Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

LETTER doi:10.

1038/nature13981

Intracellular a-ketoglutarate maintains the


pluripotency of embryonic stem cells
Bryce W. Carey1*, Lydia W. S. Finley2*, Justin R. Cross3, C. David Allis1 & Craig B. Thompson2

The role of cellular metabolism in regulating cell proliferation and (EpiSCs) could not proliferate in the absence of exogenous glutamine
differentiation remains poorly understood1. For example, most mam- (Extended Data Fig. 1f, g). However, the ability to undertake glutamine-
malian cells cannot proliferate without exogenous glutamine supple- independent growth was not limited to embryonic pluripotency; fibro-
mentation even though glutamine is a non-essential amino acid1,2. blast-derived induced pluripotent cells (iPSCs) were also able to pro-
Here we show that mouse embryonic stem (ES) cells grown under liferate in glutamine-free 2i/L medium (Extended Data Fig. 1h). These
conditions that maintain naive pluripotency3 are capable of prolif- results indicate that the GSK-3b and MAPK/ERK inhibitors in 2i-
eration in the absence of exogenous glutamine. Despite this, ES cells containing medium are both necessary and sufficient to enable prolif-
consume high levels of exogenous glutamine when the metabolite is eration of pluripotent cells in the absence of exogenous glutamine.
available. In comparison to more differentiated cells, naive ES cells The fact that cells proliferated in the absence of exogenous glutamine
utilize both glucose and glutamine catabolism to maintain a high in 2i/L medium, albeit at a slower rate than cells cultured in glutamine-
level of intracellular a-ketoglutarate (aKG). Consequently, naive ES replete medium (Extended Data Fig. 1i), indicates that these cells must
cells exhibit an elevated aKG to succinate ratio that promotes histone/ be capable of de novo glutamine synthesis. Indeed, chemical inhibition
DNA demethylation and maintains pluripotency. Direct manipula- of glutamine synthase was sufficient to block proliferation of cells in
tion of the intracellular aKG/succinate ratio is sufficient to regulate glutamine-free 2i/L medium (Extended Data Fig. 1j). Likewise, addition
multiple chromatin modifications, including H3K27me3 and ten- of cell-permeable dimethyl-a-ketoglutarate (DM-aKG), a precursor for
eleven translocation (Tet)-dependent DNA demethylation, which glutamine synthesis, was sufficient to enable glutamine-independent
contribute to the regulation of pluripotency-associated gene express- proliferation in both S/L and 2i/L conditions (Extended Data Fig. 1k),
ion. In vitro, supplementation with cell-permeable aKG directly sup- suggesting that the supply of precursors for glutamine synthesis deter-
ports ES-cell self-renewal while cell-permeable succinate promotes mines the ability of ES cells to proliferate in the absence of glutamine.
differentiation. This work reveals that intracellular aKG/succinate In support of this model, cells cultured in 2i/L preserved larger intra-
levels can contribute to the maintenance of cellular identity and cellular pools of glutamate after glutamine withdrawal than cells cul-
have a mechanistic role in the transcriptional and epigenetic state tured in S/L (Fig. 1g). These results suggest that 2i/L cells can generate
of stem cells.
Mouse ES cells can be maintained in two medium formulations: a
a ESC-V19 b ESC-1–4 c 2i/L –Q
serum-free medium reported to support a cellular phenotype that mimics 0.4 0.4 ESC-1 2i/L
S/L –Q
Cell number (× 106)
Cell number (× 106)

‘naive’ epiblast cells of the inner cell mass (containing GSK-3b and MAPK/ 0.3
2i/L –Q 0.3
ESC-2 2i/L
ESC-3 2i/L
ERK inhibitors (2i)/leukaemia inhibitory factor (LIF), hereafter 2i/L); 0.2 0.2
ESC-4 2i/L
ESC-1 S/L BF
or a serum-based medium that supports the proliferation of a more ESC-2 S/L
0.1 0.1
committed ES cell phenotype (serum/LIF, hereafter S/L)4–11. To char- ESC-3 S/L
ESC-4 S/L
0 0
acterize ES cell metabolism, we investigated whether cells cultured in 0 1 2 3 4 0 1 2 3 4
Day Day AP
these two media have different requirements for glucose and/or gluta-
mine. ES cells cultured in either medium proliferated at equivalent rates d ESC-V19 e S/L/2i –Q f ESC-V19 g Glutamate
0.25 0.3
Metabolite levels (a.u.)

S/L –Q 1.2
when glucose and glutamine were abundant and cells cultured with or 2i/L –Q S/L
Cell number (× 106)

Cell number (× 106)

0.20 S/L2i –Q BMP4/L –Q 2i/L


without 2i were unable to proliferate in the absence of glucose (Ex- 0.15
0.2
0.8
BF
tended Data Fig. 1a, b). In contrast, cells cultured in 2i/L, but not S/L, 0.10
0.1
proliferated robustly in the absence of exogenous glutamine (Fig. 1a 0.05 0.4

and Extended Data Fig. 1c). Likewise, four newly derived ES-cell lines 0
0 1 2 3 4
0
0 1 2 3 4 0
AP +Q –Q
(ESC-1–4) exhibited convincing glutamine-independent proliferation Day Day

in 2i/L medium while retaining features of pluripotent cells, including Figure 1 | 2i is necessary and sufficient to confer glutamine independence.
ES-cell-like morphology, reactivity to alkaline phosphatase (AP) and a–f, Growth curves and representative images of ES cells grown in the absence
the ability to form teratomas (Fig. 1b, c and Extended Data Fig. 1d). of glutamine (Q). a, b, Growth curves of ESC-V19 cells (a) and V6.5 ES-cell
Cells cultured in 2i medium alone could also proliferate in the absence lines (ESC-1–4) (b) cultured in glutamine-free S/L or 2i/L medium. c, e, Phase
of exogenous glutamine (Extended Data Fig. 1e). images showing ESC-1 cells cultured in glutamine-free 2i/L (c) or S/L/2i (e)
This effect was not due to differences in medium nutrient formulations medium for 3 days. Top, brightfield (BF); bottom, AP staining. Scale bars,
as supplementing S/L medium with the GSK-3b and MAPK/ERK in- 500 mm. d, Growth curve of ESC-V19 cells in glutamine-free S/L or S/L/2i
medium. f, Growth curve of ESC-V19 cells cultured without glutamine in two
hibitors present in 2i also enabled glutamine-independent proliferation serum-free media formulations containing N2 and B27 supplements, 2i/L and
while maintaining ES cell morphology and markers of pluripotency BMP4/L. g, Intracellular glutamate levels 8 h after addition of medium with or
(Fig. 1d, e). An alternative ES-cell medium containing BMP4 and LIF without glutamine. a.u., arbitrary units. Data are presented as the
added to the same serum-free formulation as in 2i/L12 failed to support mean 6 standard deviation (s.d.) of triplicate wells from a representative
glutamine-independent growth (Fig. 1f). Likewise, epiblast stem cells experiment.
1
Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, New York 10065, USA. 2Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York,
New York 10065, USA. 3Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.
*These authors contributed equally to this work.

