#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genomic Prevalence of Heterochromatic H3K9me2 and Transcription Do
Not Discriminate Pluripotent from Terminally Differentiated
Cells


Cellular differentiation entails reprogramming of the transcriptome from a

pluripotent to a unipotent fate. This process was suggested to coincide with a

global increase of repressive heterochromatin, which results in a reduction of

transcriptional plasticity and potential. Here we report the dynamics of the

transcriptome and an abundant heterochromatic histone modification,

dimethylation of histone H3 at lysine 9 (H3K9me2), during neuronal

differentiation of embryonic stem cells. In contrast to the prevailing model, we

find H3K9me2 to occupy over 50% of chromosomal regions already in stem

cells. Marked are most genomic regions that are devoid of transcription and a

subgroup of histone modifications. Importantly, no global increase occurs during

differentiation, but discrete local changes of H3K9me2 particularly at genic

regions can be detected. Mirroring the cell fate change, many genes show altered

expression upon differentiation. Quantitative sequencing of transcripts

demonstrates however that the total number of active genes is equal between stem

cells and several tested differentiated cell types. Together, these findings

reveal high prevalence of a heterochromatic mark in stem cells and challenge the

model of low abundance of epigenetic repression and resulting global basal level

transcription in stem cells. This suggests that cellular differentiation entails

local rather than global changes in epigenetic repression and transcriptional

activity.


Published in the journal: . PLoS Genet 7(6): e32767. doi:10.1371/journal.pgen.1002090
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1002090

Summary

Cellular differentiation entails reprogramming of the transcriptome from a

pluripotent to a unipotent fate. This process was suggested to coincide with a

global increase of repressive heterochromatin, which results in a reduction of

transcriptional plasticity and potential. Here we report the dynamics of the

transcriptome and an abundant heterochromatic histone modification,

dimethylation of histone H3 at lysine 9 (H3K9me2), during neuronal

differentiation of embryonic stem cells. In contrast to the prevailing model, we

find H3K9me2 to occupy over 50% of chromosomal regions already in stem

cells. Marked are most genomic regions that are devoid of transcription and a

subgroup of histone modifications. Importantly, no global increase occurs during

differentiation, but discrete local changes of H3K9me2 particularly at genic

regions can be detected. Mirroring the cell fate change, many genes show altered

expression upon differentiation. Quantitative sequencing of transcripts

demonstrates however that the total number of active genes is equal between stem

cells and several tested differentiated cell types. Together, these findings

reveal high prevalence of a heterochromatic mark in stem cells and challenge the

model of low abundance of epigenetic repression and resulting global basal level

transcription in stem cells. This suggests that cellular differentiation entails

local rather than global changes in epigenetic repression and transcriptional

activity.

Introduction

Resetting of the transcriptional program is the key driver for cell type specification during organismal development [1], [2]. While embryonic stem (ES) cells bear the fascinating ability to acquire very diverse fates, derived somatic stages are usually irreversible under physiological conditions. This unidirectionality has been suggested to depend in part on epigenetic repression of lineage unrelated genes [3], [4]. Accordingly, ES cell plasticity was suggested to rely on a low prevalence of heterochromatin and coinciding promiscuous low-level expression of many genes in stem cells [5][11]. In line with this model, distinct changes in nuclear staining had previously been observed by electron microscopy during cellular differentiation [12], [13]. Further, a subset of promoters was shown to become DNA methylated [14][16] and the repressive histone modifications H3K27me3 and H3K9me3 were reported to locally expand in differentiated cells [9].

