Epigenetic Landscaping During hESC ... - Wiley Online Library

6 downloads 7056 Views 1MB Size Report
tency, self-renewal, and in early stages of differentiation. To understand the ... of a novel bivalent domain at the promoters of the myelin. Author contributions: ...
STEM CELL EPIGENETICS, GENOMICS, AND PROTEOMICS Epigenetic Landscaping During hESC Differentiation to Neural Cells ANNA GOLEBIEWSKA,a,b STUART P. ATKINSON,a,b MAJLINDA LAKO,a,b LYLE ARMSTRONGa,b Institute of Human Genetics and bNorth East Institute for Stem Cell Research, University of Newcastle Upon Tyne, International Centre for Life, United Kingdom a

Key Words. Epigenetics • Bivalent domains • Embryonic stem cells

ABSTRACT The molecular mechanisms underlying pluripotency and lineage specification from embryonic stem cells (ESCs) are still largely unclear. To address the role of chromatin structure in maintenance of pluripotency in human ESCs (hESCs) and establishment of lineage commitment, we analyzed a panel of histone modifications at promoter sequences of genes involved in maintenance of pluripotency, self-renewal, and in early stages of differentiation. To understand the changes occurring at lineage-specific gene regulatory sequences, we have established an efficient purification system that permits the examination of two distinct populations of lineage committed cells; fluorescence activated cell sorted CD1331 CD452CD342 neural

stem cells and b-III-tubulin1 putative neurons. Here we report the importance of other permissive marks supporting trimethylation of Lysine 4 H3 at the active stem cell promoters as well as poised bivalent and nonbivalent lineage-specific gene promoters in hESCs. Methylation of lysine 9 H3 was found to play a role in repression of pluripotency-associated and lineage-specific genes on differentiation. Moreover, presence of newly formed bivalent domains was observed at the neural progenitor stage. However, they differ significantly from the bivalent domains observed in hESCs, with a possible role of dimethylation of lysine 9 H3 in repressing the poised genes. STEM CELLS 2009;27:1298–1308

Disclosure of potential conflicts of interest is found at the end of this article.

INTRODUCTION The pluripotency and developmental plasticity of embryonic stem cells (ESCs) may be largely controlled by their specific chromatin architecture [1]. The presence of numerous covalent modifications on the N-terminal tails of histones is linked to gene activities. For example, acetylation of histones H3 and H4 (H3ac and H4ac) together with the several methylation states of lysine 4 on histone H3 (H3K4me2/3) present at the gene promoters are linked with transcriptional activation. Conversely, methylation of lysines 9 and 27 on histone H3 (H3K9me2/3 and H3K27me3) are associated with gene silencing [2, 3]. Recently, unique histone modification patterns, named bivalent domains, have been reported which could contribute to the maintenance of pluripotency in both mouse and human ESCs (mESC and hESC) [4, 5]. These domains exhibit both trimethylation of lysines 27 and 4 on histone H3 (H3K27me3 and H3K4me3, respectively) over the promoter sequences of many lineage-specific genes. Bivalent domains keep important lineage-specific genes repressed due to the dominant effect of H3K27me3 over H3K4me3, while preserving their potential to become activated on differentiation. During differentiation into a particular cell lineage, they resolve according to the transcriptional status of the genes required in that specific cell type [6]. However, the bivalent histone modifications were not found at all promoter loci of multiple line-

age-control genes in either mouse or human ESCs. A number of key developmental genes were marked only by the permissive H3K4me3 and acetylation marks [7, 8] or did not exhibit either of the marks associated with bivalency at their promoters [5]. How these regions are kept silent in ESCs is still not clear. Given their important role in cell lineage determination, it is likely that these genes become permissive for activation at later developmental stages through as yet uncharacterized epigenetic mechanisms. This study is an examination of locus-specific patterns of post-translational modifications of histones in hESCs and neural progenitors that differentiate from hESC under specific culture conditions. An efficient purification system was established, in which epigenetic changes were examined in hESCderived CD133þCD45CD34 neural stem cells (NSCs) and b-III-tubulinþ putative neurons. Correlation between the gene expression profiles of these cell types and histone modification patterns at the appropriate gene promoters was established. In this study, we show the presence of other permissive histone marks supporting H3K4me3 at the active pluripotency-associated gene promoters, as well as poised bivalent and nonbivalent lineage-specific gene promoters at the undifferentiated hESC stage. H3K9 methylation was found to play a role in repressing various gene promoters on differentiation. Our data support those obtained from similar studies using mESC with the notable exception of the establishment of a novel bivalent domain at the promoters of the myelin

Author contributions: A.G.: collection and assembly of data, manuscript writing; S.P.A.: collection and assembly of data, manuscript writing; M.L.: conception and design, manuscript writing; L.A.: conception and design, manuscript writing and financial support. Correspondence: Dr. Lyle Armstrong, BSc, Ph.D., International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, U.K. Telephone: 00-44-191-241-8695; Fax: 00-44-191-241-8666; e-mail: [email protected] Received October 21, 2008; accepted for C AlphaMed Press 1066-5099/2009/$30.00/0 publication February 25, 2009; first published online in STEM CELLS EXPRESS March 12, 2009. V doi: 10.1002/stem.59

STEM CELLS 2009;27:1298–1308 www.StemCells.com

Golebiewska, Atkinson, Lako et al.

basic protein (MBP) and glial fibrillary acidic protein (GFAP) at the neural progenitor cell stage and its resolution into a repressive chromatin structure when these cells differentiate into neurons.