1 9 F E B R U A RY 2 0 1 5 | VO L 5 1 8 | N AT U R E | 4 1 3
©2015 Macmillan Publishers Limited. All rights reserved
RESEARCH LETTER

glutamate (and glutamine) from carbon sources other than glutamine cells cultured in 2i/L medium were able to use glucose-derived carbons
itself. to maintain elevated glutamate pools sufficient to support cell growth
Despite their different growth requirements, cells cultured in both (Extended Data Fig. 2b). Moreover, in comparison with their S/L coun-
S/L and 2i/L consumed high levels of glucose and glutamine and excreted terparts, 2i/L cells used more glucose-derived carbon and relatively less
similar levels of lactate, consistent with the metabolic profile of most glutamine-derived carbon to support protein synthesis (Extended Data
proliferating cells, including cancer cells and pluripotent cells (Fig. 2a)1,13. Fig. 2c), confirming that 2i promotes increased glucose-dependent
Oxidation of glucose and glutamine via the mitochondrial tricarboxylic amino acid synthesis.
acid (TCA) cycle provides a critical source of the biosynthetic precur- Diminished glutamine entry into the TCA cycle, coupled with the
sors required for cell proliferation. With the exception of aKG, steady- observed efflux of glucose-derived carbons from the TCA cycle as glu-
state levels of TCA cycle metabolites were reproducibly diminished in tamate, suggested that cells cultured in 2i/L might not be oxidizing all
ES cells cultured in 2i/L (Fig. 2b). the aKG produced from glutamine in the mitochondria. Indeed, the
In most cells, glutamine is catabolized to aKG to support TCA cycle aKG/succinate ratio was robustly elevated by 2i/L in every ES-cell line
anaplerosis (Fig. 2c). ES cells grown in S/L medium exhibited high levels tested (Fig. 3a). Cellular aKG/succinate ratios have been implicated in
of TCA cycle intermediates and virtually all intracellular glutamate, the regulation of the large family of aKG-dependent dioxygenases14. As
aKG and malate were rapidly labelled after addition of [U-13C]glutamine Jumonji C (JmjC)-domain-containing histone demethylases and the
(Fig. 2d). In contrast, a substantial fraction of these metabolites failed to Tet family of DNA demethylases comprise a major subset of these enzymes,
become labelled with glutamine in ES cells grown in 2i/L. Instead, there the elevated ratio of aKG/succinate observed in cells grown in 2i/L
was a rapid labelling of these three metabolite pools from [U-13C]glucose medium could have important implications for the regulation of chro-
(Fig. 2e). Quantification of metabolite fluxes revealed that although the matin structure.
flux of glutamine-derived carbons through aKG was similar in both Since aKG was largely derived from glutamine metabolism (Fig. 2d),
conditions, glutamine flux through malate was significantly diminished we tested whether glutamine deprivation affected histone lysine methy-
in cells cultured in 2i/L, indicating that the entry of glutamine-derived lations known to be regulated in part by aKG-dependent demethylases15.
aKG into the TCA cycle is repressed by culture in 2i/L (Fig. 2f). Instead, Cells cultured in glutamine-free medium exhibited increases in trimethy-
when cells are cultured in 2i/L, a substantial amount of both aKG and lation and decreases in monomethylation on H3K9, H3K27, H3K36 and
malate was produced from glucose (Fig. 2g). H4K20, whereas H3K4 methylations remained unchanged (Fig. 3b).
Consistent with these results, cells cultured with 2i inhibitors demon- DM-aKG reversed the increase in H3K27me3 and H4K20me3 observed
strated substantial glucose-dependent glutamate production (Extended in glutamine-deficient medium (Extended Data Fig. 3a), confirming that
Data Fig. 2a). Consequently, during conditions of glutamine depletion, these changes could be accounted for by the decline in glutamine-dependent

a Glucose uptake Glutamine uptake Lactate secretion


(mmol l–1 per mg protein)
(mmol l–1 per mg protein)

(mmol l–1 per mg protein)


Glutamine consumption

S/L
Glucose consumption

20 5 40
2i/L
Lactate production

4
15 30
3
10 20
2
5 10
1

0 0 0
Cell line: ESC-V19 ESC-1 ESC-2 ESC-3 ESC-4 Cell line: ESC-V19 ESC-1 ESC-2 ESC-3 ESC-4 Cell line: ESC-V19 ESC-1 ESC-2 ESC-3 ESC-4

Glucose
b αKG Succinate Malate Aspartate c
0.06 0.6 0.20 0.10 Pyruvate
Metabolite levels (a.u.)

Metabolite levels (a.u.)

Metabolite levels (a.u.)

Metabolite levels (a.u.)

* 0.08 Aspartate (Acetyl-CoA)


0.15
0.04 0.4 Citrate
0.06 (Oxaloacetate)
0.10
** 0.04
0.02 0.2 Malate (Isocitrate)
*** 0.05
0.02
***
0 0 0 0 Glutamine
αKG
S/L 2i/L S/L 2i/L S/L 2i/L S/L 2i/L Succinate Glutamate

d αKG Malate Glutamate f αKG Malate


Fraction labelled by 13C-Gln

Fraction labelled by 13C-Gln

Fraction labelled by 13C-Gln

1.0 1.0 1.0 2.0 2.0 **


Flux from Gln (a.u.)

Flux from Gln (a.u.)

0.8 0.8 0.8


1.5 1.5
0.6 0.6 0.6
1.0 1.0
0.4 0.4 0.4
2i/L 2i/L 2i/L 0.5 0.5
0.2 0.2 0.2
S/L S/L S/L
0 0 0 0 0
12 4 8 12 12 4 8 12 12 4 8 12 S/L 2i/L S/L 2i/L
Time (h) Time (h) Time (h)

e αKG Malate Glutamate g αKG Malate


Fraction labelled by 13C-Glc

Fraction labelled by 13C-Glc

Fraction labelled by 13C-Glc

1.0 1.0 1.0 2.0 2.0


2i/L 2i/L 2i/L
Flux from Glc (a.u.)
Flux from Glc (a.u.)

0.8 S/L 0.8 S/L 0.8 S/L 1.5 1.5


0.6 0.6 0.6 *
1.0 1.0
0.4 0.4 0.4
0.5 0.5 *
0.2 0.2 0.2
0 0 0 0 0
12 4 8 12 12 4 8 12 12 4 8 12 S/L 2i/L S/L 2i/L
Time (h) Time (h) Time (h)

Figure 2 | 2i/L alters glucose and glutamine utilization. a, Analysis of or derived from [U-13C]glucose (13C-Glc) (e) over time (0–12 h).
glucose uptake (left), glutamine uptake (centre) and lactate secretion (right). Mean 6 standard error of the mean (s.e.m.) of three independent experiments
b, Intracellular metabolite levels. Bars show mean of n 5 4 (a) or n 5 3 (b) are shown. f, g, Glutamine (f) and glucose (g) flux through aKG and malate
replicate wells 6 s.d. from representative experiments. c, Schematic of the TCA pools. Mean 6 s.e.m. of flux calculated for three independent experiments
cycle including entry points for glucose- and glutamine-derived carbons. (shown in d, e) are shown. *P , 0.05, **P , 0.005, ***P , 0.0005. P values
Isotope tracing was performed for metabolites shown in red. d, e, Fraction of were determined by unpaired two-tailed Student’s t-tests.
each metabolite labelled by 13C derived from [U-13C]glutamine (13C-Gln) (d)

4 1 4 | N AT U R E | VO L 5 1 8 | 1 9 F E B R U A RY 2 0 1 5
©2015 Macmillan Publishers Limited. All rights reserved
LETTER RESEARCH

a αKG/succinate b ESC-1 ESC-2 In ES cells, ‘bivalent domains’ are developmentally regulated genomic
0.25
S/L ***
Glutamine + – + –
16 regions characterized by the colocalization of H3K4me3 and H3K27me3
H3K9me3
2i/L *** (refs 17–19). Recent genome-wide analysis of H3K27me3 in S/L- and
αKG/succinate (a.u.)

