supporting information - pnas.org · licate sister wells were set up for each condition in 12-well...

9
Supporting Information Davidson et al. 10.1073/pnas.1118777109 SI Materials and Methods hESC Culture. For experiments where treatments were continued over several weeks, an identical sister plate was harvested as single cells via TrypLE Express (Invitrogen) to determine an accurate cell count for seeding. The experimental plate was harvested with dispase and small cell clusters were seeded at a density of 35,000 cells per centimeter squared. For multiweek experiments, trip- licate sister wells were set up for each condition in 12-well plates: 1 well was harvested with TrypLE Express to determine the cell count for each treatment and used for ow cytometric analysis; a second well was passaged with dispase and seeded at 35,000 cells per centimeter squared on the basis of the single cell count; the third well was harvested with TRIZOL for RNA isolation. To obtain total cell number of GCTM2 + /CD9 + stem cells per well, the percentage of GCTM2 + /CD9 + cells (determined by ow cytometric analysis) was multiplied by the number of cells counted per well of a 12-well plate each week. Flow Cytometry. hESCs were harvested as single cells via TrypLE Express and counted using a Nucleocounter (New Brunswick Scientic). For live ow cytometric analysis, 500,000 cells were immunolabeled with 100 μL primary antibodies, GCTM2 (hy- bridoma supernatant 1:2, a kind gift from Martin Pera, Univer- sity of Southern California, Los Angeles, CA) and CD9 (TG30 clone, 1:100; Millipore), followed by isotype-specic secondary antibodies: goat antimouse IgM-Alexa 647 (1:1,000) and goat antimouse IgG2a-biotin (1:5,000), followed by Alexa 750-R-PE- streptavidin or PE-Cy7-streptavidin (both 1:250) (all secondary antibodies and streptavidin conjugates from Invitrogen). hESCs were resuspended in 140 ng/mL DAPI in culture medium and transferred through a cell strainer before analysis on a BD FACSCanto II. Results were analyzed using FlowJo software. For H1-BAR-VENUS hESCs, the percentage of VENUS- positive cells was determined from the DAPI-negative/DsRED- positive gated population relative to H1-FUBARV-negative control line. Average mean uorescent intensity (MFI) was measured from the VENUS-positive subpopulation. For intracellular ow analysis, 1 × 10 6 hESCs were xed in 4% PFA for 20 min at room temperature, then permeabilized with 0.1% Triton for 2 min on ice. Cells were immunolabeled with 100 μL of Oct4 antibody (C-10 clone, 1:30; Santa Cruz) for 4560 min on ice, followed by goat antimouse IgG2b-Alexa 647 (1:1,000; Invitrogen). Lentiviral Transduction and Selection of hESCs. H1 hESC colonies were isolated as small clusters using dispase in the same manner as for weekly passage, then exposed to ultraconcentrated lenti- virus before seeding onto Matrigel in mouse embryonic broblast (MEF) conditional medium (CM). Transduced hESCs were cultured for a week on Matrigel, then expanded onto several 3.5- cm plates with MEFs. Each lentiviral construct contained DsRED driven by the Ubiquitin promoter as a selectable marker. After a week of expansion on MEFs, DsRED-positive areas of in- dividual colonies were mechanically isolated and transferred onto fresh MEFs. This positive selection method was repeated for another week or two as needed until entirely