MATERIALS

AND

METHODS

Cell Culture Wisconsin H9 hESC line (WiCell Research Institute, Madison, WI, www.wicell.org) was cultured as discrete colonies on feeder layers of mitotically inactivated mouse epithelial fibroblasts (MEFs) as previously described [9]. To begin differentiation into the neural lineage, H9 hESCs were plated on pregelatinized plates. Neural differentiation was induced in N2B27 serum-free medium (1  DMEM/F12 with glutamine, 1  N2, 1  B27 [PAA]; 1% MEM-NNAA [Invitrogen, Paisley, UK, http:// www.invitrogen.com]; 1% L-glutamine/penicillin-streptomycin [Sigma Aldrich, Dorset, U.K., www.sigmaaldrich.com]). After 4– 5 days the medium was changed to Neuronal base medium (DMEM/F12/Neuronal base medium 1:1; 0.5  N2; 0.5  B27 [PAA Laboratories Ltd, Pasching, Austria, http://www.paa.at]; 1% MEM-NNAA [Invitrogen]; 1% L-glutamine/penicillin–streptomycin [Sigma]) with bFGF (20 ng/ml; Invitrogen) and cultured for another 20 days. To obtain postmitotic neurons, the cultures were transferred as monolayer fragments to poly-D-lysine/laminin (Sigma/BD Biosciences, Oxford, U.K., www.bdeurope.com) plates. The cells underwent terminal differentiation in Neuronal base medium without bFGF and were collected 12–14 days after plating. Immortalized BJ foreskin human fibroblasts were obtained from LGC Promochem (LGC Promochem, Teddington, U.K., www.lgcstandards.com) and cultured in medium containing 88% Dulbecco’s modified Eagle’s medium (DMEM) without sodium pyruvate (Invitrogen); 10% FBS (Sigma); 1% MEM-NNAA (Invitrogen); 1% L-glutamine/penicillin–streptomycin (Invitrogen).

Fluorescence Activated Cell Sorting The H9 hESC-derived monolayers were dissociated into single cell suspensions in accutase solution (Chemicon Temecula, CA, http://www.chemicon.com) and used unfixed for subsequent antibody staining. The single cell suspension recovered from poly-Dlysine/laminin plates were fixed for 10 minutes in 1% formaldehyde (Sigma) and permeabilized in cold 90% methanol for 30 minutes. The cells were washed in the Lyse-Wash Assistant (LWA; BD Biosciences) and filtered using BD falcon filters (12/ 75 mm). The appropriate fluorochrome conjugated primary antibodies were added (see supporting information Table 1) and the tubes were incubated at room temperature for 30–60 minutes. Compensation for each fluorochrome was performed with use of multifluorochrome beads (BD Biosciences). Fluorescence activated cell sorting (FACS) settings were adapted to a low sheath pressure (20–25 psi), larger nozzle size (100 lm), and reduced sorting speed (1,000–3,000 events per second). Unstained cells were used as a negative control.

Chromatin Immunoprecipitation Chromatin immunoprecipitation (ChIP) was performed on H9 hESC-derived progeny according to published Q2ChiP method [10, 11] in which 10,000 cells were used for each immunoprecipitation reaction. For the list of the relevant ChIP grade antibodies see supporting information Table 2. An additional no-antibody control ChIP reaction was also included to allow for normalization for any background, if present. Q-PCR of the ChIP products was carried out in 10 ll reaction volumes in a 384-well plate system, using the Applied Biosystems 7900HT Real-time PCR machine. Genomic DNA standards (Roche Diagnostics, Basel, Switzerland, http://www.roche-applied-science.com) of know concentration (range of 100–0.01 ng/ll) were included in the Q-PCR

www.StemCells.com

1299

run to generate a standard curve from which the ChIP samples could be enumerated. A control Q-PCR with water only was used to control for PCR contamination. The DNA sequence of oligonucleotides used for ChIP analysis is shown in supporting information Table 3.

RNA Extraction/RT-PCR Total RNA was extracted from the cell populations with TRIzol (Invitrogen) according to manufacturer’s protocol and isolated by precipitation with propan-2-ol. After removal of genomic DNA contamination with DNAseI (1 IU/10 lg RNA) (37 C/30 minutes), cDNA was prepared by reverse transcription with MMLV reverse transcriptase (Promega, Southampton, U.K., http://www.promega.com) according to manufacturer’s instructions using random 15mers and then stored at 20 C. The DNA sequence of oligonucleotides used for the Q-PCR is given at the supporting information Table 4.

Immunocytochemistry Cell cultures were fixed for 10 minutes at room temperature in 4% PFA, permeabilized in 0.2% Triton X-100 for 45 minutes, and blocked in 5% BSA for 60 minutes at room temperature. After washing in 5% FBS/PBS, the cells were incubated with primary antibodies for 60 minutes at room temperature, followed by incubation with FITC conjugated secondary antibody for another 60 minutes. Finally, the cells were incubated in Hoechst solution for 5 minutes before visualization. The pictures were taken with use of fluorescence microscope Carl Zeiss Axiovert 200M. For the list of antibodies used in the study see supporting information Table 1.

RESULTS hESCs Differentiate into the Neural Lineage via Progression Through Different Stages Resembling Embryonic Neurogenesis hESCs are a valuable model of early embryogenesis and lineage-specification which yields sufficient cell numbers for investigations using ChIP. In this study, differentiation of NSCs from H9 hESCs was induced in serum-free adherent monolayer culture on pregelatinized plates (see supporting information Fig. 1). During the first 5 days of differentiation, the H9 hESC colony fragments formed typical neuroepithelial structures which organized into neural tube-like rosettes. After 4–5 days, the medium was supplemented with bFGF to promote proliferation of NSCs for another 20 days (see supporting information Figs. 1 and 2). The NSCs were induced to further differentiation by withdrawal of bFGF and plating on poly-D-lysine/laminin plates. b-III-tubulin as well as postmitotic neuronal markers (neuronal nuclear antigen [NeuN], doublecortin [DCX], microtubule-associated protein-2 (MAP2), neurofilament 200 [NF200]) were detected mainly in the densest areas, originally arising from the monolayer fragments plated on the plates (see supporting information Fig. 3A). Moreover, hESCs gave rise to a high population of cells from glial lineages. Both oligodendrocyte progenitors (A2B5, NG2, and galactocerebroside [GalC] immunopositive) were present together with astrocytic cells (S100b immunopositive) (see supporting information Fig. 3B).

