nature cell biology - mir-34 mirnas provide a barrier for somatic cell reprogramming

Upload: josh-meier

Post on 03-Jun-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    1/17

    L E T T E R S

    miR-34 miRNAs provide a barrier for somatic cell

    reprogrammingYong Jin Choi1,7, Chao-Po Lin1,7, Jaclyn J. Ho1, Xingyue He2, Nobuhiro Okada1, Pengcheng Bu1, Yingchao Zhong1,Sang Yong Kim3, Margaux J. Bennett1, Caifu Chen4, Arzu Ozturk5, Geoffrey G. Hicks5, Greg J. Hannon6

    and Lin He1,8

    Somatic reprogramming induced by defined transcription

    factors is a low-efficiency process that is enhanced by p53

    deficiency15. So far, p21 is the only p53 target shown to

    contribute to p53 repression of iPSC (induced pluripotent stem

    cell) generation1,3, indicating that additional p53 targets may

    regulate this process. Here, we demonstrate that miR-34

    microRNAs (miRNAs), particularly miR-34a, exhibit

    p53-dependent induction during reprogramming. Mir34a

    deficiency in mice significantly increased reprogramming

    efficiency and kinetics, with miR-34a and p21 cooperatively

    regulating somatic reprogramming downstream of p53. Unlike

    p53 deficiency, which enhances reprogramming at the expense

    of iPSC pluripotency, genetic ablation of Mir34apromoted

    iPSC generation without compromising self-renewal or

    differentiation. Suppression of reprogramming by miR-34a wasdue, at least in part, to repression of pluripotency genes,

    includingNanog, Sox2 andMycn(also known as N-Myc). This

    post-transcriptional gene repression by miR-34a also regulated

    iPSC differentiation kinetics. miR-34b and c similarly

    repressed reprogramming; and all three miR-34 miRNAs acted

    cooperatively in this process. Taken together, our findings

    identified miR-34 miRNAs as p53 targets that play an

    essential role in restraining somatic reprogramming.

    Differentiated somatic cells can be induced to generate pluripotent

    stem cells that functionally resemble embryonic stem cells (ESCs;

    ref. 6). This reprogramming process is rooted in the remarkable cellular

    plasticity retained during differentiation. The process can be triggered

    by exogenous expression of a set of defined ESC-specific transcription

    factors, Pou5f1 (also known as Oct4), Sox2, Klf4 and c-Myc (refs69),

    which constitute the core regulatory circuits controlling pluripotency

    and self-renewal. Enforced expression of these reprogramming factors

    1Division of Cellular and Developmental Biology, Molecular and Cell Biology Department, University of California at Berkeley, Berkeley, California 94705, USA. 2 40

    Landsdowne Street, Cambridge, Massachusetts 02139, USA. 3 Transgenetic facility, 500 Sunnyside Blvd., Woodbury, New York 11797-2924, USA. 4 Genomic Assays

    R&D, Life Technologies Corporation, 850 Lincoln Centre Dr., Foster City, California 94404, USA. 5 Manitoba Institute of Cell Biology, 675 McDermot Ave. Rm.

    ON5029, Winnipeg, MB R3E 0V9, Canada. 6 Cold Spring Harbor Laboratory, Watson School of Biological Sciences, 1 Bungtown Road, Cold Spring Harbor, New York

    11724, USA. 7 These authors contributed equally to this work.8Correspondence should be addressed to L.H. (e-mail:[email protected])

    Received 10 May 2011; accepted 22 September 2011; published online 23 October 2011; DOI: 10.1038/ncb2366

    generates iPSCs with low efficiency and slow kinetics, suggesting the

    existence of cellular and molecularbarriersto theprocess10.

    Recent studies have revealed considerable mechanistic overlap

    between somatic cell reprogramming and malignant transformation11.

    Cellular mechanisms that enhance reprogramming, including cell

    proliferation and survival, evasion of DNA-damage response, and cell

    immortalization, have long been known to promote tumorigenesis35,12.

    Several oncogenes and tumour suppressors also serve as essential

    regulators for reprogramming6,10,11. Notably, the inactivation of p53,

    one of the most important tumour suppressors, significantly enhances

    iPSC generation15,13. As a tumour suppressor, p53s transcriptional

    regulation converges onto multiple target genes that collectively

    mediate its downstream effects, including cell-cycle arrest, cellular

    senescence, apoptosis, DNA-damage response and genomic stability14.

    Similarly, p53s role in repressing reprogramming is probably alsomediated through multiple targets. To date, the cell-cycle regulator

    p21 is the only p53 target with a demonstrated role in repressing

    reprogramming. However,p21deficiency only partially phenocopies

    that ofp53(refs1,3,15), suggesting that p53 represses iPSC generation

    through as-yetunidentified mechanism(s) and target(s).

    Previous studies have identified the miR-34 microRNAs (miRNAs)

    as bona fide p53 transcriptional targets, whose overexpression

    triggers cell-cycle arrest or apoptosis in a cell-type- and context-

    dependent manner1618. miRNAs, a large family of small non-coding

    RNAs, primarily repress gene expression post-transcriptionally, by

    pairing with partially complementary messenger RNA targets19,20.

    In response to p53 activation, inducedMir34miRNAs can mediate

    p53 downstream effects by repressing specific target genes, including

    cyclin D1, cyclin E2, Cdk4, Cdk6, Bcl2 and c-Met (ref. 21).

    Although p53 is primarily characterized for its role in transcriptional

    activation, it acts as a global gene regulator that both activates

    and represses gene expression14. Direct transcriptional repression,

    NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011 1353

    mailto:[email protected]:[email protected]
  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    2/17

    L E T T E R S

    together with indirect post-transcriptional repression through miRNAs

    such as miR-34, constitute two major mechanisms for p53-

    mediated gene repression.

    The miR-34 miRNAs belong to an evolutionarily conserved family,

    with three mammalian homologues, miR-34a, b and c, localized to two

    distinct genomic loci,Mir34aandMir34b/c(Fig. 1a; ref.16). All three

    Mir34miRNAs were significantly induced when mouse embryonic

    fibroblast (MEF) reprogramming was triggered with Sox2, Oct4 and

    Klf4 in the presence or absence of c-Myc (Fig. 2a, data not shown). In

    both cases,Mir34induction, similarly to that ofp21, was dependent on

    the activation of intact p53 (Fig. 2a). We therefore investigated whether

    miR-34 miRNAs are components of the reprogramming regulatory

    circuit downstream of p53. Among all miR-34 miRNAs, miR-34a

    exhibited the highest induction level during reprogramming, whereas

    miR-34b was the lowest. We therefore focused initially on the role of

    Mir34ain iPSC induction.

    We generatedMir34aknockout mice using C57BL/6 ESCs(Fig. 1a),

    confirmed the germline transmission of the targeted allele (Fig. 1b)

    and verified the genetic ablation ofMir34a by expression studies

    (Fig. 1c). Mir34a/

    mice were born at the expected Mendelianratio, without obvious developmental or pathological abnormalities

    up to 12 months. However, when we induced the Mir34a/

    MEFs for reprogramming, we observed a significant increase in the

    reprogramming efficiency(Fig. 2b,d and Supplementary Fig. S1a).

    Three-factor-infectedMir34a/ MEFs exhibited a 4.5-fold increase

    in alkaline phosphatase-positive colonies with typical iPSC morphology

    (Fig. 2b), whereas four-factor-infected Mir34a/ MEFs yielded a

    4-fold increase (Supplementary Fig. S1a). Furthermore, when we

    plated infected MEFs into 96-well plates at a density of one cell

    per well, Mir34a deficiency caused a similar increase in alkaline

    phosphatase-positive colonies with typical iPSC morphology(Fig. 2c).

    Using MEFs carrying an Oct4Gfp knockin reporter allele22, we

    confirmed the effect of miR-34a in promoting reprogramming, scoring

    fully reprogrammed iPSCs on the basis of endogenousOct4expression

    indicated by green fluorescent protein (GFP). Consistently, a greater

    than fourfold increase in reprogramming efficiency was observed in

    Mir34a/; Oct4Gfp/+MEFs after three- or four-factor transduction

    (Fig. 2d). A significant increase in reprogramed Oct4Gfp-positive

    colonies could also be achieved using a locked nucleic acid (LNA)

    inhibitor against miR-34a (Supplementary Fig. S1b). Notably, Mir34a

    deficiency both enhanced the overall efficiency of iPSC generation and

    led to more rapid reprogramming kinetics. Small iPSC-like colonies

    first appeared 7 days post-infection in four-factor-transduced wild-type

    MEFs, but as early as post-infection day 5 inMir34a/ MEFs.