Here, we set out to test the model of widespread heterochromatinization via monitoring of the differentiation-coupled dynamics of H3K9me2, a repressive epigenetic modification, which appears to be the most abundant heterochromatic modification and has recently been reported to cover large domains in differentiated cells [17]. Unexpectedly, we found that H3K9me2 is not only highly abundant in terminally differentiated cells, but already occupies large parts of the genome in pluripotent stem cells. In this cellular state, H3K9me2 occupies most genomic regions devoid of transcription and certain histone modifications. While our analysis revealed discrete local changes particularly at gene bodies, we observed little global increase in H3K9me2 during differentiation. This unexpected finding motivated us to revisit the model of promiscuous low-level gene expression in undifferentiated cells by quantitative RNA sequencing. Remarkably, we found the actual number of low-level expressed genes, postulated hallmarks of stem cells to be equal between both developmental states. Together, our findings challenge the model of promiscuous basal gene expression as a distinct property of pluripotency and a widespread increase of heterochromatin during cellular differentiation.

Results

H3K9me2 is nearly invariant and only displays distinct local changes between developmental stages

To asses differentiation associated dynamics of the repressive histone modification H3K9me2 we made use of a highly pure and robust murine in vitro neurogenesis model [18], which we previously used to profile histone and DNA methylation [14]. Here, we generated profiles for H3K9me2 in pluripotent embryonic stem cells and derived terminally differentiated pyramidal neurons. We made use of custom tiling arrays covering 10% of the mouse genome including all well-annotated promoters, several large multi-gene loci and the complete chromosome 19 (see Figure S1 and Text S1). The chromosomal profiles for H3K9me2 revealed domains of enrichments that upon visual inspection were highly comparable between stem cells and the neuronal state (Figure 1A), which is further supported by a high overall pair-wise correlation (Figure 1B). Despite this overall similarity we noticed confined regional differences (Figure 1A), a finding which is consistent with the fact that biological replicates of H3K9me2 are more similar than the patterns between cell states (Figure 1B). We also included in our comparison a recently published dataset for H3K9me2 in a distinct ES cell line [17], which shows high correlation to our ES cell datasets despite different experimental conditions (Figure S2). Of note, analysis of the H3K9me2 dataset from Wen et al. [17] revealed that chromosome 19 behaves similar to the other chromosomes (Figure S3), suggesting that our results can be extrapolated to the entire genome. Together this demonstrates that our H3K9me2 data are reproducible and of high resolution, yet overall patterns appear to be highly similar between a pluripotent and a terminally differentiated state. Visual inspection suggests that H3K9me2 covers large domains in both ES cells and neurons (Figure 1A). To quantitatively define the actual location and sizes of domains we applied a Hidden-Markov-Model (HMM) analysis to the microarray data. This unsupervised statistical method is a widely accepted approach for unbiased data segmentation in epigenome analysis [19], [20]. The HMM analysis not only agreed with and statistically corroborated the visual impression of the raw data, but also yielded robust results under variable settings (Figure S4). It revealed that over 50% of chromosome 19 is covered by H3K9me2 in ES cells (Figure 1C). Using the same approach for the H3K9me2 data in the ES cell-derived terminally differentiated neurons, we detected a modest yet reproducible 5% increase of genomic regions covered by H3K9me2 (Figure 1C). We conclude that global coverage and size of H3K9me2 domains is nearly identical between ES cells and derived post-mitotic pyramidal neurons. In line with this finding we do not detect a significant change in global H3K9me2 levels by Western blot detection (Figure 1D). Moreover, H3K9me2 domain features of ES cells and neurons show similar size distribution and median length (Figure S4). Further analysis revealed that changes in H3K9me2 between the two examined cellular states are rare; 88% of H3K9me2 occupied regions in ES cells are also occupied in neurons (Figure 1F). Notably, regions that change in H3K9me2 state tend to be small and are below the average size of invariant domains (Figure S5). Consistent with the overall increase of 5% in H3K9me2 coverage during differentiation, regions which gain H3K9me2 are more frequent and of larger size than regions showing a loss of the mark (Figure S5 and Figure S6). Interestingly, most of the larger regions (>10 kb) that gain H3K9me2 are located within genes, starting downstream of the promoter region (Figure 2A and Figure S5). These global findings are fully reproducible in single gene controls (Figure 2B) and consistent with a focused comparison of only genic regions (Figure 2C). Importantly, this shows that our experimental and data analysis approach is indeed highly sensitive to detect differences if they do occur. Interestingly, many genes that acquire H3K9me2 show slightly reduced expression in many cases, while others increase expression upon gain of the modification (Figure 2D and Figure S5). This suggests that the gain of H3K9me2, while highly selective for gene bodies, cannot simply be explained by the silencing of gene activity.