DsRED-positive lines were isolated, after which time the stable lines were passaged with dispase as per usual. In some cases, a nal negative selection step was performed where DsRED-negative areas of colonies were mechanically cut and discarded, then the remainder of the colonies in the plate were passaged with dispase (Fig. S7). Fluorescent-Activated Cell Sorting. H1-BAR-VENUS hESCs were harvested as single cells with TrypLE Express. The DAPI-neg- ative/DsRED-positive gated population was sorted on the basis of VENUS expression. A minimum of 1 × 10 6 sorted cells was collected for downstream RNA isolation. For each experiment, samples were normalized to the same cell number on the basis of postsort cell counts before freezing in TRIZOL. siRNA Reverse Transfection of hESCs. hESCs were harvested into a single cell suspension with TrypLE Express, then counted on a NucleoCounter. Reverse transfection was carried out with 0.125 μL RNAiMAX (Invitrogen) per centimeter squared plus 20 nM siRNA according to the manufacturers instructions (this is equivalent to 0.5 μL RNAiMAX per well of a 12-well plate, or 1.25 μL per well of a 6-well plate). hESCs were seeded on Matrigel- coated plates at a density of 50,000 cells/cm 2 in MEF CM sup- plemented with 8 ng/mL bFGF, 5 μM Y-27632 and 0.5 μM thia- zovivin (both ROCK inhibitors from Tocris), and then allowed to attach overnight. The following day, cells were gently washed with knockout serum replacement (KSR) medium to remove dead cells and debris, and the culture medium was replaced with fresh MEF CM + bFGF. Cells received daily media changes thereafter for the duration of the experiment. If L or Wnt3A CM was added, cells were stimulated 48 h post-siRNA transfection. Cells were har- vested for analysis at 72 h post-siRNA transfection. siRNA sequence information (from Invitrogen): Silencer Select Negative Control 1 AXIN1, Silencer Select ID s15814 AXIN2, Silencer Select ID s15818 CTNNB1, Silencer Select ID s437 POU5F1-A, Silencer Select ID s10873 POU5F1-B, (sense) 5-AGCAGCUUGGGCUCGAGAA-3* POU5F1-C, (sense) 5-CAUGUGUAAGCUGCGGCCC-3* NANOG-A, (sense) 5-AACCAGACCUGGAACAAUUCA- 3Scrambled-NANOG, (sense) 5-AACGAGACCAUGAAC- GAUUCA-3NANOG-B, (sense) 5-AAGGGUUAAGCUGUAACAUAC- 3L and Wnt3A Conditioned Medium. L and L-Wnt3A cells (ATCC) were cultured in 10% FBS/DMEM until 90% conuent, then given a minimal volume of fresh medium (18 mL medium per 15- cm plate). This medium was incubated on cells for 48 h, then collected. A second batch of medium was incubated on the same cells for 48 h, then collected. This process was repeated a third time to yield three batches of conditioned medium (CM). Bi- ological activity of secreted Wnt3A in individual batches of CM was conrmed in 293T-BAR or A375-BAR reporter cells, then batches were pooled and ltered. L and Wnt3A CM were stored at 4 8C for up to 3 mo. Recombinant Wnt3a and Inhibitors. Recombinant Wnt3a (Millipore or Peprotech) was reconstituted at 100 μg/mL in a nal con- centration of 1% Chaps in PBS and bioactivity was conrmed for each lot number in 293T-BAR or A375-BAR cells. The follow- *POU5F1-B and POU5F1-C were previously published by Matin et al. (1). NANOG-A and Scrambled-NANOG were previously published by Hyslop et al. (2). NANOG-B was previously published by Wong et al. (3). Davidson et al. www.pnas.org/cgi/content/short/1118777109 1 of 9

