supporting information - pnas · 10. dennis g, jr, et al. (2003) david: database for annotation,...

6
Supporting Information Calvanese et al. 10.1073/pnas.1001399107 SI Materials and Methods Stem Cell Lines Culture, Differentiation, and Treatment. The Shef-1 hESC line was obtained from the Servicio de Inmunologia, Hospital Universitario Central de Asturias, and H-181 pellets were from Centro Andaluz de Biología Molecular y Medicina RegenerativaAndalusian Molecular Biology and Regenerative Medicine Cen- tre, Seville, Spain. Each laboratory cultured these lines following established protocols. Shef-1 was cultured in hESC medium [KO- DMEM, 20% KO serum replacement (Gibco), 1× nonessential amino acids, 1× glutamine (both from Lonza), 0.1 mM β-mer- captoethanol, 4 ng/mL bFGF (Peprotech)] on a mouse MEF mi- tomycin C-treated (Sigma) feeder layer, whereas H-181 was cultured on Matrigel in feeder-conditioned hESC medium. Cell lines were established from individual embryos and maintained independently, thereby ensuring the independence of our results for type of line and culture conditions. These embryos were do- nated for research in accordance with the legal requirements of the country of origin. All donors gave written informed consent. hESC were differentiated in vitro to EB and broblast-like cells (F-L) as described (1). Briey, undifferentiated colonies were de- tached by treatment with collagenase IV (Gibco) and incubated as oating aggregates for 14 d (or as indicated) in ultra-low attach- ment asks with hESC medium without bFGF or feeder condi- tioning. For further differentiation, they were attached to gelatin- coated plates and cultured in DMEM 15% FBS for an additional 1520 d, passaging with trypsin two or three times. Cells nally appeared as a monolayer of broblasts and keratinocyte-like cells. In vitro neural differentiation of the Shef-1 cell line was carried out as described (2). For the mRNA stability assay, Shef-1 cells were treated with 5 μg/mL actinomycin D (Sigma) as indicated. Mouse TC1 ES cells and the knockout ES line (SIRT1 Δex4/Δex4 ) (3) were cultured and differentiated to EB for 15 d by following protocols from the donating laboratory. Sirt1 transgenic ES cells were derived by the CNIO Transgenic Mouse Unit. Sirt1 trans- genic ( +/+ ; tg/.) and WT ES cells were established de novo from blastocysts obtained by in vitro fertilization of WT C57BL/6 oocytes with sperm of a Sirt1 transgenic ( +/+ ; tg/.) male (4) by following standard protocols (5). After fertilization, embryos were cultured in KSOM at 37 °C for 3 d until they reached the blastocyst stage, then plated in ES cell medium with feeders until outgrowth of the inner cell mass was observed. ES cell medium is composed of DMEM (high glucose) supplemented with 15% FBS, 1,000 U/mL LIF, nonessential amino acids, Glutamax, and β-mercaptoethanol. RNA Purication and Real-Time RT-PCR Analysis. RNA was isolated with TRIzol Reagent (Invitrogen) and reverse-transcribed by using the High Capacity cDNA Reverse Transcription Kit or the TaqMan MicroRNA Reverse Transcription Kit. Quantitative real-time RT- PCR (qPCR) was performed by using TaqMan Gene Expression Assays, SYBR green universal PCR master mix (see Dataset S5 for primers and assays) or the low-density TaqMan Human and Mouse Stem Cell Pluripotency Array and the ABI PRISM 7900 sequence- detection system (all from Applied Biosystems). Western Blot Analysis. Cell lysates for protein analysis were pre- pared by SDS lysis extraction and analyzed by Western blot using anti-α-tubulin (Sigma), anti-SIRT1, anti-phospho-Ser27 SIRT1, anti-p53, anti-acetyl-lysine-382 of p53, anti-OCT4, and anti-SOX2 (all from Cell Signaling Technologies), anti-acetyl-K16 of histone H4 (Active Motif), anti-HuR and anti-V5 (Invitrogen), anti- methyl-HuR (6), anti-CARM1 (Biovision), anti-NANOG (R&D Biosystems), and anti-E-cadherin antibodies (Becton Dickinson). To purify nuclear and cytoplasmic fractions, cytosolic proteins were extracted with RSB buffer by using a complete protease in- hibitor mixture (Roche), then separated from the nuclear pellet. Immunouorescence. hESC and EB cells were xed and stained (7) by using rabbit anti-SIRT1 (Cell Signaling Technologies), mouse anti-OCT4 or anti-TUBB3 antibodies (Millipore), followed by Alexa448 goat anti-rabbit IgG or Alexa594 goat anti-mouse IgG (Molecular Probes). SIRT1-TUBB3 costaining is presented as maximum projection of 10 z-stacked images (1.5 μm thick) to show the whole axon path. Flow Cytometry. EB were disaggregated by incubation in 0.4 U/mL Collagenase B (Roche; 2 h) and in enzyme-free cell dissociation buffer (Gibco; 10 min). A single-cell suspension was obtained by ltration through a 40-μm pore strainer and Histopaque 1077 (Sigma) separation of live mononuclear cells. Cells were xed, permeabilized, and stained with the Intraprep kit (Beckman Coulter). We used anti-SIRT1 (Santa Cruz), anti-OCT4, anti- MAP2 antibodies (Cell Signaling), followed by FITC-conjugated anti-mouse IgG 2B and PE-anti-rabbit IgG (H+L) (Southern Bio- tech). Cells were analyzed in a Cytomics FC 500 (Beckman Coul- ter). Because the distribution for SIRT1 staining was continuous (no positive or negative populations were distinguishable), we ar- bitrarily dened two subpopulations: cells expressing high SIRT1 levels (SIRT1-high, FITC intensity > 1.5 × 10 2 , and SIRT1-low, FITC intensity 24 × 10 1 ). To study SIRT1-associated expression of pluripotency and neuroectodermal development markers, we an- alyzed OCT4 and MAP2 expression and plotted the two cell sub- populations by PE uorescence intensity. Overlapped histograms are normalized for peak height. ChIP-on-chip, qChIP, and Data Analysis. The chromatin immuno- precipitation assay was carried out as described (8) by using anti- acK9H3 (Millipore), anti-AcK16H4, anti-SIRT1, and anti-H3 (Abcam) antibodies. For the ChIP-on-chip assay, we used the Agilent Human Promoter Array. SIRT1-immunoprecitated DNA fragments and the corresponding unbound fractions were labeled and hybridized by following manufacturers instructions. Results were analyzed by using the Agilent DNA Analytics program (9). Gene ontology was examined with DAVID (10, 11), a Web-based tool; results are shown in Dataset S2 as a GO chart and in Dataset S3 as GO clustering. For ChIP-on-chip validation, we selected the following genes: δ-like 4 (DLL4), a Notch ligand required for normal embryonic vascular development; LIM homeobox 1 (LHX1), a ho- meodomain-containing transcription factor (TF) involved in de- velopmental processes such as axon guidance; paired box 6 (PAX6), a TF involved in neuroectodermal denition and oculogenesis; member 6 of the wingless-type MMTV integration site family (WNT6), a secreted glycoprotein important in myocardial and neural crest development; bone morphogenetic protein 1 (BMP1), a metalloprotease that participates in embryonic patterning by cleaving matrix proteins and morphogens; Hairy/enhancer of split 7 (HES7), a TF involved in somite segmentation; T-box 3 (TBX3), a TF that affects developmental events such as sinoatrial node de- termination, limb bud positioning and retinal dorso-ventral pat- terning; serpin peptidase inhibitor, clade E member 1-plasminogen activator inhibitor type 1 (SERPINE1), a secreted protease inhibitor that regulates TGFβ and EGF signaling; homeobox A5 (HOXA5) a homeobox-containing TF important in lung, intestinal, and thyroid morphogenesis and blood cell differentiation; and TIMP metal- Calvanese et al. www.pnas.org/cgi/content/short/1001399107 1 of 6

