (Germain, 2002). Early immature thymocytes, such as CD4 and CD8, lack the expression of T-cell receptors (TCR) and are thus termed double-negative (DN) cells. DN cells can be subdivided into the following four stages of differentiation: DN1 (CD44+CD25−), DN2 (CD44+CD25+), DN3 (CD44−CD25+), and DN4 (CD44−CD25−) (Godfrey et al., 1993). Successive T-cell differentiation produces double positive (DP; CD4+ CD8+) αβ TCR-expressing immature cells that subsequently become positively selected and mature into cells expressing TCRs that bind self-peptide-MHC-class-I complexes (CD8+ T cells) or self-peptide-MHC-class-II ligands (CD4+ T cells) (von Boehmer et al., 1989).
The cell and tissue identities in multicellular organisms are maintained by their particular epigenome (Bernstein et al., 2007). DNA methylation of the fifth position of cytosine (5mC) is a key epigenetic marker that plays important roles in mammalian development, differentiation and maintenance of cellular identity (Kim and Costello, 2017; Smith and Meissner, 2013). 5-hydroxymethylcytosine (5hmC) is an intermediate marker that is present during passive and active demethylation (Guo et al., 2011; Serandour et al., 2012), but 5hmC has also been suggested to play an important role in gene regulation (Kim et al., 2014; Yu et al., 2012). 5hmC is enriched in the gene bodies of highly expressed genes and at active enhancers (Ichiyama et al., 2015; Tsagaratou et al., 2014).
Here, we investigated the genome-wide changes in DNA methylation and DNA hydroxymethylation and their influences on stage-specific gene expression during T-cell precursor development. We identified stage-specific differentially methylated regions (ssDMRs) and differentially hydroxymethylated regions (ssDhMRs) and found that many of these regions overlap with the binding sites of stage-specific TFs. We also identified many stage-specific splicing variants. Our study may provide a framework for the understanding of the complexity of T-cell development and lineage commitment.
C57BL/6J mice (gender- and age-matched, 6–8 weeks) were purchased from Daehan Biolink (Korea). All the animal experiments were performed according to the guidelines and regulations for rodent experiments provided by the Institutional Animal Care and Use Committee (IACUC) of KAIST. The protocols used in this study were approved by the KAIST IACUC (KA2010-21).
The antibodies used for flow cytometry were purchased from BD Biosciences, eBioscience, and BioLegend, unless otherwise indicated. The antibodies included CD4 (RM4-5), CD8α (53-6.7), CD11b (M1/70), CD11c (HL3), CD19 (1D3), CD25 (PC61), CD44 (IM7), CD45R (B220, RA3-6B2), NK1.1 (PK136), and TCRγ/δ (GL3).
Thymus cells were isolated and dispersed into single-cell suspensions by passage through a 70-μm cell strainer (SPL, Korea). The cells were blocked with anti-CD16/32 and then stained for surface molecules. DAPI (4,6-diamidino-2-phenylindole; Roche) was used for the dead cell exclusion. The data were acquired using an Aria II flow cytometer (BD Biosciences) and analyzed using FlowJo software (Tree Star).
Single-cell suspensions of thymus cells were prepared as described above (flow cytometry) and blocked with anti-CD16/32. The whole thymus cell suspensions were enriched via the depletion of CD4+ or CD8+ cells by staining with a biotin-conjugated anti-CD4 or CD8α antibody, followed by incubating with streptavidin-microbeads (Miltenyi) and magnetic separation. The enriched cells were stained for surface molecules, and the dead cells were excluded by staining with DAPI. Sorting was performed using an Aria II system (BD Biosciences) and an 85-μm nozzle.
To isolate cells during the early stage of T-cell differentiation, cells were obtained from the thymus of C57BL/6J mice and stained with antibodies. The obtained cells were separated using the following process: 1) DAPI-integrated dead cells were excluded via FSC-A versus DAPI plots; 2) the cells were gated for singlet cells from FSC-A versus FSC-H plots; and 3) among the cells that were negative for lineage (B220-CD11b-CD11c-CD19-NK1.1-) and TCRγ/δ expression, the double-negative (DN), double-positive (DP), and single-positive (SP) T-cell subsets were isolated based on the CD4 and CD8 expression level. The DN3 and DN4 T cells were further identified according to the CD25 and CD44 expression (
MBD-seq was performed as previously described (Kim et al., 2014). Methylated DNA was precipitated using a Methyl-Miner methylated DNA enrichment kit (Invitrogen). The purified methylated DNA fragments were ligated to a pair of adaptors for sequencing using an Illumina NextSeq500 to generate 75 bp single-end reads.