Upload: phungcong

Post on 15-Aug-2019

214 views

Category:

Documents


0 download

TRANSCRIPT

Supporting InformationDavidson et al. 10.1073/pnas.1118777109SI Materials and MethodshESC Culture. For experiments where treatments were continuedover several weeks, an identical sister plate was harvested as singlecells via TrypLE Express (Invitrogen) to determine an accuratecell count for seeding. The experimental plate was harvested withdispase and small cell clusters were seeded at a density of 35,000cells per centimeter squared. For multiweek experiments, trip-licate sister wells were set up for each condition in 12-well plates:1 well was harvested with TrypLE Express to determine the cellcount for each treatment and used for flow cytometric analysis;a second well was passaged with dispase and seeded at 35,000 cellsper centimeter squared on the basis of the single cell count; thethird well was harvested with TRIZOL for RNA isolation.To obtain total cell number of GCTM2+/CD9+ stem cells per

well, the percentage of GCTM2+/CD9+ cells (determined byflow cytometric analysis) was multiplied by the number of cellscounted per well of a 12-well plate each week.

Flow Cytometry. hESCs were harvested as single cells via TrypLEExpress and counted using a Nucleocounter (New BrunswickScientific). For live flow cytometric analysis, 500,000 cells wereimmunolabeled with 100 μL primary antibodies, GCTM2 (hy-bridoma supernatant 1:2, a kind gift from Martin Pera, Univer-sity of Southern California, Los Angeles, CA) and CD9 (TG30clone, 1:100; Millipore), followed by isotype-specific secondaryantibodies: goat antimouse IgM-Alexa 647 (1:1,000) and goatantimouse IgG2a-biotin (1:5,000), followed by Alexa 750-R-PE-streptavidin or PE-Cy7-streptavidin (both 1:250) (all secondaryantibodies and streptavidin conjugates from Invitrogen). hESCswere resuspended in 140 ng/mL DAPI in culture medium andtransferred through a cell strainer before analysis on a BDFACSCanto II. Results were analyzed using FlowJo software.For H1-BAR-VENUS hESCs, the percentage of VENUS-

positive cells was determined from the DAPI-negative/DsRED-positive gated population relative to H1-FUBARV-negativecontrol line. Average mean fluorescent intensity (MFI) wasmeasured from the VENUS-positive subpopulation.For intracellular flow analysis, 1 × 106 hESCs were fixed in 4%

PFA for 20 min at room temperature, then permeabilized with0.1% Triton for 2 min on ice. Cells were immunolabeled with100 μL of Oct4 antibody (C-10 clone, 1:30; Santa Cruz) for 45–60min on ice, followed by goat antimouse IgG2b-Alexa 647(1:1,000; Invitrogen).

Lentiviral Transduction and Selection of hESCs. H1 hESC colonieswere isolated as small clusters using dispase in the same manneras for weekly passage, then exposed to ultraconcentrated lenti-virus before seeding onto Matrigel in mouse embryonic fibroblast(MEF) conditional medium (CM). Transduced hESCs werecultured for a week on Matrigel, then expanded onto several 3.5-cm plates withMEFs. Each lentiviral construct contained DsREDdriven by the Ubiquitin promoter as a selectable marker. Aftera week of expansion on MEFs, DsRED-positive areas of in-dividual colonies were mechanically isolated and transferred ontofresh MEFs. This positive selection method was repeated foranother week or two as needed until entirely DsRED-positivelines were isolated, after which time the stable lines were passagedwith dispase as per usual. In some cases, a final negative selectionstep was performed where DsRED-negative areas of colonieswere mechanically cut and discarded, then the remainder of thecolonies in the plate were passaged with dispase (Fig. S7).

Fluorescent-Activated Cell Sorting. H1-BAR-VENUS hESCs wereharvested as single cells with TrypLE Express. The DAPI-neg-ative/DsRED-positive gated population was sorted on the basis ofVENUS expression. A minimum of 1 × 106 sorted cells wascollected for downstream RNA isolation. For each experiment,samples were normalized to the same cell number on the basis ofpostsort cell counts before freezing in TRIZOL.

siRNA Reverse Transfection of hESCs. hESCs were harvested intoa single cell suspension with TrypLE Express, then counted ona NucleoCounter. Reverse transfection was carried out with 0.125μL RNAiMAX (Invitrogen) per centimeter squared plus 20 nMsiRNA according to the manufacturer’s instructions (this isequivalent to 0.5 μLRNAiMAXper well of a 12-well plate, or 1.25μL per well of a 6-well plate). hESCs were seeded on Matrigel-coated plates at a density of 50,000 cells/cm2 in MEF CM sup-plemented with 8 ng/mL bFGF, 5 μM Y-27632 and 0.5 μM thia-zovivin (both ROCK inhibitors from Tocris), and then allowed toattach overnight. The following day, cells were gently washed withknockout serum replacement (KSR)medium to remove dead cellsand debris, and the culture medium was replaced with fresh MEFCM+bFGF.Cells received dailymedia changes thereafter for theduration of the experiment. If L or Wnt3A CM was added, cellswere stimulated 48 h post-siRNA transfection. Cells were har-vested for analysis at 72 h post-siRNA transfection.siRNA sequence information (from Invitrogen):