    As miR-34b and c share the miR-34a seed sequence and are similarlyinduced during reprogramming(Fig. 2a), they probably also regulate

    iPSC generation. We generatedMir34b/cknockout mice (Fig. 1ac). As

    with miR-34a deficiency,Mir34b/cknockout alone promoted somatic

    reprogramming, although to a lesser degree (Fig. 2e). Interestingly,

    MEFs deficient for all three miR-34 miRNAs exhibited an even

    greater increase in iPSC generation(Fig. 2e), suggesting a cooperative

    effect among these genes, although the exact molecular and cellular

    mechanisms underlying this cooperation still remained unclear. As the

    Mir34a/;Mir34b/c/ MEFs did not completely phenocopyp53/

    MEFs, extra mechanisms may act downstream of p53 to mediate the

    suppression of reprogramming.

    Previous studies have identified p21 as an important mediator

    of p53 suppression of reprogramming1,12. Mir34 and p21 both

    exhibitedp53-dependent induction during reprogramming (Fig. 2a).

    p21 inductionalso wasobservedinMir34a/ MEFs(Fig. 3a). Whereas

    MEFs deficient for Mir34aalone or p21 alone showed comparable

    increases in iPSC generation, MEFs deficient for both Mir34aandp21

    exhibited a cooperative increase that recapitulated a significant fraction

    of the p53 effect (Fig. 3c). Given the functional similarities among

    the three miR-34 miRNAs, the cooperative effects among p21 and all

    miR-34 miRNAs could be even greater. Thus, the miR-34 miRNAs,

    together with p21, constitute important downstream effectors of p53

    to mediate the repression of reprogramming.

    p21 represses reprogramming efficiency primarily through its

    repression of cell proliferation12. Although miR-34a and p21 repress

    reprogramming efficiency similarly (Fig. 3c), their effects on cell

    proliferation are quite different. The increased cell proliferation in

    p21/ MEFs was highly significant (Fig. 3b), yet within the time

    frame of our reprogramming experimentsMir34a/ MEFs exhibited

    little increase up to passage 6. We cannot completely exclude the

    possibility that a mild increase in Mir34a/

    MEF proliferationcontributed to the increased reprogramming. Yet, unlike p21, whose

    effects on reprogramming are largely attributed to its inhibition

    of cell proliferation12, miR-34a is likely to act mostly through a

    separate mechanism independent of cell proliferation. These two

    distinct mechanisms may underlie the cooperative regulation of

    reprogramming by miR-34a and p21.

    Mir34a/ iPSCs resemble wild-type iPSCs and ESCs, exhibiting

    ESC-like morphology (Fig. 4a and Supplementary Fig. S1c) and

    expressing key molecular markers for pluripotency. High levels of Oct4,

    Nanog and SSEA1 were detected (Fig. 4b and Supplementary Fig. S1d).

    Mir34a/ iPSCs injected into immunocompromised nude mice

    yielded differentiated teratomas, containing terminally differentiated

    cell types from all three germ layers (Fig. 4c,d and Supplementary

    Fig. S1e). Furthermore, three independent lines of four-factor-induced

    Mir34a/ iPSCs all yielded healthy adult chimaeric mice with a high

    percentage of iPSC contribution (Fig. 4e and Supplementary Fig. S1g,

    Table S1). Taken together,Mir34adeficiency enhances the efficiency of

    iPSC generation without compromising self-renewal or pluripotency.

    Although p53deficiency induced reprogramming more efficiently

    than Mir34a deficiency, the p53/ iPSCs exhibited compromised

    self-renewal and differentiation capacity1,4. As also reported in

    ref. 1, we have observed that four-factor-induced p53/ iPSCs

    lost ESC-like morphology after five to six passages in culture,

    and failed to generate highly differentiated teratomas. In contrast,

    four-factor-induced Mir34a/ iPSCs remained stable beyondpassage 26, with no significant differences in self-renewal when

    compared with wild-type iPSCs(Supplementary Fig. S1f).Furthermore,

    generation of healthy adult chimaeras from p53/ iPSCs was

    difficult. The percentage of p53/ iPSC contribution was low,

    and the majority of such chimaeras succumbed to tumorigenesis

    before 7 weeks1. In contrast,Mir34a/ iPSCs exhibited functional

    pluripotency: all three four-factor-induced Mir34a/ iPSC lines

    tested gave rise to healthy adult chimaeras with a high percentage

    of iPSC contribution (Fig. 4e and Supplementary S1g, Table S1),

    which remained tumour free for 6 months (to the time of

    manuscript preparation).

    1354 NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    3/17

    L E T T E R S

    +/++//+/

    ++//+/

    ++//

    ES #3ES #3

    +/++//

    +/++//

    +/++//

    No. 1 No. 2 No. 3

    No. 1 No. 2 No. 3

    8.6 kb10.7 kb

    11 kb

    8.3 kb

    34 a +/+ 34b/c+/+

    Foldu

    pregulation

    34 a +/ 34b/c+/34 a/ 34b/c/

    : loxP

    E P

    X5probe

    5p

    robe

    5p

    robe

    3p

    robe

    3p

    robe

    5probe 3probe

    3probeMir34a E

    ELacZ

    PNeo

    E P

    Kozaksequence : FRT E : EcoRI P : PsiI X : XhoI

    X E

    M : MfeI S : SpeI

    : loxP

    M

    M

    M

    S

    S

    S

    M

    M

    M

    S

    S

    SPGK-Neo

    Mir34b

    Mir34c

    11.8 kb

    10.7 kb

    9.0 kb6.6 kb6.4 kb5.3 kb

    +/+

    +/ /

    +/+

    +/flN

    eo

    +/fl

    +/

    miR-34a

    Mir 34a Mir34b/c

    Mir34a

    miR-34b miR-34c

    Mir 34a Mir34b/c

    Mir34b/c

    Mir34aWT

    Mir34a

    Mir34b/cWT

    Mir34b/cflNeo

    Mir34b/c

    0

    0.25

    0.50

    0.75

    1.00

    1.25

    Fold

    upregulation

    0

    0.25

    0.50

    0.75

    1.00

    1.25

    34b/c+/+ 34b/c+/ 34b/c/Foldu

    pregulation

    0

    0.25

    0.50

    0.75

    1.00

    1.25

    a

    b

    c

    FRT

    Figure 1Generation ofMir34aand Mir34b/cknockout MEFs. (a) Diagrams

    of endogenousMir34aand Mir34b/cgene structure (WT) and the knockout

    construct (). Using recombineering, we engineered the Mir34atargeting

    vector with a 6-kilobase (kb) homologous arm on both 5 and 3

    ends, flanking a Kozak sequence, a lacZ complementary DNA and an

    FRTNeoFRT cassette. The Mir34b/c targeting vector (Neo) contains

    a 6-kb homologous arm at each end, with the Mir34b/c gene and a

    Neo selection cassette flanked by loxP sites. (b) Validating the germline

    transmission of the Mir34a and Mir34b/c targeted allele using Southern

    analysis. Putative wild-type, Mir 34a+/ and Mir 34a/ animals derived

    from three independently targeted ESC lines were analysed by Southern

    blotting using probes either 5 or 3 to the homologous arms (left). Similar

    validation was carried out for wild-type, Mir 34b/c/+ and Mir 34b/c+/

    animals (right). (c) Confirming loss of miR-34 expression in Mir34a and

    Mir34b/cknockout MEFs. Littermate-controlled wild-type,Mir 34a+/ and

    Mir 34a/ MEFs were analysed by real-timePCR to quantify theexpression

    of Mir34a. Whereas wild-type MEFs showed robust miR-34a induction

    on culture stress, no miR-34a expression was detected in Mir 34a/

    MEFs. The miR-34a level in Mir 34a+/ MEFs was approximately half

    that of wild-type MEFs. Similar validation was carried out for Mir 34b/c/

    MEFs. Error bar, s.d., n= 3. Uncropped images of blots are shown in

    Supplementary Fig. S6.

    Consistent with these differences, we observed that four-factor-

    induced p53/ iPSCs, but not Mir34a/ or wild-type iPSCs,

    failed to silence retroviral transgenes, thus exhibiting lower levels

    of the corresponding endogenous genes (Supplementary Fig. S3ac).

    The exogenous expression of reprogramming factors, particularly

    c-Myc (Supplementary Fig. S3e), may contribute to the impaired

    self-renewal and differentiation (Supplementary Fig. S3d), and

    promote tumorigenesis1. In contrast, pluripotency was established

    easily in Mir34a/ iPSCs by the endogenous transcription factor

    circuitry, leaving the exogenous transgenes dispensable for the

    maintenance of pluripotency.