hMeDIP-seq was performed as previously described (Kim et al., 2014). Fragmented DNA was end-repaired, A-tailed, and ligated to paired-end adapters. The pre-adapted DNA was subjected to immunoprecipitation using a hMeDIP kit (Diagenode). The immunoprecipitated DNA was amplified by 18 cycles of PCR using Illumina PCR primers and then size fractioned on a 2% agarose gel to obtain 200–300 bp fragments. The sequencing was performed using an Illumina NextSeq500 to generate 75 bp single-end reads.
RNA-seq was performed as previously described (Baek et al., 2016). The RNA sequencing library was prepared using a TruSeq RNA Sample Prep Kit (Illumina, USA), and the sequencing was performed using Illumina NextSeq500 to generate 100-bp paired-end reads.
FastQC (FastQC v0.11.3) (A Andrews, 2010) was used to filter the low-quality sequencing reads. Then, the reads were mapped to mouse genome build mm10 using TopHat (TopHat v2.0.11) (Trapnell et al., 2009) with the default parameters. DESeq (DESeq v3.1) (Anders and Huber, 2010) was used to select the differentially expressed genes among the five different developmental stages (fold-change>2 and
The sequenced MBD-seq and hMeDIP-seq reads were mapped to the mouse reference genome (mm10) using Bowtie (Bowtie v1.1.1) (Langmead et al., 2009) (parameters: -m 1), indexed and sorted using SAMtools (v.0.1.19) (Li et al., 2009). To identify the differentially methylation regions (DMRs) and differentially hydroxymethylated regions (DhMRs), we used MEDIPS (MEDIPS v1.18.0) (Lienhard et al., 2014) (parameters: extend = 200, shift = 0, window size = 200, and unique = TRUE) and various preprocessing steps (saturation estimation, sequence pattern coverage, CpG enrichment bias and GC bias normalization). The DMRs and DhMRs were selected based on the following parameters: diff.method = edgeR, prob.method = poisson, MeDIP = T, CNV = F, type = RPKM, and
To identify the enhancer regions, we obtained the ChIP-seq data of active chromatin markers (H3K4me1, H3K4me3, H3K27ac, and Pol II) at the DP stage from the NCBI GEO database (GSE20898, GSE47995, and GSE63732). Each ChIP-seq data set was mapped to the reference genome (mm10), and the peaks were identified using HOMER. We identified the regions overlapping with the H3K4me1, H3K27ac and Pol II peaks and then filtered the H3K4me3-enriched regions because H3K4me3 peaks are enriched at promoter regions.
To identify the TF binding motifs at stage-specific DMR or DhMR regions, we used the findMotifsGenome.pl command in HOMER. This command identifies motifs enriched in specific regions compared with randomly selected background regions (enrichment threshold:
To investigate the gene expression changes, we performed an RNA-seq analysis. Based on the read per kilobase per million mapped reads (RPKM) ≥ 1, approximately 17,500 genes were expressed among 20,861 reference transcripts (mm10). According to the pairwise comparisons of the gene expression changes between two stages along the T-cell developmental process, the most dramatic changes occurred between the DN4 and DP stages, involving 1,874 (11.4%) differentially expressed genes (DEGs; fold-change ≥ 2,
To understand the patterns of the gene expression changes during each stage of development, we performed hierarchical clustering of 2,688 genes with a greater than twofold change in expression between any stages (FC ≥ 2,
Subsequently, we focused on the changes in the TF expression levels. We obtained a list of 1,646 TFs from the GO term “DNA-dependent regulation of transcription” (GO:0006350) after removing genes with ambiguous annotation (Zhang et al., 2012). Among the 1,646 TFs, 150 genes were selected (FC ≥ 2,
We performed GO and pathway analyses of the lists of DEGs at each stage of T-cell development (
Alternative splicing is a regulatory mechanism of gene expression that produces two or more distinct mRNA species from a single gene. We used the MISO method to identify 435 exons differentially spliced during early T-cell development (PSI > 10% and Bayes factor > 5) (Wang et al., 2008). Exon-skipping was the most frequent event (Fig. 2A), and, among the five stages, exon-skipping occurred most frequently during the DP stage. Interestingly, many alternative splicing events occurred in genes that play an important role in the development and activation of T cells. For example, the
Also, we observed changes in DNA methylation and hydroxymethylation of alternative promoters of isoforms to observe epigenetic changes of the alternative first exons discovered during T-cell development. We found that several genes important for T-cell development showed DN3 stage-specific hypermethylation in the alternative promoters of the coding region. For example,
To understand how epigenetic changes, such as DNA methylation and hydroxymethylation, are associated with the gene expression changes during T-cell development, we first observed the expression patterns of the Dnmt and Tet family proteins, which mediate DNA methylation and hydroxymethylation (Figs. 3A and 3B). During each stage of T-cell development, significant changes in DNA methylation and hydroxymethylation were observed over the course of the T-cell lineage, and the most significant changes occurred between DN3 and DN4 (Fig. 3C). Most changes in DNA methylation and hydroxymethylation occurred at intragenic regions (