Silencer Select Negative Control 1AXIN1, Silencer Select ID s15814AXIN2, Silencer Select ID s15818CTNNB1, Silencer Select ID s437POU5F1-A, Silencer Select ID s10873POU5F1-B, (sense) 5′-AGCAGCUUGGGCUCGAGAA-3′*POU5F1-C, (sense) 5′-CAUGUGUAAGCUGCGGCCC-3′*NANOG-A, (sense) 5′-AACCAGACCUGGAACAAUUCA-3′†Scrambled-NANOG, (sense) 5′-AACGAGACCAUGAAC-GAUUCA-3′†NANOG-B, (sense) 5′-AAGGGUUAAGCUGUAACAUAC-3′‡

L and Wnt3A Conditioned Medium. L and L-Wnt3A cells (ATCC)were cultured in 10% FBS/DMEM until ∼90% confluent, thengiven a minimal volume of fresh medium (18 mL medium per 15-cm plate). This medium was incubated on cells for 48 h, thencollected. A second batch of medium was incubated on the samecells for 48 h, then collected. This process was repeated a thirdtime to yield three batches of conditioned medium (CM). Bi-ological activity of secreted Wnt3A in individual batches of CMwas confirmed in 293T-BAR or A375-BAR reporter cells, thenbatches were pooled and filtered. L and Wnt3A CM were storedat 4 8C for up to 3 mo.

Recombinant Wnt3a and Inhibitors.Recombinant Wnt3a (Milliporeor Peprotech) was reconstituted at 100 μg/mL in a final con-centration of 1% Chaps in PBS and bioactivity was confirmed foreach lot number in 293T-BAR or A375-BAR cells. The follow-

*POU5F1-B and POU5F1-C were previously published by Matin et al. (1).†NANOG-A and Scrambled-NANOG were previously published by Hyslop et al. (2).‡NANOG-B was previously published by Wong et al. (3).

Davidson et al. www.pnas.org/cgi/content/short/1118777109 1 of 9

ing compounds were reconstituted in DMSO: XAV939 (Tocris),CHIR99021 (AxonMedChem), and BIO (Calbiochem).

Luciferase Reporter Assays. The lentiviral constructs used to gen-erate reporter 293T-BAR or A375-BAR are described elsewhere(4). Nonpluripotent cells were seeded in triplicate for eachcondition in 48-well plates and stimulated with Wnt3A or othertreatments for 16–18 h. Each well was then lysed in 50 μL 1×passive lysis buffer (Promega). Ten microliters per well of lysatewas assayed using the Dual Luciferase Assay kit (Promega) andnormalized to Renilla counts (Renilla is driven constitutively byEF1-α promoter in these cells). For H1-BAR-Luciferase hESCs,cells were seeded in sextuplicate for each condition and stimu-lated for 72 h before being assayed for Firefly luciferase. At-tempts to integrate EF1-α driven Renilla into H1-BAR-Luciferase cells resulted in rapid silencing of the EF1-α promoterwithin 3 wk postlentiviral transduction; thus Renilla normaliza-tion was not performed for hESC luciferase reporter lines.

RNA Isolation, cDNA Synthesis, and Quantitative PCR.TotalRNAwasextractedviaTRIZOLaccording to themanufacturer’sprotocol.Atotal of 2.5 μg RNA was used to generate cDNA using RevertAidFirst Strand cDNA Synthesis kit (Fermentas). cDNA was dilutedto 25 ng/μL, and quantitative PCR (qPCR) was carried out using 2μL of cDNAper 10 μL reaction (in duplicates for each run). qPCRwas performed using Applied Biosystems SYBR Green-baseddetection (Applied Biosystems) according to the manufacturer’sprotocol on a Roche Lightcycler 480 instrument. All primer pairswere validated for specificity to human and confirmed to not yielda signal with MEF cDNA. Transcript copy numbers were nor-malized to GAPDH for each sample, then fold expression overcontrol was calculated for each gene of interest. Primer sequencesare listed in Table S1.