0.20 16
*** H3K9me1
0.15
*** 2i/L-cultured ES cells revealed that H3K27me3 was specifically depleted
16
0.10
H3K27me3 at bivalent domain gene promoters in 2i/L-cultured cells11. Our data sug-
16
H3K27me1 gest that the observed increase in aKG might promote aKG-dependent
0.05
H3K27me3 demethylation in 2i/L ES cells. Indeed, cells cultured in
0 H4K20me3
ESC-1 ESC-2 ESC-3 ESC-4
6 2i/L exhibited a greater increase in H3K27me3 at bivalent domain
H4K20me1
c
6 promoters when incubated with the H3K27me3 demethylase inhibitor
O2 JMJD3 CO2
H3K36me3
16 GSK-J4 than cells cultured in S/L (Fig. 3e and Extended Data Fig. 3b, c).
UTX
H3K36me1
16 The average fold change across the 14 bivalent promoters tested showed
αKG Succinate a highly significant increase in 2i/L-cultured ES cells compared with
16
GSK-J4 H3K4me3 S/L-cultured ES cells (Fig. 3e). Similarly, two independent cell lines
d 16 with mutations in the Jumonji domain of the H3K27me3 demethylase
H3K4me1
GSK-J4 (μM) 0 1 2.5 5 7.5 10
H3K27me3
16
16
JMJD3 (JMJD3D/D-1 and JMJD3D/D-2) (Extended Data Fig. 4a–c) demon-
H3K27me1
16
H3
strated increases in H3K27me3 levels relative to control lines that were
16
H4
6 significantly elevated in cells cultured in 2i/L, reflecting enhanced demethy-
H3 H3
H2B
lation at these loci in ES cells cultured in 2i/L (Fig. 3f). Furthermore,
Direct blue H2A
H4 treatment with GSK-J4, but not the inactive isomer GSK-J5, increased
e f the aKG/succinate ratio in cells cultured in 2i/L (Fig. 3g). These results
ESC-1 JMJD3Δ/Δ-1 JMJD3Δ/Δ-2
2.0 2.0 2.5 indicate that 2i/L rewires glutamine metabolism to maintain aKG
pools favouring active demethylation of a variety of histone marks.
(JMJD3Δ/Δ/control)
(JMJD3Δ/Δ/control)
(GSK-J4/control)

1.5
Fold change
Fold change

In addition to reduced H3K27me3 at bivalent domain promoters,


Fold change

1.5 2.0
1.0 cells cultured in 2i/L exhibit DNA hypomethylation5,7–9. Incubating cells
0.5
1.0 1.5 with ascorbic acid, a cofactor for aKG-dependent dioxygenases, acti-
P < 0.0001 P < 0.005 P < 0.05 vates Tet-dependent gene expression and promotes DNA demethyla-
0
S/L 2i/L
0.5
S/L 2i/L
1.0
S/L 2i/L tion20. Therefore, we tested whether aKG treatment could exert similar
effects (Extended Data Fig. 5a). Total DNA methylation was reduced in
g
1.2
Ctrl ** cells cultured with cell-permeable aKG (Extended Data Fig. 5b) and
GSK-J4, active
GSK-J5, inactive
treatment with aKG, but not succinate, induced expression of inner-
αKG/succinate (a.u.)

0.8 cell-mass- and germline-associated genes previously identified as tar-


gets of Tet-mediated activation (Extended Data Fig. 5c)20,21. The effects
0.4 of aKG persisted upon extended passaging (Extended Data Fig. 5d) and
were largely abrogated in Tet1/Tet2 double-knockout ES cells (Extended
0
Dose (μM): – 1 1 5 5 – 1 1 5 5
Data Fig. 5e). These results suggest that intracellular aKG production
S/L 2i/L
may stimulate the activity of multiple aKG-dependent dioxygenases
to regulate coordinately the epigenetic marks characteristic of naive
Figure 3 | Histone demethylation is regulated by intracellular aKG in ES pluripotency.
cells. a, Gas chromatography–mass spectrometry (GC–MS) analysis of the
To test whether modulation of the aKG/succinate ratio can influence
aKG/succinate ratio in ESC-1–4 cells grown in either S/L or 2i/L medium.
b, Western blot of ESC-1 and ESC-2 cells grown in 2i/L medium with or pluripotent cell fate decisions, we performed colony-formation assays
without glutamine for 3 days. Molecular weight marker (in kDa) is shown. with S/L-cultured ES cells in the presence of aKG or succinate. Colonies
c, Simplified schematic of the reaction mechanism of aKG-dependent formed in S/L medium supplemented with DM-aKG (S/L1DM-aKG)
dioxygenases. d, ESC-1 cells grown in S/L in the presence of increasing amounts had brighter AP staining and retained the compact colony morphology
of GSK-J4 for 24 h. e, H3K27me3 chromatin immunoprecipitation followed by typical of undifferentiated ES cells (Fig. 4a). Although the total number
quantitative polymerase chain reaction (ChIP-qPCR) of ESC-1 cells cultured of colonies was similar in all three conditions, the S/L1DM-aKG wells
in S/L or 2i/L medium containing 30 mM of GSK-J4 for 5 h. Values represent contained more than double the number of fully undifferentiated col-
fold change (GSK-J4/control) at individual bivalent domain genes (n 5 14). onies compared with S/L only and S/L1DM-succinate (Fig. 4b). As a
f, H3K27me3 ChIP-qPCR of CRISPR/Cas9 edited cells JMJD3D/D-1 (left) and
further test of the ability of aKG to promote maintenance of ES cells,
JMJD3D/D-2 (right) cultured in S/L or 2i/L. Values represent fold change
(JMJD3D/D cells relative to control cells) at individual bivalent domain genes we used a knock-in Nanog–green fluorescent protein (GFP) reporter
(n 5 10). Bars represent mean values. P values were determined by unpaired line22 and found that aKG was sufficient to enhance Nanog expression
two-tailed Student’s t-test (e, f). g, The ratio of aKG/succinate in ESC-1 cells in a dose-dependent manner (Fig. 4c and Extended Data Fig. 6). These
grown in S/L or 2i/L medium with 1 mM or 5 mM of GSK-J4 or GSK-J5 for 3 h. results support the conclusion that aKG promotes the self-renewal of
Ctrl, control. **P , 0.001, ***P , 0.0001, as determined by two-way ES cells in vitro.
analysis of variance (ANOVA) with Sidak’s multiple comparisons These data demonstrate that the cellular aKG/succinate ratio con-
post-test (a, g). Data are presented as the mean 6 s.d. (a) or s.e.m. (g) of tributes to the ability of ES cells to suppress differentiation. The rewir-
triplicate wells from a representative experiment. ing of cellular metabolism by inhibitors of GSK-3b and MAPK/ERK
signalling results in a reprogramming of glucose and glutamine meta-
aKG. Treatment with GSK-J4 (ref. 16), a cell-permeable inhibitor that bolism; in turn, this leads to accumulation of aKG and favours demethy-
preferentially inhibits UTX and JMJD3, the two H3K27me3-specific lation of repressive chromatin marks such as DNA methylation and
JmjC-family histone demethylases (Fig. 3c), induced a dose-dependent H3K9me3, H3K27me3 and H4K20me3 (see Supplementary Discussion).
increase in H3K27me3 with a concomitant reduction of H3K27me1 Future studies will investigate the mechanisms through which these
that was comparable in magnitude to the difference observed when inhibitors influence the nuclear/cytosolic accumulation of aKG derived
cells were cultured in the presence or absence of glutamine (Fig. 3b, d). from glucose and glutamine. While we cannot rule out chromatin-
These data indicate that the methylation of certain histone lysines, independent effects of aKG supplementation on ES cells, our results
including H3K27, are actively suppressed by aKG-dependent histone support the notion that chromatin in pluripotent ES cells is responsive
demethylases in ES cells maintained in 2i/L medium. to alterations in intracellular metabolism. Indeed, recent clonal analysis
1 9 F E B R U A RY 2 0 1 5 | VO L 5 1 8 | N AT U R E | 4 1 5
©2015 Macmillan Publishers Limited. All rights reserved
RESEARCH LETTER

a 4. Nichols, J., Silva, J., Roode, M. & Smith, A. Suppression of Erk signalling promotes
ground state pluripotency in the mouse embryo. Development 136, 3215–3222
(2009).
5. Smith, Z. D. et al. A unique regulatory phase of DNA methylation in the early
mammalian embryo. Nature 484, 339–344 (2012).
6. Wray, J., Kalkan, T. & Smith, A. G. The ground state of pluripotency. Biochem. Soc.
Trans. 38, 1027–1032 (2010).
7. Leitch, H. G. et al. Naive pluripotency is associated with global DNA
hypomethylation. Nature Struct. Mol. Biol. 20, 311–316 (2013).
8. Ficz, G. et al. FGF signaling inhibition in ESCs drives rapid genome-wide
demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13,
S/L+DMSO S/L+DM-αKG S/L+DM-succinate 351–359 (2013).
9. Habibi, E. et al. Whole-genome bisulfite sequencing of two distinct interconvertible
b S/L
c Nanog–GFP
DNA methylomes of mouse embryonic stem cells. Cell Stem Cell 13, 360–369
70
Undifferentiated
(2013).
5,000
60 Mixed 10. Borgel, J. et al. Targets and dynamics of promoter DNA methylation during early
Number of colonies

50 Differentiated 4,000 mouse development. Nature Genet. 42, 1093–1100 (2010).


40 11. Marks, H. et al. The transcriptional and epigenomic foundations of ground state
3,000
M.F.I.