Two Stages of Lineage Specification Can Be Purified During hESC Differentiation into the Neural Lineage Examination of the histone modification patterns at the promoters of individual lineage specific genes is not possible in a

1300

Epigenetic Landscaping During hESC Differentiation

Figure 1. H9 hESC-derived neural lineage purification system. (The images shown in [A–D] are representative of at least five independent experiments). H9 hESCs were differentiated into the neural lineage for 25 days on pregelatinized plates. Proliferation of NSCs was induced by bFGF. (A): FACS sorting of H9 hESC-derived NSCs from within the monolayer culture. The NSC population was isolated by sorting the CD133þCD34CD45 population from H9 hESC-derived monolayers. CD133þ population ranged between 2% and 3% across experiments. Any spectral overlaps between fluorochromes were compensated and the gates were set up manually according to the patterns of unstained cells. The CD133þ population is shown in blue (left panel) and investigated further for the coexpression of the CD34 (left panel) and CD45 (right panel) epitopes. A distinct population of CD34þ cells was observed within the H9 hESC-derived monolayer culture; however, neither CD133þCD34þ nor CD34þCD45þ populations were detected. (B): CD133 expression on PAX7 expressing myogenic progenitors is low during early hESC differentiation. (C): H9 hESC-derived b-III-tubulinþ neurons. (A): Fluorescence activated cell sorted (FACS) CD133þCD34CD45 cells underwent further differentiation whereas plated on poly-D-lysine/laminin plates giving rise to b-III-tubulinþ cells in bFGF-depleted medium. (B): H9 hESCderived monolayers were transferred to poly-D-lysine/laminin plates in bFGF-depleted medium for 12–14 days. b-III-tubulinþ putative neurons were observed in mainly in dense areas of the plate originating from original monolayer fragments. (D): FACS sorting of H9 hESC-derived neurons. b-III-tubulinþ putative neurons were sorted from the differentiated cell populations on poly-D-lysine/laminin plates (13% positive cells). Because of the lack of an adequate isotype control unstained fixed and permeabilized cells were used as a negative control. Abbreviations: APC, allophyocyanin; DAPI, 40 , 6-diamidino-2-phenylindole; hESCs, human embryonic stem cells; NSC, neural stem cells; PE, phycoerthyrin.

heterogeneous population of differentiated cells; therefore, FACS was used to enrich NSCs and terminally differentiated neurons. H9 hESC-derived monolayers, after 25 days of bFGF induced proliferation, were used as a source of NSCs (Fig. 1A). The CD133 (prominin-1/2) epitope was chosen as a specific NSC marker because expression of this has been reported for hESC-derived NSCs in culture [12], putative NSCs of the cerebellum [13], and embryonic neuroepithelial cells [14]. As CD133 can be expressed by immature hematopoietic stem cells [15], and hESC-derived endothelial progenitors (M. Lako, personal communication) CD133 positive cells were sorted only from the CD34 and CD45 negative population (CD133þ) (Fig. 1A) as both of these progenitor cell types express CD34. A distinct population of CD34þ cells was observed within H9 hESC-derived monolayers, but they did

not coexpress CD133, whereas no CD45þ cells were detected. Skeletal muscle satellite cells may also express CD133 at some stages of their development [16] but some evidence suggests that these cells are also CD34 positive [17] and therefore would be eliminated from the CD133 positive and CD34/ CD45 negative population. Our data from other work suggest that differentiation of hESC over similar timescales to those employed in our neuronal differentiation model did not produce significant numbers of PAX7 positive skeletal muscle satellite cells that were also CD133 positive (Fig. 1B). In view of these observations, we are confident that the FACS strategy employed in this work is able to generate highly enriched populations of NSCs. When plated on poly-D-lysine/ laminin precoated plates, sorted CD133þ cells underwent further differentiation and gave rise to neurons expressing b-III-

Golebiewska, Atkinson, Lako et al.

1301

Figure 2. Expression profile of H9 human embryonic stem cells (hESCs) and changes during differentiation into the neural lineage. The expression of key pluripotency markers and genes involved in neurogenesis as well as other lineage markers were examined in undifferentiated H9 hESCs (H9); H9-derived NSCs (CD133þ) and H9 hESC-derived putative neurons (b-IIItubulinþ). Human BJ fibroblasts (BJ) were used as a control representing a nonpluripotent, terminally differentiated cell line. Threshold cycle (Ct) values were obtained by Q-PCR analysis with the use of Applied Bioscience 7900HT Fast Real-Time PCR. The relative expression calculations were performed with use of QBase software where GAPDH was used as a reference gene in each sample and external mRNA reference was used as an inter-run calibrator. The data are presented as mean  SEM (n ¼ 4). Abbreviations: GFAP, glial fibrillary acidic protein; hTERT, telomerase reverse transcriptase subunit; MBP, myelin basic protein; NES, nestin; VIM, vimentin.

tubulin (Fig. 1C). Similarly, of the whole aggregates plated on poly-D-lysine/laminin, not all of the CD133þ progeny expressed b-III-tubulin, which supports the multipotency of CD133þ NSCs. H9 hESC-derived neurons were purified from differentiated populations grown on poly-D-lysine/laminin plates under differentiation conditions for 12–14 days. b-IIItubulin was chosen as a marker for neurons generated in this differentiation protocol due to the lack of specific cell surface markers for postmitotic neurons (Fig. 1D). Other markers have been used for FACS enrichment of neurons such as neural cell adhesion molecule (NCAM, CD56) [18] but it is difficult to use this system in heterogeneous populations of cells derived from differentiating hESC because CD56 is expressed on other early progenitor cells such as those of skeletal muscle (J. Parris and L. Armstrong, personal communication).

Expression Profiling of Undifferentiated hESC, CD1331 NSCs and Differentiated b-III-Tubulin1 Neurons H9 hESC exhibited unambiguous expression of a set of important genes associated with pluripotency, including OCT4, NANOG, SOX2, GDF3, LIN28, and TERT (Fig. 2). The expression levels of these genes decreased gradually on hESC differentiation although low levels were still detected in www.StemCells.com

CD133þ cells apart from SOX2, whose expression is highly abundant in neuroepithelial cells and neural progenitors [19, 20]. It has been reported that hESCs express many lineagespecific genes at very low levels [21] and from all examined lineage-specific genes representing all three embryonic germ layers examined, only GFAP was not detected in H9 hESCs (Fig. 2). However, the expression of such lineage-specific genes was significantly lower in hESCs compared with the differentiated cells in which they are functionally expressed (data not shown). All markers of neural stem/progenitor cells were upregulated in CD133þ cells and further downregulated in the b-III-tubulinþ population. The expression profile observed in CD133þ populations indicates a neuroepithelial status. Unlike adult central nervous system (CNS) NSCs, radial glial (RG) cells do not express GFAP [22, 23]. The expression of OLIG2 and PAX7 together with neurogenic RG markers, such as PAX6, NES (Nestin), and VIM (Vimentin) could indicate that these cells do not correlate to a specific regional identity and CD133þ cells could retain the potential to differentiate into a wide range of neural precursors. An increase in the expression levels of the markers of more mature neural lineages such as MBP and TUBB3 (b-III-Tubulin) in the CD133þ population could indicate that these cells are more permissive for the ‘‘leaky’’ expression of latter neural markers in NSCs compared with hESCs. Importantly, only