Western Blots. Cells were lysed on ice in 1× RIPA buffer (50 mMTris pH 7.5, 150 mM NaCl, 0.1% Nonidet P-40, 0.2% deoxy-cholate) freshly supplemented with protease and phosphataseinhibitor mixtures (Roche). Cleared lysates were normalized byprotein concentration as determined by BCA assay (Pierce)before diluting with sample buffer + 20 mM DTT. To remove

junctional (cadherin-bound) β-catenin, some lysates were alsoConA stripped. Cleared lysates containing 750 μg protein in 200μL were incubated with 50 μL (packed volume) of ConA-se-pharose beads (Amersham) overnight at 4 8C with gentle end-over-end agitation. Lysates were normalized again by proteinconcentration using a BCA assay before adding sample buffer. Atotal of 10 μg protein was loaded per lane for Westerns. Blotswere blocked in 4% (wt/vol) skim milk powder, then incubatedwith the following primary antibodies: goat antihuman Axin1(1:1,000; R&D Systems), rabbit antihuman Axin1 (1:1,000; CellSignaling; no. 2087), rabbit antihuman β-catenin (1:1,000; CellSignaling), mouse antihuman β-catenin (1:1,000; BD Trans-duction Laboratories), rabbit anti-GFP (1:50,000; Abcam), ratantihuman HSP90 (1:20,000; Abcam), rabbit antihuman Nanog(1:1,000; Cell Signaling), goat antihuman Oct4 (1:500; R&DSystems), mouse antihuman Oct4 (C-10 clone, 1:1,000; SantaCruz), and mouse antihuman β-tubulin I (1:20,000; Sigma).Primary antibodies were diluted in 1% (wt/vol) BSA, 0.02% (wt/vol) sodium azide in TBS-T. Species-specific HRP-conjugatedsecondary antibodies were used, followed by ECL-based de-tection (Pierce).

Plasmid DNA Transient Transfections. HEK293T cells were trans-fected in a 6-well plate format with 10 μL per well lipofectamine2000 according to the manufacturer’s protocol (Invitrogen) usingthe following combination of plasmids per well:

240 ng pGL3-BARL,240 ng pRL-TK-Renilla,0.8–1,000 ng pLM-vexGFP-Oct4, pLenti-PGK-GFP-Puro, and/or pMXs-hNANOG, and

pCS2+ up to 2.4 μg total DNA.

Twenty-four hours posttransfection, cells from each well weresplit into duplicate wells of a 12-well plate and sextuplicate wellsof a 96-well plate. Three to four hours later, cells were stimulatedwith 50% L or Wnt3A CM for an additional 15–18 h. The 96-wellplate was then lysed in 25 μL per well of 1× passive lysis bufferfor the dual luciferase assay. The 12-well plate was lysed in 80 μLper well of 1× RIPA buffer for Western blots.

1. Matin MM, et al. (2004) Specific knockdown of Oct4 and beta2-microglobulinexpression by RNA interference in human embryonic stem cells and embryoniccarcinoma cells. Stem Cells 22:659–668.

2. Hyslop L, et al. (2005) Downregulation of NANOG induces differentiation of humanembryonic stem cells to extraembryonic lineages. Stem Cells 23:1035–1043.

3. Wong RC, et al. (2011) L1TD1 is a marker for undifferentiated human embryonic stemcells. PLoS ONE 6:e19355.

4. Biechele TL, Moon RT (2008) Assaying beta-catenin/TCF transcription with beta-catenin/TCF transcription-based reporter constructs. Methods Mol Biol 468:99–110.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 2 of 9

70

60

50

40

30

10

20

0

Lu

cife

rase

Act

ivit

y (F

old

ove

r L

CM

co

ntr

ol)

