adplus-dvertising


TRANSDIFFERENCIATION 2 PDF


À propos / Télécharger Lire le document
Nom original: TRANSDIFFERENCIATION 2.pdf
Auteur: Steve

Ce fichier au format PDF 1.6 a été généré par Acrobat PDFMaker 10.1 for Word / Adobe PDF Library 10.0, et a été envoyé sur Fichier-PDF le 25/08/2014 à 16:08, depuis l'adresse IP 212.195.x.x. La présente page de téléchargement du fichier a été vue 1193 fois.
Taille du document: 8.5 Mo (32 pages).
Confidentialité: fichier public





Aperçu du document


www.sciencemag.org/content/345/6198/826/suppl/DC1

Supplementary Material for
Sequential histone-modifying activities determine the robustness of
transdifferentiation
Steven Zuryn, Arnaud Ahier, Manuela Portoso, Esther Redhouse White, Marie-Charlotte
Morin, Raphaël Margueron, Sophie Jarriault*

*Corresponding author. E-mail: sophie@igbmc.fr
Published 15 August 2014, Science 345, 826 (2014)
DOI: 10.1126/science.1255885
This PDF file includes:

Materials and Methods
Figs. S1 to S16
Table S1
Full Reference List

Materials and Methods
C. elegans strains and culture
RB1304 (wdr-5.1(ok1417)III), RB1025 (set-2(ok952)III), MT14851 (set2(n4582)III), VC912 (jmjd-3.1(gk384)X), VC936 (jmjd-3.1(gk387)X), VC31 (spat3(gk22)X), OH7317 (F21H12.1(ot86) rol-6(e187) II; ntIs1) and wild-type (N2) C.
elegans were provided by the CGC, which is funded by NIH Office of Research
Infrastructure Programs (P40 OD010440). FXO3463 (F21H12.1(tm3463)II),
FXO1905 (ash-2(tm1905)II), FXO3121 (jmjd-3.2(tm3121)X) and FXO3197 (jmjd3.3(tm3197)X) were obtained from the National BioResource Project. PFR366 (set2(bn129)III; ash-2(tm1905)II) and the wdr-5.1p::wdr-5.1::gfp integrated strain were
gifts from F. Palladino. ZR159 (utx-1(tm3118) jmjd-3.2(tm3121) jmjd-3.3(tm3197)X;
Ex[utx-1p::utx-1DD::gfp rol-6(+)]) and ZR332 (jmjd-3.1(gk384) jmjd-3.2(tm3121)
jmjd-3.3(tm3197)X) were a gift from L. Salcini. The following transgenes were used:
syIs63 [cog-1p::gfp;unc-119(+)]IV, wyIs75 [unc-47p::DsRed; exp-1p::GFP; odr1p::RFP]III, bxIs7[egl-5p::gfp; lin-15(+)], kuIs36 [egl-26p::gfp; unc-119(+)];
mcIs46 [dlg-p::dlg-1::rfp]; xnIs96 [hmr-1p::hmr-1::gfp], gaIs245 [col-34p::his24::mCherry], fpIs13 [egl-26p::mCherry::PH], mcIs17 [lin-26p::gfp::nls], edIs6
[unc-119p::gfp], ncIs2 [tag-168p::gfp], fpIs1 [ace-4p::gfp]. All three alleles of set-2
(ok952, n4582 and bn129) were examined and exhibited similar Y-to-PDA defects;
and the two F21H12.1(ot86 and tm3463) exhibited similar Y-to-PDA defects. Unless
specified, the set-2(bn129), wdr-5.1(ok1417) and jmjd-3.1(fp15) null alleles were
used. C. elegans were handled using standard methods (24).
Transgenic strains
Transgenic lines were generated by microinjection into the gonadal arms of adult
animals using standard methods (25). In the majority of cases, 50 ng·µl-1 of the odr1p::dsRed (26) co-injection plasmid was mixed with transgenic plasmids and injected.
In a few cases, 5 ng·µl-1 of the myo-2p::dsRed co-injection plasmid was used instead.
For all transgenic plasmids containing the jmjd-3.1 coding sequence, 20 ng·µl-1 was
used. For the egl-5p and col-34p driven constructs, either 10 ng·µl-1 or 20 ng·µl-1 was
injected, with consistent concentrations in matched controls. 10 ng·µl-1 of hsp16.2p::wdr-5.1, 5 ng·µl-1 of the jmjd-3.1 fosmid, and 30 ng·µl-1 of jmjd-3.1p::gfp
were injected. Two to five independent transgenic lines corresponding to each
transgene were examined for fluorescence expression and effects on Y-to-PDA
transdifferentiation, and non-transgenic counterparts were always scored alongside
transgenic animals from the same feeding plates.
Constructs
pSJ901, a derivative of the pPD95.75 plasmid with an additional multiple
cloning site in place of the gfp coding sequence was used as a base vector in which to
generate the transgenic plasmids described here, unless otherwise stated. The coding
sequences of jmjd-3.1, set-2, wdr-5.1, spr-5, his-72, and unc-3 were cloned from a
complementary DNA (cDNA) library prepared from WT (N2) RNA that had been
extracted using RNA-Solv. All final constructs were sequenced verified before use.
jmjd-3.1 plasmids
The jmjd-3.1p::jmjd-3.1::mCherry transgenic plasmid included 2.7 kb of jmjd3.1 promoter driving the full-length jmjd-3.1 isoform b (longest variant) cDNA with
mCherry fused to the C-terminus. jmjd-3.1 cDNA was amplified by PCR using the
following primers F: ATGCAAGGGAAAAATTCACACTTGAC and R:
2

ATTTTTGATGCCTACGTTCCTAAT and sub-cloned into pGEM-T easy. mCherry
was
amplified
from
pCFJ104
with
the
primers
F:
GCCTAGCTAGCATGGTCTCAAAGGGTGAAGAAG
and
R:
CGGATGCTAGCCTACTTATACAATTCATCCATGCCAC and cloned into the
Xba1 site of pGEM-T to fuse, in frame, with the C-terminus of jmjd-3.1. jmjd3.1::mCherry was then cloned into the Not1 site of pSJ901, creating pSJ926. The
jmjd-3.1 promoter was amplified from N2 genomic DNA using the following primers
F:
TCCCCCCGGGCTGTGACACGCCACGTACTCG
and
R:
TCCCCCCGGGACGATGGTAAATGCTGGGGAAC and cloned into the Xma1 site
of pSJ901, upstream of jmjd-3.1::mCherry.
Derivatives of this construct as well as others constructs modified below were
generated using standard PCR-based mutagenesis cloning techniques.
The jmjd-3.1p::jmjd-3.1(∆JmjC domain)::mCherry transgenic plasmid was
generated with the following primers F: AATATGCTCATTGGGAACCAGGC and
R: GAACAAATATCGACTTGCTTTCCGAA.
The jmjd-3.1p::jmjd-3.1(H811T, E813G)::mCherry transgenic plasmid was
generated
with
the
following
primers
F:
CAGCAACCATGGGAAACTCACTCATGGCTAGCATCAACTGGA
and
R:
TGAGTTTCCCATGGTTGCTGGAGTGCGTGATCCGATTG.
The jmjd-3.1p::jmjd-3.1(C998S, C1001S, C1025S, C1027S)::mCherry
transgenic
plasmid
was
made
with
the
following
primers
F:
CTTGCTGATCGCGTTCAACGATCTAAATACTCTACATCTG
and
R:
GACAATGTTGAAGATTTCAGATGTAGAGTATTTAGATCGTTGAAC as well
as F: CAGTAGATTCGTACCCCTTCTCCTCTCATTCTCATGTAG and R:
CCTTTTTGTATTCGTTTGCTACATGAGAATGAGAGGAGAAGG.
A putative multipartite nuclear localization signal (NLS) was identified in the
JMJD-3.1 protein sequence (amino acids: 369-717) using Motif Scan
(http://myhits.isb-sib.ch/). The jmjd-3.1p::jmjd-3.1(∆NLS)::mCherry transgenic
plasmid
was
generated
with
the
following
primers
F:
and
R:
ATTAATTATCAAATTGAATTTCGTG
TCGACCACGCTTTTTTATTGGT.
The jmjd-3.1p::jmjd-3.1(∆NLS)::mCherry::2XSV40NLS transgenic plasmid was
generated by amplifying two tandem viral SV40 NLS sequences from the p625
vector, kindly provided by Iva Greenwald (Columbia University, New York, NY),
using
the
following
primers
F:
TGGCATGGATGAATTGTATAAGTCGGCTAGAGCAATGACCGCTCCAAAGA
AGAAACG
and
R:
GACAAGTTGGTAATGGTAGCGACCGGCGCTCTATTTTTCTACCGGTACTTT
GCGTTTC. We next used this product to fuse the 2XSV40 NLS onto the C-terminus
of mCherry in the jmjd-3.1p::jmjd-3.1(∆NLS)::mCherry plasmid.
The jmjd-3.1p::jmjd-3.1::Dendra2 transgenic plasmid was generated by
replacing the mCherry in jmjd-3.1p::jmjd-3.1::mCherry with Dendra2 amplified by
PCR from pML2005 (M. Labouesse) using the following primers F:
tGATGCCTACGTTCCTAATTCTAGCATGAACCTTATTAAGGAAGATATGAG
AGT
and
R:
GCGAATTCACTAGTGATTGCTCTAGCTTACCATGCTTGACTTGGTAGAG.
The hsp-16.2p::jmjd-3.1::mCherry transgenic plasmid generated by amplifying
the hsp-16.2 promoter(27) from pPD49.78 (Andrew Fire plasmid kit) and replacing
the jmjd-3.1 promoter of jmjd-3.1p::jmjd-3.1::mCherry using PCR based cloning with
the
following
primers
F:
3