30 pluripotency. Cell 149, 590–604 (2012).


*** 12. Ying, Q. L., Nichols, J., Chambers, I. & Smith, A. BMP induction of Id proteins
2,000
20
suppresses differentiation and sustains embryonic stem cell self-renewal in
10 1,000
collaboration with STAT3. Cell 115, 281–292 (2003).
0
0 13. Zhang, J., Nuebel, E., Daley, G. Q., Koehler, C. M. & Teitell, M. A. Metabolic regulation
e

te
KG
cl

S/L S/L in pluripotent stem cells during reprogramming and self-renewal. Cell Stem Cell
na
hi

ci
Ve

+DM-αKG
M

uc

11, 589–595 (2012).


D

-s
M

14. Kaelin, W. G. Jr. Cancer and altered metabolism: potential importance of hypoxia-
D

inducible factor and 2-oxoglutarate-dependent dioxygenases. Cold Spring Harb.


Figure 4 | aKG promotes the maintenance of pluripotency. a, b, Colony Symp. Quant. Biol. 76, 335–345 (2011).
formation assay using ESC-1 cells. Cells were plated at clonal density and media 15. Cloos, P. A., Christensen, J., Agger, K. & Helin, K. Erasing the methyl mark: histone
were changed to experimental media containing either DM-aKG or DM- demethylases at the center of cellular differentiation and disease. Genes Dev. 22,
succinate on day 2 and then analysed 4 days later by AP staining and scored 1115–1140 (2008).
for number of differentiated, mixed and undifferentiated colonies. 16. Kruidenier, L. et al. A selective jumonji H3K27 demethylase inhibitor modulates
the proinflammatory macrophage response. Nature 488, 404–408 (2012).
a, Representative brightfield images of AP-stained colonies. Vehicle, DMSO, 17. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental
dimethylsulphoxide. b, Quantification of colonies. DM-aKG has more genes in embryonic stem cells. Cell 125, 315–326 (2006).
undifferentiated colonies than vehicle- or DM-succinate-treated wells. 18. Boyer, L. A. et al. Polycomb complexes repress developmental regulators in murine
***P , 0.0001, calculated by two-way ANOVA with Tukey’s multiple embryonic stem cells. Nature 441, 349–353 (2006).
comparisons post-test. c, Mean GFP intensity of Nanog–GFP cells treated for 19. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and
lineage-committed cells. Nature 448, 553–560 (2007).
3 days with or without DM-aKG. M.F.I., mean fluorescence intensity. Data are
20. Blaschke, K. et al. Vitamin C induces Tet-dependent DNA demethylation and a
presented as the mean 6 s.e.m. (b) or 95% confidence intervals (c) of triplicate blastocyst-like state in ES cells. Nature 500, 222–226 (2013).
wells from a representative experiment. 21. Hackett, J. A. et al. Synergistic mechanisms of DNA demethylation during
transition to ground-state pluripotency. Stem Cell Reports 1, 518–531 (2013).
22. Faddah, D. A. et al. Single-cell analysis reveals that expression of Nanog is biallelic
of pluripotent cells revealed that DNA methylation is highly dynamic, and equally variable as that of other pluripotency factors in mouse ESCs. Cell Stem
balancing the antagonistic processes of removal and addition23. Together, Cell 13, 23–29 (2013).
these results suggest that continued elucidation of the interconnections 23. Shipony, Z. et al. Dynamic and static maintenance of epigenetic memory in
pluripotent and somatic cells. Nature 513, 115–119 (2014).
between signal transduction and cellular metabolism will shed import-
ant light on stem cell biology, organismal development and cellular Supplementary Information is available in the online version of the paper.
differentiation. Acknowledgements B.W.C. is a Howard Hughes Medical Institute fellow of the Jane
Coffin Childs Memorial Research Fund. L.W.S.F. is the Jack Sorrell Fellow of the Damon
Online Content Methods, along with any additional Extended Data display items Runyon Cancer Research Foundation (DRG-2144-13). This work was funded by grants
and Source Data, are available in the online version of the paper; references unique from the National Institutes of Health/National Institute of General Medical Sciences
to these sections appear only in the online paper. (C.D.A.) and from the National Cancer Institute (C.B.T.). We thank C. Li for assistance
with FACS analysis and M. Dawlaty, D. Faddah and R. Jaenisch for sharing cell lines
Received 29 April; accepted 20 October 2014. used in this study.
Published online 10 December 2014. Author Contributions B.W.C. and L.W.S.F. designed and performed all experiments in
the study under the guidance of C.D.A. and C.B.T. J.R.C. contributed material support.
1. Lunt, S. Y. & Vander Heiden, M. G. Aerobic glycolysis: meeting the metabolic B.W.C., L.W.S.F., C.D.A. and C.B.T. wrote the manuscript.
requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 27, 441–464 (2011).
2. Eagle, H., Oyama, V. I., Levy, M., Horton, C. L. & Fleischman, R. The growth response Author Information Reprints and permissions information is available at
of mammalian cells in tissue culture to L-glutamine and L-glutamic acid. J. Biol. www.nature.com/reprints. The authors declare competing financial interests: details
Chem. 218, 607–616 (1956). are available in the online version of the paper. Readers are welcome to comment on
3. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, the online version of the paper. Correspondence and requests for materials should be
519–523 (2008). addressed to C.B.T. (thompsonc@mskcc.org) or C.D.A. (alliscd@mail.rockefeller.edu).

4 1 6 | N AT U R E | VO L 5 1 8 | 1 9 F E B R U A RY 2 0 1 5
©2015 Macmillan Publishers Limited. All rights reserved
LETTER RESEARCH