    Similar toMir34a/ iPSCs,Mir34a/;Mir34b/c/ iPSCs also

    exhibited robust self-renewal and differentiation capacity, with typical

    ESC morphology, key pluripotency markers and the ability to generate

    differentiated teratomas (Supplementary Fig. S2a

    c). Their ability togenerate chimaeras remains to be determined.

    The increased reprogramming efficiency and compromised pluripo-

    tency ofp53/ MEFs may result from aberrant apoptosis and DNA

    damage response4. However,Mir34adeficiency failed to protect cells

    from apoptosis or DNA-damage response during reprogramming

    (Supplementary Fig. S4ac). Thus, the enhanced reprogramming effi-

    ciency observed inMir34a/ MEFs may reflect a mechanism largely

    independent of apoptosis and DNA damage response. These findings

    contrast with the ability ofMir34aoverexpression to trigger apoptosis

    in specific tumour cells17,18. These Mir34a effects on apoptosis are

    probably cell-type dependent, differing between primary fibroblasts16

    and specific cancer cell lines17,18. Cells deficient forMir34amay exhibit

    apoptotic defects under conditions other than reprogramming. It is

    also possible that the lack of apoptotic protection in reprogramming

    Mir34a/ MEFs reflectsthe functional redundancy ofmiR-34b andc.

    Enhanced reprogramming in p53-deficient cells has also been at-

    tributed to cell immortalization and increased cell proliferation15,12,15.

    Whereas cell immortalization greatly promotes iPSC generation from

    p53/ MEFs (ref.5), increased cell proliferation is a key mechanism

    driving stochastic reprogramming ofp53KD B cells12. AlthoughMir34

    overexpression induced growth arrest and cellular senescence in pri-

    mary fibroblasts16, no defects in senescence response were observed in

    Mir34a/ MEFs during reprogramming (Supplementary Fig. S4d and

    data not shown). As described above, we did not observe a significant

    MEF proliferation difference caused byMir34adeletion within the

    time frame of our reprogramming experiment. Presumably, miR-34aregulates reprogramming efficiency largely through a mechanism

    independent of cell proliferation and cell senescence.

    To better define the molecular mechanism of miR-34a in reprogram-

    ming, we initiated a search for its targets, specifically by examining

    genes that promotes the iPSC generation for possible miR-34a-binding

    sites. RNA22 identified a number of such genes with predicted miR-34a

    sites23 (Supplementary Fig. S5d). Of all the genes tested,Nanog,Sox2

    andMycn(N-Myc) emerged as top candidates(Fig. 5a and Supplemen-

    tary Fig. S5a). All three exhibitedMir34a-dependent repression (Fig. 5b

    and Supplementary Fig. S5c), and each had potent effects in promoting

    reprogramming24,25. ESCs over-expressing miR-34a, miR-34b or

    NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011 1355

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    4/17

    L E T T E R S

    WT

    WT

    Mir34a/

    p53/

    2,500 infected

    cells per well2,500infectedcells perwell

    No.

    1

    No.

    2

    No.

    1

    No.

    2No.

    1

    No.

    2

    No.

    1

    No.

    2

    WT 34a/ WT p53/

    P< 0.01 P< 0.01

    NumberofAP-positivecolonies

    NumberofAP-positivecolonies

    WT 34a/ p53/

    NumberofAP-positivecolonies

    P< 0.05

    P< 0.005

    P< 0.0001

    n= 3

    n= 3

    n= 3

    pri-miR-34b/c

    Foldupregulation

    Mature miR-34b

    WT WT34

    a/

    34b/c

    /

    34a/

    34b/c/

    WT WT34

    a/

    34b/

    c/

    34a/

    34b/

    c/

    WT34

    a/

    34b/c/

    WT34

    a/

    34b/c

    /

    Foldupregulation

    Foldupregulation

    Mature miR-34c

    Foldupregulation

    pri-miR-34a

    Mature

    miR-34a p21

    WT uninfected

    WT infected

    p53/uninfected

    p53/infected

    0

    1

    23

    4

    5

    6

    7

    8

    0

    0.5

    1.01.5

    2.0

    2.5

    3.0

    3.5

    4.0

    0

    2

    4

    6

    1

    2

    3

    4

    0

    12

    24

    36

    48

    60

    0

    30

    60

    90

    120

    150

    0

    4

    8

    12

    16

    20

    a

    b

    d e

    c

    No.

    1

    No.

    2

    No.

    1

    No.

    2

    WT 34a/

    NumberofOct4Gfppositivecolonies

    OSKM

    0

    3

    6

    9

    12

    15

    34a /

    No.

    1

    No.

    2

    No.

    1

    No.

    2

    WT

    NumberofOct4Gfppo

    sitivecolonies

    OSK

    0

    1

    2

    3

    4

    5

    6

    7

    8

    NumberofOct4-p

    ositivecolonies

    n= 2

    n= 2

    n= 2

    n= 4

    OSKM

    WT34a

    /

    34b/

    c/

    34a/ ;3

    4b/c

    /0

    5

    10

    15

    20

    25

    30

    NumberofOct4-p

    ositivecolonies

    n= 2

    n= 2

    n= 2

    n= 4

    OSK

    WT34

    a/

    34b/

    c/

    34a/ ;

    34b/

    c/

    0

    4

    8

    12

    16

    20WT

    Phase Oct4Gfp

    Mir34a/

    Uninfected Infected Uninfected Infected Uninfected Infected

    P< 0.01 P< 0.02

    Figure 2Deficiency of miR-34 miRNAs increases reprogramming efficiency.

    (a) Four reprogramming factors triggered p53-dependent induction of

    miR-34 miRNAs. Three days after transduction, pri-miR-34a (the precursor

    transcript from the Mir34alocus), mature miR-34a and p21 were measuredin uninfected and four-factor-induced wild-type (WT) and p 53

    /MEFs.

    Induction of pri-miR-34a was dependent on the intact p53 response, and was

    comparable to that of p21. Induction of pri-miR-34b/c and mature miR-34b

    and miR-34c was determined in wild-type, Mir 34a/ andMir 34b/c/

    MEFs. Error bar, s.d., n=3. (b) Mir34adeficiency significantly enhanced

    three-factor-induced MEF reprogramming. 2,500 three-factor-infected

    wild-type,Mir 34a/ orp 53/ MEFs were plated to score reprogramming

    by alkaline phosphatase (AP)-positive colonies with characteristic ESC

    morphology. A representative image and quantitative analysis is shown

    out of five independent experiments, comparing littermate-controlled

    wild-type and Mir 34a/ MEFs (left, P< 0.01), as well as wild-type and

    p53/

    MEFs (right, P< 0.01). Error bar, s.d., n=4. (c) Single-sorted,

    four-factor-infected MEFs were cultured at a density of one cell per well.

    Four weeks post-plating, alkaline phosphatase-positive colonies with typical

    iPSC morphology were scored for wild-type, Mir 34a/ and p53/

    iPSCs. Four independent experiments confirmed this finding. P< 0.05

    for comparison between wild-type and Mir34a/ MEFs. Error bar, s.e.m.,

    n= experiments with independent MEF lines. (d) Mir34adeficiencysignificantly enhanced MEF reprogramming as measured by Oct4Gfp

    reporter expression. Three- or four-factor-infected wild-type and Mir 34a/

    MEFs that carry an Oct4Gfpallele were sorted at the density of 2,500 cells

    per well and 1,000 cells per well, respectively. Reprogramming efficiency

    was quantified by GFP-positive clones. Images of Oct4Gfp-positive iPSCs

    are shown on the left. A quantitative analysis for reprogramming efficiency

    triggered by four factors (left, P< 0.01) or three factors (right, P< 0.02)

    is shown. Scale bar, 100 m. Error bar, s.d., n=3. OSKM, Oct4, Sox2, Klf4

    and Myc; OSK, Oct4, Sox2 and Klf4. (e) miR-34a, b and c cooperatively

    regulate somatic reprogramming. Deficiency in Mir34a or Mir34b/calone

    significantly promoted somatic reprogramming, yet deficiency in all Mir34

    miRNAs exhibited further increase. Two independent experiments confirmed

    this finding. n, experiments with independent MEFs. All P-values were

    calculated on the basis of a two-tailed Students t-test.

    1356 NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    5/17

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    6/17

    L E T T E R S

    Tuj1/DAPI SMA/DAPI Foxa2/DAPI

    Oct4 DAPI SSEA1 DAPI

    Wild type

    Wildtype

    Wildtype

    M

    ir34a

    /

    Mir34a

    /

    Mir34a/

    Mir34a/

    Oct4/DAPI SSEA1/DAPI

    Epidermis Cart ilage Cil liated epithelium

    Neural rosette Bone-like Gut-like epithelium

    Ectoderm Mesoderm Endoderm Ectoderm Mesoderm Endoderm

    Epidermis Cartilage Gut-l ike epithel ium

    Hair foll icle Muscle Cil liated epithelium

    Tuj1/DAPI SMA/DAPI Foxa2/DAPI

    Wild type

    a e

    b

    c

    d

    Figure 4 Mir34a/ iPSCs functionally resemble wild-type iPSCs. (a) iPSCs

    derived from both wild-type and Mir 34a/ MEFs exhibited ESC-like

    morphology in culture, with robust alkaline phosphatase expression.