Upload: others

Post on 13-Jul-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

Supporting InformationCalvanese et al. 10.1073/pnas.1001399107SI Materials and MethodsStem Cell Lines Culture, Differentiation, and Treatment. The Shef-1hESClinewasobtained fromtheServiciode Inmunologia,HospitalUniversitario Central de Asturias, and H-181 pellets were fromCentro Andaluz de Biología Molecular y Medicina Regenerativa–Andalusian Molecular Biology and Regenerative Medicine Cen-tre, Seville, Spain. Each laboratory cultured these lines followingestablished protocols. Shef-1 was cultured in hESC medium [KO-DMEM, 20% KO serum replacement (Gibco), 1× nonessentialamino acids, 1× glutamine (both from Lonza), 0.1 mM β-mer-captoethanol, 4 ng/mL bFGF (Peprotech)] on a mouse MEF mi-tomycin C-treated (Sigma) feeder layer, whereas H-181 wascultured on Matrigel in feeder-conditioned hESC medium. Celllines were established from individual embryos and maintainedindependently, thereby ensuring the independence of our resultsfor type of line and culture conditions. These embryos were do-nated for research in accordancewith the legal requirements of thecountry of origin. All donors gave written informed consent.hESC were differentiated in vitro to EB and fibroblast-like cells

(F-L) as described (1). Briefly, undifferentiated colonies were de-tached by treatment with collagenase IV (Gibco) and incubated asfloating aggregates for 14 d (or as indicated) in ultra-low attach-ment flasks with hESC medium without bFGF or feeder condi-tioning. For further differentiation, they were attached to gelatin-coated plates and cultured in DMEM 15% FBS for an additional15–20 d, passaging with trypsin two or three times. Cells finallyappeared as a monolayer of fibroblasts and keratinocyte-like cells.In vitro neural differentiation of the Shef-1 cell line was carried outas described (2). For the mRNA stability assay, Shef-1 cells weretreated with 5 μg/mL actinomycin D (Sigma) as indicated.Mouse TC1 ES cells and the knockout ES line (SIRT1Δex4/Δex4)

(3) were cultured and differentiated to EB for 15 d by followingprotocols from the donating laboratory. Sirt1 transgenic ES cellswere derived by the CNIO Transgenic Mouse Unit. Sirt1 trans-genic (+/+; tg/.) and WT ES cells were established de novo fromblastocysts obtained by in vitro fertilization of WT C57BL/6oocytes with sperm of a Sirt1 transgenic (+/+; tg/.) male (4) byfollowing standard protocols (5). After fertilization, embryoswere cultured in KSOM at 37 °C for 3 d until they reached theblastocyst stage, then plated in ES cell medium with feeders untiloutgrowth of the inner cell mass was observed. ES cell medium iscomposed of DMEM (high glucose) supplemented with 15%FBS, 1,000 U/mL LIF, nonessential amino acids, Glutamax, andβ-mercaptoethanol.

RNA Purification and Real-Time RT-PCR Analysis. RNA was isolatedwith TRIzol Reagent (Invitrogen) and reverse-transcribed by usingtheHighCapacity cDNAReverseTranscriptionKitor theTaqManMicroRNAReverse TranscriptionKit. Quantitative real-timeRT-PCR (qPCR) was performed by using TaqMan Gene ExpressionAssays, SYBR green universal PCRmastermix (seeDataset S5 forprimers and assays) or the low-density TaqManHuman andMouseStemCell Pluripotency Array and theABI PRISM7900 sequence-detection system (all from Applied Biosystems).

Western Blot Analysis. Cell lysates for protein analysis were pre-pared by SDS lysis extraction and analyzed by Western blot usinganti-α-tubulin (Sigma), anti-SIRT1, anti-phospho-Ser27 SIRT1,anti-p53, anti-acetyl-lysine-382 of p53, anti-OCT4, and anti-SOX2(all from Cell Signaling Technologies), anti-acetyl-K16 of histoneH4 (Active Motif), anti-HuR and anti-V5 (Invitrogen), anti-methyl-HuR (6), anti-CARM1 (Biovision), anti-NANOG (R&D

Biosystems), and anti-E-cadherin antibodies (Becton Dickinson).To purify nuclear and cytoplasmic fractions, cytosolic proteinswere extracted with RSB buffer by using a complete protease in-hibitor mixture (Roche), then separated from the nuclear pellet.