GCCTGCAGGTCGACTCTAGCTGGCTTCCTCTGAGCCCGCT
and
R:
CGATATCGAGCTCCTAGGTACCCTCCAAGGGTCCTCCTGAAAATGTTC.
The egl-5p::jmjd-3.1::mCherry transgenic plasmid was made by cutting a 6.5 kb
long egl-5 promoter sequence from pSJ676 (12) with XbaI and inserting it into the
compatible AvrII site in pSJ926 (see above).
The egl-5p::jmjd-3.1::mCherry::2XSV40NLS transgenic plasmid was made by
amplifying a 2XSV40NLS sequences from the p625 vector (see above) using the
following
primers
F:
TGGCATGGATGAATTGTATAAGTCGGCTAGAGCAATGACCGCTCCAAAGA
AGAAACG
and
R:
GACAAGTTGGTAATGGTAGCGACCGGCGCTCTATTTTTCTACCGGTACTTT
GCGTTTC. We next used this product to fuse the 2XSV40 NLS onto the C-terminus
of mCherry in the egl-5p::jmjd-3.1::mCherry plasmid.
The egl-5p::jmjd-3.1(H811T, E813G)::mCherry::2XSV40NLS transgenic
plasmid
was
generated
using
the
following
primers
F:
CAGCAACCATGGGAAACTCACTCATGGCTAGCATCAACTGGA
and
R:
TGAGTTTCCCATGGTTGCTGGAGTGCGTGATCCGATTG.
For expression in HeLA cells, jmjd-3.1 cDNA was amplified with PCR using the
following primers F: CTCGAGCGATGAACTCCACCATGAGGCC and R:
CTCGAGTTAATTAGGAACGTAGGCATCAAAAAT and cloned into the XhoI site
of pEYFP-C1 (Clontech) in frame with a C-terminal HA-tag. The same strategy was
employed for generating the jmjd-3.1(fp15) variant except that the following reverse
primer was used instead R: CTCGAGCTAGTTGATGCTAGCCATGAGTG.
Molecular dissection of the jmjd-3.1-pEYFP-C1 construct was performed with
the
following
primers
jmjd-3.1(∆760-1061)
F:
TATCCGTATGATGTTCCTGATTATGCT
and
R:
TTCGGAAAGCAAGTCGATATTTGTTC;
jmjd-3.1(∆418-1061)
F:
TATCCGTATGATGTTCCTGATTATGCT
and
R:
TCGACCACGCTTTTTTATTGGT;
jmjd-3.1(∆1-368,760-1061)
F:
AATTTCAAGAAGCAGATGAATGAAATCG
and
R:
TCGACCACGCTTTTTTATTGGT.
The gfp-tagged jmjd-3.1 fosmid (8481741786659566_B04) was kindly provided
by Mihail Sarov (Max Planck Insitute of Molecular Cell Biology and Genetics,
Dresden, Germany).
set-2 and wdr-5.1 plasmids
pSJ671 (egl-5p::MCS::SL2::mCherry) was used as a base construct for cloning
genes to be expressed in the Y cell. pSJ671 included 6.5 kb of the egl-5 promoter
fused to a delta pes-10 minimal promoter sequence as well as a SL2 bicistronic
element and mCherry (12). Full-length set-2 cDNA was amplified by PCR using the
following primers F: GCAAGGTACCATGTCCACACATGATATGAACCA and R:
GCAAGGTACCTCAATTAAGATATCCACGACACGTC and cloned into the
Acc65I site of pSJ671.
The egl-5p::set-2(Y1485A, Y1487A)::SL2::mCherry transgenic plasmid was
generated
using
the
primers
F:
GAAATCACGGCTGACGCCAAGTTCCCAATTGAAGACGAC
and
R:
GGAACTTGGCGTCAGCCGTGATTTCTTCACCCT.
The hsp-16.2p::wdr-5.1::mCherry transgenic plasmid was generated by
replacing the jmjd-3.1 cDNA of the hsp-16.2p::jmjd-3.1::mCherry construct with
wdr-5.1 cDNA via PCR based cloning using the following primers F:

4

GGGAATTCGATTCCGCTCGAGATGGATACCAGCGAAAATGCTGC and R:
GGAACCTGTCCTTTTTGTATTCGTTTGAAACATCCGAGCGCCATATATGA.
For expression in HeLA cells, wdr-5.1 cDNA was amplified with PCR using the
following primers F: CGGAATTCTATGGATACCAGCGAAAATGCTGC and R:
GCTCTAGACGAACATCCGAGCGCCATATATGAATC and cloned in between
the EcoRI and XbaI sites of pECFP-C1 (Clontech) in frame with a C-terminal FLAGtag. We used the same strategy to clone wdr-5.1 cDNA in frame with a C-terminal
HA-tag in the pEYFP-C1 (Clontech) vector except that the reverse primer
GCTCTAGAAACATCCGAGCGCCATATATGAATCTTG was used instead.
H3.3 plasmids
The egl-5p::FLAG::his-72::SL2::mCherry transgenic plasmid contains fulllength his-72 cDNA encoding the H3.3 variant, which was amplified by PCR using
the
following
primers
F:
CCGGGTACCAGAAAAAATGGACTACAAGGACGACGATGACAAGATGGCT
CGTACCAAGCAAACCG
and
R:
CCGGGTACCTTAAGCACGTTCTCCTCGGATACGTC and subcloned into
pJet1.2. The forward primer incorporates a FLAG tag sequence in frame with his-72.
FLAG::his-72 was then cut out with Acc65I and cloned into the Acc65I site of
pSJ671. Since the Y cell is postmitotic and does not undergo DNA replication during
transdifferentiation (10), we chose to over-express a mutant version of the C. elegans
H3.3 variant, the Drosophila counterpart of which has been shown to be deposited in
chromatin at all stages of the cell cycle, unlike H3, incorporation of which is tightly
restricted to the S-phase during cell division.
The egl-5p::FLAG::his-72(K4A)::SL2::mCherry transgenic plasmid was
generated with the following primers F: CAAACCGCTCGTAAATCCACCG and R:
AGCGGTACGAGCCATCTTGTCATCGTC.
spr-5 and rbr-2 plasmids
The egl-5p::spr-5::SL2::mCherry transgenic plasmid contains full length spr-5
cDNA, which was amplified by PCR using the following primers F:
GCCGGTACCATGTCGTCTGACACGGGTTCC
and
R:
GCCGGTACCCTATTCAACTGTTGGCACTAGTGGC and cloned into the Acc65I
site of pSJ671.
The col-34p::spr-5::SL2::mCherry transgenic plasmid was generated as above
except that spr-5 cDNA was cloned into the Acc65I site of pSJ6144 (after egl-27
cDNA was excised from this site), a construct in which the egl-5 promoter has been
replaced with 400 bp of the col-34 promoter(12). This promoter also drives
expression in the Y cell.
The egl-5p::spr-5(K519A)::SL2::mCherry transgenic plasmid was generated
with
the
following
primers
F:
GAAGTGTGAATGCCTGTATTCTCGAATTTGATCGAGTTTTTTGG and R:
CGAGAATACAGGCATTCACACTTCCACAGCCAATATTTCG.
The egl-5p::rbr-2::gfp::SL2::mCherry transgenic plasmid contains full length
rbr-2 cDNA, which was amplified by PCR from the pSMprbr-2::rbr-2::gfp (28)
plasmid kindly provided by Anne Brunet (Stanford University, Stanford, CA) using
the
following
primers
F:
TTGGCGCGCCAGAAAAAATGCGTGCACGTCGTCAAGAG
and
R:
AAGGCGCGCCCTATTTGTATAGTTCATCCATGCCATGTG. The resulting PCR
product was cloned into the AscI site of pSJ671.
unc-3 plasmids