METHODS 37 uC for 30 min. Samples were analysed using an Agilent 7890A GC coupled to
Cell lines. ESC-1–4 lines are V6.5 ES cells derived from C57BL/6 3 129S4/SvJae Agilent 5975C mass selective detector. The GC was operated in splitless mode with
F1 embryos in 2i/L medium. Cells were derived from embryonic day (E)3.5 blas- constant helium gas flow at 1 ml min21. One microlitre of derivatized metabolites
tocysts following standard ES-cell isolation procedures24. Flushed blastocysts were was injected onto an HP-5MS column and the GC oven temperature ramped from
plated onto laminin-coated dishes (20 mg ml21, Stemgent 06-0002) in 2i/L med- 60 uC to 290 uC over 25 min. Peaks representing compounds of interest were
ium. Mice were purchased from Jackson Laboratories (C57BL/6 JAX 000664 and extracted and integrated using MassHunter software (Agilent Technologies) and
129S4/SvJae JAX 009104). Tet1/2 double-knockout ES cells25, V19 ES cells (ESC- then normalized to both the internal standard (d5-2HG) peak area and the protein
V19) and OKS iPSCs26 were a gift from R. Jaenisch. All cells were routinely tested content of duplicate samples as determined by a BCA protein assay (Thermo
for mycoplasma contamination. Mice were maintained at The Rockefeller University. Scientific). Ions used for quantification of metabolite levels are as follows: d5-
All animal procedures were designed following National Institutes of Health 2HG m/z 354; aKG, m/z 304; aspartate, m/z 334; glutamate, m/z 363; malate,
guidelines and approved by the Institutional Animal Care and Use Committee m/z 335; and succinate, m/z 247. All peaks were manually inspected and verified
at The Rockefeller University. relative to known spectra for each metabolite. For isotope tracing studies, experi-
Cell culture. Maintenance media for ES cells were as follows: serum/LIF (S/L) ments were set up as described earlier using glucose- and glutamine-free DMEM:
maintenance medium contained Knockout DMEM (Gibco) supplemented with NB media base supplemented with 12C-glucose (Sigma) and 12C-glutamine (Gibco)
15% ES-cell-qualified FBS (Gemini), penicillin/streptomycin (Life Technologies), or the 13C versions of each metabolite, [U-13C]glucose or [U-13C]glutamine (Cambridge
0.1 mM 2-mercaptoethanol, 2 mM L-glutamine (Life Technologies) and LIF plated Isotope Laboratories). Enrichment of 13C was assessed by quantifying the abund-
onto irradiated feeder mouse embryonic fibroblasts (MEFs); 2i/LIF (2i/L) main- ance of the following ions: aKG, m/z 304–315; aspartate, m/z 334–346; glutamate,
tenance conditions used a base medium made from a 1:1 mix of DMEM/F12 (Life m/z 363–377; and malate, m/z 335–347. Correction for natural isotope abundance
Technologies 11302-033) and Neurobasal medium (Life Technologies 21103-049) was performed using IsoCor software27. Flux was calculated as the product of the
containing N2 and B27 supplements (Life Technologies 17502-048 and 17504- first order rate constant of the kinetic labelling curve and relative metabolite pool
044, 1:100 dilutions), penicillin/streptromycin, 0.1 mM 2-mercaptoethanol, 2 mM size (normalized to mean S/L values for each experiment)28. The flux from glucose-
L-glutamine, LIF, CHIR99021 at 3 mM (Stemgent) and PD0325901 at 1 mM (Stem- and glutamine-derived carbons was calculated for each of three independent
gent). Experimental media used for all experiments (except growth curves without experiments and the average flux for each metabolite was shown. Flux experiments
glucose, 13C isotope tracing experiments and 14C labelling experiments) contained represent the average of three independent experiments; all other experiments were
a 1:1 mix of glutamine-free DMEM (Life Technologies 11960-051) and Neurobasal performed independently at least twice and a representative experiment is shown.
medium (Life Technologies 21103-049) with or without 2 mM glutamine. With Protein labelling. ES cells were plated at 7.5 3 105 cells per 6-well plate into ex-
the exception of 15% dialysed FBS (Gemini 100-108) in S/L experimental medium, perimental medium (S/L or 2i/L) containing 0.01% unenriched D-[U-14C]-glucose
all other supplements were equivalent to maintenance media (S/L or 2i/L). For (Perkin Elmer NEC042V250UC) or L-[U-14C]-glutamine (Perkin Elmer NEC45
growth curves without glucose, 13C isotope tracing experiments and 14C labelling 1050UC). Forty-eight hours later, cells were washed with PBS, scraped and pel-
experiments, media contained a 1:1 mix of glutamine- and glucose-free DMEM leted at 4 uC. Protein pellets devoid of lipid fractions were isolated according to the
(Invitrogen A14430-01) and glutamine- and glucose-free Neurobasal medium Bligh-Dyer method29. Briefly, pellets were resuspended in 200 ml distilled H2O,
(Invitrogen 0050128DJ) containing either 20 mM [U-13C]glucose or 2 mM [U-13C] 265 ml 100% methanol and 730 ml of chloroform. Samples were vortexed for 1 h at
glutamine (Cambridge Isotope Laboratories) and either 20 mM unlabelled glucose 4 uC. The organic phase was removed and the remaining sample washed with 13
or 2 mM unlabelled glutamine as necessary; all supplements were the same as in ex- volume of methanol and spun at 14,200g for 5 min. The supernatant was discarded
perimental media described above (S/L or 2i/L). All experiments were performed and the pellet was resuspended in 6 M guanidine hydrochloride at 65 uC for 30–
using feeder-free conditions. ESC-1 EpiSCs were cultured feeder-free on fibronec- 45 min. Samples were quantified using Beckman LS 60001C instrument. Values
tin (Sigma)-coated plates in EpiSC maintenance medium including DMEM/F12, represent the average from four wells normalized to protein of duplicate samples.
N2 and B27 supplements, penicillin/streptromycin, 0.1 mM 2-mercaptoethanol, Labelling experiments were performed twice.
21
L-glutamine, 75 mg ml BSA (Gibco) supplemented with human activin A (20 ng ml ;
21 Growth curves. ES cells or EpiSCs were plated in maintenance medium at a
Peprotech) and bFgf (10 ng ml21; Invitrogen). EpiSCs were passaged 1:2 or 1:4 concentration of 375,000 cells per 12-well plate. The following day cells were
using Accutase every other day. For ES-cell to EpiSC differentiation, ESC-1 cells washed with PBS and media were changed to experimental media (for S/L con-
were plated onto fibronectin-coated dishes. Twenty-four hours after plating, the ditions this included dialysed FBS) with or without individual metabolites. Cells
medium was changed to EpiSC maintenance medium supplemented with 6 mM were counted each day using a Beckman Coulter Multisizer 4. All growth curves
JAK inhibitor (Calbiochem) for five passages. Analysis was performed on passage were performed independently at least two times.
7 EpiSCs. GSK-J4 and GSK-J5 were purchased from Tocris Bioscience. ChIP. Native ChIP assays (histones) were performed with approximately 6 3 106
Teratomas. ESC-1 cells were plated in maintenance medium at a concentration of ES cells per experiment. Cells were subject to hypotonic lysis and treated with
2.5 3 105 cells per T25 dish. The following day medium was changed to 2i/L micrococcal nuclease to recover mono- to tri-nucleosomes. Nuclei were lysed by
experimental medium with or without glutamine. 72 h later, 1 3 106 cells were brief sonication and dialysed into N-ChIP buffer (10 mM Tris pH 7.6, 1 mM
harvested from each group and mixed 1:1 with experimental medium (without EDTA, 0.1% SDS, 0.1% Na-Deoxycholate, 1% Triton X-100) for 2 h at 4 uC.
glutamine) plus Matrigel Basement Membrane Matrix (BD) or experimental med- Soluble material was incubated overnight at 4 uC after addition of 0.5–1 mg of
ium alone and injected into the flanks of recipient SCID mice aged 8–12 weeks antibody bound to 25 ml protein A Dynal magnetic beads (Invitrogen), with 5%
(NOD scid gamma JAX 005557 purchased from Jackson Laboratories). All con- kept as input DNA. Magnetic beads were washed, chromatin was eluted and ChIP
ditions produced tumours in 4–8 weeks. Mice were euthanized before tumour size DNA was dissolved in 10 mM Tris pH 8 for quantitative PCR reactions (see later).
exceeded 1.5 cm in diameter. Tumours were excised and fixed in 4% paraformalde- Three separate ChIP experiments were performed on replicate biological samples.
hyde overnight at 4 uC. Tumours were paraffin-embedded and sections were stained The data shown are the average qRT–PCR values (n 5 3).
with haematoxylin and eosin according to standard procedures by Histoserv. ChIP-qPCR. Primers are listed below. All qPCR was performed using an Applied
Glucose, glutamine and lactate measurements. Glucose, glutamine and lactate Biosystems StepOnePlus system and Power SYBR Green PCR master mix. ChIP
levels in culture medium were measured using a YSI 7100 multichannel biochem- samples were diluted 1:100 in H2O and 5 ml was used per reaction. ChIP-qPCR signals
istry analyser (YSI Life Sciences). Fresh medium was added to 12-well plates of were calculated as per cent input. Primers were as follows: Gata6, forward, 59-
sub-confluent cells and harvested 48 h later. Changes in metabolite concentrations CGCAGCACACAGGTACAGTT-39, reverse, 59-GGGATCCAAGCAGATTGA
relative to fresh media were normalized to the protein content of each well. These AA-39; Pax9, forward, 59-AGGTGTGCGACAGCTAAAGG-39, reverse, 59-ATC
experiments were performed independently at least two times. AACCCGGAGTGATCAAG-39; Lhx1, forward, 59-TGCCAGGCACCATTACA
Metabolite profiling. For all metabolite experiments, cells were seeded in their GT-39, reverse, 59-AGGCAAAGGAAAAACCATGA-39; Hoxa2, forward, 59- CC
standard culture medium in 6-well plates and the next day were changed into AATGACAATTTGGGCTTT-39;reverse, 59-TGAGGCGTTCCTTTCTGACT-39;
experimental medium. Medium was changed again at the indicated time before Hoxc9, forward, 59-TTCTTCCCTTTGGCCTTTTT-39; reverse, 59-AGGGTGTC
harvest (usually 1–24 h). Metabolites were extracted with 1 ml ice-cold 80% methanol TTGGCTCTCTCA-39; Evx1, forward, 59-GCCAGGTGATCTGGGTGGGGA-39,
supplemented with 20 mM deuterated 2-hydroxyglutarate (D-2-hydroxyglutaric- reverse, 59-TGAGAACCGGCCTTGTGTGCT-39; Fgf5, forward, 59-GGGATCTC
2,3,3,4,4-d5 acid (d5-2HG)) as an internal standard. After overnight incubation at CTGTGCCTGGGGT-39; reverse, 59-AGGCCTGTACTGCAGCCACATTT-39;
280 uC, lysates were harvested and centrifuged at 21,000g for 20 min to remove Ascl2, forward, 59-GCTCCAGAAGCAGTTCTCCCCTGA-39, reverse, 59-GATA
protein. Extracts were dried in an evaporator (Genevac EZ-2 Elite) and resuspended GAGCCAGAGCCCAAGCCCC-39; Lrat, forward, 59-CCAAGTCCTTCAGTCT
by incubation at 30 uC for 2 h in 50 ml of 40 mg ml21 methoxyamine hydrochloride CTTGCCCC-39, reverse, 59-GGCCACACAGGCTGCTTCCA-39; Lhx5, forward,
in pyridine. Metabolites were further derivatized by addition of 80 ml of MSTFA 59-AACCCTTAGGCCCCAGCCCC-39; reverse, 59-CGTGGGCCTGGAGGGG
plus 1% TMCS (Thermo Scientific) and 70 ml ethyl acetate (Sigma) and incubated at AGAA-39; Sox17, forward, 59-GTCTCCCCATGTAGCTCTCCTGCC-39, reverse,