Immunofluorescence. hESC and EB cells were fixed and stained (7)by using rabbit anti-SIRT1 (Cell Signaling Technologies), mouseanti-OCT4 or anti-TUBB3 antibodies (Millipore), followed byAlexa448 goat anti-rabbit IgG or Alexa594 goat anti-mouse IgG(Molecular Probes). SIRT1-TUBB3 costaining is presented asmaximumprojection of 10 z-stacked images (1.5 μm thick) to showthe whole axon path.

Flow Cytometry. EB were disaggregated by incubation in 0.4 U/mLCollagenase B (Roche; 2 h) and in enzyme-free cell dissociationbuffer (Gibco; 10 min). A single-cell suspension was obtained byfiltration through a 40-μm pore strainer and Histopaque 1077(Sigma) separation of live mononuclear cells. Cells were fixed,permeabilized, and stained with the Intraprep kit (BeckmanCoulter). We used anti-SIRT1 (Santa Cruz), anti-OCT4, anti-MAP2 antibodies (Cell Signaling), followed by FITC-conjugatedanti-mouse IgG2B and PE-anti-rabbit IgG (H+L) (Southern Bio-tech). Cells were analyzed in a Cytomics FC 500 (Beckman Coul-ter). Because the distribution for SIRT1 staining was continuous(no positive or negative populations were distinguishable), we ar-bitrarily defined two subpopulations: cells expressing high SIRT1levels (SIRT1-high, FITC intensity > 1.5 × 102, and SIRT1-low,FITC intensity 2–4×101).To studySIRT1-associatedexpressionofpluripotency and neuroectodermal development markers, we an-alyzed OCT4 and MAP2 expression and plotted the two cell sub-populations by PE fluorescence intensity. Overlapped histogramsare normalized for peak height.

ChIP-on-chip, qChIP, and Data Analysis. The chromatin immuno-precipitation assay was carried out as described (8) by using anti-acK9H3 (Millipore), anti-AcK16H4, anti-SIRT1, and anti-H3(Abcam) antibodies. For the ChIP-on-chip assay, we used theAgilent Human Promoter Array. SIRT1-immunoprecitated DNAfragments and the corresponding unbound fractions were labeledand hybridized by following manufacturer’s instructions. Resultswere analyzed by using the Agilent DNA Analytics program (9).Gene ontology was examined with DAVID (10, 11), a Web-basedtool; results are shown inDataset S2 as aGOchart and inDataset S3as GO clustering. For ChIP-on-chip validation, we selected thefollowinggenes:δ-like4 (DLL4), aNotch ligand required fornormalembryonic vascular development; LIM homeobox 1 (LHX1), a ho-meodomain-containing transcription factor (TF) involved in de-velopmental processes such as axon guidance; paired box 6 (PAX6),a TF involved in neuroectodermal definition and oculogenesis;member 6 of the wingless-type MMTV integration site family(WNT6), a secreted glycoprotein important in myocardial andneural crest development; bone morphogenetic protein 1 (BMP1),a metalloprotease that participates in embryonic patterning bycleaving matrix proteins andmorphogens; Hairy/enhancer of split 7(HES7), a TF involved in somite segmentation; T-box 3 (TBX3),a TF that affects developmental events such as sinoatrial node de-termination, limb bud positioning and retinal dorso-ventral pat-terning; serpin peptidase inhibitor, clade E member 1-plasminogenactivator inhibitor type 1 (SERPINE1), a secreted protease inhibitorthat regulates TGFβ and EGF signaling; homeobox A5 (HOXA5)a homeobox-containingTF important in lung, intestinal, and thyroidmorphogenesis and blood cell differentiation; and TIMP metal-

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 1 of 6

Page 2: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

lopeptidase inhibitor 1 (TIMP1), a matrix protein that regulatesproteases in development.ForqChIP,PCRswerecarriedoutwithSYBR-greenPCRmaster

mixandanalyzedbyusing the7900HTFastReal-TimePCRSystem(all from Applied Biosystems). Primers for each promoter areshown in Dataset S5. The enrichment factor refers to the copynumber of a gene of interest in the bound fraction after ChIPwith the appropriate antibody, divided by the copy number of thatgene in the unbound form for that antibody. For SIRT1, ChIP dataare expressed as the percent enrichment of the SIRT1 immuno-precipitated sample relative to thenegative control. ForhistonemarkChIP, where two samples were compared, data were further nor-malized for the total histone H3 signal.