BIODMSO

KSRLCMWnt3A

H1-BAR-Luc

0

500

1000

2000

1500

2500

LCM

Wnt3

A CM

Chaps

% V

EN

US

+ ce

lls

10

5

0

15

20

30

25

35

40LC

MW

nt3A

CMCha

ps

rWnt3A

Mea

n F

luo

resc

ent

Inte

nsi

ty

C

rWnt3A

A B

H1-BAR-VENUS H1-BAR-VENUS

Chaps

3

2.5

2

1.5

1

0

0.5GC

TM

2+/C

D9+

cel

lsp

er w

ell (

x10^

6)

wk1 wk2 wk3

DMSOrWnt3A BIO

Fig. S1. Recombinant Wnt3A and GSK3-β inhibitor, BIO, both fail to maintain undifferentiated hESCs for 3 wk. (A) The number of GCTM2+/CD9+ cells per wellwas calculated by multiplying the percent of GCTM2+/CD9+ cells by the number of cells counted each week from 1 well of a 12-well plate. For recombinantWnt3A (rWnt3A) treated cells: concentrations tested were 100 ng/mL and 400 ng/mL; vehicle control was 1% (wt/vol) Chaps. For BIO-treated cells: concen-trations tested were 2 μM and 4 μM; vehicle control was DMSO. (B) H1-BAR-Luciferase reporter hESCs treated with BIO (2 μM and 4 μM) with or without 50%CM. Graph is mean + SEM of experimental replicates from a representative experiment. (C) Flow cytometric quantification of VENUS expression and meanfluorescent intensity for H1-BARV cells after 72-h stimulation with CM or with recombinant Wnt3a. Concentration of L and Wnt3A CM, 50%; rWnt3A, 100, 400,and 1,200 ng/mL. Graphs are mean + SEM from two independent experiments. Fifty percent Wnt3A CM yields a similar reporter response as 400 ng/mL rWnt3A.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 3 of 9

AXIN1

β-TUBULIN I

CM:XAV:

- L 3A 3A - L 3A 3A - L 3A 3A - - - + - - - + - - - +

6 days 72h 24h

KS

R

LCM Wnt3A CM

DMSOXAV

14

12

10

8

6

4

0

2

Luci

fera

se A

ctiv

ity

(fold

ove

r LC

M c

ontr

ol)

A B H1-BAR-Luc

KSR + DMSO LCM + DMSO WNT3A + DMSO WNT3A +2.5uM XAVWNT3A +0.2uM XAV

C

BA

R-V

ENU

SB

AR

-VEN

US

Ubi

q-D

sRED

Fig. S2. XAV939 stabilizes AXIN and blocks Wnt3A-induced activation of β-catenin reporter in hESCs. (A) Western blot showing AXIN1 expression in hESCscultured with KSR medium, 50% LCM, 50% Wnt3A CM, or 50% Wnt3A CM + 2.5 μM XAV for the period noted. All conditions without XAV received DMSOvehicle control. (B) XAV inhibits Luciferase reporter activation induced by Wnt3A in H1-BAR-Luc hESCs in a dose-dependent manner. Wnt3A CM, 50%; XAV, 100nM, 250 nM, and 2.5 μM). (C) Representative images of H1-BAR-Venus hESCs cultured in the conditions listed for 6 d. XAV inhibits VENUS reporter expression(green) and morphological differentiation (brightfield) in a dose-dependent manner. L and Wnt3A CM both are 50% in KSR medium. (Scale bar, 400 μm.)

Davidson et al. www.pnas.org/cgi/content/short/1118777109 4 of 9

POU5F1TROY

TKDR

GATA6MIXL

FOXA2SOX17

SOX1PAX6

0.0 6.8

Relative Expression (log2)

- 5.0

KSR LCM Wnt3A CM0days: 2 3 4 2 3 4 2 3 4

0.0 6.0

Relative Expression (log2)

- 0.5

1% Chaps recombinant Wnt3A

POU5F1TROYCDX1

T

KDRGATA4

MSX1

GATA6MIXL

FOXA2SOX17

SOX1PAX6

A B

Fig. S3. Similar gene expression profiles obtained for hESCs cultured with recombinant Wnt3A versus Wnt3A CM. (A) qRT-PCR results for H1 cells cultured for72 h with increasing concentrations of recombinant Wnt3A protein (50, 100, 200, 400, 800, and 1200 ng/mL). Heatmaps represent fold expression over vehiclecontrol. (B) Transcriptional effects of Wnt3A CM on hESCs cultured for 2, 3, or 4 d.