Fig. 1.

Fig. 2.

H3K9me2 and H3K27me3 are mutually exclusive

Given the high prevalence of H3K9me2, we next asked how its presence relates to a distinct repressive chromatin modification, namely trimethylation of H3K27 (H3K27me3). This mark is set by the Polycomb pathway and often occurs in domains of several kilobases [9], [21], [22]. We find that both heterochromatic histone modifications occur mutually exclusive even when in direct neighborhood as illustrated by the sharp boundaries of the H3K9me2 signal next to H3K27me3 peaks (Figure 3A, 3B and 3C). This is consistent with a previous study in human embryonal carcinoma cells that was limited to promoters [23].

Fig. 3.

H3K9me2 is largely exclusive with active chromatin

We further related H3K9me2 occupancy to regions with transcriptional activity or presence of the active modification H3K4me2. Active regions are mutually exclusive with H3K9me2 in ES cells but surprisingly to a lesser extent in neurons (Figure 3D). The compatibility of H3K9me2 and gene expression in neurons is however limited to gene bodies and does not occur in the promoters of expressed genes, consistent with the former regions gaining H3K9me2 during differentiation (Figure 2A and Figure S7). We find the majority of H3K4me2 regions to be mutually exclusive with H3K9me2 in stem cells (Figure 3D). In neurons, a small number of regions become co-occupied, again most of these being within transcribed genes (Figure 3D). Importantly, an HMM independent analysis confirms that regions with high H3K9me2 enrichment do not overlap with transcribed genes in stem cells, yet a subset does in neurons (Figure 3E). We conclude that gain of H3K9me2 during differentiation has only a minor effect on the overall chromosomal coverage of the modification, yet it occurs highly localized and preferentially at genic regions.

Prevalent low-level transcription is not a stem cell–specific feature

Our finding of surprising conservation of heterochromatin patterns in a refined model of differentiation let us to revisit the transcriptome in a quantitative manner using high throughput RNA sequencing (RNAseq). RNAseq in ES cells and derived neurons revealed the expected down regulation of stem cell specific genes and induction of neuron specific genes (Figure 4A). Further, when counting RNA molecules after gene mapping and normalization, both cell types displayed a characteristic bimodal distribution. This reflects a group of genes that is in a clear off state with no detectable RNA molecules and a second peak of expressed genes (Figure 4B). Separating these two groups of genes by a stringent cutoff revealed that out of 35′606 transcription units, 45% are expressed in ES cells. Interestingly, we identified a slightly higher number (50%) of expressed genes in terminally differentiated neurons, indicating that differentiation of stem cells is not coinciding with a reduced number of highly expressed genes. This agrees with a recent report that suggested that stem cells and somatic cells do mainly differ in the number of low-level expressed genes due to a global reduction of basal gene activity in the course of lineage-commitment and loss of pluripotency [10]. To test this in our in vitro differentiation system, we grouped the genes that could not clearly be assigned to the on or off state, into a separate class of genes expressed at low to background level (Figure 4B). This analysis reveals that in stem cells 16% of all transcription units show a basal expression level. Surprisingly however, the proportion of genes expressed at such low level (14%) is very similar in neurons. This unexpected finding prompted us to conduct an additional RNAseq experiment in a second fully differentiated somatic murine cell type; primary mouse embryonic fibroblasts (MEFs). Interestingly, also fibroblasts display a similar transcriptional landscape as stem cells, with 46% of all transcription units being highly expressed and 13% being expressed at basal levels. Hence, this qualitative similarity of expression patterns is not specific to the neuronal subtype we generated in vitro, but appears to be a more general property of both undifferentiated and differentiated cells. Thus, while transcripts expressed at low levels show little overlap between stem cells and somatic cells (Figure S8), their numbers are remarkably similar. Stem cells do also not show an increased number of highly expressed genes. Based on additional analysis we can exclude that this similarity of the transcriptional landscape is a consequence of insufficient sampling (Figure S9). Moreover, it is not limited to genic regions as the abundance of transcripts generated from diverse classes of endogenous repeat is comparable between stem cells and neurons (Figure 4C).