1302

Epigenetic Landscaping During hESC Differentiation

TUBB3 expression was detected in b-III-tubulinþ populations. No GFAP expression was detected in any of the examined cell populations, indicating a lack of contamination by astrocytic cells from differentiated H9 hESC-derived aggregates. GFAP expression is regulated by DNA methylation present at its promoter in neuroepithelial cells during mouse development and is thus activated only in mature astrocytes [24]. Except for baseline levels of KRT19 (Cytokeratin19), none of examined non-neural markers was detected in H9 hESCderived progeny. The epithelial marker P63 was not present, which suggests that the expression of KRT19 was not indicative of an epithelial cell fate. VIM was also present in BJ fibroblasts. The baseline expression levels of NES, TUBB3, and KRT19 in BJ fibroblasts could reflect low levels of cellular stress response [25–27].

trols for the chromatin immunoprecipitations were also performed. Glyceraldehdye 3-phosphate dehydrogenase (GAPDH) is ubiquitously expressed in all human cells and therefore a fragment of its promoter (215 bp before the transcription start site to 20 bp after) is used as a positive control for activating histone modifications (see supporting information Table 3 and associated figure). This section of the GAPDH promoter does not have repressive histone modifications in any of the cell types analyzed. These data support those of previous groups in which a lack of H3K27me3 has been reported at the Oct4 and Sox2 promoters in mouse neuronal precursor cells [31], indicating that proteins other than Polycomb complexes play a role in silencing pluripotency genes at the early stages of differentiation.

Changes in Histone Modification Patterns at the Promoters of Pluripotency-Associated Genes Reveals the Repression of Their Promoters During hESC Differentiation

Bivalent Promoters of Lineage-Specific Genes Resolve at the Progenitor Stage and Are Further Repressed upon Latter Differentiation

ChiP gives information about the spatio-temporal occupancy of a DNA sequence by a native protein in its in vivo native chromatin environment [28]. The presence of H3ac, H4ac, and H3K4me at gene promoters has been strongly associated with potential transcriptional activation. In particular, H3K4me3 is associated with active transcription [29]. All examined permissive marks were found to be present at the promoters of the examined pluripotency-associated genes (Fig. 3). Low levels of H3K4me2 were detected at the pluripotency markers in BJ fibroblasts, which confirmed a wider abundance of this mark among silent sections of the genome; but importantly, no H3K4me3 was present. Generally, pluripotency-associated gene promoters did not display the H3K9ac mark in BJ fibroblasts, as this mark is associated with transcriptional competence. Because of the difficulty of obtaining high numbers of cells after differentiation and FACS sorting, the presence of only two permissive histone modifications was assessed during hESC differentiation (H3K4me2 and H3K4me3). A gradual decrease in expression levels was associated with the loss of high levels of permissive marks at promoters of the pluripotency-associated genes examined herein (Fig. 3). CD133þ cells did not display H3K4me3 at the pluripotency-associated gene promoters correlating with their much lower expression levels in these cells; however, significant levels of H3K4me2 were still detected at the pluripotency-associated gene promoters in CD133þ and bIII-tubulinþ cells. H3K9me and H3K27me at the gene promoters are mainly associated with transcriptional repression and these marks are generally associated with facultative and constitutive heterochromatin respectively [30]. The promoters of pluripotency markers in H9 hESC did not display significant levels of H3K9me2, H3K9me3, or H3K27me3 (Fig. 3), whereas all three were present in BJ fibroblasts although the level of H3K27 trimethylation at the promoter of LIN28 is very low. The presence of H3K27me3 at pluripotency-associated gene promoters has been reported previously in murine T-cells [4] and mouse fibroblasts [6]. H3K9me2 was identified first at pluripotency-associated gene promoters in CD133þ NSCs, and its repressive activity was supported additionally by H3K9me3 and H3K27me3 in the b-III-tubulinþ population. The yield and efficiency of chromatin immunoprecipitation varies between antibodies directed against different histone modifications so it is difficult to make direct comparisons between absolute levels of H3K9 dimethylation, H3K9 trimethylation, and H3K27 trimethylation although comparisons of relative levels can be made (Fig. 3B). Con-

The presence of bivalent domains at the promoters of lineagespecific genes has been reported in ESCs, which consist of the permissive H3K4me3 mark together with the repressive H3K27me3 mark present at the same genomic loci [4, 5]. Studies of such domains have shown that lineage-specific transcription factors and genes involved in neurogenesis are particularly abundant among this group [32, 33]. Our data confirm the presence of such bivalent domains in hESC and show a correlation between histone modification patterns and gene expression. All of the examined neural lineage-specific transcription factors within this group, with the exception of GLI3, displayed bivalent domains at their promoters in hESC (Fig. 4). Resolution of these bivalent domains is expected on hESC differentiation and all bivalent domains were indeed resolved in CD133þ NSCs, in which only H3K4me3 remained at the promoters of genes upregulated during neurogenesis (PAX6, PAX7, SOX1, SOX3, and OLIG2). Markers of other lineages, such as NODAL and GATA4, lost H3K4me3 and displayed increased levels of H3K27me3, which remained also at high levels in the b-III-tubulinþ neurons. During further differentiation, the H3K27me3 mark was redisplayed at the bivalent promoters of neural stem/progenitor markers in b-III-tubulinþ cells, together with decrease or even loss of H3K4me3. Surprisingly, despite a lack of expression of OLIG2 and SOX3 in b-III-tubulinþ cells significant levels of H3K4me3 marks were still detected. In other more highly differentiated cells, resolution of bivalency should have taken place, and this is supported by the apparent lack of the H3K4me3 modification and the continued presence of H3K27me3 at the promoters of these same genes in the BJ fibroblasts, which are used as an example of a mostly differentiated cell type in which neuronal specific genes are not expressed. Some bivalent domains seem to persist (such as at the promoters of PAX6 and OLIG2) in BJ fibroblasts which supports the observations of other groups [6, 34]. Our data, however, suggest that this may not be an exact equivalent of the bivalency observed in both mESCs and hESCs because the levels of bivalent histone modifications detected in BJ fibroblasts are different than those of hESCs, with higher levels of the repressive H3K27me3 mark and lower levels of H3K4me3. The reasons for the apparent retention of bivalent chromatin domains in BJ fibroblasts are not clear at present. Interestingly, H3K4me3 was not the only permissive mark observed at the bivalent promoters in H9 hESCs and significant levels of H3K4me2, H3K9ac, H3ac, and H4ac were also present (Fig. 4). This indicates that H3K4me3 may be supported by other permissive marks, despite insignificant levels