RNA Interference. RNA interference was performed by usinga protocol slightly modified from that of Braam et al. (12). Briefly,confluent hESC colonies were disaggregated by using Accutase(1:10 in PBS) and filtered through a 40-μm strainer to obtaina single-cell suspension, then seeded inMatrigel-coated plates withmTESR1medium (Stem Cell Technologies). The siRNA complexwas prepared with Lipofectamine RNAiMAX (Invitrogen) byfollowing the manufacturer’s protocol and mixed with the cells atthe time of seeding. Cells were maintained in mTESR1, which waschanged daily, and collected at the indicated time. For EB for-mation, cells were scraped 2 d after transfection and aggregatesresuspended in EB formation medium, as above.

RNA Immunoprecipitation. mRNA was immunoprecipitated as de-scribed (13) by using anti-HuR (Santa Cruz), anti-methyl-HuR,and anti-V5 antibodies (Invitrogen). Cell pellets were homoge-nized in lysis buffer (100 mM KCl, 5 mMMgCl2, 10 mMHepes atpH 7.0, 0.5% Nonidet P-40, 1 mM DTT, RNase OUT and com-plete protease inhibitor mixture), precleared with IgG1 mouse

control (BDPharmingen) andProteinASepharose beads (Sigma),then incubated with beads and IgG1 or with anti-HuR antibody,washed with NT2 buffer, and incubated with RNase-free DNase I(Invitrogen; 15 min, 37 °C). RNA was examined by qPCR.

HuR Phosphorylation Assay. Phosphorylated-HuR (Ser) was immu-noprecipitated from 100 μg of total protein by using an anti-phosphoserine–agarose-conjugated antibody (Sigma). Immunopre-cipitated phospho-HuR was detected by using total HuR antibody(Santa Cruz).

Cloning of HuR Wild-Type, HuR(R217K) and HuR(R217A), PlasmidConstruction, and Transfection. Full-length HuR cDNA wasobtained from the RZPD Deutsches Ressourcenzentrum für Ge-nomforschung. HuR(WT)-V5 was constructed by PCR amplifica-tion using the 5′ oligonucleotide containing the V5 tag sequence,and subcloned into pCDNA 3.3 TOPO vector (Invitrogen). HuR(R217K)-V5 and HuR(R217A)-V5 mutants were constructed byusing theQuikChange kit for site-directedmutagenesis (Stratagene)with two complementary oligonucleotides, and the pCDNA-HuR(WT)-V5 plasmid as the template. Primers are shown inDataset S5.For plasmid transfection, Shef-1 hESC was prepared as a single-cellsuspension (see RNA Interference above). Cells were pelleted foreach reaction and transfected with plasmid by using the AmaxaNucleofector kit 2 (Lonza), following manufacturer’s protocols.

Bisulphite Sequencing of Multiple Clones. DNA methylation wasquantified by PCR analysis following bisulphite modification ofDNA.Bisulphite genomic sequencing was carried out as described(14). Primers were designed by using Methyl Primer ExpressSoftware (Applied Biosystems); primer sequences are shown inDataset S5.

1. Li SS, et al. (2009) Target identification of microRNAs expressed highly in humanembryonic stem cells. J Cell Biochem 106:1020–1030.

2. Pankratz MT, et al. (2007) Directed neural differentiation of human embryonic stemcells via an obligated primitive anterior stage. Stem Cells 25:1511–1520.

3. Cheng HL, et al. (2003) Developmental defects and p53 hyperacetylation in Sir2homolog (SIRT1)-deficient mice. Proc Natl Acad Sci USA 100:10794–10799.

4. Pfluger PT, Herranz D, Velasco-Miguel S, Serrano M, Tschop MH (2008) Sirt1 protectsagainst high-fat diet-inducedmetabolic damage. ProcNatl Acad Sci USA 105:9793–9798.

5. Nagy A, Gertsenstein M, Vintersten K, Behringer R (2003) Manipulating the MouseEmbryo: A Laboratory Manual (Third Ed) (Cold Spring Harbor Lab Press, Cold SpringHarbor, NY).