Fig. 4.

Discussion

Embryonic stem cells are characterized by their potential to differentiate into any cell type of the three germ layers in the developing embryo, while somatic cells lose this developmental plasticity upon lineage-commitment. Despite its relevance for our understanding of development and disease, the molecular determinants of pluripotency are still not fully understood and the factors responsible for this uniqueness of stem cells are actively debated [24]. Our study of gene expression and an abundant heterochromatin mark reveal surprising conservation of the transcriptome and epigenome landscape between pluripotent and unipotent cells. H3K9me2 is already highly prevalent in ES cells, arguing that the pathways that mediate H3K9me2 are highly active in stem cells, and serve similar functions as in somatic cells, which only show a slight increase of the mark (from 53 to 58%). Interestingly, the observed gain occurs very localized at gene bodies and does not necessarily coincide with lower transcription of the corresponding gene. The analysis of regions that acquire H3K9me2 during differentiation further revealed a differentiation specific coexistence of H3K9me2 and transcriptional activity, which is not detected in the pluripotent state and which could be involved in modulating expression in the differentiated cell. Nevertheless, despite the subtle increase we detected, an involvement of H3K9me2 in globally regulating cell-type specific gene repression appears unlikely. The limited dynamics as compared to massive transcriptome reprogramming and the limited correlation between expression and gain of H3K9me2 at target genes argue against H3K9me2 as being a major player in setting up gene expression programs.

These findings disagree with a recent report that suggested absence of large H3K9me2 domains in ES cells and found a striking increase in differentiated cells [17]. Notably, our ES cell profile for H3K9me2 is similar to the one generated previously (Figure 1B and Figure S2), making data analysis a likely explanation for the discrepancy. We applied an unbiased approach that is insensitive to variations between arrays and which we show to lead to similar results under various parameter settings (Figure 3 and Figure S10). As already discussed by Fillion and van Steensel [25], the previous study relied on defined thresholds, which can be prone to false estimation of differences particular in the absence of biological replicates [25].

Widespread low-level expression in stem cells has previously been reported and interpreted as a sign of pluripotency [5], [6], [10]. It has been speculated that this basal promiscuous activity would poise genes for rapid induction upon receipt of differentiation cues [5], [6], [10]. Using mRNA sequencing in our differentiation paradigm does not confirm this model. We do not find evidence of elevated transcription throughout the genome or on specific chromosome (Figure S8; [10]). A likely explanation for these discrepancies is that microarrays, which were used in the previous studies, overestimate low level signal due to cross-hybridization [26]. In the present study we utilized RNAseq, which permits an actual counting of RNA molecules and thus enables accurate discrimination between very low and no expression. Notably, RNA sequencing experiments have recently put other findings in question that relied on quantifying small transcriptome differences detected by microarrays. For example, recent RNAseq data challenged the presence of pervasive intergenic transcription [26] and the existence of transcriptional dosage compensation of the single male X chromosome [27]. In addition to the increased sensitivity of RNAseq that can explain the differences to previous studies, the presence of a small fraction of differentiated cells in suboptimal conditions of stem cell culture could similarly contribute to an overestimation of the number of genes that are actually expressed in stem cells. Notably, the ES cell differentiation protocol applied by us is optimized to reduce the number of differentiated cells in the culture [18].