    Scale bar, 20m for the left panel, 100 m for the middle and right

    panels. (b) Both wild-type and Mir 34a/ iPSCs expressed pluripotency

    markers, including nucleus-localized Oct4 and membrane-localized SSEA1.

    Scale bar, 20 m. (c,d) Wild-type and Mir 34a/ iPSCs both generated

    differentiated teratomas. Teratomas derived from four-factor-inducedwild-type (left) and Mir 34a/ (right) iPSCs were collected from nude

    mice 46 weeks after subcutaneous injection. Haematoxylin and eosin

    staining (c), as well as immunofluorescence staining (d), revealed

    terminally differentiated cell types derived from all three germ layers.

    Scale bar in c, 25 m; in d, 50 m. (e) Four-factor-induced Mir 34a/

    iPSCs efficiently contribute to adult chimaeric mice. We injected three

    independent lines of passage 7 Oct4Gfp/+, Mir 34a/ iPSCs into

    albino-C57BL/6/cBrd/cBrd/cr blastocysts. The iPSC contribution to adult

    chimaeric mice was determined by coat colour pigmentation. DAPI,4,6-diamidino-2-phenylindole.

    global gene expression31. Along with transcription factors, miRNAs

    have emerged as essential gene regulators in self-renewal and

    differentiation of pluripotent stem cells32,33. ESCs with deficient

    global miRNA biogenesis exhibit proliferation and differentiation

    defects34. In addition, the RNA-binding protein LIN28, which

    induces reprogramming by suppressing let-7 biogenesis8,3537, has been

    identified as a major reprogramming factor in humans. Beside let-7

    miRNAs, a number of miRNAs are specifically enriched or depleted in

    pluripotent stem cells and are demonstrated to regulate self-renewal

    and differentiation30,32,34,3840. Here, we identify the miR-34 miRNAs as

    regulators that suppress reprogramming downstream of p53, providing

    direct evidence that modulation of miRNA abundance could constitute

    a critical step in establishing pluripotency. As miRNA functions

    can be manipulated by oligonucleotide-based inhibitors or mimics,

    our findings suggest a paradigm for promoting reprogramming by

    manipulating specific miRNA functions.

    Although p53 directly regulates hundreds of target genes14, miR-34

    miRNAs and p21 are the main downstream targets that cooperatively

    1358 NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    7/17

    L E T T E R S

    a

    c

    d

    e

    b

    No. 1 No. 2 No. 3 No. 1 No. 2 No. 3

    OSKM OSK

    No. 1 No. 2 No. 3 No. 1 No. 2 No. 3 No. 1 No. 2 No. 3

    0.3

    Wild type Mir34a/;

    Mir34b/c/

    OSK

    0.16

    43

    43

    34

    72

    43

    34

    55

    43

    34

    72

    43

    34

    55

    43

    34

    34

    55

    Mr(K)

    Mr(K) Mr(K)

    Wildtype

    Wildtype

    Mir34a

    /

    LIF feeder(day 2)Untreated

    Wild type

    Wild type

    Mir34a/

    Mir34a/

    (Days) (Days)

    Nanog Oct4 Sox2

    Mir34a

    /

    Rela

    tivemRNAlevels

    (Days) (Days)

    RelativemRNAlevels

    Rela

    tivemRNAlevels

    Relat

    ivemRNAlevels

    RelativemRNAlevels

    RelativemRNAlevels

    Nanog

    UGUUGGUCGAU-----UCUGUGA

    AGACACU--CCAUC

    UUC

    GGUC

    Sox2

    UG UG UCGAUUCU GACGGU

    UU AU AGCUGAGA UUGCCA

    GUU

    U

    G

    A AU

    miR-34a

    N-Myc

    UC GUGACGGU

    CCA CGG AG CACUGCCA

    UGU GUCUG

    AUUG

    CACUGUAGCUG

    miR-34a

    miR-34a

    AC UCA

    Oct4

    Tub

    Nanog

    Sox2

    N-Myc

    0.04 0.05 0.02 0.06

    1 0.93

    1.12 0.84

    0.95

    0.99 0.95 1.05 1.10 1.06 1.14

    1.08 0.98 1.85 1.57 1.78

    1.09 1.07 3.54 2.98 3.17 0.87

    0.96 1.03 1.01 1.52 1.40

    1.16

    0.96 1.05 0 .981 .20 1.18 1.16

    1.07 0.76 1.74 1.70 1.72

    1.46

    0 .85 1.32 2 .97 2.95 3.06

    1.16 1.11 0.73 1.79 2.04 1.93

    0.93

    1.03 0.99 0.98 1.06 1.11 1.03

    0.97 0.920.961.10 1.04

    1.07 0.80 2.01 2.45 1.28

    0.53 0.59 0.49

    1 0.97 0.70 0.53 0.61

    1 0.87 0.95 0.90 0.83

    1 1.11 1.06 1.08 1.21

    Nanog

    Oct4

    Tub

    Sox2

    Wild type Mir34a/ Mir34a/Wild type

    No. 1 No. 2 No. 3

    Nanog

    Oct4

    Tub

    Sox2

    N-Myc

    0.03

    0.02 0.03

    0.12 0.01

    0.010.03

    Mock

    GFPsiR

    NA

    miR

    -34a

    miR

    -34b

    miR

    -34c

    0.29 0.14 0.04 0.01

    0.05 0.07 0.00 0.06

    0.04 0.11 0.03 0.04

    0.07

    0.040.05

    0.14 0.04

    0.03 0.02

    0.09

    0.09 0.20.09

    0 1 2 3

    (Days)

    40

    0.2

    0.4

    0.6

    0.8

    1.0

    0 1 2 3 0

    0.4

    0.8

    1.2

    1.6

    0 1 2 3 40

    0.5

    1.0

    1.5

    0

    0.2

    0.4

    0.6

    0.8

    1.0

    0 1 2 3

    (Days)

    0 1 2 30

    0.3

    0.6

    0.9

    1.2

    1.5

    0

    0.2

    0.4

    0.6

    0.8

    1.0

    0 1 2 3

    LIF feeder + RA

    (day 2)Untreated

    Nanog Oct4 Sox2

    1 1.05 0.62 0.45 0.53

    0.94 0.99 1.07 1.16 1.11 1.13

    1.00 1.03 0.97 1.13 1.08 1.15

    Figure 5 Mir34a represses Nanog, Sox2 and N-Myc expression

    post-transcriptionally. (a) Schematic representation of the Nanog, Sox2and N-Myc 3UTR, and the predicted miR-34 binding sites. The mouse

    Nanog, Sox2 and N-Myc each contains one putative miR-34a-binding

    site within its 3UTR. (b) Enforced expression of Mir34a, Mir34b

    and Mir34c in ESCs reduced the protein levels of Nanog, Sox2 and

    N-Myc, but not Oct4. Feeder-free ESCs were transfected with miRNA

    mimics for miR-34a, miR-34b and miR-34c, and a negative control,

    Gfp short interfering RNA (siRNA). At 48 h post transfection, western

    analysis indicated a significant reduction in the protein levels of Nanog,

    Sox2 and N-Myc, but not Oct4. The value of each band indicates the

    relative expression level normalized by the internal control, -tubulin,

    averaged between two independent experiments, and presented as mean

    s.e.m. (c) Derepression of Nanog, Sox2 and N-Myc was observed in

    Mir34-deficient iPSCs. A significant increase of Nanog and Sox2, but

    not Oct4, was observed in four-factor-induced Mir 34a/ iPSCs, when

    compared with passage-matched, littermate-controlled wild-type iPSCs. A

    similar comparison was made for passage-matched, three-factor-inducedwild-type and Mir 34a/; Mir 34b/c/ double-knockout iPSCs, where

    derepression of Nanog, Sox2 and N-Myc was observed. For this western

    analysis, the quantitation of each band was carried out by Quantity One

    software, and was normalized against its own internal tubulin control. The

    s.e.m. of three independent iPSC lines are shown for each genotype. ( d,e)

    Mir34a-deficient iPSCs exhibited slower kinetics during differentiation.