We further investigated the relationships between gene expression and DNA methylation or hydroxymethylation. A negative correlation was observed between promoter methylation and gene expression during all stages. In contrast, a positive correlation was observed between gene-body hydroxymethylation and gene expression during most stages. Thus, increased promoter hydroxymethylation is likely associated with increased gene expression, while promoter hypermethylation is likely associated with decreased gene expression. The average density profile of 5hmC and 5mC around transcription start sites (TSSs) and gene-body regions clearly showed that the 5mC density is depleted from the TSSs of stage-specific DEGs, whereas 5hmC is enriched in the gene bodies of stage-specific DEGs (Figs. 4A and 4B).
We selected stage-specific differentially methylated regions (ssDMRs) and hydroxymethylated regions (ssDhMRs) at each stage (Figs. 4C and 4D). Two examples (
Many stage-specific, hypomethylated DMRs were located in the regulatory regions of stage-specific TFs (Fig. 5A). For example, many DN3-specific TFs, such as
Genome-wide changes in DNA methylation and hydroxymethylation associated with T cells have been previously reported (Ko et al., 2015; Lee et al., 2001; Wilson et al., 2005; Zhao et al., 2016). Sellars et al. demonstrated that the dynamic regulation of DNA methylation plays an important role in enhancing the differential expression of
We observed a dynamic network of genes and TF expression that was previously reported in transcriptome analysis during the development of T-cell. During the developmental transition from DN3 to CD4 and CD8, we classified the patterns of gene expression into 24 distinct clusters (
We analyzed DNA methylation and DNA hydroxymethylation to observe epigenetic changes that regulate these gene expression changes. Similar to previously reported tissues and cells, we observed reverse correlation between promoter and methylation during development and positive correlation between gene body and hydroxymethylation. But alternative splicing events were more prevalent during the DP stage than during the other stages, and genes known to mediate alternative splicing were also highly expressed during this stage. We classified hundreds of alternative splicing events occurring during the early T-cell developmental stages into six categories and observed the relationship between these genes and the DNA methylation. During T-cell development, we have identified splicing variants of various genes, such as contrastive differential expression between isoforms or specific exons, and the presence of DMR in the gene body region of these genes and stage-specific hypermethylation of alternative promoters. We also compared DNA methylation of genes with or without splicing variants during the T-cell developmental stages to examine the potential role of DNA methylation in the alternative splicing (
We observed changes in stage-specific methylation in many cis-regulatory elements. Many DMRs and DhMRs were stage-specific and associated with the expression of corresponding genes. A recent study analyzing the methylation patterns of 17 normal tissues showed that many tissue-specific 5mC regions are located in distal regulatory regions (Hon et al., 2013; Lister et al., 2009). We also found that many stage-specific DMRs were located in intergenic regions rather than the promoter or gene body and that these DMRs overlapped with enhancer elements defined by H3K27ac peaks. Interestingly, these genes were enriched with GO terms related to T-cell development, such as T-cell differentiation and T-cell selection. Interestingly, a recent study reported that the stable CD4 expression in CD4+ T cells is regulated by the enhancer-dependent demethylation of the
In conclusion, we systematically analyzed the changes in gene expression, DNA methylation/hydroxymethylation, and stage-specific splicing variant from the initial T-cell differentiation stage to the post-T-cell fate stage. Deletion of alternative promoters and exons by CRISPR technology will validate the functional roles of alternative promoter expression in thymocyte development, and the mechanisms mediating selective promoter activity suggested by our study. Recently, rapid developments in methods used in single-cell analyses have enabled the analysis of gene expression and epigenomic changes at the single-cell level (Kakaradov et al., 2017; Villani et al., 2017). Considering the recent technical advances, our study is limited because it is based on an analysis of cell populations. Integrated analyses of transcriptomes and epigenomes (i.e., DNA methylation, hydroxymethylation, ATAC-seq, and Hi-C) at the single-cell level could be highly valuable for elucidating the complex molecular networks underlying T-cell lineage commitment.