5

For expression in HeLA cells, full-length unc-3 cDNA was amplified with PCR
using the following primers F: CCGCTCGAGATGAGTTTGACAGCTCCGC and R:
GCTCTAGACGAGACAGACGGGACGACG and cloned in between the XhoI and
XbaI sites of pECFP-C1 (Clontech) in frame with a C-terminal FLAG-tag.
Isolation and cloning of jmjd-3.1 and egl-27 mutants
fp15, fp11, fp13, and fp25 mutants in which PDA was absent, as indicated by the
lack of expression of the syIs63 transgenic marker, were isolated from an EMS screen
(4). The fp15 mutant was backcrossed and underwent EMS variant deep sequence
mapping as previously described (4). Deep sequencing mapping or manual
sequencing led to the identification and characterization of the three other alleles of
jmjd-3.1 (fp11, fp13, and fp25). The egl-27(fp20) mutant was isolated from the same
screen.
Staging, microscopy equipment, and fluorescent marker scoring
For fluorescence marker and immunofluorescence analysis in larvae and adults,
examination of the gonad was used to assess developmental stage. Apart from
immunofluorescence experiments, all imaging was performed on live animals
mounted on a 2% agarose pad on glass slides with 25 mM NaN3. We visualized
fluorescence using a Zeiss Z1 imager microscope and Hamamatsu ORCA-ER camera
with AxioVision40 v4.8.2.0 software. Nomarski optics was used to examine nuclear
morphology and cell position. Attachment to neighboring hindgut cells was imaged
with a Leica TCS SP5 confocal microscope and 3D rendering performed using Imaris
software (Olympus). Consistent with a role for JMJD-3.1 in buffering the Td process
against variations, we found the penetrance of jmjd-3.1 mutants to vary between 4.7%
and 18.3% at the permissive temperature of 20°C under normal growth conditions.
Immunofluorescence
The H3K4me3 antibody (CS-003-100) was obtained from Diagenode. The
H3K27me3 antibody (#6523) was received from T. Jenuwein. The LMN-1 antibody
(29) was obtained from the Developmental Studies Hybridoma Bank. The FLAG-tag
antibody was obtained from Sigma. Whole-mount immunofluorescence was
performed on synchronized L1, L2 or L3 worms using a slightly modified FinneyRuvkun procedure (10). Fluorescence was quantified using Image J 1.47v software.
Protein production and purification
Recombinant C. elegans Flag and His tagged JMJD-3.1 WT, fp15, fp11 and fp25
mutant proteins and Flag G9a protein (kindly provided by D. Reinberg) were all
produced in SF9 insect cells after infection with the corresponding baculoviruses and
purified as described previously (30). His tagged hJMJD2D (kindly provided by R.
Schneider) was expressed in BL21(DE3) cells and purified with nickel affinity gel.
V-SET (Kindly provided by R. Klose) protein was expressed in BL21(DE3) cells and
purified as previously described (31).
In vitro demethylation assays
To prepare H3-labelled methyl-histone octamers, histone methyltransferase
assays (HMT) were performed with purified V-SET or G9a proteins as previously
described (31). After the HMT reaction, the reaction mixtures were dialysed in
histone storage buffer (10 mM HEPES-KOH (pH7.5), 10 mM KCl, 10% glycerol, 0.1
mM PMSF). H3 labelled dialyzed histones were incubated with 4.5 µg (fig. S3) of
6

purified Flag and His tagged JMJD-3.1 WT, Flag and His tagged JMJD-3.1 mutant
version proteins or His tagged hJMJD2D protein in histone demethylation buffer (50
mM HEPES-KOH (pH 8), 0.023 mM Fe(NH4)2(SO4)2, 1 mM α-ketoglutarate, 2 mM
ascorbate) at 37°C for 1 h. Demethylation was analysed by the modified NASH
protocol as described in (32).
Mononucleosome isolation and immunoprecipitation
Mixed stage worms were grown on peptone enriched plates seeded with HB101
E. coli at 25°C and then washed three times in M9 buffer and resuspended in 2
volumes of nuclear isolation buffer (0.5 M Sucrose, 25 mM HEPES, pH7.5, 25 mM
KCl, 0.1 mM EDTA, 0.15 mM Spermine, 0.15 mM Spermidine, 10 mM MgCl2, 10
mM DTT, Complete protease inhibitor (Roche)). The solution was then flash frozen
in N2 (l) before being pulverized in a mortar and pestle and dounced. Debris was
pelleted with gentle centrifugation and the supernatant subsequently centrifuged at
high-speed to pellet nuclei. Nuclear extract was obtained by incubating the nuclei in
the hypotonic Buffer I (3 mM EDTA, 0.2 mM EGTA, 10 mM DTT, Complete
protease inhibitor (Roche)) for 30 min at 4°C after which chromatin was pelleted by
high-speed centrifugation. Micrococcal nuclease (MNase) treatment was performed as
follows. Chromatin was resuspended in Buffer II (10 mM PIPES pH 6.8, 5 mM
MgCl2, 1 mM CaCl2, 50 mM NaCl, 0.1 mM PMSF, 10 mM DTT, Complete protease
inhibitor (Roche)) and 4U of MNase was added and incubated at 37°C for 15, 30, 45,
or 60 min depending on the efficiency of digestion (as judged by fragment size of
DNA extracted from each reaction. see fig. S14A).
Immunoprecipitation of Mononucleosomes was performed as previously
described (20) with the following modifications. Mononucleosomes were extracted
with 10 mM Tris (pH8), 500 mM NaCl, 4 mM EDTA and dialyzed against 20 mM
HEPES-KOH (pH 8), 50 mM KCl, 1 mM EDTA, 3% Glycerol, 0.1 mM PMSF. 20 µg
of dialyzed mononucleosome were immunoaffinity purified by incubation with
H3K4me3 (A. motif 39159) or H3K27me3/me2 (clone 7B11/G5) antibodies and
protein A agarose (pre-blocked with 1 mg·ml-1 BSA and 0.1 mg·ml-1 salmon sperm
DNA) in IP buffer (50 mM HEPES-KOH (pH 8), 50 mM KCl, 50 mM NaCl, 4 mM
EDTA, 0.5% NP-40 ; 0 .1% N-lauroyl sarcosine) overnight at 4°C. After washing
three times with IP buffer, histones were eluted by boiling in SDS sample buffer,
separated by SDS-PAGE and western blot was revealed with H3K4me3 (A. motif
39159) or H3K27me3 (A. motif 39155) antibodies.
RNA interference
For the RNAi screen, we sequence-verified clones from the Ahringer library (a
gift from M. Labouesse). RNAi was performed by injecting double stranded RNA
(dsRNA) directly into worms, a more effective means of suppressing gene expression
in the Y cell when compared to performing RNAi by the feeding method. To this end,
the insert of each clone was PCR amplified using T3 and SP6 primers. For some
genes that were not present in the library or did not correspond to the correct library
position, a segment of the gene from a cDNA library made from RNA extracted from
N2 worms was PCR amplified with primers containing flanking T7 promoter
sequences. We next performed in vitro transcription using the PCR products as
templates and either T3 and SP6, or T7 RNA polymerase, depending on the flanking
sequence of each product. Single stranded RNA was allowed to anneal to form
dsRNA by gradually lowering the temperature of the sample from 65°C. rrf3(pk1426);syIs63(cog-1p::gfp) adults were microinjected with dsRNA and the
7