Golebiewska, Atkinson, Lako et al.

1303

Figure 3. Histone modification patterns at pluripotency-associated gene promoters in H9 human embryonic stem cells (hESCs) and their differentiated counterparts. (A): Presence of permissive and repressive histone modification marks was examined at the promoters of pluripotency-associated gene promoters in H9 hESCs (H9) and differentiated H9 hESC-derived neural stem cells (NSCs) (CD133þ) and H9 hESC-derived putative neurons (b-III-tubulinþ). Human BJ fibroblasts (BJ) were used as a control representing a nonpluripotent cell line. ChiP experiments were performed with use 10,000 cells per immunoprecipitation reaction. Levels of immunoprecipitated fragments of DNA were validated by Q-PCR and results have been displayed as a percentage of an input chromatin sample to normalize between reactions. The data are presented as mean  SEM (n ¼ 4). Background was controlled for during the ChIP procedure by using a no-antibody control. (B): The repressive histone marks dimethyl H3K9, trimethyl H3K9, and trimethyl H3K27 are shown in detail to compare the relative levels of these modifications between H9 hESCs (H9), differentiated H9 hESC-derived NSCs (CD133þ), H9 hESC-derived putative neurons (b-III-tubulinþ), and BJ fibroblasts. Abbreviation: hTERT, telomerase reverse transcriptase subunit.

of transcription in hESCs. H3K4me2 and histone H3 acetylation levels were significantly decreased or totally lost in BJ fibroblasts at the promoters of most of the lineage-specific genes not expressed in this cell line. As obtaining high number of FACS sorted populations is difficult, only the permissive mark examined at bivalent promoters during neural difwww.StemCells.com

ferentiation was H3K4me2. An increase in H3K4me2 levels was detected at most of the bivalent neural promoters activated in CD133þ cells. Significant levels of H3K4me2 were also observed at the promoters of non-neural genes in the H9 hESC-derived progeny, which could reflect the loss of H3K4me3 at the same residue upon resolution of bivalency.

1304

Epigenetic Landscaping During hESC Differentiation

Figure 4. Histone modification patterns at bivalent promoters of lineage-specific genes in H9 human embryonic stem cells (hESCs) and their differentiated counterparts. Presence of permissive and repressive histone modification marks was examined at the promoters of lineage-specific gene promoters H9 hESCs (H9) and differentiated H9 hESC-derived neural stem cells (CD133þ) and H9 hESCderived putative neurons (b-III-tubulinþ). Human BJ fibroblasts (BJ) were used as a control representing a nonpluripotent cell line. ChiP experiments were performed with use of 10,000 cells per immunoprecipitation reaction. Levels of immunoprecipitated fragments of DNA were validated by Q-PCR and results have been displayed as a percentage of an input chromatin sample to normalize between reactions. The data are presented as mean  SEM (n ¼ 4). Background was controlled for during the ChIP procedure by using a noantibody control.

H3K4me2 was still detected at non-neural gene promoters also in b-III-tubulinþ cells. Low baseline levels of H3K9me2 and H3K9me3 were detected at the bivalent promoters in H9 hESCs, possibly due to the persistence of spontaneously differentiated cells within the hESC culture (Fig. 4). Both H3K9me2 and H3K9me3 were significantly increased at the silent bivalent promoters in BJ fibroblasts used as a typical differentiated cell reference. In the hESC-derived CD133þ NSCs, H3K27me3 present at the promoters of non-neural markers was supported by H3K9me2 and H3K9me3 appeared additionally in further differentiated b-III-tubulinþ cells. Both H3K9me3 and H3K9me2 were present together with H3K27me3 in b-III-tubulinþ cells at the promoters of silenced

early neural stem/progenitor markers. This indicates that Polycomb group (PcG) proteins important for establishing the H3K27me3 mark are not the only regulators that cause repression of bivalent domain promoters at lineage-specific genes during differentiation.

Nonbivalent Promoters of Lineage-Specific Genes Exhibit Changes in Permissive and Repressive Histone Marks Associated with Transcriptional Status of Genes Several genes upregulated in the CD133þ NSCs do not demonstrate bivalent chromatin domains in the preceding H9

Golebiewska, Atkinson, Lako et al.

1305

Figure 5. Histone modification patterns at nonbivalent promoters of lineage-specific genes in H9 human embryonic stem cells (hESCs) and their differentiated counterparts. Presence of permissive and repressive histone modification marks was examined at the promoters of lineage-specific gene promoters in H9 hESCs (H9) and differentiated H9 hESC-derived neural stem cells (CD133þ) and H9 hESC-derived putative neurons (b-III-tubulinþ). Human BJ fibroblasts (BJ) were used as a control representing a nonpluripotent cell line. ChiP experiments were performed with use of 10,000 cells per immunoprecipitation reaction. Levels of immunoprecipitated fragments of DNA were validated by Q-PCR and results have been displayed as a percentage of an input chromatin sample to normalize between reactions. The data are presented as mean  SEM (n ¼ 4). Background was controlled for during the ChIP procedure by using a no-antibody control. Abbreviations: GFAP, glial fibrillary acidic protein; MBP, myelin basic protein; NES, nestin; TUBB3, b-IIITubulin; VIM, vimentin.