©2015 Macmillan Publishers Limited. All rights reserved


RESEARCH LETTER

59-AGAAGAGTCACTGTGGAGGTGAGGG-39; brachyury, forward, 59-GCCA GCG-39, reverse, 59-CGGACCCCAAGAACCATCAC-39; Jmjd3 gRNA #2, for-
CTGCTTTCCCGAGACCC-39; reverse, 59-CCAGGACAGGCAGGGTAGGGG-39; ward, 59-TGGCCTGCAGAGGGAGATAG-39, reverse, 59-ATTTCGTCGGCAT
Gata4, forward, 59-ACGTGTGGTGTTAATGTGCAAGCC-39, reverse, 59-TGCC TCCTGTG-39.
CACAAGCCTGCGATCC-39; Sox21, forward, 59-AACAGACATGCCAGTCAG FACS. Nanog–GFP ES cells22 were cultured in S/L experimental medium for three
CAGTG-39, reverse, 59-TTAGCATCGCACCACCCAGAGTC-39; Pou5f1, for- passages and 2.5 3 104 cells were plated into a 6-well plate. Twenty-four hours
ward, 59-GAGGTCAAGGCTAGAGGGTGG-39, reverse 59-AGGGACGGTTTC later medium was changed to S/L medium containing vehicle control or DM-aKG.
ACCTCTCC-39. Media were subsequently changed 48 h later and cells were harvested the following
qRT–PCR. RNA was isolated using the RNeasy kit (Qiagen). After DNase treat- day. FACS analysis was performed at The Rockefeller University Flow Cytometry
ment, 1–2 mg RNA was used for cDNA synthesis using the First-Strand Synthesis Resource Center using a BD LSR II. Data were generated using FlowJo. Experiments
kit (Invitrogen). Quantitative RT–PCR analysis was performed in biological trip- were performed two independent times and a representative experiment depicting
licate using an ABI Prism 7000 (Applied Biosystems) with Platinum SYBR green. triplicate biological wells is shown.
All data were generated using cDNA from three wells for each condition. Primers Western blot analysis. Lysates were extracted in 13 Laemmli buffer, separated by
used were as follows: Pou5f1, forward, 59-ACATCGCCAATCAGCTTGG-39, reverse, SDS–PAGE and transferred to Immobilon PVDF (Millipore) membranes. Mem-
59-AGAACCATACTCGAACCACATCC-39; Nanog, forward, 59-AAGATGCGG branes were blocked in 5% milk prepared in phosphate-buffered saline (PBS) plus
ACTGTGTTCTC-39, reverse, 59-CGCTTGCACTTCATCCTTTG-39; Esrrb, for- 0.1% Tween 20 (PBS-T), incubated with primary antibodies overnight at 4 uC and
ward, 59-TTTCTGGAACCCATGGAGAG-39, reverse, 59-AGCCAGCACCTCC with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h the
TTCTACA-39; Klf2, forward, 59-TAAAGGCGCATCTGCGTACA-39, reverse, 59- next day. After ECL application (Millipore), imaging was performed using Lumimager
CGCACAAGTGGCACTGAAAG-39; Nr0b1, forward, 59-TCCAGGCCATCAA LAS-3000 (FujiFilm). The following antibodies were used for western blotting: H3
GAGTTTC-39, reverse, 59-ATCTGCTGGGTTCTCCACTG-39; Fgf5, forward, 59- (Abcam 1791), H3K4me3 (Active Motif 39159), H3K4me1 (Millipore 07-436),
AAACTCCATGCAAGTGCCAAAT-39, reverse, 59-TCTCGGCCTGTCTTTTC H3K9me1 (gift from T. Jenuwein), H3K9me3 (Active Motif 39161), H4 (Abcam
AGTTC-39; Zfp42, forward, 59-CGAGTGGCAGTTTCTTCTTGG-39, reverse, 59- 0158), H4K20me1 (Abcam 9051), H4K20me3 (Millipore 07-463), H3K27me1
CTTCTTGAACAATGCCTATGACTCACTTCC-39; actin, forward, 59-TGGCG (Millipore 07-448), H3K27me3 (Millipore 07-449), H3K36me3 (Abcam 9050)
CTTTTGACTCAGGAT-39, reverse, 59-GGGATGTTTGCTCCAACCAA-39; Asz1, and H3K36me1 (Millipore 07-548). All antibodies were used at a dilution of
forward, 59-GAGTGGGCTTCTCCCAGAAA-39, reverse, 59-GGTCATTTTCCC 1:1,000. H3K27me3 antibody used for ChIP-qPCR, Cell Signaling 9733BF.
GCTCATTC-39; Wdfc15a, forward, 59-TGTGTGGAACCCTGGACAAC-39, reverse, Self-renewal assays. ES cells free from feeder MEFs were plated at 100 cells per
59-GCCAATGCCGTCGTTATTTT-39; Daz1, forward, 59-CAACTGTTAACTAC well in 6-well plates coated with 20 mg ml21 mouse laminin (Stemgent 06-0002) in
CACTGCAG-39, reverse, 59-CAAGAGACCACTGTCTGTATGC-39; Gapdh, for- maintenance S/L medium. The next day, media were changed to S/L experimental
ward, 59-TTCACCACCATGGAGAAGGC-39, reverse, 59-CCCTTTTGGCTCCA medium containing dimethyl-a-ketoglutarate (4 mM, Sigma 349631), dimethyl-
CCCT-39. succinate (4 mM, Sigma W239607) or DMSO vehicle control. Four days later, cells
DNA methylation. Genomic DNA was extracted from ES-cell samples using were washed with PBS and stained for alkaline phosphatase using Vector Red
Puregene Core Kit A (Sigma). DNA methylation was measured using the colori- Alkaline Phosphatase Kit (Vector Labs) according to manufacturer’s instructions.
metric MethylFlash Methylated DNA quantification kit (Epigentek) according to Self-renewal assays were performed independently at least two times.
manufacturer instructions. ELISA experiments were performed independently Statistics. Comparisons were made using unpaired two-tailed Student’s t-tests or
two times. two-way ANOVA with appropriate post-test (determined using GraphPad Prism)
CRISPR/Cas9 ES cells. A Cas9-2A-PURO plasmid was purchased from Addgene as indicated. Experiments were performed with three or four replicates as is the
(Addgene plasmid 48139)30. Two gRNAs targeting exon 17 of mouse Jmjd3 were standard in the field. Variation is shown as s.d., s.e.m. or 95% confidence intervals
designed using the online software (http://crispr.mit.edu) resource from the Zhang as indicated in figure legends.
Laboratory and were cloned into Cas9-2A-Puro using the BbsI restriction enzyme
sites. ESC-1 cells cultured in 2i/L medium were transfected with either Cas9-2A- 24. Markoulaki, S., Meissner, A. & Jaenisch, R. Somatic cell nuclear transfer and
Puro control or Jmjd3 gRNA-containing plasmids using Lipofectamine 2000 (Life derivation of embryonic stem cells in the mouse. Methods 45, 101–114 (2008).
Technologies). After 24 h, cells were changed to fresh medium containing 1 mg ml21 25. Dawlaty, M. M. et al. Combined deficiency of Tet1 and Tet2 causes epigenetic
puromycin for 48 h. After selection, cells were cultured for 24 h in 2i/L medium and abnormalities but is compatible with postnatal development. Dev. Cell 24,
310–323 (2013).
then split to clonal density. After approximately 7 days, colonies were picked and 26. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming
expanded for analysis. Genomic DNA was purified from individual clones and reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222
used for PCR amplification of regions surrounding each gRNA target site. gRNA (2012).
#1 product is 367 bp and gRNA #2 is 317 bp. Cloning of PCR products was per- 27. Millard, P., Letisse, F., Sokol, S. & Portais, J. C. IsoCor: correcting MS data in
formed using pGEM-T Easy (Promega). Mutants were identified by Sanger sequen- isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).
cing (Genewiz). gRNA oligonucleotides were as follows: Jmjd3 gRNA #1, forward, 28. Yuan, J., Bennett, B. D. & Rabinowitz, J. D. Kinetic flux profiling for quantitation of
cellular metabolic fluxes. Nature Protocols 3, 1328–1340 (2008).
59-CACCTGTGGATGTTACCCGCATGA-39, reverse, 59-AAACTCATGCGGG 29. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification.
TAACATCCACA-39; Jmjd3 gRNA #2, forward, 59-CACCGTCCCTGGCAGCC Can. J. Biochem. Physiol. 37, 911–917 (1959).
GAACGCC-39, reverse, 59-AAACGGCGTTCGGCTGCCAGGGAC-39. PCR pri- 30. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nature
mers were as follows: Jmjd3 gRNA #1, forward, 59-GGCTAAGGCCTAAGAGT Protocols 8, 2281–2308 (2013).