In summary, our analysis suggests to revisit the model of massive heterochromatinization during cellular differentiation via a global increase in repressive histone marks [5][9], [17] and coinciding repression of basal gene activity [5], [6], [10].

Our data together with previous reports on dynamics of DNA methylation, H3K9me3 and the Polycomb pathway between pluripotent and somatic cells [9], [14], [15], [28][31] support a model whereby repressive chromatin is already highly active in stem cells and that epigenome reprogramming entails localized changes of repressive histone modifications and DNA methylation at regulatory regions that specify and stabilize lineage specification and terminal differentiation [32]. It will be interesting to determine if these local differences account for the observed changes in nuclear morphology [33]. Notably, epigenetic repression can be overcome by the local activity of transcription factors upon strong induction cues during normal differentiation or artificially during generation of induced pluripotent stem cells (iPS) [34] and might therefore safeguard rather than actively channel development via direct transcriptome regulation.

Methods

Cell culture

Wild-type embryonic stem cells (129Sv-C57Bl/6) were cultured and differentiated as previously described [14], [35]. Fibroblasts were isolated from wild-type embryos (C57Bl/6).

Western blot and peptide dot blot analysis

Peptide sequences can be found in Table S2. Western blot analysis was performed with acid extracts using 1/1000 dilutions of either anti-H3K9me2 (Abcam no. 1220) or anti-H4 (Upstate, no. 07–108) antibodies. Blots were developed with ECL reagent (GE Healthcare).

Chromatin-IP (ChIP)

ChIP experiments were performed as described before [14], starting with 70 µg of chromatin and 5 µg of the following antibodies: anti-dimethyl-H3K9 (LP Bio, no. AR-0108), anti-dimethyl-H3K9 (Abcam no. 1220), anti-trimethyl-H3K27 (Upstate, no. 07–449), anti-dimethyl-H3K4 (Upstate, no. 07–030). H3K9me2 ChIP samples were amplified using the WGA2 kit (Sigma) and hybridized to a custom tiling microarray (NimbleGen Systems Inc., see below). H3K27me3 and H3K4me2 ChIP libraries for Illumina sequencing were prepared with the Illumina ChIP-Seq DNA Sample Prep Kit (Cat# IP-102-1001) according to Illumina's instructions and sequenced on the Genome Analyzer II following the manufacturer's protocols. ChIP-real time PCR was performed using SYBR Green chemistry (ABI) and 1/40 of ChIP or 20 ng of input chromatin per PCR reaction. Primers are listed in Table S1.

Microarray design

H3K9me2 ChIP samples were hybridized to custom designed microarrays representing all well-annotated promoters, several large multi-gene loci and the complete chromosome 19 with an average probe spacing of 100 bp and a total of 2.1 million features (HD2.1, NimbleGen Systems Inc).

Microarray hybridization and analysis

Sample labeling, hybridization and array scanning were performed by NimbleGen Systems Inc. according to standard procedures. For analysis, raw fluorescent intensity values were used to calculate log2 of the bound/input ratios for each individual oligo. Subsequently, for comparison all arrays were normalized to a median log2 = 0 and scale normalized to have the same median absolute deviation using the “LIMMA” R/Bioconductor package [36], [37].

RNAseq data analysis

RNA from ES cells, neurons and fibroblasts of two independent biological replicates each was used for cDNA preparation using oligo dT primers followed by sequencing on an Illumina GA II analyzer. Reads were mapped to the Mus musculus transcriptome and normalized to transcript length and sequencing library size (for details see Text S1).

Bioinformatics

Unless otherwise stated, H3K9me2 enriched regions were identified by HMM and H3K4me2 and H3K27me3 peaks using MACS peak finder [38]. Active regions were defined as RefSeq transcription units with a normalized RNAseq log2 read count above 5 (for details see Text S1). Microarray design, hybridization and analysis, ChIPseq and RNAseq analysis and additional references are described in Text S1.