AXIN1

NANOG

β-CATENIN

β-TUBULIN

β-CATENIN

whole cell lysate

ConA-stripped lysate

CHIRXAV

KSR (untre

ated)

DMSOLCM

Wnt3A CM

Wnt3A CM +CHIR

Wnt3A CM + XAV

OCT4

A B

AXIN1

β-TUBULIN

β-CATENIN

β-TUBULIN

β-CATENIN

whole cell lysate

ConA-stripped lysate

AXIN1+2

CTNNB1

MockContro

l

POU5F1-A

POU5F1-B

OCT4

siRNA:

GFP

β-TUBULIN

Fig. S4. Western blots showing protein expression in hESC following activation or inhibition of Wnt/β-catenin pathway. (A) Western blots from H1 cellscultured for on MEFs in KSR media for 72 h with the following treatments: untreated (control), DMSO (vehicle control), 5 μM CHIR99021 (CHIR), 2.5 μM XAV939(XAV), 50% LCM, 50% Wnt3A CM, Wnt3A CM + 5 μM CHIR, and Wnt3A CM +2.5 μM XAV. Results show increased levels of nonmembrane-bound β-catenin(ConA-stripped lysates) under conditions that activate the Wnt pathway compared with the respective controls. Conversely, Axin levels are reduced underconditions that activate Wnt/β-catenin signaling and increased by the inhibitor, XAV939. (B) Western blots from H1-BAR-VENUS cells at 72 h post-siRNAtransfection on Matrigel in MEF CM. POU5F1-A siRNA results in a greater level of stabilized β-catenin protein in the nonmembrane-bound fraction (ConA-stripped) compared with control siRNA. The GFP antibody detects VENUS protein levels, indicative of BAR reporter activation.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 5 of 9

KSRH

1-B

AR

-VEN

US

LCM Wnt3A CM Culture for 6 days in KSR, L CM, or Wnt3a CM

KSR

H1-

BA

R-V

ENU

S

LCM-6d, KSR-8d Wnt3A CM-6d, KSR-8d Replate onto MEFs in KSR (no CM), culture for 8 days

Cel

l Cou

nts

VENUS

KSR LCM-6d, KSR-8d Wnt3A CM-6d, KSR-8d

A

C

D

CD9

GC

TM2

EKSR LCM-6d, KSR-8d Wnt3A CM-6d, KSR-8d

8.2% 7.8% 9.2% 0.2%

H1-FuBAR-VENUS(negative control)

negative isotype control

B

Cel

l Cou

nts

VENUS

KSR LCM Wnt3A CM

9.6% 17.2% 29.8% 0.3%

H1-FuBAR-VENUS(negative control)

Fig. S5. Wnt-induced BAR-VENUS expression is reversible in hESCs. (A) Images of H1-BAR-VENUS cells cultured on MEFs for 6 d in the following conditions: KSRmedia (untreated), 50% LCM in KSR (control), or 50% Wnt3A CM in KSR. (Upper) VENUS fluorescence. (Lower) Brightfield images. (Scale bar, 400 μm.) (B) Flowcytometric quantification of percent VENUS expression in the samples pictured in A, with H1-FuBAR-VENUS as gating reference. (C) Following 6 d of culturewith KSR, LCM, or Wnt3a CM, the H1-BAR-VENUS cells were replated onto MEFs in KSR media (no CM) and subsequently cultured for 8 d. Images show thathESCs previously cultured with Wnt3a CM revert back to VENUS-negative colonies. (D) Flow cytometric quantification of percent VENUS expression in thesamples pictured in C, with H1-FUBAR-VENUS as a gating reference. (E) Flow cytometric plots showing GCTM2/CD9 coexpression of hESCs from C and D.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 6 of 9

H1-BAR-VENUS

% V

enus

+ ce

lls

50

40

30

20

10

0

60

siRNA:

OCT4

NANOG

HSP90

70

Moc

k

Con

trol

PO

U5F

1-A

PO

U5F

1-B

NA

NO

G-A

NA

NO

G-B

Scr

ambl

ed

NA

NO

G

short exp.

long exp.