animals were allowed to recover at 25°C. F1 progeny derived from these adults were
scored for the presence of PDA.
Cell transfection procedures and biochemical analyses
HeLa cells were cultured with Gibco DMEM-5% FCS. Transfection was
performed with 10 µg of plasmid DNA added to 30 µl of X-tremeGENE 9 DNA
transfection reagent (Roche) in Gibco optiMEM media. α-HA-tagHRP (Cell
Signalling) and α-FLAG-tagHRP (Sigma) antibodies (Western blot) or α-HA-tag and
α-FLAG-tag coupled to agarose beads from Sigma (Immunoprecipitations) were
used. Because we found the ZnBD of JMJD-3.1 to interact nonspecifically during coimmunoprecipitation experiments, we used a truncated version of JMJD-3.1 for
physical interaction experiments that mimicked the fp15 allele.
Rescue experiments
Rescue in jmjd-3.1 mutant background: When assessing for the capability of a
construct to rescue jmjd-3.1 single mutant Y-to-PDA defects, the worms were grown
at the slightly stressful temperature of 25°C to enhance the penetrance of mutant
phenotypes.
Heat shock rescue: Animals were synchronized to appropriate stages by timed
egg pulses at 25°C. Heat shock was administered by immersion of feeding plates in a
34°C water bath followed by 20 min recovery at 20°C and then grown at 25°C until
scored. We found that we achieved the best results when hsp-16.2p::jmjd3.1::mCherry transgenic animals were heat-shocked for one hour and hsp-16.2p::wdr5.1::mCherry transgenic were heat-shocked for 30 min. Fluorescent detection of
JMJD-3.1::mCherry and WDR-5.1::mCherry fusion proteins in the Y/PDA nucleus
was achieved within 30 min after treatment at all stages tested (JMJD-3.1::mCherry:
L1 100%, n = 17; L2 100%, n = 18; L3 90%, n = 20; L4 100%, n = 22; WDR5.1::mCherry: L1 90.9%, n = 22; L2 95.2%, n = 21; L3 94.7%, n = 19; L4 100%, n =
20) and disappeared 3 h after treatment (JMJD-3.1::mCherry: L1 100%, n = 14; L2
84.6%, n = 13; L3 77.7%, n = 9; L4 86.7%, n = 15; WDR-5.1::mCherry: L1 88.8%, n
= 18; L2 89.5%, n = 19; L3 100%, n = 15; L4 93.7%, n = 16). Because heat shock
pulses were delivered at the beginning of each stage tested, the presence of nuclear
protein for each construct was restricted to the stage intended. Animals were assessed
for rescuing of transdifferentiation by examining for the presence of the PDA fate
marker cog-1p::gfp and by Nomarski observations of nuclei morphology and position.
Dendra2 conversion
Individual transgenic embryos or newly hatched L1 animals were mounted on
2% agarose pads on glass slides with M9 (embryos) or 10 mM NaN3 (L1).
Photoconversion was carried out under 40X magnification using a 405 nm filter and
an OSRAM 103 W/2 lamp with 100% output for 8 s. The Y cell was imaged before
and after photoconversion. Individuals were then recovered on feeding plates and
allowed to develop at 25°C until the L3 stage at which point they were re-imaged
with the same microscope settings. Fluorescence intensities were analyzed with Image
J 1.47v software.
Stress assays
Worms were grown for multiple generations at 20°C, synchronized to the L1
stage and exposed to 10 J/m2 of UV on NGM agar plates containing OP50 bacteria in

8

a CL-1000 Ultraviolet Crosslinker (UVP), or seeded into 96 well plates containing
M9 buffer, concentrated OP50, and either 2% DMSO (Thermo Scientific) or 0.6 mM
Paraquat (Sigma-Aldrich) (total volume 40 µl). Worms were scored for Td defects
once they reached the L3 stage or older. Control experiments were always performed
alongside each treatment in the same conditions except without irradiation or
chemical exposure. For temperature stress experiments, worms were grown at either
20°C or 25°C for at least three generations before being scored.
Statistical analyses
Two-way analysis of variance (ANOVA) (33) was used to test the interaction
effect observed between two mutant alleles. Student’s t-test was used to test the
significance between a single variable treatment versus non-treatment (e.g. transgenic
versus non-transgenic worms).

9

10

Fig. S1.
Several mutant alleles of jmjd3.1 result in a low penetrance Y-to-PDA TD
defects. (A) Logic for identifying factors required for invariant somatic
transdifferentiation (Td), through an unbiased genetic screen. The Y cell is established
as a hindgut cell ~300 minutes after the first cell cleavage in the embryo. Y forms an
integrated cross-half section of the six-cell rectal tube used for defecation (see Fig.
1A). At the end of the L1 larval stage, Y retracts from the rectal tube and changes into
a motor neuron referred to as PDA. Td is completed by the L3 stage and occurs with
100% efficiency. Worms carrying a marker of terminal PDA fate (cog-1p::gfp
integrated array [syIs63]), were subjected to ethyl methanesulfonate (EMS) treatment
and assayed for a Td defect whereby the Y cell is present during L1, but the final
PDA cell was no longer present as indicated by (B) absence of cog-1p::gfp and other
markers in PDA’s usual position (See also fig S10). Next, progeny from these animals
were assessed for Td– penetrance to identity genes specifically involved in producing
an invariant conversion outcome. We thus focused on mutants with a low penetrance
(fp15, fp11, fp13, and fp25), where Td was executed in the majority of the cases but
had lost its perfect efficiency, as these are more likely to harbor mutations in factors
that determine invariant Td, provided that it was not due to functional redundancy or
residual activity. Pink arrow head, PDA cell body; Green arrowhead, PDA axon. Red
asterisk, putative VA11 neuron; Yellow asterisk, anal sphincter muscle; Black scale
bar, 5 µm. (C) Deep mapping reveals mutations in the jmjd-3.1 gene. The causal
mutation behind the low penetrance Td– phenotype in fp15 mutants was localized to a
region of the genome on chromosome X by EMS variant mapping based deep
sequencing (4, 34). At the centre of this region, we identified a homozygous G-to-A
nucleotide substitution in the 8th exon of jmjd-3.1(F18E9.5b), which translated into a
premature stop codon (W822Stop) in the encoded protein. Alleles fp11, fp13, and
fp25 were subsequently identified as mutations in jmjd-3.1 (Fig. 1C) (D) jmjd-3.1 WT
locus rescues fp15 Y-to-PDA mutant phenotype. A PCR product (green line)
amplified from wild-type N2 genomic DNA incorporating the entire jmjd-3.1 open
reading frame as well as 5’ and 3’ cis-regulatory sequences was microinjected into
fp15 mutants and found to be sufficient to rescue Td in 5/5 independent transgenic
lines. Non-transgenic counterparts were scored alongside transgenics for each line.
Scoring was performed at the slightly stressful temperature of 25°C in order to
increase the penetrance of the Td– phenotype in fp15 mutants (see Fig. 4B). We also
demonstrated mutant phenocopy with the available jmjd-3.1(gk384) null deletion
mutant (fig. S2). Columns indicate mean of 5 independent transgenic lines. n >150,
***P<0.0001. Error bars, S.E.M.; statistical analysis was performed using Student’s ttest.

11

Fig. S2
No overlapping role for the other H3K27me3/2 demethylases in Y-to-PDA
conversion. To uncover any possible redundancy between C. elegans H3K27
demethylase homologs, we compared for Td defects genetic deletion mutants of all
H3K27me3/2 demethylases: wild type (n =156), jmjd-3.1(gk384) (n =89), jmjd3.2(tm3121) (n =81), and jmjd-3.3(tm3197) (n =89) single mutants. We also
compared for defects in jmjd-3.1(gk384) jmjd-3.2(tm3121) jmjd-3.3(tm3197) (n =116)
triple mutants and utx-1(tm3118) jmjd-3.2(tm3121) jmjd-3.3(tm3197);Ex[utx1p::UTX-1DD::GFP] (n =104) triple mutants whereby utx-1(tm3118) lethality was
rescued with a catalytically inactivated utx-1 transgene (35). All four homologs are
encoded on chromosome X and utx-1 and jmjd-3.1 are situated immediately next to
each other rendering genetic recombination, and hence the construction of a
quadruple mutant near impossible. However, we performed RNAi of utx-1 in a jmjd3.1 background (alongside a dpy-8 RNAi control, n =176) and found no significant
increase in Td defect (n =184). These results suggest that there is no redundancy
between these genes during Td. Each of the alleles used here (except fp15, which is
enzymatically null; fig. S5) contain large deletions in their coding sequences and are
most likely genetic null mutations (35). Thus, the low penetrance Td defects of
jmjd3.1 mutants reflect a role in promoting the invariant efficiency of Y cell type
conversion. Columns represent means of 3 experiments. Error bars, S.E.M.