©2015 Macmillan Publishers Limited. All rights reserved


LETTER RESEARCH

a ESC-V19 b 0.20
ESC-1 c x108
serum/LIF x108
2i/LIF
15
S/L -glc +gln +gln
1.4 - gln - gln

Cell number (x 106)


1.4
S/L /2i -glc glutamine glutamine
Doubling time (h)

total ion count

total ion count


0.15 1.2 1.2
10 1.0 1.0
0.10 0.8 0.8
0.6 0.6
5
0.05 0.4 0.4
0.2 0.2
0 0.00 0.0 0.0
S/L 2i/L 0 1 2 3 4 16.12 16.16 16.2 16.24 16.28 16.12 16.16 16.2 16.24 16.28
Day retention time (m) retention time (m)

d Ectoderm Endoderm Mesoderm

e ESC-1 f 1000 g EpiSC h OSK iPSC


0.3 0.8 0.3
Relative expression

2i/L -Q 100 Fgf/ActA +Q S/L-Q


Cell number (x 106)

Cell number (x 106)


Cell number (x 106)

2i -Q Fgf/ActA -Q 2i/L-Q
10 0.6
0.2 0.2
1
0.4
0.1 0.100 0.1
0.2
0.010

0.0 0.001 0.0 0.0


0 1 2 3 4 0 1 2 3 0 1 2 3 4
N 3/4

f5
Es 2
rrb
N f2
b1
Zf g
p4
o

Day Day Day


Fg
Kl
r0
an
ct
O

i ESC-1 j ESC-V19: 2i/L k ESC-V19: S/L ESC-V19: 2i/L


30 0.3 2i/L-Q 0.8 0.8 2i/L -Q
S/L -Q
Cell number (x 106)

S/L -Q+DMαKG 2i/L -Q+DMαKG


Cell number (x 106)

2i/L-Q+MSO
Doubling time (h)

0.6 0.6
20 0.2
0.4 0.4
10 0.1
0.2 0.2

0 0.0 0.0 0.0


+gln -gln 0 1 2 3 0 1 2 3 0 1 2 3
Day Day Day

Extended Data Figure 1 | Pluripotent stem cells can proliferate in the transcription (qRT–PCR), normalized to Gapdh and expressed as a ratio of
absence of glutamine when cultured in 2i/LIF medium. a, Doubling time of values of mouse ES cells cultured in 2i/L medium. g, Growth curve of EpiSCs
ESC-V19 cells cultured in S/L or 2i/L. b, Growth curve of ESC-1 cells cultured in cultured in serum-free epiblast medium (serum-free medium containing FGF
S/L or S/L/2i medium devoid of glucose. c, Samples of S/L (left) and 2i/L (right) and activin A (Fgf/ActA)) with or without glutamine. h, Growth curve of an
media with and without glutamine were analysed by GC–MS. Representative iPSC line derived from fibroblasts using Oct3/4 (O), Klf4 (K) and Sox2 (S)
chromatograms of the total ion count reveal a clear glutamine (Q) peak in 1Q cultured in glutamine-free S/L or 2i/L medium. i, Doubling time of ESC-1 cells
media (grey) and no detectable glutamine in 2Q media (red). m, minutes. cultured in 2i/L medium in the presence and absence of glutamine. j, Growth
d, Teratoma formation from ES cells grown in 2i/L medium without glutamine curve of ESC-V19 cells cultured in glutamine-free 2i/L medium in the presence
for 3 days. Representative images of haematoxylin and eosin staining reveal or absence of 1 mM methylsulphoxide (MSO). k, ESC-V19 cells grown in
neural tissue (ectoderm), hepatocytes and pancreatic acinar cells (endoderm) glutamine-free S/L (left) or 2i/L (right) medium with or without 4 mM DM-
and smooth muscle (mesoderm). Scale bar, 200 mm. e, Growth curve of ESC-1 aKG. For growth curve experiments, cells were seeded on day 0 in complete
cells grown in glutamine-free 2i/L or 2i medium. f, Gene expression analysis medium and then were changed to experimental medium on day 1 (indicated
confirms that EpiSCs, which represent post-implantation pluripotency, were by red arrow). Data are presented as the mean 6 s.d. of triplicate wells from a
generated from ESC-1 cells by culture with Fgf and activin A. Transcript levels representative experiment.
were assessed by quantitative real-time polymerase chain reaction with reverse

©2015 Macmillan Publishers Limited. All rights reserved


RESEARCH LETTER

a glutamate b glucose-derived c 14
C-glc 14
C-gln
glutamate

incorporated into protein


incorporated into protein
(normalized to mg protein, a.u.)