Fig. S6. NANOG does not contribute to repression of β-catenin reporter activity in hESCs. H1-BAR-VENUS cells were transfected with siRNAs, then analyzed forVENUS expression by flow cytometry at 72 h posttransfection. Graph of mean + SEM from three independent experiments. Western blots below show OCT4and NANOG protein reduction resulting from RNAi-mediated knockdown for each siRNA from a representative experiment.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 7 of 9

week 1

passage 1

Passage cells with dispase as normal onto matrigel in the presence of ultraconcentrated lentivirus (12-well plate format)

Passage each well with dispase into a 6cm plate with MEFs & expand

week 2

passage 2

manually select & transfer DsRED+ cells onto 3.cm plate with MEFs

week 3

passage 3

manually select & transfer DsRED+ cells to fresh MEFs; freeze heterogeneous line

week 4

passage 4

manually select & transfer DsRED+ cells to fresh MEFs; freeze homogeneous line

week 5

passage 5

If needed manually select & dis-card DsRED-negative cells; pas-sage remaining DsRED+ cells with dispase as per normal

Passage 1 + 2 daysPassage 1 + 9 days

Passage 2 Passage 2 for transfer

repeat manual selection until line is >90% DsRED+

Passage 5Selected DsRED+ line

Fig. S7. Method of generating and mechanically selecting stable hESC lines using fluorescent protein expression.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 8 of 9

Table S1. Primer sequences used for real-time quantitative PCR

Gene Primer –forward Primer -reverse Reference

AFP GAAGCAAGAGTTTCTCATTAACC AATTTAAACTCCCAAAGCAGCAAXIN2 GCGATCCTGTTAATCCTTATCAC AATTCCATCTACACTGCTGTCCD34 GCGCTTTGCTTGCTGAGTTT GCCATGTTGAGACACAGGGTCDX1 GGTAAGACTCGGACCAAGGA GATCTTCACCTGCCGTTCAGFOXA2 GACAAGTGAGAGAGCAAGTG ACAGTAGTGGAAACCGGAG (1)FST AGACCAATAATGCCTACTGTG ACTTTGCTTTGATACACTTTCCGAPDH TGAAGGTCGGAGTCAACGGA CCATTGATGACAAGCTTCCCGGATA4 CCAATCTCGATATGTTTGACGA TTGATGCCGTTCATCTTGTGGATA6 AACTTCCACCTCTTCTAACTCAG CATCTTGACCCGAATACTTGAGKDR CAAAGACTTGCTCTGGTAATAGC GAGACTGACGCCTATGTAGAGMIXL1 GGTACCCCGACATCCACTT GCCTGTTCTGGAACCATACCT (2)MSX1 TTAACCCTCACACTGCTCC CTCTAGCTCTGTTCAACTGTCNANOG AGAAGGCCTCAGCACCTAC GGCCTGATTGTTCCAGGATT (3)PAX6 CTTCACCATGGCAAATAACC GAAATGAGTCCTGTTGAAGTGPOU5F1 GGGAAGGTATTCAGCCAAACG GGTTCGCTTTCTCTTTCGGGSOX1 GAAACACAATCGCTGAACCA TCGTCAGGAATAATGAACAAGGSOX17 CCTGGGTTTTTGTTGTTGCT TCGTCAGGAATAATGAACAAGGT CTCCTTCAGCAAAGTCAAGC TTAAGAGCTGTGATCTCCTCGTROY GGAGTTGTCTAAGGAATGTGG GCTGAACAATTTGCCTTCTGTUBB3 CGGTGGTGGAGCCCTACAAC AGGTGGTGACTCCGCTCAT

1. Cai J, et al. (2006) Assessing self-renewal and differentiation in human embryonic stem cell lines. Stem Cells 24:516–530.2. Rosa A, Spagnoli FM, Brivanlou AH (2009) The miR-430/427/302 family controls mesendodermal fate specification via species-specific target selection. Dev Cell 16:517–527.3. Hyslop L, et al. (2005) Downregulation of NANOG induces differentiation of human embryonic stem cells to extraembryonic lineages. Stem Cells 23:1035–1043.

Davidson et al. www.pnas.org/cgi/content/short/1118777109 9 of 9