    Wild-type and Mir 34a/ iPSCs were both triggered to differentiate

    by withdrawal of LIF in the absence (d) or presence (e) of retinoic

    acid treatment. Images of a typical iPSC culture two days after each

    differentiation condition are shown on the left, and quantitative analyses

    of the decline of Nanog, Sox2 and Oct4 transcripts in response to these

    differentiating conditions are shown on the right. Error bar, s.e.m., n=3.P

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    8/17

    L E T T E R S

    repress iPSC generation. Previous studies indicate that p53 effects on

    reprogramming reflect changes in cell proliferation, immortalization,

    apoptosis and DNA-damage response4,5,12. p21, a key downstream

    target of p53, represses cell proliferation. Interestingly, Mir34a

    deficiency alters MEF reprogramming not through cell proliferation

    but, at least in part, by posttranscriptional derepression of pluripotency

    genes. Thus, miR-34 miRNAs, together with other p53 targets,

    including p21, collectively mediate the suppression of somatic

    reprogramming through multiple critical cellular pathways.

    METHODS

    Methods and any associated references are available in the online

    version of the paper athttp://www.nature.com/naturecellbiology

    Note: Supplementary Information is available on the Nature Cell Biology website

    ACKNOWLEDGEMENTS

    We thank B. Zaghi, A. Basila, A. Perez, G. Lai, M. Foth, A. Kemp, A. Fresnoza,

    A. Valeros, A. Fritz, D. Schichnes and H. Noller for technical assistance and

    A. Economides for discussions and input. We also thank J. M. Halbleib, D. Schaffer

    andR. M.Harland forreading themanuscripts, andI. Lemischka, Q. Li,M. Hemann

    and B. Vogelstein for sharing reagents. In particular, we are grateful for the

    pathological expertise R. Van Andel offered us. L.H. is a Searle Scholar, and is

    supported by a new faculty award by California Institute for Regenerative Medicine(RN2-00923-1) and an R01 (R01 CA139067) and an R00 grant (R00 CA126186)

    from the National Cancer Institute (NCI). G.J.H. is a Howard Hughes Medical

    Institute investigator, and is supported by a program project grant from the NCI.

    C-P.L. is supported by a Siebel postdoctoral fellowship.

    AUTHOR CONTRIBUTIONS

    Y.J.C., C-P.L. and L.H. designed all experiments and carried out the majority of

    the experiments shown in all figures and supplementary figures. J.J.H. and Y.Z.

    carried out immunofluorescence analyses and teratoma analyses to characterize

    the pluripotency of the iPSCs. L.H., X.H., P.B. and G.J.H. generated knockout

    constructs for Mir34a and Mir34b/c, and identified the correctly targeted ESC

    clones forMir34a. N.O. and P.B. identified the correctly targeted ESC clone for

    Mir34b/c, validated Mir34a and Mir34b/ctargeting in mice by Southern blotting

    and generated Mir34a/, Mir34b/c/ andMir34 triple knockout MEFs. S.Y.K.

    and G.J.H. carried out the blastocyst injection for Mir34a

    +/

    and Mir34b/c

    +/

    ESC clones. A.O., S.Y.K. and G.G.H. characterized the pluripotency of iPSCs using

    chimaera assays. M.J.B. and C.C. contributed to the identification of miR-34a

    targets.

    COMPETING FINANCIAL INTERESTS

    The authors declare no competing financial interests.

    Published online at http://www.nature.com/naturecellbiology

    Reprints and permissions information is available online at http://www.nature.com/

    reprints

    1. Hong, H. et al. Suppression of induced pluripotent stem cell generation by the

    p53p21 pathway. Nature 460, 11321135 (2009).

    2. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic cell

    reprogramming.Nature 460,11401144 (2009).

    3. Li, H.et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460,

    11361139 (2009).

    4. Marion, R. M.et al. A p53-mediated DNA damage response limits reprogrammingto ensure iPS cell genomic integrity. Nature 460, 11491153 (2009).

    5. Utikal, J. etal. Immortalization eliminates a roadblock during cellular reprogramming

    into iPS cells. Nature 460, 11451148 (2009).

    6. Yamanaka, S. & Takahashi, K. Induction of pluripotent stem cells from mouse

    fibroblast cultures. Tanpakushitsu Kakusan Koso 51,23462351 (2006).

    7. Takahashi, K.et al. Induction of pluripotent stem cells from adult human fibroblasts

    by defined factors. Cell 131, 861872 (2007).

    8. Yu, J.et al. Induced pluripotent stem cell lines derived from human somatic cells.

    Science 318, 19171920 (2007).

    9. Park, I. H.et al. Reprogramming of human somatic cells to pluripotency with defined

    factors. Nature 451, 141146 (2008).

    10. Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and

    applications.Genes Dev. 24, 22392263 (2010).

    11. Krizhanovsky, V. & Lowe, S. W. Stem cells: the promises and perils of p53. Nature

    460, 10851086 (2009).

    12. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to

    acceleration. Nature 462, 595601 (2009).

    13. Zhao, Y. et al. Two supporting factors greatly improve the efficiency of human iPSC

    generation. Cell Stem Cell 3, 475479 (2008).

    14. Riley, T., Sontag, E., Chen, P. & Levine, A. Transcriptional control of human

    p53-regulated genes.Nat. Rev. Mol. Cell Biol. 9, 402412 (2008).

    15. Banito, A.et al. Senescence impairs successful reprogramming to pluripotent stem

    cells.Genes Dev. 23, 21342139 (2009).

    16. He, L. et al. A microRNA component of the p53 tumour suppressor network. Nature

    447, 11301134 (2007).

    17. Raver-Shapira, N. et al. Transcriptional activation of miR-34a contributes to

    p53-mediated apoptosis. Mol. Cell26, 731743 (2007).

    18. Chang, T. C. et al. Transactivation of miR-34a by p53 broadly influences gene

    expression and promotes apoptosis. Mol. Cell 26,745752 (2007).

    19. Ambros, V. The functions of animal microRNAs. Nature 431, 350355 (2004).

    20. Bartel, D. P. MicroRNAs: genomics, biogenesis, mechanism, and function.Cell116,

    281297 (2004).

    21. Hermeking, H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 17,

    193199 (2009).

    22. Lengner, C. J. et al. Oct4 expression is not required for mouse somatic stem cell

    self-renewal.Cell Stem Cell 1, 403415 (2007).

    23. Miranda, K. C. et al . A pattern-based method for the identification of

    MicroRNA binding sites and their corresponding heteroduplexes. Cell 126,

    12031217 (2006).

    24. Yamanaka, S. & Blau, H. M. Nuclear reprogramming to a pluripotent state by three

    approaches. Nature 465, 704712 (2010).

    25. Nakagawa, M., Takizawa, N., Narita, M., Ichisaka, T. & Yamanaka, S. Promotion of

    direct reprogramming by transformation-deficient Myc. Proc. Natl Acad. Sci. USA

    107, 1415214157 (2010).

    26. Wei, J. S. et al. The MYCN oncogene is a direct target of miR-34a. Oncogene 27,

    52045213 (2008).

    27. Cole, K. A. et al . A functional screen identifies miR-34a as a candidate

    neuroblastoma tumor suppressor gene. Mol. Cancer Res. 6, 735742 (2008).

    28. Kuijk, E. W. et al. PTEN and TRP53 independently suppress Nanog expression in

    spermatogonial stem cells.Stem Cells Dev. 19, 979988 (2009).

    29. Lin, T. et al. p53 induces differentiation of mouse embryonic stem cells by

    suppressing Nanog expression. Nat. Cell Biol. 7, 165171 (2005).

    30. Tarantino, C. et al. miRNA 34a, 100, and 137 modulate differentiation of mouseembryonic stem cells. FASEB J. 24, 32553263 (2010).

    31. Filipowicz, W., Bhattacharyya, S. N. & Sonenberg, N. Mechanisms of post-

    transcriptional regulation by microRNAs: are the answers in sight? Nat. Rev. Genet.

    9, 102114 (2008).

    32. Melton, C., Judson, R. L. & Blelloch, R. Opposing microRNA families regulate

    self-renewal in mouse embryonic stem cells. Nature 463, 621626 (2010).

    33. Wang, Y.et al. Embryonic stem cell-specific microRNAs regulate the G1-S transition

    and promote rapid proliferation. Nat. Genet. 40, 14781483 (2008).

    34. Wang, Y., Medvid, R., Melton, C., Jaenisch, R. & Blelloch, R. DGCR8 is essential for

    microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat. Genet.

    39,380385 (2007).

    35. Heo, I. et al. Lin28 mediates the terminal uridylation of let-7 precursor MicroRNA.

    Mol. Cell 32, 276284 (2008).

    36. Newman, M. A., Thomson, J. M. & Hammond, S. M. Lin-28 interaction with

    the Let-7 precursor loop mediates regulated microRNA processing. RNA 14,

    15391549 (2008).