12

Fig. S3
Mosaic analysis reveals that jmjd-3.1 acts cell autonomously. jmjd-3.1(fp15)
mutants carrying functional jmjd-3.1p::JMJD-3.1::mCherry transgenic arrays were
scored for Td– phenotypes. Transgenic (+) worms (as assessed by the presence of the
co-injection marker odr-1p::rfp, n = 146) were scored alongside non-transgenic (-)
counterparts (n = 79). In transgenic animals, the presence or absence of JMJD3.1::mCherry in the nucleus of the PDA or the Td– cell was also assessed. When
JMJD-3.1::mCherry was not detected in the cell (n = 23), the fraction of Td– events
was similar to non-transgenic worms as opposed to when JMJD-3.1::mCherry was
detected (n = 123) resulting in rescuing of Td. Columns represent means of three
independent replicates under the slightly stressful temperature of 25°C. Error bars,
S.E.M.; *P >0.05. Statistical analysis was performed with a Student’s t-test.

13

Fig. S4
Perturbing JMJD-3.1’s lysine demethylase activity and histone H3 tail
recognition disrupts its Td function. (A) Alignment of the amino acid sequences of
KDM6B family members JMJD-3.1 and UTX-1 from C. elegans (Ce) and their
orthologs in Human (Hs), and Drosophila (Dm). A green bar represents the conserved
JmjC domain and a yellow bar represents the zinc-binding domain (ZnB). Pink
asterisks indicate the amino acids mutated in order to disrupt Fe2+ coordination
(H811T, E813G), which is necessary for lysine demethylation. Yellow asterisks
indicate conserved cysteine residues mutated in order to disrupt Zn2+ coordination and
H3 tail recognition (C998S, C1001S, C1025S, C1027S) (36). Scale bar, 1 μm. (B)
Transgene schematics of constructs expressed in jmjd-3.1(fp15) mutants and
corresponding fluorescent micrographs indicate that mutated versions of JMJD-3.1
are correctly expressed and localized in the PDA/Td– cell. The cell’s outline is
indicated by a broken white line and the nucleus by a broken blue line. (C) Effect of
each transgene on Td in jmjd-3.1(fp15) mutants. n >100. Grey columns show nontransgenic counterparts scored alongside transgenic animals. Columns represent
means of at least 3 independent transgenic lines for each construct. Error bars,
S.E.M.; ***P<0.001. Statistical comparisons were made by Student’s t-test.

14

Fig. S5
Mutations in JMJD-3.1 corresponding to alleles isolated from screen abolish
H3K27me3 demethylase activity in vitro. (A) Western blot shows that an equal
amount of expressed and purified JMJD-3.1 and variant proteins were used for
demethylation assays. We also purified the human JMJD2D H3K9 demethylase as a
control for the assay. (B) Amino acid changes corresponding to Td– mutant alleles
abolish JMJD-3.1’s H3K27me3 demethylase activity on in vitro methylated
recombinant histones (see materials and methods) (C) JMJD2D had no effect on
H3K27 methylated substrates, but did demethylate H3K9 methylated substrates, as
expected. Error bars, S.E.M.; **P<0.01; ns, not significant compared to - . Statistical
comparisons were made by Student’s t-test.

15

Fig. S6
Mutation in spat-3 suppresses the Td– phenotype in jmjd-3.1 mutants. spat3(gk22) (n =151) mutants displayed no defects in Td. Td defects in jmjd-3.1(fp15) (n
=109) mutants were significantly suppressed in spat-3(gk22) jmjd-3.1(fp15) (n =118)
double mutants. Note that gk22 has been described as a partial loss-of-function allele
(7). Columns represent means of three independent replicates under the slightly
stressful temperature of 25°C. Error bars, S.E.M. **P >0.01. Statistical analysis was
performed with a Student’s t-test.

16

Fig. S7
RNA interference (RNAi) screen of genes predicted to encode histone
demethylases and methyltransferases. (A) We found that RNAi performed using
the bacterial feeding method lead to low penetrance Y-to-PDA defects of genes for
which null alleles exhibit a total loss of PDA. To increase the chances of identifying
genes leading to a low penetrance Td– phenotype, we instead injected double stranded
RNA (dsRNA) synthesized from RNAi plasmid templates of the Ahringer RNAi
library that were firstly sequence-confirmed (37). Furthermore, dsRNA was injected
into the gonads of adult rrf-3(pk1426);cog-1p::gfp mutants that are hypersensitive to
RNAi and then transferred to the slightly stressful temperature of 25°C where F1
progeny were scored under high magnification. (B) A list of genes screened and their
orthologs can be found in Table S1. Only dsRNA targeting set-2 resulted in a Td–
phenotype. RNAi against jmjd-3.1 or dpy-8 were used as positive and negative
controls respectively.

17

Fig. S8
Analysis of redundancy and alternative C. elegans H3K4 methyltransferases.
Double ash-2(tm1905);set-2(bn129) mutants exhibit no enhancement in Td defects
compared to single mutants, suggesting that Set1 complex activity is deficient in each
single mutant. Also, mutation of the MLL-related H3K4 methyltransferase set-16
(38), does not affect Td. This suggests that the sole H3K4 methyltransferase acting
during Td is the set-2 based Set1 complex. Error bars, S.E.M.; n >100.

18

Fig. S9
Overexpression of active H3K4 demethylases or H3.3K4A in the Y cell
phenocopies Set1 deficiencies. (A) Overexpression of functional antagonists of Set1
complex’s H3K4 methyltransferase activity. spr-5 (Lsd1 in humans) encodes an
amine oxidase domain containing H3K4me2 demethylase enzyme (39) and rbr-2
19

(Jarid in humans) encodes a JmjC domain containing H3K4me3 demethylase enzyme
(40). egl-5 and col-34 5’ cis-regulatory sequences were used to drive expression in the
Y cell during the L1 larval stage as controlled by a transcriptional mCherry reporter
that was separated from each demethylase protein by an SL2 sequence (middle
panels). Each promoter is expressed in Y during the L1 stage. spr-5 was catalytically
inactivated by mutating K519 into an alanine (K661A in human Lsd1) (41).
Representative photographs of transgenic L4 worms showing the effect on Td caused
by over-expressing spr-5 or rbr-5 in the Y cell during L1 (panels on right). (B)
Quantification of the effect on Td by overexpressing SPR-5, SPR-5K519A, or RBR-2 in
the Y cell during L1 (n > 100 for each transgene). (C) Quantification of the effect on
Td by overexpressing H3.3 (HIS-72) or H3.3K4A in the Y cell during L1 (n > 100 for
each transgene). (D) Representation of a nucleosome and the approximate position of
lysine 4 on H3.3. This residue was changed to an unmethylable alanine. (E)
Immunofluorescence of whole transgenic animals with a FLAG antibody and
corresponding DAPI staining indicate that wild-type and mutant H3.3 proteins are
expressed in Y and localize to DNA in the nucleus. (F) Schematics detail the
transgenic sequences constructed in order to over-express, in the Y cell during L1,
wild type and mutant versions of his-72 (H3.3) fused to an N terminal FLAG tag. SL2
mCherry was used to visualize expression of the transgene in Y (middle panels).
Representative photographs of transgenic L4 worms showing the effect on Td caused
by over-expressing H3.3 or H3.3(K4A) in the Y cell during L1 (panels on right). (GJ) Global analysis of H3K4me3 and H3K27me3 levels during Td. (G) Representative
micrograph of the tail of an L3 worm co-stained for αLMN-1 (lamin) and
αH3K4me3. (H) Representative micrographs of individual Y/PDA nuclei during the
stages depicted. Quantification was calculated as a ratio of αH3K4me3 fluorescence
intensity to αLMN-1 fluorescence intensity, which marks nuclei ubiquitously acting
as an internal control for staining. We observed no global change in H3K4me3 levels
at different stages of Td (L1, n =12; L2, n =12; L3, n =17). (I) We also did not
observe any global changes in H3K27me3 levels (L1, n =15; L2, n =17; L3, n =12).
These results are consistent with a model in which SET-1 and JMJD-3.1 activities are
addressed to relevant target loci through their association with step-specific
transcription factors (Fig. 3), rather than acting in a global genome-wide manner. (J)
As a positive control for these experiments, we performed immunofluorescence of
wdr-5.1 and set-2 mutants for αH3K4me3 and observed a marked loss in H3K4me3
levels, as expected (WT, n =20; wdr-5.1(ok1417), n =10; set-2(bn129), n =15). White
scale bars, 1 μm; yellow scale bar 30 μm; black scale bar, 25 μm. Columns, mean
values; Error bars, S.E.M.; *P < 0.05, **P<0.001. Statistical comparisons were made
by Student’s t-test.