hESC (Fig. 5) supporting published observations of several groups [32, 33]. All examined nonbivalent domain promoters, with the exception of GLI3, belong to proteins important in cytoskeleton formation in differentiated progeny. In case of the NES and VIM promoters, H3K4me3 was detected in pluripotent cells indicating that these genes are also poised for transcription in the early stages of differentiation; however, the mechanisms which keep these genes silent are likely to be Polycomb-independent because they lack H3K27me3. Neither H3K4me3 nor H3K27me3 were displayed at the TUBB3, GFAP, MBP, and GLI3 promoters in H9 hESC. Expression of GFAP and MBP are induced on terminal differentiation of glial lineages and possibly there is little requirement for them www.StemCells.com

to be poised for transcriptional activation in hESCs yet. Other permissive histone marks were found to be present at the lineage-specific genes, which did not display bivalency in H9 hESC (Fig. 5). Interestingly, H3K4me2, H3K9ac, H3ac, and H4ac were present also at the GFAP, MBP, and GLI3 promoters. Only baseline levels of H3K27me3 were displayed at the promoters of NES, GFAP, and MBP in BJ fibroblasts, indicating that other mechanisms independent of Polycomb proteins are required for silencing these genes in non-neural cells. VIM is abundantly expressed in fibroblast cells and a significant increase of H3K4me3 and other permissive marks was observed at its promoter in BJ fibroblasts, with no H3K27me3 detected. Low levels of NES and TUBB3

Epigenetic Landscaping During hESC Differentiation

1306

expression in BJ fibroblasts were reflected in the histone modification pattern at their promoters. Permissive histone marks were at similar levels as in H9 hESCs and no H3K27me3 was displayed at these parts of the genome. As pluripotent cells differentiated to CD133þ NSCs, levels of H3K4me3 and H3K4me2 increased at the promoters of NES, VIM, TUBB3, MBP, and GFAP. Low levels of H3K27me3 also appeared at the promoters of GFAP and MBP, which resembles the establishment of a form of bivalent domain especially because these genes are not yet expressed at significant levels in CD133þ NSCs. Monh et al. reported the appearance of new bivalent domains in mouse neuronal progenitors at the promoters of genes expressed during the latter stages of differentiation (34). Whether the results observed in this study truly report bivalent domains appearing in NSCs is uncertain, as the reported levels of H3K27me3 were much lower than the documented levels of classic bivalent domains present in hESCs. As MBP and GFAP were downregulated in the b-IIItubulinþ neuronal population only the H3K27me3 mark remained at their promoters which is reminiscent of the resolution of bivalent chromatin domains and thus lends support to the possible bivalent status of their promoters in the CD133þ NSCs. As expected, H3K4me3 and H3K4me2 levels were strongly increased at the TUBB3 promoter in b-IIItubulinþ putative neurons, with no H3K27me3 detected. Additionally, significant levels of H3K4me3 were present at the NES and VIM promoters, concomitant with H3K27me3 displayed in b-III-tubulinþ cells, although again not at the levels equivalent to bivalent domains in hESCs. Threshold levels of the H3K9me2 and H3K9me3 marks were observed at several nonbivalent domain promoters in H9 hESCs (Fig. 5). Only the VIM promoter showed no H3K9me in BJ fibroblasts. Interestingly, the levels of H3K9me2 and especially H3K9me3 present at other silent lineage-specific promoters seem to be lower compared with most of the examined bivalent domain promoters, which could again support the prediction that these genes are repressed by other mechanisms than classic repressive histone modifications. A significant increase of the H3K9me2 mark was detected in CD133þ cells at the MBP and GFAP promoters (Fig. 5). It is possible that this mark might play a more important role in keeping the poised genes in progenitor cells in the repressed form, rather than H3K27me3. With the exception of the TUBB3 gene, all other markers were repressed in terminally differentiated cells, with H3K9me2 and H3K9me3 marks detected at their promoters in b-III-tubulinþ cells. Interestingly, the levels of repressive marks were much higher compared with BJ fibroblasts. This could indicate differences between two differentiated cell types, especially with regard to silencing neural stem/ progenitor markers during final differentiation. However, further investigation must be performed to exclude the influence of in vitro conditions in regulation of epigenetic changes during hESC differentiation. Only the TUBB3 promoter remained in a fully permissive state during all stages of differentiation.

DISCUSSION In this study, we tried to establish the importance of locusspecific post-translational modifications of histones in regulation of transcription during differentiation of hESCs. The presence of a panel of histone modifications at regulatory sequences were correlated with transcriptional status of the examined genes. Given that most studies to date have concentrated on not more than two histone modifications, a full appreciation of the changes occurring at the promoters during

differentiation is often not permitted, which may lead to misleading interpretations. One of the main limitations of epigenetic examination of changes occurring during mESC and hESC differentiation lies in the heterogeneity of cell populations produced during differentiation protocols. An efficient purification system was established to examine changes at the promoters of lineage specific genes occurring during differentiation. Two steps of lineage-specification CD133þ NSCs and b-III-tubulinþ putative neurons were found to be an excellent system to examine temporal changes at certain lineage-specific genes during progress of differentiation. Despite genome-wide examination these have not been addressed and the clear trends observed in this study clear elucidate this process in more detail. It may be argued that such trends are specific to the H9 hESC line but because other hESC lines are capable of neuronal differentiation [35] we believe that the hESC used in this work are representative of pluripotent cell differentiation. The histone modification pattern present at pluripotencyassociated gene promoters was positively correlated with transcriptional status. All pluripotency-associated gene promoters displayed only permissive histone marks in hESCs. We report that H3K9me2 could play a role in initiation of silencing and is followed by H3K27me3 and H3K9me3 during terminal differentiation into neuronal lineages. Additionally, significant levels of permissive histone marks were still observed after in vitro differentiation at silent gene promoters, which on the contrary were not present in human BJ fibroblasts, which suggests that certain levels of permissive marks can still be present at already silent gene promoters, despite the lack of gene expression and that repressive histone modifications exhibit a dominant effect in maintaining the repressed state of the promoters. BJ fibroblasts are a terminally differentiated cell type that we believe represent an opposite state of differentiation to hESC and are a useful comparison for intermediate differentiation states such as the CD133 positive NSCs. It needs further investigation to elucidate whether observed differences arise from the different cell types examined or whether certain mechanisms are not fully functional under in vitro conditions. Bivalent domains were observed at many early lineagespecific gene promoters, mainly transcription factors, in hESCs. Interestingly, we show that other permissive marks support H3K4me3 in poising the promoters in hESCs. During differentiation bivalent domains were resolved according to the transcriptional status of the gene and repression of the promoters was followed additionally by the elevated levels of H3K9 methylation. Interestingly, the study shows for the first time that in case of bivalent domain gene promoters resolved in the neural progenitor state to favor gene repression upon terminal differentiation, the H3K27me3 mark was redisplayed together with H3K9me2 and H3K9me3 supporting the final repression state. These results are consistent with previous reports, in that expression of only relatively small numbers of genes in human embryonic fibroblasts is controlled by PcG proteins (which establish the H3K27me3 mark) [36], suggesting that the majority of them are permanently silenced by other mechanisms. It has been reported that bivalent domain promoters together with house-keeping gene promoters and pluripotency-associated gene promoters can be characterized by the presence of high-density CpG islands, which are more likely to be hypermethylated during in vitro differentiation [37]. The association between H3K9 methylation and DNA methylation has been already well documented [38, 39]. A substantial class of genes in hESCs lacks both H3K4me3 and H3K27me3 [33, 34]. As their promoters lack H3K27me3, the relative inactivity of the genes appears to be