    37. Viswanathan, S. R., Daley, G. Q. & Gregory, R. I. Selective blockade of microRNA

    processing by Lin28. Science 320, 97100 (2008).38. Anokye-Danso, F. et al. Highly efficient miRNA-mediated reprogramming of mouse

    and human somatic cells to pluripotency. Cell Stem Cell 8, 376388 (2011).

    39. Miyoshi, N.et al. Reprogramming of mouse and human cells to pluripotency using

    mature microRNAs. Cell Stem Cell 8, 633638 (2011).

    40. Samavarchi-Tehrani, P. e t al . Functional genomics reveals a BMP-driven

    mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming.

    Cell Stem Cell 7, 6477 (2010).

    1360 NATURE CELL BIOLOGY VOLUME 13 | NUMBER 11 | NOVEMBER 2011

    http://www.nature.com/naturecellbiology/http://www.nature.com/naturecellbiologyhttp://www.nature.com/reprintshttp://www.nature.com/reprintshttp://www.nature.com/reprintshttp://www.nature.com/reprintshttp://www.nature.com/naturecellbiologyhttp://www.nature.com/naturecellbiology/
  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    9/17

    DOI: 10.1038/ncb2366 M E T H O D S

    METHODSAnimals.Using recombineering, we engineered the Mir34a knockout constructwith a 6-kb homologous arm on both the 5 and 3 ends, which flanked a Kozak

    sequence, a LacZ cDNA and an FRTNeoFRT cassette. The Mir34b/c targeting

    vector contains a 6-kb homologous arm at each end, with the Mir34b/c gene

    and a Neo selection cassette flanked byloxPsites. The deleted region forMir34a

    knockoutcontainedtheMir34a pre-miRNAand 20-base pair(bp) flankingsequenceat each end. The deleted region for Mir34b/c knockout contained Mir34b and c

    pre-miRNA with a 200-bp flanking sequence at each end. To facilitate homologous

    recombination, theMir34aandMir34b/cknockout constructs were electroporatedinto Bruce4 ESCs and v6.5 ESCs, respectively. 300 Neo-resistant ESC colonies for

    each knockout construct were screened for homologous recombination. Correctly

    targeted ESCs were validated with Southern analysis using both 5 and 3 probes.

    Three independently targeted ESC clones for Mir34a and two for Mir34b/c were

    injected into blastocysts, all of which gave rise to chimaeric animals able to produce

    germline transmission.

    Oct4Gfpmice were acquired from Jackson Laboratory (catalogue no. 008214),

    and maintained on a mixed C57BL/6 and 129S4Sv/Jae genetic background. The

    ACTFLP (catalogue no. 005703) and the p21/ mice (catalogue no.003263)

    were purchased from the Jackson Laboratory. Mir34a, p53 and p21 knockout

    mice were bred and maintained on the isogenic C57BL/6J background to generate

    littermate-controlled MEFs.NCr-nu/nufemale athymic mice were purchased fromTaconic (catalogue no. NCRNU).

    Cell culture.Primary MEFs were isolated from littermate-controlled E13.5

    embryos. MEFs and ecotropic Phoenix cells were cultured in DMEM (Invitrogen,catalogue no. 11995-073) with 10% fetal bovine serum and 1% 100 penicillin

    and streptomycin (Invitrogen, catalogue no. 15140-163). iPSCs were cultured onto

    irradiated MEF feeder layers in the ESC medium, which contained Knockout

    DMEM (Invitrogen, catalogue no. 10829-018), 15% ES-grade fetal bovine serum

    (Invitrogen, catalogue no. 16141079), 2 mM l-glutamine (Invitrogen, catalogue no.

    25030-164), 1104 M MEM non-essential amino acids (Invitrogen, catalogue no.

    11140-076), 1104 M 2-mercaptoethanol (Sigma, catalogue no. M3148) and 1%

    100 penicillin and streptomycin. Feeder-free ESCs, provided by I. Lemischka,were

    cultured in the same ESC medium on gelatin-coated plates 41. Dicer/ HCT116

    cells, provided by B. Vogelstein, were cultured in McCoy 5A medium (Invitrogen,catalogue no. 16600082) with 10% fetal bovine serum and 1% 100 penicillin and

    streptomycin42.

    iPSC induction and reprogramming efficiency. Ecotropic phoenix cells weretransfected with the pMX retroviral vectors encoding mouse Oct4, Sox2, c-Myc

    and Klf4 (Addgene, catalogue nos 13366, 13367, 13375 and 13370) by the calcium

    phosphate transfection method43. One day before infection, MEFs were seeded at1.3105 cells per well in a six-well plate. MEFs were infected twice, and cultured for

    48 h before we sorted 1,000four-factor-infectedMEFs or 2,500three-factor-infected

    MEFs into each well ofa 12-wellplate.Oncethe iPSC-likecoloniesappeared,alkaline

    phosphatase staining or Oct4 staining was carried out to evaluate reprogramming

    efficiency. To establish stable iPSC lines, single iPSC-like colonies were each picked

    into one well of a 96-well plate, before expanding on irradiated MEF feeders. To

    determine reprogramming efficiency usingOct4Gfp/+ MEFs, we collected 1,000

    four-factor-infectedMEFs or 2,500 three-factor-infectedMEFs. The reprogrammediPSC colonies were scored by GFP expression.

    To determine if serial transfection of miR-34a LNA inhibitor promotes somatic

    reprogramming, we transfected miR-34a LNA oligo (Exiqon, catalogue no. 41840-

    00) and a control LNA oligo (Exiqon, catalogue no. 199004-00) into Oct4Gfp/+MEFsevery 6 daysduring the course of somatic reprogramming.The reprogrammed

    colonies were scored by GFP-positive clones with characteristic ESC morphology.

    Alkaline phosphatase and immunofluorescent staining.Alkaline phosphatasestaining was carried out using an Alkaline Phosphatase Detection Kit (Millipore,

    catalogue no. SCR004) according to the manufacturers protocol. For immunocyto-

    chemical staining, iPSCs were fixed with 4% paraformaldehyde and incubated with

    blocking solution (0.1%Triton X-100 and5% normalgoat serumin PBS) for30 min

    at room temperature. To detect the expression of pluripotency markers, cells were

    stained with antibody against Oct3/4 (1:100, Santa Cruz Biotechnology, catalogue

    no. sc-5279), SSEA1 (1:200, Santa Cruz Biotechnology, catalogue no. sc-21702) andNanog (1:100, Abcam, ab80892). To measure DNA-damage response, wild-type,

    Mir34a/ andp53/ MEFs infected with four reprogramming factors were col-

    lectedon days 3,6 and9 post-infection.After beingfixedwith4% paraformaldehyde,

    cells were stained with antibody against -H2AX (1:200, Millipore, catalogue no.

    05-636).

    Teratoma formation and chimaera generation. 1 106 wild-type, Mir34a/

    orMir34a/ Mir34b/c/ iPSCs were injected into dorsal flanks of 67-week-old

    immune-deficient nu/nu mice. Resulting teratomas were fixed in 10% formalin,

    embedded in paraffin, sectioned to 6 m and stained with haematoxylin and eosin.

    To detect differentiated cell types, we stained the cryosection slides with antibody

    against Tuj1 (1:100, Abcam,cat.no. ab7751), SMA(1:100, Abcam,cat.no. ab18147)

    and Foxa2 (1:500, Abcam, cat. no. ab40874) to detect cell lineages derived from

    ectoderm, mesoderm and endoderm, respectively.

    To determine the ability ofMir34a/ iPSCs to contribute to adult chimaeras,

    we generated Oct4Gfp/+, Mir34a/, Aw/a iPSCs on a mixed C57BL/6and 129S4Sv/Jae genetic background. Three stable Mir34a/ iPSC lines were

    established and injected into albino-C57BL/6/cBrd/cBrd/cr blastocysts at passage

    7. Chimeric blastocysts were subsequently transferred to day 2.5 pseudopregnantrecipient CD-1 females, and the percentage of chimaera contribution was estimated

    by scoring the level of coat colour pigmentation.

    iPSC differentiation assay.We triggered differentiation of wild-type andMir34a/ iPSCs with LIF withdrawal in the presence or absence of retinoic acid4,34.

    To remove the feeder cells, iPSCs cultured in the complete media with LIF were

    trypsinized and plated on 0.1% gelatin-coated plates for 1 h before the collection.

    Subsequently, feederless iPSCs were plated on gelatin-coated plates in LIF-free

    media. 1M retinoic acid was added to iPSC cultures at day 1. The differentiating

    iPSCs were collecteddailyin thecourse of 4 days, andthe expression of pluripotency

    genes was analysed by real-time PCR.