20

Fig. S10
Set1 and jmjd-3.1 mediate different phases of cell conversion (A) Micrographs of
nuclear appearance and quantification. α, Not determined. (B) Representative
micrographs in live L1 worms (before Td is initiated) show that Y is present and
expresses correct markers in wdr-5.1(ok1417) and jmjd-3.1(fp15) mutants. However,
in each mutant in later larvae, cells that do not Td display abnormal marker
expression. In wdr-5.1(ok1417) mutants, Td– cells with fried egg nuclear morphology
have persistent expression of hindgut markers. In wdr-5.1(ok1417) and jmjd-3.1(fp15)
mutants, Td– cells with speckled nuclear morphology correctly lose expression of
hindgut markers, but do not properly activate PDA neural markers. (C) Confocal
images and 3D reconstruction of the adherens junction marker (DLG-1::RFP) shows
that Td– cells with an epithelial nuclear appearance in wdr-5.1(ok1417) mutants do
not detach from the rectal tube. In wild type animals, the Y cell detaches from the
rectal tube and another cell called P12.pa moves into position to replace it as a section
of the tube. In wdr-5.1(ok1417) mutants, P12.pa and the Td– cell both attach to the
rectal tube via adherens junctions. The white dashed line marks the ventral side of the
worm. The yellow dashed line represents the rectal slit. Adherens junctions in the 3D
reconstruction are color-coded based on the corresponding cell. In the bottom panels
are graphical illustrations of the relative positions of the cells with adherens junctions
highlighted by darker color. Scale bars, 1 μm.

21

Fig. S11
Expression patterns of Set1 complex subunits (set-1 and wdr-5.1) and jmjd-3.1.
(A) Representative confocal photographs of functional translational fusion proteins
driven by native promoters for the Set1 complex components set-2 and wdr-5.1 (42),
as well as jmjd-3.1. A double transgenic line was generated to show co-expression
patterns for WDR-5.1::GFP and JMJD-3.1::mCherry. Blue broken lines encircle the
nucleus of the Y/PDA cell at different developmental stages in live animals. Set1
complex components are present throughout development and during Td into PDA in
the Y cell nucleus. In contrast, JMJD-3.1 is absent during L1, when the Y cell
dedifferentiates, but is detected beforehand in the embryo and during redifferentiation
into PDA. In some cases we observed faint additional WDR-5.1::GFP staining in the
cytoplasm (see L1 panel). (B) Representative micrographs of a transcriptional jmjd3.1p::GFP reporter and an engineered fosmid reporter with GFP inserted at the Cterminus. Since a GFP reporter driven by jmjd-3.1 promoter is continuously expressed
from Y birth to PDA formation, the absence of JMJD-3.1 protein from the L1 nucleus
of the Y cell is likely not due to a regulation of its transcription. (C) Quantification of
the presence and subcellular localization of different jmjd-3.1 reporters in the Y/PDA
cell at different cellular stages. Columns represent mean data from ≥3 independent
transgenic lines for each construct. Error bars, S.E.M.; scale bars, 1 μm.

22

Fig. S12
Dendra2 conversion experiments. (A) jmjd-3.1(fp15) mutants injected with the
jmjd-3.1p::jmjd-3.1::Dendra2 transgene are rescued for defects in Td (at 25°C)
indicating that the JMJD-3.1 fusion protein is functional (n =139). (B) Representative
fluorescence photographs of the Y/PDA cell (nucleus is outlined by a blue broken
line), imaged with red and green channels before and after photoconversion of JMJD3.1::Dendra2 in L1 larvae (see materials and methods). The same animal was imaged
with the same microscope settings after Y underwent Td. (C) Histogram showing the
quantification of the ratio of green to red fluorescence intensity. These results and
those in Fig. 2D indicate that JMJD-3.1::Dendra2 is not present in the Y cell during
dedifferentiation (L1) and that all photoconverted protein (red channel) is degraded
prior to this step and re-synthesized thereafter. Compared to Fig. 2D, here
photoconversion was performed during dedifferentiation rather than before. This
confirmed that JMJD-3.1::Dendra2 is not present at this time and is resynthesized
after dedifferentiation. n =11. Thus JMJD3.1 levels are regulated post-translationally
in a step-specific manner. Columns represent mean values. Scale bar, 1 μm; error
bars, S.E.M. *p <0.01, **p<0.001. Statistical comparisons were made by Student’s ttest.

23

Fig. S13
Forced JMJD-3.1 nuclear localization in the Y cell during the dedifferentiation
phase of Td results in Y-to-PDA defects. (A) Quantification of the fluorescence
intensity of jmjd-3.1 and egl-5 transcriptional reporters in the Y cell during the L1
larval stage indicates that the promoter of egl-5 is stronger than that of jmjd-3.1. We
therefore used the egl-5 promoter to strongly overexpress JMJD-3.1::mCherry during
dedifferentiation. (B) Representative photograph of a transgenic worm in which
forced JMJD-3.1 nuclear localization during Y cell dedifferentiation resulted in a Td–
cell that remained attached to the rectal tube and persistently expressed the epithelial
hindgut specific marker lin-26p::GFP. Thus, ectopic JMJD-3.1 can inhibit
dedifferentiation of hindgut identity. (C) In some other cases, the Y cell underwent
dedifferentiation but converted into abnormal neurons with aberrant axonal migration.
Left panel shows a wild-type axon in a non-transgenic worm (-); the right panel
shows aberrant axonal migration in a transgenic (+) worm. Red arrowhead, cell body;
yellow arrowhead, axon. (D) Quantification of cellular phenotypes in transgenic lines.
The Td defect is largely mitigated by catalytic inactivation of JMJD-3.1. Together
with Fig 2E, where JMJD3.1 nuclear localization is modulated through the deletion of
a putative NLS and/or addition of viral SV40 NLS’, these results show that JMJD-3.1
degradation in L1 animals is nuclear-dependent, sufficient for eliminating jmjd-3.1
activity, and functionally important. 3 independent lines were assessed for each
transgene; n >75; Yellow scale bar, 50 μm; white scale bar, 1 μm; black scale bar, 5
μm. Error bars, S.E.M.

24

Fig. S14
The UNC-3 interaction domain of JMJD-3.1 is required for its functionality. (A)
Schematic of JMJD-3.1 protein fragments used for transgenic rescue experiments
(below). (B) Amino acids 418-759 of JMJD-3.1, which specifically interact with
UNC-3 (Fig. 3C), are required for the rescuing ability of a jmjd-3.1p::JMJD3.1::mCherry transgene injected into jmjd-3.1(fp15) mutants. Each JMJD-3.1 variant
was expressed normally and localized to the nucleus of PDA (panels below). ∆,
deletion of the indicated amino acids. Columns represent the means of at least three
independent transgenic lines. Scale bar, 1 μm; error bars, S.E.M; n > 100.

25

Fig. S15
Detection of bivalent mononucleosomes in C. elegans. (A) Mononucleosomes were
isolated from worms (see materials and methods). DNA was extracted (middle lane)
from mononucleosome samples and resolved on 2% agarose alongside intact
mononucleosomes (right lane). DNA size (~147 bp for extracted DNA and ~350bp
for intact mononucleosomes) indicates that samples are mainly homogenous
mononucleosomes. (B) Immunoprecipitation of mononucleosomes containing
H3K4me3 reveals the co-presence of H3K27me3. In the reciprocal experiment,
immunoprecipitation of mononucleosomes containing H3K27me3/me2 reveals the
co-presence of H3K4me3. Blots shown are representative of at least three
independent experiments.