Golebiewska, Atkinson, Lako et al.

PcG-independent, yet how they are repressed in ESCs remains unknown. Interestingly, we show that all examined nonbivalent gene promoters displayed significant levels of permissive histone marks, including H3K4me3 in certain cases in hESCs. Given their important role in cell lineage determination, it is likely that these genes become permissive for activation at later developmental stages through as yet uncharacterized epigenetic mechanisms. Data arising from many reports suggest that many genes are regulated mainly by levels of histone acetylation. In many cases, simple inhibition of histone deacetylases is sufficient for activation of expression [40–42]. Moreover, genes encoding cytoskeleton proteins were found to be abundant within repressor element 1-silencing transcription factor (REST) regulated genes during neurogenesis [43], and correlation between REST and histone deacetylase has been well documented [44, 45]. It is possible that only primary lineagecontrol genes are required for the very early developmental stages and bivalent domains enable their immediate activation. Importantly, during increases in gene expression, the gene promoters displayed increased levels of permissive marks. Upon repression presence of repressive marks, particularly H3K9me2 and H3K9me3 was detected; however, the increase was much less significant compared with the high levels detected at repressed bivalent domain promoters. Another question is emerging in recently published data, whether bivalency observed at certain promoters of lineagespecific genes in differentiated cells is equivalent to the same phenomenon in hESCs. In this study, we report that few lineage-specific genes still displayed both H3K4me3 and H3K27me3 in BJ fibroblasts and b-III-tubulinþ neurons; however, the overall relation between two marks was different compared with bivalent domains observed in hESCs. Similarly, the presence of newly formed bivalent domains in CD133þ NSCs is questionable for similar reason. Nevertheless, the possible role of H3K9me2 in repressing certain

REFERENCES Shamblott MJ, Axelman J, Wang S et al. Derivation of pluripotent stem cells from cultured human primordial germ cells. Proc Natl Acad Sci USA 1998;95:13726–13731. 2 Roh TY, Cuddapah S, Cui K et al. The genomic landscape of histone modifications in human T cells. Proc Natl Acad Sci USA 2006; 103:15782–15787. 3 Guenther MG, Levine SS, Boyer LA et al. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 2007;130:77–88. 4 Azuara V, Perry P, Sauer S et al. Chromatin signatures of pluripotent cell lines. Nat Cell Biol 2006;8:532–538. 5 Pan G, Tian S, Nie J et al. Whole-genome analysis of histone H3 lysine 4 and lysine 27 methylation in human embryonic stem cells. Cell Stem Cell 2007;1:299–312. 6 Mikkelsen TS, Ku M, Jaffe DB et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 2007;448: 553–560. 7 Szutorisz H, Canzonetta C, Georgiou A et al. Formation of an active tissue-specific chromatin domain initiated by epigenetic marking at the embryonic stem cell stage. Mol Cell Biol 2005;25:1804– 1820. 8 Chakrabarti SK, Francis J, Ziesmann SM et al. Covalent histone modifications underlie the developmental regulation of insulin gene transcription in pancreatic beta cells. J Biol Chem 2003;278: 23617–23623. 9 Tilgner K, Atkinson SP, Golebiewska A et al. Isolation of primordial germ cells from differentiating human embryonic stem cells. Stem Cells 2008;26:3075–3085. 10 Dahl JA, Collas P. A quick and quantitative chromatin immunoprecipitation assay for small cell samples. Front Biosci 2007;12: 4925–4931. 11 Dahl JA, Collas P. Q2ChIP, a quick and quantitative chromatin immunoprecipitation assay, unravels epigenetic dynamics of developmentally

1307

poised genes in progenitor state should be further approached. The inhibition of gene expression in multipotent state by H3K9 methylation has been reported on many genes expressed in later stages of differentiation such as opioid receptors [46] or genes encoding synaptic vesicle proteins [47] that are important in neuronal cells. It should be noted that data arising in the literature is a result of different ChiP protocols with different detection techniques and importantly various threshold levels. Furthermore, only a few bivalent targets have been rigorously confirmed by sequential ChiP to have concomitant H3K4me3 and H3K27me3 marks on the same chromatin segment. The evaluation of the presence of different marks on the same DNA sequence is crucial in making final conclusions. The question whether the bivalency is still a valid phenomenon in lineage-restricted cells still remains open.

ACKNOWLEDGMENTS The authors are grateful to Marcin Jurga and Colin McGuckin of Newcastle University for the kind gift of antibodies used for immunostaining in this study and to Ian Dimmick and Rebecca Stewart of Newcastle University for their assistance with FACS. The authors also like to thank ONE North East Regional Developmental Agency for funding this work.

DISCLOSURE

POTENTIAL CONFLICTS INTEREST

The authors indicate no potential conflicts of interest.