Mol. Cells 2018; 41(11): 953-963
Published online November 30, 2018 https://doi.org/10.14348/molcells.2018.0213
Copyright © The Korean Society for Molecular and Cellular Biology.
Byoung-Ha Yoon1,2,4, Mirang Kim1,2,4, Min-Hyeok Kim3, Hee-Jin Kim2, Jeong-Hwan Kim2, Jong Hwan Kim1,2, Jina Kim1,2, Yong Sung Kim1,2, Daeyoup Lee3, Suk-Jo Kang3, and Seon-Young Kim1,2,*
1Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Korea, 2Genome Editing Research Center, Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea, 3Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
Correspondence to:*Correspondence: kimsy@kribb.re.kr
The stepwise development of T cells from a multipotent precursor is guided by diverse mechanisms, including interactions among lineage-specific transcription factors (TFs) and epigenetic changes, such as DNA methylation and hydroxymethylation, which play crucial roles in mammalian development and lineage commitment. To elucidate the transcriptional networks and epigenetic mechanisms underlying T-cell lineage commitment, we investigated genome-wide changes in gene expression, DNA methylation and hydroxymethylation among populations representing five successive stages of T-cell development (DN3, DN4, DP, CD4+, and CD8+) by performing RNA-seq, MBD-seq and hMeDIP-seq, respectively. The most significant changes in the transcriptomes and epigenomes occurred during the DN4 to DP transition. During the DP stage, many genes involved in chromatin modification were up-regulated and exhibited dramatic changes in DNA hydroxymethylation. We also observed 436 alternative splicing events, and approximately 57% (252) of these events occurred during the DP stage. Many stage-specific, differentially methylated regions were observed near the stage-specific, differentially expressed genes. The dynamic changes in DNA methylation and hydroxymethylation were associated with the recruitment of stage-specific TFs. We elucidated interactive networks comprising TFs, chromatin modifiers, and DNA methylation and hope that this study provides a framework for the understanding of the molecular networks underlying T-cell lineage commitment.
Keywords: DNA hydroxymethylation, DNA methylation, T-cell development
The lineage-specific differentiation of immature T cells into CD4+ or CD8+ mature T cells is a stepwise process with well-defined sequential developmental stages (Germain, 2002). Early immature thymocytes, such as CD4 and CD8, lack the expression of T-cell receptors (TCR) and are thus termed double-negative (DN) cells. DN cells can be subdivided into the following four stages of differentiation: DN1 (CD44+CD25−), DN2 (CD44+CD25+), DN3 (CD44−CD25+), and DN4 (CD44−CD25−) (Godfrey et al., 1993). Successive T-cell differentiation produces double positive (DP; CD4+ CD8+) αβ TCR-expressing immature cells that subsequently become positively selected and mature into cells expressing TCRs that bind self-peptide-MHC-class-I complexes (CD8+ T cells) or self-peptide-MHC-class-II ligands (CD4+ T cells) (von Boehmer et al., 1989).
The cell and tissue identities in multicellular organisms are maintained by their particular epigenome (Bernstein et al., 2007). DNA methylation of the fifth position of cytosine (5mC) is a key epigenetic marker that plays important roles in mammalian development, differentiation and maintenance of cellular identity (Kim and Costello, 2017; Smith and Meissner, 2013). 5-hydroxymethylcytosine (5hmC) is an intermediate marker that is present during passive and active demethylation (Guo et al., 2011; Serandour et al., 2012), but 5hmC has also been suggested to play an important role in gene regulation (Kim et al., 2014; Yu et al., 2012). 5hmC is enriched in the gene bodies of highly expressed genes and at active enhancers (Ichiyama et al., 2015; Tsagaratou et al., 2014).