6. Li H, et al. (2002) Lipopolysaccharide-induced methylation of HuR, an mRNA-stabilizing protein, by CARM1. Coactivator-associated arginine methyltransferase. JBiol Chem 277:44623–44630.

7. Ungrin MD, Joshi C, Nica A, Bauwens C, Zandstra PW (2008) Reproducible, ultra high-throughput formation of multicellular organization from single cell suspension-derived human embryonic stem cell aggregates. PLoS ONE 3:e1565.

8. Fraga MF, et al. (2005) Epigenetic differences arise during the lifetime of monozygotictwins. Proc Natl Acad Sci USA 102:10604–10609.

9. Lee TI, et al. (2006) Control of developmental regulators by Polycomb in humanembryonic stem cells. Cell 125:301–313.

10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, andIntegrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3.

11. Huang DW, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis oflarge gene lists using DAVID Bioinformatics Resources. Nat Protoc 4:44–57.

12. Braam SR, et al. (2008) Feeder-free culture of human embryonic stem cells inconditioned medium for efficient genetic modification. Nat Protoc 3:1435–1443.

13. Abdelmohsen K, et al. (2007) Phosphorylation of HuR by Chk2 regulates SIRT1expression. Mol Cell 25:543–557.

14. Fraga MF, et al. (2005) Loss of acetylation at Lys16 and trimethylation at Lys20 ofhistone H4 is a common hallmark of human cancer. Nat Genet 37:391–400.

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 2 of 6

Page 3: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

A

B

Pluripotency (OCT4)

Developmental (MAP2)

Even

ts(R

elat

ive

units

) SIRT1-high

SIRT1-low

100 101 102 103100 101 102 103

Fluorescence intensity

50 µm

50 µm50 µm

SIRT1

SIRT1

Merge

Merge

OCT4

TUBB3

Plu

ripot

ency

Dev

elop

men

tal

50 m

50 m

0

20

40

60

80

100

120

SIRT1

Tubulin

ES EB7 EB15 F-L feeders

Shef-1

SIR

T1/G

APD

H

C

m

m

Fig. S1. Characterization of SIRT1 down-regulation during EB differentiation. (A) Flow cytometry analysis of pluripotency (OCT4) and neural differentiation(MAP2) markers in disaggregated EB cells. We arbitrarily defined a gate for cells with FITC intensity > 1.5 × 102 (SIRT1-high subpopulation) and for cells witha FITC intensity from 2 to 4 × 101 (SIRT1-low), and plotted these subpopulations by PE fluorescence intensity. Graphs show the overlap of these histograms,scaled to the same total intensity, to visualize peak displacement. (B) Confocal fluorescence microscopy assay showing cell localization of SIRT1 (green) andOCT4 or TUBB3 (red) in Shef-1 15-d EB. (C) Time-course expression study of SIRT1 by qPCR (relative to GAPDH) and WB during Shef-1 hESC differentiation.α-tubulin was used as loading control. Mitomycin C-treated mouse embryonic fibroblasts used as feeder cells showed no murine Sirt1. Real-time PCR values aremean ± SD of three independent experiments.

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 3 of 6

Page 4: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

BA SIRT1 promoter

H-181 ES

H-181 EB

Shef-1 ES

Shef-1 EB

-207 bp

PBL

+45 bp

0

20

40

60

80

100

120

140

IgG AcH4 AcH3 H3

rDNA

0

100

200

300

400

500

600

IgG AcH4 AcH3 H3

SIRT1-Pr

0

100

200

300

400

500

600

700

IgG AcH4 AcH3 H3

POU5F1

ES

EB

0

100

200

300

400

500

600

700

IgG AcH4 AcH3 H3

SIRT1-Ex1

ES

EB

0

20

40

60

80

100

120

140

ES EB7 EB14

% m

iR34

a/R

NU

19 e

xpre

ssio

nre

lativ

eto

ES

miR34aC D E

H-181

0

20

40

60

80

100

120

140

160

180

200

ES EB7 EB14 F-L

HuR/

GAP

DH

ES EB14

Shef-1

Shef-1

0

10

20

30

40

50

60

70

80

ES EB14

% p

hosp

hoSe

r-Hu

R/to

tal

HuR

GF

Phospho-S27 SIRT1

SIRT1

TUBULIN

ES EB7 EB14

Shef-1

Shef-1

0

10

20

30

40

50

60

70

ES EB F-L

Rel

a�ve

mR

NA

leve

ls

(-c

aten

in/G

APD

H)