Datasets

Microarray and deep sequencing data were deposited at NCBI's Gene Expression Omnibus and are accessible through GEO Series accession number GSE27866 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE27866).

Supporting Information

Attachment 1

Attachment 2

Attachment 3

Attachment 4

Attachment 5

Attachment 6

Attachment 7

Attachment 8

Attachment 9

Attachment 10

Attachment 11

Attachment 12

Attachment 13


Zdroje

1. BoyerLAMathurDJaenischR

2006

Molecular control of pluripotency.

Curr Opin Genet Dev

16

455

462

2. EgliDBirkhoffGEgganK

2008

Mediators of reprogramming: transcription factors and transitions

through mitosis.

Nat Rev Mol Cell Biol

9

505

516

3. ReikWDeanWWalterJ

2001

Epigenetic reprogramming in mammalian

development.

Science

293

1089

1093

4. BernsteinBEMeissnerALanderES

2007

The mammalian epigenome.

Cell

128

669

681

5. NiwaH

2007

Open conformation chromatin and pluripotency.

Genes Dev

21

2671

2676

6. MeshorerEMisteliT

2006

Chromatin in pluripotent embryonic stem cells and

differentiation.

Nat Rev Mol Cell Biol

7

540

546

7. Gaspar-MaiaAAlajemAPolessoFSridharanRMasonMJ

2009

Chd1 regulates open chromatin and pluripotency of embryonic stem

cells.

Nature

460

863

868

8. PerssonJEkwallK

2010

Chd1 remodelers maintain open chromatin and regulate the

epigenetics of differentiation.

Exp Cell Res

316

1316

1323

9. HawkinsRDHonGCLeeLKNgoQListerR

2010

Distinct epigenomic landscapes of pluripotent and

lineage-committed human cells.

Cell Stem Cell

6

479

491

10. EfroniSDuttaguptaRChengJDehghaniHHoeppnerDJ

2008

Global transcription in pluripotent embryonic stem

cells.

Cell Stem Cell

2

437

447

11. Gaspar-MaiaAAlajemAMeshorerERamalho-SantosM

2011

Open chromatin in pluripotency and reprogramming.

Nat Rev Mol Cell Biol

12

36

47

12. FrancastelCSchubelerDMartinDIGroudineM

2000

Nuclear compartmentalization and gene activity.

Nat Rev Mol Cell Biol

1

137

143

13. AhmedKDehghaniHRugg-GunnPFussnerERossantJ

2010

Global chromatin architecture reflects pluripotency and lineage

commitment in the early mouse embryo.

PLoS ONE

5

e10531

doi:10.1371/journal.pone.0010531

14. MohnFWeberMRebhanMRoloffTCRichterJ

2008

Lineage-specific polycomb targets and de novo DNA methylation

define restriction and potential of neuronal progenitors.

Mol Cell

30

755

766

15. MeissnerAMikkelsenTSGuHWernigMHannaJ

2008

Genome-scale DNA methylation maps of pluripotent and

differentiated cells.

Nature

454

766

770

16. FarthingCRFiczGNgRKChanCFAndrewsS

2008

Global mapping of DNA methylation in mouse promoters reveals

epigenetic reprogramming of pluripotency genes.

PLoS Genet

4

e1000116

doi:10.1371/journal.pgen.1000116

17. WenBWuHShinkaiYIrizarryRAFeinbergAP

2009

Large histone H3 lysine 9 dimethylated chromatin blocks

distinguish differentiated from embryonic stem cells.

Nat Genet

41

246

250

18. BibelMRichterJSchrenkKTuckerKLStaigerV

2004

Differentiation of mouse embryonic stem cells into a defined

neuronal lineage.