Here, we investigated the genome-wide changes in DNA methylation and DNA hydroxymethylation and their influences on stage-specific gene expression during T-cell precursor development. We identified stage-specific differentially methylated regions (ssDMRs) and differentially hydroxymethylated regions (ssDhMRs) and found that many of these regions overlap with the binding sites of stage-specific TFs. We also identified many stage-specific splicing variants. Our study may provide a framework for the understanding of the complexity of T-cell development and lineage commitment.
C57BL/6J mice (gender- and age-matched, 6–8 weeks) were purchased from Daehan Biolink (Korea). All the animal experiments were performed according to the guidelines and regulations for rodent experiments provided by the Institutional Animal Care and Use Committee (IACUC) of KAIST. The protocols used in this study were approved by the KAIST IACUC (KA2010-21).
The antibodies used for flow cytometry were purchased from BD Biosciences, eBioscience, and BioLegend, unless otherwise indicated. The antibodies included CD4 (RM4-5), CD8α (53-6.7), CD11b (M1/70), CD11c (HL3), CD19 (1D3), CD25 (PC61), CD44 (IM7), CD45R (B220, RA3-6B2), NK1.1 (PK136), and TCRγ/δ (GL3).
Thymus cells were isolated and dispersed into single-cell suspensions by passage through a 70-μm cell strainer (SPL, Korea). The cells were blocked with anti-CD16/32 and then stained for surface molecules. DAPI (4,6-diamidino-2-phenylindole; Roche) was used for the dead cell exclusion. The data were acquired using an Aria II flow cytometer (BD Biosciences) and analyzed using FlowJo software (Tree Star).
Single-cell suspensions of thymus cells were prepared as described above (flow cytometry) and blocked with anti-CD16/32. The whole thymus cell suspensions were enriched via the depletion of CD4+ or CD8+ cells by staining with a biotin-conjugated anti-CD4 or CD8α antibody, followed by incubating with streptavidin-microbeads (Miltenyi) and magnetic separation. The enriched cells were stained for surface molecules, and the dead cells were excluded by staining with DAPI. Sorting was performed using an Aria II system (BD Biosciences) and an 85-μm nozzle.
To isolate cells during the early stage of T-cell differentiation, cells were obtained from the thymus of C57BL/6J mice and stained with antibodies. The obtained cells were separated using the following process: 1) DAPI-integrated dead cells were excluded via FSC-A versus DAPI plots; 2) the cells were gated for singlet cells from FSC-A versus FSC-H plots; and 3) among the cells that were negative for lineage (B220-CD11b-CD11c-CD19-NK1.1-) and TCRγ/δ expression, the double-negative (DN), double-positive (DP), and single-positive (SP) T-cell subsets were isolated based on the CD4 and CD8 expression level. The DN3 and DN4 T cells were further identified according to the CD25 and CD44 expression (
MBD-seq was performed as previously described (Kim et al., 2014). Methylated DNA was precipitated using a Methyl-Miner methylated DNA enrichment kit (Invitrogen). The purified methylated DNA fragments were ligated to a pair of adaptors for sequencing using an Illumina NextSeq500 to generate 75 bp single-end reads.
hMeDIP-seq was performed as previously described (Kim et al., 2014). Fragmented DNA was end-repaired, A-tailed, and ligated to paired-end adapters. The pre-adapted DNA was subjected to immunoprecipitation using a hMeDIP kit (Diagenode). The immunoprecipitated DNA was amplified by 18 cycles of PCR using Illumina PCR primers and then size fractioned on a 2% agarose gel to obtain 200–300 bp fragments. The sequencing was performed using an Illumina NextSeq500 to generate 75 bp single-end reads.
RNA-seq was performed as previously described (Baek et al., 2016). The RNA sequencing library was prepared using a TruSeq RNA Sample Prep Kit (Illumina, USA), and the sequencing was performed using Illumina NextSeq500 to generate 100-bp paired-end reads.