12

1

www.StemCells.com

OF OF

13 14

15 16 17 18 19 20

21 22 23

regulated genes in human carcinoma cells. Stem Cells 2007;25: 1037–1046. Kania G, Corbeil D, Fuchs J et al. Somatic stem cell marker prominin-1/CD133 is expressed in embryonic stem cell-derived progenitors. Stem Cells 2005;23:791–804. Lee A, Kessler JD, Read TA et al. Isolation of neural stem cells from the postnatal cerebellum. Nat Neurosci 2005;8:723. Marzesco AM, Janich P, Wilsch-Brauninger M et al. Release of extracellular membrane particles carrying the stem cell marker prominin-1 (CD133) from neural progenitors and other epithelial cells. J Cell Sci 2005;118:2849–2858. Miraglia S, Godfrey W, Yin AH et al. A novel five-transmembrane hematopoietic stem cell antigen: Isolation, characterization, and molecular cloning. Blood 1997;90:5013–5021. Alessandri G, Pagano S, Bez A et al. Isolation and culture of human muscle-derived stem cells able to differentiate into myogenic and neurogenic cell lineages. Lancet 2004;364:1872–1883. Sacco A, Doyonnas R, Kraft P et al. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008;456:502–506. Pruszak J, Sonntag KC, Aung MH et al. Markers and methods for cell sorting of human embryonic stem cell-derived neural cell populations. Stem Cells.2007;25:2257–2268. Avilion AA, Nicolis SK, Pevny LH et al. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev 2003;17:126–140. Zappone MV, Galli R, Catena R et al. Sox2 regulatory sequences direct expression of a (b)-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells. Development 2000;127: 2367–2382. Efroni S, Duttagupta R, Cheng J et al. Global transcription in pluripotent embryonic stem cells. Cell Stem Cell 2008;2:437–447. Noctor SC, Martinez-Cerdeno V, Ivic L et al. Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases. Nat Neurosci 2004;7:136–144. Kriegstein AR, Noctor SC. Patterns of neuronal migration in the embryonic cortex. Trends Neurosci 2004;27:392–399.

Epigenetic Landscaping During hESC Differentiation

1308

24 Takizawa T, Nakashima K, Namihira M et al. DNA methylation is a critical cell-intrinsic determinant of astrocyte differentiation in the fetal brain. Dev Cell 2001;1:749–758. 25 Neuhuber B, Gallo G, Howard L et al. Reevaluation of in vitro differentiation protocols for bone marrow stromal cells: Disruption of actin cytoskeleton induces rapid morphological changes and mimics neuronal phenotype. J Neurosci Res 2004;77:192–204. 26 Croft AP, Przyborski SA. Formation of neurons by non-neural adult stem cells: Potential mechanism implicates an artifact of growth in culture. Stem Cells 2006;24:1841–1851. 27 Raedt R, Pinxteren J, Van Dycke A et al. Differentiation assays of bone marrow-derived multipotent adult progenitor cell (MAPC)-like cells towards neural cells cannot depend on morphology and a limited set of neural markers. Exp Neurol 2007;203:542–554. 28 Orlando V. Mapping chromosomal proteins in vivo by formaldehyde-crosslinked-chromatin immunoprecipitation. Trends Biochem Sci 2000;25: 99–104. 29 Schneider R, Bannister AJ, Myers FA et al. Histone H3 lysine 4 methylation patterns in higher eukaryotic genes. Nat Cell Biol 2004;6: 73–77. 30 Zinner R, Albiez H, Walter J et al. Histone lysine methylation patterns in human cell types are arranged in distinct three-dimensional nuclear zones. Histochem Cell Biol 2006;125(1–2):3–19. 31 Boyer LA, Plath K, Zeitlinger J et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 2006;441:349–353. 32 Pan G, Tian S, Nie J et al. Whole-genome analysis of histone H3 lysine 4 and lysine 27 methylation in human embryonic stem cells. Cell Stem Cell 2007;1:299–312. 33 Zhao XD, Han X, Chew JL et al. Whole-genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells. Cell Stem Cell 2007;1:286–298. 34 Mohn F, Weber M, Rebhan M et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol Cell 2008;30:755–766. 35 Ma W, Tavakoli T, Derby E et al. Cell-extracellular matrix interactions regulate neural differentiation of human embryonic stem cells. BMC Dev Biol 2008;8:90–103.

36 Bracken AP, Dietrich N, Pasini D et al. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev 2006;20:1123–1136. 37 Meissner A, Mikkelsen TS, Gu H et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 2008;252: 766–770. 38 Sato N, Kondo M, Arai K. The orphan nuclear receptor GCNF recruits DNA methyltransferase for Oct-3/4 silencing. Biochem Biophys Res Commun 2006;344:845–851. 39 Feldman N, Gerson A, Fang J et al. G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat Cell Biol 2006;8:188–194. 40 Han S, Lu J, Zhang Y et al. HDAC inhibitors TSA and sodium butyrate enhanced the human IL-5 expression by altering histone acetylation status at its promoter region. Immunol Lett 2007;108:143–150. 41 Van Lint C, Emiliani S, Verdin E. The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr 1996;5:245–253. 42 McCool KW, Xu X, Singer DB et al. The role of histone acetylation in regulating early gene expression patterns during early embryonic stem cell differentiation. J Biol Chem 2007;282:6696–6706. 43 Bruce AW, Donaldson IJ, Wood IC et al. Genome-wide analysis of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes. Proc Natl Acad Sci USA 2004;101:10458–10463. 44 Huang Y, Myers SJ, Dingledine R. Transcriptional repression by REST: Recruitment of Sin3A and histone deacetylase to neuronal genes. Nat Neurosci 1999;2:867–872. 45 Naruse Y, Aoki T, Kojima T et al. Neural restrictive silencer factor recruits mSin3 and histone deacetylase complex to repress neuron-specific target genes. Proc Natl Acad Sci USA 1999;96:13691–13696. 46 Park SW, He Y, Ha SG et al. Epigenetic regulation of kappa opioid receptor gene in neuronal differentiation. Neuroscience 2008;151: 1034–1041. 47 Ekici M, Hohl M, Schuit F et al. Transcription of genes encoding synaptic vesicle proteins in human neural stem cells: Chromatin accessibility, histone methylation pattern, and the essential role of rest. J Biol Chem 2008;283:9257–9268.

See www.StemCells.com for supporting information available online.