Nat Neurosci

7

1003

1009

19. BirneyEStamatoyannopoulosJADuttaAGuigoRGingerasTR

2007

Identification and analysis of functional elements in 1%

of the human genome by the ENCODE pilot project.

Nature

447

799

816

20. KochCMAndrewsRMFlicekPDillonSCKaraozU

2007

The landscape of histone modifications across 1% of the

human genome in five human cell lines.

Genome Res

17

691

707

21. RingroseLParoR

2007

Polycomb/Trithorax response elements and epigenetic memory of

cell identity.

Development

134

223

232

22. SchwartzYBPirrottaV

2007

Polycomb silencing mechanisms and the management of genomic

programmes.

Nat Rev Genet

8

9

22

23. O'GeenHSquazzoSLIyengarSBlahnikKRinnJL

2007

Genome-wide analysis of KAP1 binding suggests autoregulation of

KRAB-ZNFs.

PLoS Genet

3

e89

doi:10.1371/journal.pgen.0030089

24. HannaJHSahaKJaenischR

2010

Pluripotency and cellular reprogramming: facts, hypotheses,

unresolved issues.

Cell

143

508

525

25. FilionGJvan SteenselB

2010

Reassessing the abundance of H3K9me2 chromatin domains in

embryonic stem cells.

Nat Genet 42: 4; author reply 5–6

26. van BakelHNislowCBlencoweBJHughesTR

2010

Most "dark matter" transcripts are associated with known

genes.

PLoS Biol

8

e1000371

doi:10.1371/journal.pbio.1000371

27. XiongYYChenXSChenZDWangXZShiSH

2010

RNA sequencing shows no dosage compensation of the active

X-chromosome.

Nature Genetics

42

1043-U1029

28. MikkelsenTSKuMJaffeDBIssacBLiebermanE

2007

Genome-wide maps of chromatin state in pluripotent and

lineage-committed cells.

Nature

448

553

560

29. PasiniDBrackenAPHansenJBCapilloMHelinK

2007

The polycomb group protein Suz12 is required for embryonic stem

cell differentiation.

Mol Cell Biol

27

3769

3779

30. PanGTianSNieJYangCRuottiV

2007

Whole-Genome Analysis of Histone H3 Lysine 4 and Lysine 27

Methylation in Human Embryonic Stem Cells.

Cell Stem Cell

1

299

312

31. ListerRPelizzolaMDowenRHHawkinsRDHonG

2009

Human DNA methylomes at base resolution show widespread

epigenomic differences.

Nature

462

315

322

32. MohnFSchubelerD

2009

Genetics and epigenetics: stability and plasticity during

cellular differentiation.

Trends Genet

25

129

136

33. MeisterPMangoSEGasserSM

2011

Locking the genome: nuclear organization and cell

fate.

Curr Opin Genet Dev. In press

34. TakahashiKYamanakaS

2006

Induction of pluripotent stem cells from mouse embryonic and

adult fibroblast cultures by defined factors.

Cell

126

663

676

35. BibelMRichterJLacroixEBardeYA

2007

Generation of a defined and uniform population of CNS progenitors

and neurons from mouse embryonic stem cells.

Nat Protoc

2

1034

1043

36. SmythGK

2004

Linear models and empirical bayes methods for assessing

differential expression in microarray experiments.

Stat Appl Genet Mol Biol

3

Article3

37. SmythGKSpeedT

2003

Normalization of cDNA microarray data.

Methods

31

265

273

38. ZhangYLiuTMeyerCAEeckhouteJJohnsonDS

2008

Model-based analysis of ChIP-Seq (MACS).

Genome Biol

9

R137

39. FeldmanNGersonAFangJLiEZhangY

2006

G9a-mediated irreversible epigenetic inactivation of Oct-3/4

during early embryogenesis.

Nat Cell Biol

8

188

194

Štítky
Genetika Reprodukční medicína

Článek vyšel v časopise

PLOS Genetics


2011 Číslo 6

Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#