26

Table S1.
Table S1. Predicted C. elegans histone demethylase and methyltransferase genes targeted by RNAi
Gene
Name
Mammalian ortholog
Predicted or established function

Td
defects
(n/tot.)

Histone Demethylases
Jumonji domain-containing
F18E9.5
D2021.1
F23D12.5
C29F7.6
T26A5.5
F29B9.2
F43G6.6
ZK593.4
T28F2.4
Y48B6A.11
C06H2.3
C27F2.5
F29B9.4

jmjd-3.1
utx-1
jmjd-3.2
jmjd-3.3
jhdm-1
jmjd-1.2
jmjd-1.1
rbr-2
jmjc-1
jmjd-2
jmjd-5
vps-22
psr-1

JMJD3
UTX/UTY
JMJD3/UTY/UTX
JMJD3/UTY/UTX
FBXL10/FBXL11
PHF8
JHDM1D/PHF8
JARID1A/B/C/D
C14orf169
JMJD2A/B/C
JMJD5
LOC339123
PTDSR/JMJD6

H3K27me3/me2 demethylation
H3K27me3/me2 demethylation
H3K27me3/me2 demethylation
H3K27me3/me2 demethylation
Unknown
H3K9me2 & H3K27me2 demethylation
Unknown
H3K4me3 demethylation
N/A
H3K9me3/me2 & H3K36me3/me2 demethylation
Unknown
Unknown
Unknown

5/89
0/168
0/213
0/176
0/147
0/110
0/160
0/62
0/73
N/A
0/60
0/45
0/66

H3K4me2/me1 demethylation
H3K4me2 demethylation
Unknown
CoREST complex interacts with spr-5

0/182
0/206
0/78
0/96

Polycomb group ortholog
H3K27 methylation – PRC2 component
H3K27 methylation – PRC2 component
H3K27 methylation – PRC2 component
H3K27 methylation – PRC2 component

0/36
0/66
0/16
0/65
N/A

Unknown
H3K4 methylation

2/73*
36/71*

Unknown
H3K36me2 methylation
H3K36 and H3K9 trimethylation
H3K36 and H3K9 trimethylation
Unknown
H3K4me3 methylation
Unknown
H4K20 methylation
Unknown

0/127
0/176
0/139
0/91
0/114
2/110
0/113
0/162
0/91
0/131

Amine-oxidase domain-containing
T08D10.2
Y40B1B.6
R13G10.2
D1014.8

lsd-1
spr-5
amx-1
spr-1

LSD1/KDM1
LSD1/KDM1
LSD1/KDM1
CoREST

Histone methyltransferases
Polycomb group
C50E10.4
R06A4.7
F54C1.3
C09G4.5
K07A1.12

sop-2
mes-2
mes-3
mes-6
lin-53

N/A
EZH2
N/A
EED
RBBP7/4 (RbAp46/48)

Trithorax-related group
F42A9.2
T12F5.4

lin-49
lin-59

BRD1/BRPF1
ASH1L

Set domain-containing
F25D7.3
Y2H9A.1
C43E11.3
R05D3.11
T26A5.7
C26E6.9
C07A9.7
C32D5.5
C47E8.8

blmp-1
mes-4
met-1
met-2
set-1
set-2
set-3
set-4
set-5

C49F5.2

set-6

F02D10.7
F15E6.1
F33H2.7

set-8
set-9
set-10

BLIMP-1/PRDI-BF1
NSD1
HYPB/Set2
SETDB1
SETD8
SET1/SET1A/B
SMYD4
SUV420H1
MLL5
EHMT2/EHMT1/SETD2/SU
V39H1/SUV39H2
SETD2/EHMT2/EHMT1/MLL
/SUV39H1/SUV39H2
SETD5/MLL5/ASH1L
SMYD3/SMYD2

H3K9 methylation

0/37

Unknown
Unknown
Unknown

N/A
0/260

27

F34D6.4
K09F5.5
K12H6.11
R06F6.4
R11E3.4
T12D8.1
T21B10.5
T22A3.4

set-11
set-12
set-13
set-14
set-15
set-16
set-17
set-18

W01C8.3
W01C8.4

set-19
set-20

Y24D9A.2
Y32F6A.1
Y41D4B.12
Y43F11A.5

set-21
set-22
set-23
set-24

Y43F4B.3
Y51H4A.12
Y71H2AM.8
Y73B3B.2
Y92H12BR.6
ZC8.3
C15H11.5
C41G7.4

set-25
set-26
set-27
set-28
set-29
set-30
set-31
set-32

EHMT1 & EHMT2
SETD2
SETMAR/SUV39H1/EHMT1
SMYD1/SMYD3/SMYD2
EHMT1/EHMT2/SETMAR
MLL1/2/3
PRDM11/PRDM7/PRDM9
SMYD1/SMYD3
SUV39H1/SETD2/WHSC1a
ndSCARNA22
EHMT2/SETMAR
EHMT1/EHMT2/SUV39H2/S
UV39H2/SUV39H1/SETMA
R
MLL5
SETMAR
SETD5
EHMT2/EHMT1/SUV39H2/S
UV39H1
SETD5/MLL5/ASH1L
SETD3
MLL/SETD5/MLL5
SETD4
SMYD1
MLL5/DMXL2
EHMT1/EHMT2

H3K9me1/me2 methylation & H3K27me2
methylation
H3K9 methylation
Unknown
Unknown
Unknown
H3K4 methylation
Unknown
Unknown

0/143
0/104
0/146
0/158
0/44
0/160
0/24
N/A

Unknown
H3K9 methylation

N/A
N/A

H3K9 methylation
Unknown
H3K9 methylation
Unknown

0/96
N/A
N/A
0/175

H3K9 methylation
H3K9me3 methylation
Unknown
Unknown
Unknown
H3K4me1/me2 methylation
Unknown
H3K9 methylation

N/A
N/A
N/A
N/A
0/137
N/A
N/A

*RNAi targeted against lin-49 and lin-59 resulted in gross morphological defects in the tail of animals
confounding interpretation of the Td– phenotype, which may have been caused by unspecific cellular
effects. N/A, unavailable RNAi construct.

Author Contributions: SZ and SJ designed the project, analysed data and wrote the
paper. SJ supervised the project and SZ conducted most of the experiments. AA,
ERW, MCM and MP (under supervision from RM) performed experiments and
analyzed data.

28

References and Notes
1. C. Jopling, S. Boue, J. C. Izpisua Belmonte, Dedifferentiation, transdifferentiation and
reprogramming: Three routes to regeneration. Nat. Rev. Mol. Cell Biol. 12, 79–89 (2011).
Medline doi:10.1038/nrm3043
2. E. M. Tanaka, P. W. Reddien, The cellular basis for animal regeneration. Dev. Cell 21, 172–
185 (2011). Medline doi:10.1016/j.devcel.2011.06.016
3. S. Jarriault, Y. Schwab, I. Greenwald, A Caenorhabditis elegans model for epithelial-neuronal
transdifferentiation. Proc. Natl. Acad. Sci. U.S.A. 105, 3790–3795 (2008). Medline
doi:10.1073/pnas.0712159105
4. S. Zuryn, S. Le Gras, K. Jamet, S. Jarriault, A strategy for direct mapping and identification of
mutations by whole-genome sequencing. Genetics 186, 427–430 (2010). Medline
doi:10.1534/genetics.110.119230
5. K. Agger, P. A. Cloos, J. Christensen, D. Pasini, S. Rose, J. Rappsilber, I. Issaeva, E. Canaani,
A. E. Salcini, K. Helin, UTX and JMJD3 are histone H3K27 demethylases involved in
HOX gene regulation and development. Nature 449, 731–734 (2007). Medline
doi:10.1038/nature06145
6. F. De Santa, M. G. Totaro, E. Prosperini, S. Notarbartolo, G. Testa, G. Natoli, The histone H3
lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated
gene silencing. Cell 130, 1083–1094 (2007). Medline doi:10.1016/j.cell.2007.08.019
7. O. Karakuzu, D. P. Wang, S. Cameron, MIG-32 and SPAT-3A are PRC1 homologs that
control neuronal migration in Caenorhabditis elegans. Development 136, 943–953
(2009). Medline doi:10.1242/dev.029363
8. N. J. Francis, R. E. Kingston, C. L. Woodcock, Chromatin compaction by a polycomb group
protein complex. Science 306, 1574–1577 (2004). Medline doi:10.1126/science.1100576
9. A. Shilatifard, The COMPASS family of histone H3K4 methylases: Mechanisms of regulation
in development and disease pathogenesis. Annu. Rev. Biochem. 81, 65–95 (2012).
Medline doi:10.1146/annurev-biochem-051710-134100
10. J. P. Richard, S. Zuryn, N. Fischer, V. Pavet, N. Vaucamps, S. Jarriault, Direct in vivo
cellular reprogramming involves transition through discrete, non-pluripotent steps.
Development 138, 1483–1492 (2011). Medline doi:10.1242/dev.063115
11. J. Liang, M. Wan, Y. Zhang, P. Gu, H. Xin, S. Y. Jung, J. Qin, J. Wong, A. J. Cooney, D.
Liu, Z. Songyang, Nanog and Oct4 associate with unique transcriptional repression
complexes in embryonic stem cells. Nat. Cell Biol. 10, 731–739 (2008). Medline
doi:10.1038/ncb1736
12. K. Kagias, A. Ahier, N. Fischer, S. Jarriault, Members of the NODE (Nanog and Oct4associated deacetylase) complex and SOX-2 promote the initiation of a natural cellular
reprogramming event in vivo. Proc. Natl. Acad. Sci. U.S.A. 109, 6596–6601 (2012).
Medline doi:10.1073/pnas.1117031109
13. I. Chambers, S. R. Tomlinson, The transcriptional foundation of pluripotency. Development
136, 2311–2322 (2009). Medline doi:10.1242/dev.024398