FastQC (FastQC v0.11.3) (A Andrews, 2010) was used to filter the low-quality sequencing reads. Then, the reads were mapped to mouse genome build mm10 using TopHat (TopHat v2.0.11) (Trapnell et al., 2009) with the default parameters. DESeq (DESeq v3.1) (Anders and Huber, 2010) was used to select the differentially expressed genes among the five different developmental stages (fold-change>2 and
The sequenced MBD-seq and hMeDIP-seq reads were mapped to the mouse reference genome (mm10) using Bowtie (Bowtie v1.1.1) (Langmead et al., 2009) (parameters: -m 1), indexed and sorted using SAMtools (v.0.1.19) (Li et al., 2009). To identify the differentially methylation regions (DMRs) and differentially hydroxymethylated regions (DhMRs), we used MEDIPS (MEDIPS v1.18.0) (Lienhard et al., 2014) (parameters: extend = 200, shift = 0, window size = 200, and unique = TRUE) and various preprocessing steps (saturation estimation, sequence pattern coverage, CpG enrichment bias and GC bias normalization). The DMRs and DhMRs were selected based on the following parameters: diff.method = edgeR, prob.method = poisson, MeDIP = T, CNV = F, type = RPKM, and
To identify the enhancer regions, we obtained the ChIP-seq data of active chromatin markers (H3K4me1, H3K4me3, H3K27ac, and Pol II) at the DP stage from the NCBI GEO database (GSE20898, GSE47995, and GSE63732). Each ChIP-seq data set was mapped to the reference genome (mm10), and the peaks were identified using HOMER. We identified the regions overlapping with the H3K4me1, H3K27ac and Pol II peaks and then filtered the H3K4me3-enriched regions because H3K4me3 peaks are enriched at promoter regions.
To identify the TF binding motifs at stage-specific DMR or DhMR regions, we used the findMotifsGenome.pl command in HOMER. This command identifies motifs enriched in specific regions compared with randomly selected background regions (enrichment threshold:
To investigate the gene expression changes, we performed an RNA-seq analysis. Based on the read per kilobase per million mapped reads (RPKM) ≥ 1, approximately 17,500 genes were expressed among 20,861 reference transcripts (mm10). According to the pairwise comparisons of the gene expression changes between two stages along the T-cell developmental process, the most dramatic changes occurred between the DN4 and DP stages, involving 1,874 (11.4%) differentially expressed genes (DEGs; fold-change ≥ 2,
To understand the patterns of the gene expression changes during each stage of development, we performed hierarchical clustering of 2,688 genes with a greater than twofold change in expression between any stages (FC ≥ 2,
Subsequently, we focused on the changes in the TF expression levels. We obtained a list of 1,646 TFs from the GO term “DNA-dependent regulation of transcription” (GO:0006350) after removing genes with ambiguous annotation (Zhang et al., 2012). Among the 1,646 TFs, 150 genes were selected (FC ≥ 2,
We performed GO and pathway analyses of the lists of DEGs at each stage of T-cell development (
Alternative splicing is a regulatory mechanism of gene expression that produces two or more distinct mRNA species from a single gene. We used the MISO method to identify 435 exons differentially spliced during early T-cell development (PSI > 10% and Bayes factor > 5) (Wang et al., 2008). Exon-skipping was the most frequent event (Fig. 2A), and, among the five stages, exon-skipping occurred most frequently during the DP stage. Interestingly, many alternative splicing events occurred in genes that play an important role in the development and activation of T cells. For example, the
Also, we observed changes in DNA methylation and hydroxymethylation of alternative promoters of isoforms to observe epigenetic changes of the alternative first exons discovered during T-cell development. We found that several genes important for T-cell development showed DN3 stage-specific hypermethylation in the alternative promoters of the coding region. For example,
To understand how epigenetic changes, such as DNA methylation and hydroxymethylation, are associated with the gene expression changes during T-cell development, we first observed the expression patterns of the Dnmt and Tet family proteins, which mediate DNA methylation and hydroxymethylation (Figs. 3A and 3B). During each stage of T-cell development, significant changes in DNA methylation and hydroxymethylation were observed over the course of the T-cell lineage, and the most significant changes occurred between DN3 and DN4 (Fig. 3C). Most changes in DNA methylation and hydroxymethylation occurred at intragenic regions (
We further investigated the relationships between gene expression and DNA methylation or hydroxymethylation. A negative correlation was observed between promoter methylation and gene expression during all stages. In contrast, a positive correlation was observed between gene-body hydroxymethylation and gene expression during most stages. Thus, increased promoter hydroxymethylation is likely associated with increased gene expression, while promoter hypermethylation is likely associated with decreased gene expression. The average density profile of 5hmC and 5mC around transcription start sites (TSSs) and gene-body regions clearly showed that the 5mC density is depleted from the TSSs of stage-specific DEGs, whereas 5hmC is enriched in the gene bodies of stage-specific DEGs (Figs. 4A and 4B).