RNA IP: HuR

b

Fig. S2. Mechanisms of SIRT1 transcript and protein regulation. (A) Bisulfite genomic sequencing of multiple clones of the SIRT1 promoter in two hESC lines(H-181, Shef-1), the corresponding differentiated sample (EB), and peripheral blood lymphocytes. Black box, methylated CpG; white box, unmethylated CpG.(B) qChIP of global acetylated histones H4 and H3 in Shef-1 hESC and EB. qPCR corresponding to promoters of rDNA (negative control), POU5F1 (OCT4; positivecontrol of down-regulation of activation markers in differentiation), and two regions of the SIRT1 promoter located up- (SIRT1-Pr) and downstream (SIRT1-Ex1)of the transcriptional start site. IgG, negative IP control. Results are expressed as the percentage bound/unbound ratio of DNA copy number for each IP, furthernormalized with respect to total H3 enrichment (considered constant in chromatin). (C) Time-course expression study of miR34a by qPCR (relative to RNU19,a small RNA whose expression is considered constant) in hESC Shef-1 (ES) and in 7- and 14-d EB (EB7 and EB14). (D) Western blot analysis of hESC line Shef-1(ES), and 7- and 14-d EB (EB7 and EB14) with anti-phosphorylated-Ser27 of SIRT1, -total SIRT1 and -α-tubulin (loading control) antibodies. (E) HuR binding toβ-catenin RNA. RNA IP of HuR in hESC (ES), 15-d EB (EB) and fibroblast-like cells (F-L) of the Shef-1 hESC line. β-catenin levels were detected by qPCR (relative toGAPDH). (F) Time-course expression study of HuR by qPCR (relative to GAPDH) in hESC Shef-1 (ES), 7- and 14-d EB (EB7 and EB14), and fibroblast-like cells (F-L)(Left), and H-181 hESC and 14-d EB (Right). (G) Densitometric quantification of phosphorylated-HuR detected after IP with anti-phospho-Ser antibody and WBanalysis in Shef-1 ES and 14-d EB. Data relative to total HuR levels. Data are expressed as mean ± SD of three independent experiments.