(normalized to mg protein, a.u.)


Fraction labeled by 13C-Glc

0.20 0.15 4 * 15

Relative abundance
0.15 3
0.10 10
0.10 2

0.05 5
0.05 1

14C-Gln
14C-Glc
0.00 0.00 0 0
S/L 2i/L S/L 2i/L S/L 2i/L
L

2i

/L
S/

L/

2i

-gln
S/

Extended Data Figure 2 | Mouse ES cells cultured with 2i demonstrate glucose-derived carbons is shown. c, Incorporation of 14C derived from
altered glucose and glutamine utilization. a, 2i enables glutamate synthesis [U-14C]glucose (14C-glc) (left) or derived from [U-14C]glutamine (14C-gln)
from glucose-derived carbons. ESC-1 cells cultured in S/L, S/L/2i or 2i/L (right) into total cellular protein after 48 h incubation. P , 0.05 for 14C-glc,
medium were incubated with medium containing [U-13C]glucose for 4 h P 5 0.1 for 14C-gln, calculated by unpaired two-tailed Student’s t-test. Data are
and the fraction of glutamate containing glucose-derived carbons is shown. presented as the mean 6 s.d. of triplicate wells (a, b) or 6 s.e.m. of
b, ESC-1 cells were cultured for 4 h in glutamine-free S/L or 2i/L medium quadruplicate wells (c) from a representative experiment.
containing [U-13C]glucose and the total amount of glutamate labelled by

©2015 Macmillan Publishers Limited. All rights reserved


LETTER RESEARCH

a 2i/LIF b serum/LIF
glutamine + - - 40 ctrl
DMαKG - - + GSK-J4
30
H3K27me3

% input
H3K27me1 20

10
H4K20me3
H4K20me1 0

1
T

6
f5

H 4
at

x1
H l2

x9
c9

So 2
G 7
So 5

x1
x2

A
x1
x
c

a
Fg

a
Lr

Ev
Pa
ox

Lh

Lh

ox
As

at

at
G
H3K4me3
H3K4me1 c 2i/LIF
40 ctrl
H3 GSK-J4
30
H4
% input
20 * *
* *
10 * * * * *

1
T
f5

H 4

6
at

x1
H l2

x9
c9

So 2
G 7
So 5

x1
x2

A
x1
x
c

Fg

a
Lr

Ev
Pa
ox

Lh

Lh

ox
As

at

at
G
Extended Data Figure 3 | Regulation of histone methylation in 2i/LIF cells. ESC-1 cells cultured in S/L (b) or 2i/L (c) medium with or without 30 mM GSK-
a, Western blot analysis of ESC-1 cells grown in glutamine-free 2i/L medium J4 for 5 h. Data are presented as the mean 6 s.e.m. of triplicate samples.
for 24 h with supplementation as indicated. b, c, H3K27me3 ChIP-qPCR of *P , 0.05 by unpaired Student’s two-tailed t-test.

©2015 Macmillan Publishers Limited. All rights reserved


RESEARCH LETTER

a Jmjd3 exon 17
PAM
5’ - TGCC TGTGGATGTTACCCGCATGA AGGCGGG - 3’

3’ - ACGGACACCTACAATGGGCGTACA TCCGCCC - 5’

Jmjd3 gRNA #1 5’ - TGTGGATGTTACCCGCATGA - 3’


PAM
5’ - GAAGGTCCCTGGCAGCCGAACGCCAGG TGTG - 3’

3’ - CTTC CAGGGACCGTCGGCTTGCGGTCCACAC - 5’

Jmjd3 gRNA #2 5’ - GTCCCTGGCAGCCGAACGCC - 3’

b Indels in Jmjd3
In-frame stop
WT 5’ TGTGGATGTTACCCGCATGAAGG 3’
JMJΔ/Δ−1 Allele 1 5’ TGTGGATGTTACCCG - - TGAAGG 3’ -2 a.a #5 in exon 18
JMJΔ/Δ−1 Allele 2 5’ TGTGGATGTTACCC - - - - GAAGG 3’ -4 a.a. #16 in exon 17

WT 5’ GTCCCTGGCAGCCGAAC - GCCAGG 3’ In-frame stop


Δ/Δ−
JMJ 2 Allele 1 5’ GTCCCTGGCAGCCGAACAGCCAGG 3’ +1 a.a #5 in exon 18
JMJΔ/Δ−2 Allele 2 5’ GTCCCTGGCAGCCGAACCGCCAGG 3’ +1 a.a #5 in exon 18

c JMJD3Δ/Δ-2 +1bp

A/C
Extended Data Figure 4 | Generation of JMJD3 mutant cells. a, Schematic of protospacer adjacent motif (PAM) sequences are identified in green. Predicted
targeting strategy for guide RNAs (gRNAs) to mouse Jmjd3 exon 17. gRNA cut site is indicated by red triangle. Location of in-frame downstream stop is
sequences are highlighted in blue. b, Representative sequences from two clones indicated on the right. c, An example chromatogram for clone JMJD3D/D-2
used in this study. Sanger sequencing revealed indels as shown in schematic. showing single-base-pair insertions at the predicted Cas9 cleavage site.
Red dashes, deleted bases; red bases, insertions. gRNA is highlighted in blue and

©2015 Macmillan Publishers Limited. All rights reserved


LETTER RESEARCH

a b 0.4

Relative % 5-mC
O2 CO2 0.3
TET1/2

0.2
αKG succinate
0.1

0.0
S/L S/L
+DM-αKG

c vehicle
d
5 5 S/L
DM-αKG
Relative expression

Relative expression
S/L+DM-αKG, passage 4
4 DM-succinate 4

3 3

2 2

1 1

0 0
4

4
z1

z1
5a

5a
l

l
az

az
3/

3/
As

As
c1

c1
D

D
ct

ct
O

O
df

df
W

e
4 Asz1 4 Dazl vehicle
Relative expression

Relative expression

DM-αKG
3 3 DM-succinate

2 2

1 1

0 0
TET1/2 WT TET1/2 KO TET1/2 WT TET1/2 KO

Extended Data Figure 5 | aKG increases Tet activity in mouse ES cells. expression in ESC-1 cells cultured in S/L medium with or without DM-aKG for
a, Simplified schematic of the reaction mechanism of Tet1/2 enzymes. four passages. e, Gene expression in wild-type or Tet1/Tet2 double-knockout
b, Relative per cent 5-methylcytosine (% 5-mC) in ESC-1 cells cultured in S/L (KO) mouse ES cells cultured with DM-aKG or DM-succinate for 72 h. qRT–
medium with or without DM-aKG for 24 h. Each data point represents a PCR data (c–e) was normalized to actin or Gapdh and samples were normalized
sample from triplicate wells of a representative experiment. c, Gene expression to the control group. Oct3/4 is not expected to change and is included as a
in ESC-1 cells cultured with DM-aKG or DM-succinate for 3 days. d, Gene control. Data are presented as the mean 6 s.e.m. of triplicate wells.

©2015 Macmillan Publishers Limited. All rights reserved


RESEARCH LETTER

a S/L + vehicle
100
S/L + αKG

80
% of Max

60

40

20

0
0 10 2 10 3 10 4 10 5
GFP

b
8000

6000
M.F.I.

4000

2000

S/L 4.0 3.0 2.0 1.0 0.5


+DMαKG (mM)
Extended Data Figure 6 | aKG increases Nanog expression.
a, Representative histogram of GFP intensity of Nanog–GFP cells treated
with or without DM-aKG for 3 days. Grey represents background staining.
b, ESC-1 cells were cultured in S/L medium with DM-aKG for four passages
and then switched to medium containing the indicated amounts of DM-aKG
(0.5–4 mM) or vehicle control (S/L) for 3 days. GFP expression (M.F.I.) was
determined by fluorescence-activated cell sorting (FACS). Data are presented
as the mean 6 s.d. of triplicate wells from a representative experiment.

©2015 Macmillan Publishers Limited. All rights reserved

You might also like