We selected stage-specific differentially methylated regions (ssDMRs) and hydroxymethylated regions (ssDhMRs) at each stage (Figs. 4C and 4D). Two examples (
Many stage-specific, hypomethylated DMRs were located in the regulatory regions of stage-specific TFs (Fig. 5A). For example, many DN3-specific TFs, such as
Genome-wide changes in DNA methylation and hydroxymethylation associated with T cells have been previously reported (Ko et al., 2015; Lee et al., 2001; Wilson et al., 2005; Zhao et al., 2016). Sellars et al. demonstrated that the dynamic regulation of DNA methylation plays an important role in enhancing the differential expression of
We observed a dynamic network of genes and TF expression that was previously reported in transcriptome analysis during the development of T-cell. During the developmental transition from DN3 to CD4 and CD8, we classified the patterns of gene expression into 24 distinct clusters (
We analyzed DNA methylation and DNA hydroxymethylation to observe epigenetic changes that regulate these gene expression changes. Similar to previously reported tissues and cells, we observed reverse correlation between promoter and methylation during development and positive correlation between gene body and hydroxymethylation. But alternative splicing events were more prevalent during the DP stage than during the other stages, and genes known to mediate alternative splicing were also highly expressed during this stage. We classified hundreds of alternative splicing events occurring during the early T-cell developmental stages into six categories and observed the relationship between these genes and the DNA methylation. During T-cell development, we have identified splicing variants of various genes, such as contrastive differential expression between isoforms or specific exons, and the presence of DMR in the gene body region of these genes and stage-specific hypermethylation of alternative promoters. We also compared DNA methylation of genes with or without splicing variants during the T-cell developmental stages to examine the potential role of DNA methylation in the alternative splicing (
We observed changes in stage-specific methylation in many cis-regulatory elements. Many DMRs and DhMRs were stage-specific and associated with the expression of corresponding genes. A recent study analyzing the methylation patterns of 17 normal tissues showed that many tissue-specific 5mC regions are located in distal regulatory regions (Hon et al., 2013; Lister et al., 2009). We also found that many stage-specific DMRs were located in intergenic regions rather than the promoter or gene body and that these DMRs overlapped with enhancer elements defined by H3K27ac peaks. Interestingly, these genes were enriched with GO terms related to T-cell development, such as T-cell differentiation and T-cell selection. Interestingly, a recent study reported that the stable CD4 expression in CD4+ T cells is regulated by the enhancer-dependent demethylation of the
In conclusion, we systematically analyzed the changes in gene expression, DNA methylation/hydroxymethylation, and stage-specific splicing variant from the initial T-cell differentiation stage to the post-T-cell fate stage. Deletion of alternative promoters and exons by CRISPR technology will validate the functional roles of alternative promoter expression in thymocyte development, and the mechanisms mediating selective promoter activity suggested by our study. Recently, rapid developments in methods used in single-cell analyses have enabled the analysis of gene expression and epigenomic changes at the single-cell level (Kakaradov et al., 2017; Villani et al., 2017). Considering the recent technical advances, our study is limited because it is based on an analysis of cell populations. Integrated analyses of transcriptomes and epigenomes (i.e., DNA methylation, hydroxymethylation, ATAC-seq, and Hi-C) at the single-cell level could be highly valuable for elucidating the complex molecular networks underlying T-cell lineage commitment.
Haejeong Heo, Hee-Jin Kim, Keeok Haam, Hyun Ahm Sohn, Yang-Ji Shin, Hanyong Go, Hyo-Jung Jung, Jong-Hwan Kim, Sang-Il Lee, Kyu-Sang Song, Min-Ju Kim, Haeseung Lee, Eun-Soo Kwon, Seon-Young Kim, Yong Sung Kim, and Mirang Kim
Mol. Cells 2023; 46(5): 298-308 https://doi.org/10.14348/molcells.2023.2148Uijin Kim and Dong-Sung Lee
Mol. Cells 2023; 46(2): 86-98 https://doi.org/10.14348/molcells.2023.0013Sangrea Shim, Hong Gil Lee, and Pil Joon Seo
Mol. Cells 2021; 44(10): 746-757 https://doi.org/10.14348/molcells.2021.0160