    Real-time PCR analyses.TaqMan miRNA assays (Applied Biosystems) were usedto measure the level of mature miR-34 miRNAs. The mRNA and pri-miRNA levels

    were determined by real-time PCR with SYBR (Kapa Biosystems, catalogue no.

    KK4604). The real-time PCR primers used for endogenous and exogenous Oct4,Sox2andKlf4have been described in ref. 44. The primers for total Sox2,Oct4,Klf4

    and c-Mychave been describedin ref. 45.The primers forNanoghavebeen described

    in ref. 43. The primers for pri-miR-34a, pri-miR-34b/c, p21 and Actin have been

    described in ref. 16.

    Western analyses. Annealed miR-34a, miR-34b and miR-34c oligos and con-trol siRNA oligos were each transfected into ESCs at a final concentration

    of 50 nM using Lipofectamine 2000 (Invitrogen, catalogue no. 11668027). 48 h

    post-transfection, ESCs transfected with miR-34a, miR-34b, miR-34a and con-

    trol Gfp siRNA, as well as the mock transfected control, were collected intoradioimmunoprecipitation assay buffer, 20 mM Tris at pH 7.5, 150 mM NaCl,

    1% Nonidet P-40, 0.5% sodium deoxycholate, 1 mM EDTA and 0.1% SDS with

    protease inhibitor cocktail (Roche, catalogue no. 11836153001). Cell lysates were

    subjected to western analyses. For iPSC collection, trypsinized iPSCs and feeder

    MEFs were both plated on a gelatin-coated plate. After one hour of culture,

    feeder MEFs were largely attached to the plates, whereas most iPSCs remained

    in the supernatant or lightly attached to the MEF feeders. iPSCs were thenseparated from the feeders, and subjected to western analyses. Antibodies against

    mouse Nanog (Abcam, ab80892), Sox2 (Millipore, ab5603), Oct3/4 (Santa Cruz,

    sc-5279), N-Myc (Abcam, ab18698), Klf4 (Abcam, ab26648), p21 (Santa Cruz,

    sc-6246) and p53 (Vector Laboratories, CM5) were used at 1:1,000 dilution.

    -tubulin (Sigma, clone B-5-1-2) was used at a 1:4,000 dilution as a loading

    control. The quantitation of all western analysis was carried out with Quantity

    One (Bio-Rad).

    Proliferation, apoptosis and senescence assay. Cell proliferation was measuredby the cumulative population doublings. For each passage, cells were plated at a

    concentration of 6104 cells per well of a 12-well plate. After every two days of

    culture, cells were trypsinized, counted and seeded at the same concentration.

    To determinethe percentageof apoptoticcells in infectedMEFs, cellswere stained

    with APC-annexin V (BD Pharmingen, catalogue no. 559925) in 10 mM HEPES at

    pH7.4,140mMNaCl, 2.5 mMCaCl2, then incubatedwith7-AAD(BD Pharmingen,

    catalogue no. 550475). The percentages of early-apoptotic cells, late-apoptotic cells

    and necrotic cells were determined by FACS analysis.

    The senescent cells were determined by senescence-associated -galactosidase

    (SA--Gal) staining. Infected MEFs were fixed in 2% (v/v) formaldehyde and

    0.2% (w/v) glutaraldehyde, then stained with freshly prepared staining solution

    (5mM K3Fe(CN)6; 5mM K4Fe(CN)6; 30 mM citric acid/phosphate buffer, 150 mM

    NaCl, 2 mM MgCl2; 1 mgml1 X-Gal at pH 6.0) at 37 C for 1620 h (ref. 46).

    The percentages of SA--Gal-positive cells were calculated in three independentrepresentative fields, and the total number of cells counted for each measurement

    exceeded 400.

    Luciferase assays. The mouse Sox2 3UTR containing one miR-34a-bindingsite was cloned into the PENTR/TOPO-D vector (Invitrogen, catalogue no.

    K2400-20, forward primer, 5-CACCGGAGAAGGGGAGAGATTTTCAAAG-3;

    reverse primer, 5-TACATGGATTCTCGGCAGCCTGAT-3). Similarly, a fragment

    containing the mouseNanog3 UTR and a small portion of its open reading frame

    was cloned (forward primer, 5-CACCAACCAAAGGATGAAGTGCAAGCGG-3;

    NATURE CELL BIOLOGY

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    10/17

    M E T H O D S DOI: 10.1038/ncb2366

    reverse primer, 5-TCAGGAGGCAAAGATAAGTGGGCA-3). Mutagenesis of the

    miR-34a-binding sites within these 3UTR fragments was carried out using the

    following primers:Nanogforward, 5-ACTGTAGCTGTCTTCGAAGAGGGCGTC-

    AGATCTTGTTACG-3;Nanogreverse, 5-TCTGACGCCCTCTTCGAAGACAGC-

    TACAGTGTACTTACAT-3;Sox2forward, 5-ATGTCCATTGTTTATGGCGCGC-

    CAATATATTTTTCGAGGAAAGGGTTCTTG-3; Sox2reverse, 5-TTCCTCGAA-

    AAATATATTGGCGCGCCATAAACAATGGACATTTGATTGCCA-3. Wild-typeand mutated 3UTR constructs were each cloned downstream of a firefly luciferase

    (Luc) reporter. Each Luc construct (100 ng) was transfected intoDicer-deficient

    HCT116 cells together with aRenillaluciferase construct (10ng) as a normalizationcontrol, and either 50 nM control Gfp siRNA or miR-34a mimics, respectively.

    The same experiment was carried out in feeder-free ESCs. The firefly and Renilla

    luciferase activity of each transfection was determined by dual luciferase assay

    (Promega, catalogue no. E1960) 48 or 72 h post-transfection.

    41. Schaniel, C. et al. Smarcc1/Baf155 couples self-renewal gene repression with

    changes in chromatin structure in mouse embryonic stem cells. Stem Cells 27,

    29792991 (2009).

    42. Cummins, J. M.et al. The colorectal microRNAome. Proc. Natl Acad. Sci. USA103,

    36873692 (2006).

    43. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from

    mouse embryonic and adult fibroblast cultures by defined factors. Cell 126,

    663676 (2006).

    44. Judson, R. L., Babiarz, J. E., Venere, M. & Blelloch, R. Embryonic stem cell-specific

    microRNAs promote induced pluripotency.Nat. Biotechnol. 27, 459461 (2009).

    45. Brambrink, T. et al. Sequential expression of pluripotency markers during direct

    reprogramming of mouse somatic cells. Cell Stem Cell 2, 151159 (2008).

    46. Debacq-Chainiaux, F., Erusalimsky, J. D., Campisi, J. & Toussaint, O. Protocols to

    detect senescence-associated-galactosidase (SA-betagal) activity, a biomarker of

    senescent cells in culture and in vivo. Nat. Protoc. 4, 17981806 (2009).

    NATURE CELL BIOLOGY

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    11/17

    S U P P LE M E N T A RY I N F O R M AT I O N

    WWW.NATURE.COM/NATURECELLBIOLOGY 1

    DOI: 10.1038/ncb2366

    Figure S1 mir-34deficiency significantly enhanced reprogramming without

    compromising iPSC pluripotency. a.1000 four-factor infected WT and mir-

    34a-/-MEFs (left), or WT and p53-/-MEFs (right) were collected by FACS

    sorting and plated in each well of a 12 well plate. Alkaline phosphatase-

    positive colonies with ESC-like morphology were counted 2 weeks post-

    infection. A representative image and quantitative analysis are shown out of

    five independent experiments comparing littermate-controlled WT and mir-

    34a-/-(left, **P< 0.01), as well as WT andp53-/-MEFs (right, **P< 0.01)

    for reprogramming efficiency. Error bar, standard deviation, n=4. b. Inhibition

    of miR-34aby LNA promoted somatic reprogramming.Oct4Gfp/+MEFs were

    transfected with 100nM miR-34aLNA oligos or a control LNA oligo every 6

    days during reprogramming. A representative quantitative analysis was shown

    out of two independent experiments. Error bar, standard deviation, n=3.

    Scale bar, 20mm. c.Representative images of three- (left) or four- (right)

    factor induced iPSCs derived from both WT and mir-34a-/-MEFs. Three

    independently derived iPSC lines were shown for each genotype, all of which

    exhibited ES-like morphology. OSKM, Oct4, Sox2, Klf4 and c-Myc; OSK, Oct4,

    Sox2 and Klf4. Scale bar, 100mm. d.Both WT and mir-34a-/-iPSCs induced

    by three reprogramming factors expressed pluripotency markers, exhibiting

    nuclear expression of Oct4 and Nanog, as well as membrane expression of

    SSEA1. Scale bar, 20mm. e.Three-factor induced WT (left) and mir-34a-/-

    iPSCs (right) gave rise to terminally differentiated teratomas. H&E staining

    revealed terminally differentiated tissue types from all three germ layers,

    including epidermis and neural rosette from ectoderm, cartilage and muscle

    from mesoderm, and gut-like epithelia and ciliated epithelia from endoderm.