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 4 of 6

Page 5: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

CAiSIRT1SCR

ol2

iHuR

ol1

iCARM1

CARM1

HuR

SIRT1

OCT4

NANOG

TUBULIN

ol2ol1

Shef-1 SCR day3

Shef-1 iSIRT1 day3

% e

xpre

ssio

nre

lativ

eto

ES

BMP1

HES7

LHX1

0

5000

10000

15000

0200400600800

1000

0

500

1000

1500

0

5000

10000

15000

20000

0500

1000150020002500

0500

1000150020002500

0

200

400

600

05000

10000150002000025000

HOXA5

DLL4

TBX3

WNT6

PAX6

AcH4K16

SCR iSIRT1

SIRT1

Tubulin

Ac-p53

p53

E-cadherin

SCR iSIRT1

SIRT1

Tubulin

Nanog

Sox2

Oct4

D

F

G

0

100

200

300

400

0100200300400500600700

0100200300400500600

0100200300400500

0

50

100

150

200

0

100

200

300

400

0

200

400

600

800ES

EB

0

100

200

300

400

050

100150200250

0

50

100

150

200

0

50

100

150

0

50

100

150

qChIP

% e

nric

hmen

tS

IRT1

ChIP-chip qChIP

%B

ound

/Unb

ound

rRNA

HES7

LHX1

HOXA5

TIMP1

BMP1

B

E

Fig. S3. SIRT1 ChIP-on-chip and RNAi validation. (A) qChIP of SIRT1 in Shef-1 hESC. qPCR was used to study enrichment relative to a chromatin sample im-munoprecipitatedwithout antibody (NAB) for the SIRT1-bound regions ofBMP1,HES7, LHX1,HOXA5, and TIMP1. Results expressed as Fig. 3A. The ribosomal RNAgene (rDNA) promoter was included as a negative control for SIRT1 binding and histone modifications. (B) qChIP of acetyl-lysine 16 of histone H4 (AcH4K16) andacetyl-lysine 9 of histone H3 (AcH3K9) in Shef-1 hESC and EB. qPCR corresponds to the same genomic regions as above. Results are expressed as in Fig. 3B. (C)Western blot of CARM1, HuR, SIRT1, POU5F1 (OCT4), NANOG, and α-tubulin in Shef-1 cells, 3 d after transfection with control siRNA (SCR), SIRT1-specific siRNA(iSIRT1 ol1, ol2), HuR1-specific siRNA, or CARM1-specific siRNA (iCARM1ol1, ol2). (D) Phase-contrastmicroscopy photographs of hESC Shef-1, 3 d after transfectionwith scrambled siRNA (SCR) or iSIRT1 ol1 siRNA (iSIRT1). (E) Expression of SIRT1-bound genes during EB and neural differentiation. Gene expressionwasmeasuredby qPCR in Shef-1 hESC and 7- and 14-d EB (EB7 and EB14), and in the neuron-enriched sample derived from in vitro differentiation to neuroectodermal cells. Dataare normalized to GAPDH expression, relative to the undifferentiated hESC sample (GAPDH expression is set to 100%). qPCR values are mean ± SD of threeindependent experiments.(F)WBof SIRT1, acetyl-lysine 382 of p53, total p53, acetyl-lysine 16 of histoneH4, and α-tubulin in Shef-1 cells 3 d after transfectionwithcontrol siRNA (SCR) or SIRT1-specific siRNA (iSIRT1). (G) Western blot using the same samples, for pluripotency markers Oct4, E-cadherin, Nanog, and Sox2.

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 5 of 6

Page 6: Supporting Information - PNAS · 10. Dennis G, Jr, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 10.1186/gb-2003-4-5-p3. 11

Sirt1

TUBULIN

Undifferen�ated mESC

Sirt1

TUBULIN

ES EB3 EB14EB7 ES EB3 EB14EB7 ES EB3 EB14EB7 ES EB3 EB14EB7

WT1 Sirt1 KO WT2 SuperSirt1

A B

C

D

Fig. S4. mES cell lines characterization. (A) WB analysis of Sirt1 levels in mES lines of TC1 (WT1), TC1 with deletion of Sirt1 exon 4 (SIRT KO), mES clone with thenormal copy number of the Sirt1 gene (WT2; Materials and Methods) and an ES clone bearing extra copies of the Sirt1 gene (SuperSirt1). (B) WB of Sirt1 in thesame mESC lines differentiated into EB for 3, 7, and 14 d (EB3, EB7, EB14). α-tubulin was used as a loading control. (C) Pluripotency and differentiation markerexpression in Sirt1 KO (Sirt1−/−) or SuperSirt1 (Sirt1+++) mESC differentiation. Five pluripotency (Fgf4, Rest, Tdgf1, Nodal, Tcfcp2l1), five neuroectodermal (Syp,Nes Isl1, Sfrp2, Hlxb9), two mesodermal (Brachyury, Col2A1), and two endodermal (Lama, Fn1) cell markers were measured by qPCR in mESC (ES) and 15-d EB.Data are normalized with respect to Gapdh expression, relative to the undifferentiated ES sample for each line. qPCR values are mean ± SD of three in-dependent experiments. (D) qChIP of Sirt1 in TC1 mESC. qPCR was used to analyze Sirt1 enrichment relative to a chromatin sample immunoprecipitatedwithout antibody, for the Nanog, Oct4, Nes, Neurod1, and Sfrp2 promoters. Results are expressed as percent enrichment of the bound/unbound ratio (DNAcopy number) of Sirt1 relative to the NAB immunoprecipitate.

Other Supporting Information Files

Dataset S1 (XLS)Dataset S2 (XLS)Dataset S3 (XLS)Dataset S4 (XLSX)Dataset S5 (XLSX)

Calvanese et al. www.pnas.org/cgi/content/short/1001399107 6 of 6