    Scale bar, 25mm. f.Four-factor-inducedOct4-Gfp/+, mir-34a-/-iPSCs can

    be maintained in culture for 20 passages without losing the characteristic

    morphological features (phase-contrast image, left) and Oct4expression (Oct4-

    Gfpexpression, right). Scale bar, 100mm. g.Representative image of chimeric

    animals derived from three independent Oct4-Gfp/+, mir-34a-/-iPSC lines.

    a

    0

    40

    80

    120

    160

    200

    40

    60

    80

    100

    WT

    mir-34a-/-

    1000 infected

    cells/well

    WT 34a-/-

    #1

    #2

    #1

    #2

    #ofAPpositivecolonies

    #ofAPpositivecolonies

    p53-/-

    WT p53-/-

    #1

    #2

    #1

    #2

    WT

    1000 infected

    cells/well

    ** P

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    12/17

    S U P P LE M E N T A RY I N F O R M A T I O N

    2 WWW.NATURE.COM/NATURECELLBIOLOGY

    Figure S2 34a-/-; mir-34b/c-/- iPSCs functionally resemble WT iPSCs.

    a.Representative phase-contrast images of three- factor induced

    iPSCs derived from mir-34a-/-; mir-34b/c-/- MEFs. Two independently

    derived iPSC lines were shown to exhibit ES-like morphology. Scale

    bar, 100mm. b. mir-34a-/-; mir-34b/c-/- iPSCs induced by three

    reprogramming factors expressed pluripotency markers, including

    Oct4 and SSEA1. Scale bar, 20mm. c. Three-factor induced mir-34a-/-;

    mir-34b/c-/- iPSCs gave rise to differentiated teratomas. H&E staining

    revealed terminally differentiated tissue types from all three germ

    layers. Scale bar, 25mm.

    mir-34a ; mir-34b/c

    a

    Line

    #1

    Line#

    2

    -/- -/-

    b

    c

    SSEA-1 / DAPISSEA-1DAPI

    Oct4 / DAPIOct4DAPI

    Cartilage Cilliated EpitheliumNeural Rosette

    Ectoderm Mesoderm Endoderm

    mir-34a ; mir-34b/c iPSCs-/- -/-

    mir-34a;

    mir-34b/c-/- -/-

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    13/17

    S U P P LE M E N T A RY I N F O R M AT I O N

    WWW.NATURE.COM/NATURECELLBIOLOGY 3

    Figure S3 Endogenous, exogenous and total levels of reprogramming factors

    in four-factor induced iPSCs.RNAs were prepared from WT, mir-34a-/-and

    p53-/-MEFs, four-factor induced iPSCs, as well as V6.5 ES cells. The levels of

    endogenous (middle), exogenous (right) and total (left) Sox2(a), Klf4 (b) and

    Oct4(c) were each determined by real-time PCR analyses. The levels of total

    Nanog (d) and c-Myc (e) were also determined. Error bar, standard deviation, n=3.

    a

    b

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    c

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    WT

    34a

    p53

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    Foldu

    pregulation

    Fold

    upregulation

    Foldupregulation

    Fo

    ldupregulation

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    Foldu

    pregulation

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    Nanog

    d e

    Fold

    upregulation

    WT

    34a-/-

    p53

    #1 #

    2#3

    #1

    #2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    Total c-Myc

    Foldupregulation

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6 Exogenous Sox2

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    1.6Exogenous Klf4

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    EndogenousOct4

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2 Total Oct4

    0

    1

    2

    3

    4

    5

    6

    7 Total Sox2

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    -/-

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    WT

    34a

    p53

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    EndogenousSox2

    Foldupregulation

    0

    1

    2

    3

    4

    5

    6

    7

    8

    WT

    34a-/-

    p53

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    -/-

    Total Klf4

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

    EndogenousKlf4

    Fo

    ldupregulation

    Fo

    ldupregulation

    Fo

    ldu

    pregulation

    ExogenousOct4

    0

    0.5

    1

    1.5

    2

    2.5

    WT

    34a-/-

    p53-/-

    #1

    #2

    #3 #

    1#2

    #3 #

    1#2

    #3

    ES

    MEF WT iPS 34a iPS-/-

    p53 iPS-/-

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    14/17

    S U P P LE M E N T A RY I N F O R M A T I O N

    4 WWW.NATURE.COM/NATURECELLBIOLOGY

    Figure S4Deficiency of mir-34adoes not significantly impair apoptoticresponse, DNA damage response, and senescence during reprogramming.

    a. Three-factor transduced WT, mir-34a-/-and p53-/-MEFs were collected

    before infection and 9 days post-infection. The apoptotic response of

    each cell line was measured using Annexin-V and 7-amino-actinomycin

    D (7-AAD) staining. Representative FACS profiles of MEFs with three

    different genotypes were shown before and after the transduction of three

    reprogramming factors. b.A quantitative analysis was carried out to compare

    the percentage of early and late apoptotic populations of each genotype

    before infection and 9 days after infection. A representative bar graph

    was shown out of four independent experiments. Although loss of p53

    significantly impaired the apoptotic response in three-factor infected MEFs,

    mir-34a-/-MEFs exhibited no evasion of apoptosis during reprogramming.

    c.Four-factor transduced WT, mir-34a-/-and p53-/-MEFs were collected

    six days post-infection, and the DNA damage was measured by the extent

    of DNA double strand breaks usingg-H2AX (red) staining. The nuclei were

    stained by DAPI (blue). Although loss of p53significantly impaired the DNA

    damage response in the four-factor infected MEFs, both WT and mir-34a-/-

    MEFs exhibited very little g-H2AX staining. Scale bar, 20mm. d.Wildtype,

    mir-34a-/-, and p53-/-MEFs were each infected with three reprogramming

    factors. SA-b-Gal staining was performed 3 days post infection to determine

    the percentage of senescent population for each genotype. The quantitative

    analyses were shown for SA-b-Gal staining in MEFs with three different

    genotypes.

    ba

    WT mir-34a-/- p53-/-

    Annexin V

    7-AAD

    Uninfected

    OSK,

    D9p

    ostinfection

    0

    2

    4

    6

    8

    10

    12

    Uninfected D9 post infection

    WT #1

    WT #2

    34a #1

    34a #2

    p53

    Percentageofearlyand

    lateapoptoticcells(%) -/-

    -/-

    -/-

    c d

    Percen

    tageofSA--gal

    pos

    itivecells(%)

    WT#1

    WT#2

    34a#1-/-

    34a#2-/-

    p53

    0

    10

    20

    30

    40

    50

    60

    WT mir-34a p53-/- -/-

    H

    2AX/DAPI

    -/-

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    15/17

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    16/17

    S U P P LE M E N T A RY I N F O R M A T I O N

    6 WWW.NATURE.COM/NATURECELLBIOLOGY

    Figure S6 The uncropped western blots shown in the main figures.a. The

    uncropped western blots shown in Fig. 3a. with the locations of molecular

    weight markers. b. The uncropped western blots shown in Fig. 5b with the

    locations of molecular weight markers. c, d, e. The uncropped western blots

    shown in the left (c), middle (d), and right (e) panel of Fig. 5c with the

    locations of molecular weight markers. MW, molecular weight.

    a b

    c

    d e

  • 8/12/2019 Nature Cell Biology - MiR-34 MiRNAs Provide a Barrier for Somatic Cell Reprogramming

    17/17

    S U P P LE M E N T A RY I N F O R M AT I O N

    Table S1 Contributions of mir-34adeficientiPSCs to chimeric animals.

    9

    mir-34a-/- iPSC line #3 92

    mir-34a-/-

    iPSC line #1

    10

    Supplementary Table S1. Contributions of mir-34a-/-

    iPSCs to chimeric animals

    Percentage of

    Chimerism

    50% (2/3)

    60% (1/3)

    40% (1/2)

    35% (1/2)

    Genetic background for

    blastocysts

    C57BL/6J

    C57BL/6-cBrd/cBrd/Cr and

    C57BL/6J

    No. of live

    Chimeras

    3 (33%)

    2 (20%)

    10% (2/17)

    20% (2/17)

    30% (2/17)

    40% (1/17)50% (6/17)

    60% (1/17)

    70% (3/17)

    iPS cell line

    mir-34a-/-

    iPSC line #2

    # of injected

    blastocysts

    # of live

    pups

    45 17 (38%) C57BL/6-cBrd/cBrd/Cr

    20

    34