1

14. P. Kratsios, A. Stolfi, M. Levine, O. Hobert, Coordinated regulation of cholinergic motor
neuron traits through a conserved terminal selector gene. Nat. Neurosci. 15, 205–214
(2012). Medline doi:10.1038/nn.2989
15. I. Issaeva, Y. Zonis, T. Rozovskaia, K. Orlovsky, C. M. Croce, T. Nakamura, A. Mazo, L.
Eisenbach, E. Canaani, Knockdown of ALR (MLL2) reveals ALR target genes and leads
to alterations in cell adhesion and growth. Mol. Cell. Biol. 27, 1889–1903 (2007).
Medline doi:10.1128/MCB.01506-06
16. Y. W. Cho, T. Hong, S. Hong, H. Guo, H. Yu, D. Kim, T. Guszczynski, G. R. Dressler, T. D.
Copeland, M. Kalkum, K. Ge, PTIP associates with MLL3- and MLL4-containing
histone H3 lysine 4 methyltransferase complex. J. Biol. Chem. 282, 20395–20406 (2007).
Medline doi:10.1074/jbc.M701574200
17. S. A. Miller, A. C. Huang, M. M. Miazgowicz, M. M. Brassil, A. S. Weinmann, Coordinated
but physically separable interaction with H3K27-demethylase and H3K4methyltransferase activities are required for T-box protein-mediated activation of
developmental gene expression. Genes Dev. 22, 2980–2993 (2008). Medline
doi:10.1101/gad.1689708
18. B. E. Bernstein, T. S. Mikkelsen, X. Xie, M. Kamal, D. J. Huebert, J. Cuff, B. Fry, A.
Meissner, M. Wernig, K. Plath, R. Jaenisch, A. Wagschal, R. Feil, S. L. Schreiber, E. S.
Lander, A bivalent chromatin structure marks key developmental genes in embryonic
stem cells. Cell 125, 315–326 (2006). Medline doi:10.1016/j.cell.2006.02.041
19. P. Voigt, W. W. Tee, D. Reinberg, A double take on bivalent promoters. Genes Dev. 27,
1318–1338 (2013). Medline doi:10.1101/gad.219626.113
20. P. Voigt, G. LeRoy, W. J. Drury 3rd, B. M. Zee, J. Son, D. B. Beck, N. L. Young, B. A.
Garcia, D. Reinberg, Asymmetrically modified nucleosomes. Cell 151, 181–193 (2012).
Medline doi:10.1016/j.cell.2012.09.002
21. A. A. Mansour, O. Gafni, L. Weinberger, A. Zviran, M. Ayyash, Y. Rais, V. Krupalnik, M.
Zerbib, D. Amann-Zalcenstein, I. Maza, S. Geula, S. Viukov, L. Holtzman, A. Pribluda,
E. Canaani, S. Horn-Saban, I. Amit, N. Novershtern, J. H. Hanna, The H3K27
demethylase Utx regulates somatic and germ cell epigenetic reprogramming. Nature 488,
409–413 (2012). Medline doi:10.1038/nature11272
22. Y. S. Ang, S. Y. Tsai, D. F. Lee, J. Monk, J. Su, K. Ratnakumar, J. Ding, Y. Ge, H. Darr, B.
Chang, J. Wang, M. Rendl, E. Bernstein, C. Schaniel, I. R. Lemischka, Wdr5 mediates
self-renewal and reprogramming via the embryonic stem cell core transcriptional
network. Cell 145, 183–197 (2011). Medline doi:10.1016/j.cell.2011.03.003
23. W. Zhao, Q. Li, S. Ayers, Y. Gu, Z. Shi, Q. Zhu, Y. Chen, H. Y. Wang, R. F. Wang, Jmjd3
inhibits reprogramming by upregulating expression of INK4a/Arf and targeting PHF20
for ubiquitination. Cell 152, 1037–1050 (2013). Medline doi:10.1016/j.cell.2013.02.006
24. S. Brenner, The genetics of Caenorhabditis elegans. Genetics 77, 71–94 (1974). Medline
25. C. C. Mello, J. M. Kramer, D. Stinchcomb, V. Ambros, Efficient gene transfer in C. elegans:
Extrachromosomal maintenance and integration of transforming sequences. EMBO J. 10,
3959–3970 (1991). Medline

2

26. M. P. Klassen, K. Shen, Wnt signaling positions neuromuscular connectivity by inhibiting
synapse formation in C. elegans. Cell 130, 704–716 (2007). Medline
doi:10.1016/j.cell.2007.06.046
27. E. G. Stringham, D. K. Dixon, D. Jones, E. P. M. Candido, Temporal and spatial expression
patterns of the small heat shock (hsp16) genes in transgenic Caenorhabditis elegans. Mol.
Biol. Cell 3, 221–233 (1992). Medline doi:10.1091/mbc.3.2.221
28. E. L. Greer, T. J. Maures, A. G. Hauswirth, E. M. Green, D. S. Leeman, G. S. Maro, S. Han,
M. R. Banko, O. Gozani, A. Brunet, Members of the H3K4 trimethylation complex
regulate lifespan in a germline-dependent manner in C. elegans. Nature 466, 383–387
(2010). Medline doi:10.1038/nature09195
29. G. Hadwiger, S. Dour, S. Arur, P. Fox, M. L. Nonet, A monoclonal antibody toolkit for C.
elegans. PLOS One 5, e10161 (2010). Medline doi:10.1371/journal.pone.0010161
30. R. Margueron, N. Justin, K. Ohno, M. L. Sharpe, J. Son, W. J. Drury 3rd, P. Voigt, S. R.
Martin, W. R. Taylor, V. De Marco, V. Pirrotta, D. Reinberg, S. J. Gamblin, Role of the
polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–
767 (2009). Medline doi:10.1038/nature08398
31. H. Wei, M. M. Zhou, Dimerization of a viral SET protein endows its function. Proc. Natl.
Acad. Sci. U.S.A. 107, 18433–18438 (2010). Medline doi:10.1073/pnas.1009911107
32. Y. Tsukada, J. Fang, H. Erdjument-Bromage, M. E. Warren, C. H. Borchers, P. Tempst, Y.
Zhang, Histone demethylation by a family of JmjC domain-containing proteins. Nature
439, 811–816 (2006). Medline doi:10.1038/nature04433
33. B. K. Slinker, The statistics of synergism. J. Mol. Cell. Cardiol. 30, 723–731 (1998).
Medline doi:10.1006/jmcc.1998.0655
34. S. Zuryn, S. Jarriault, Deep sequencing strategies for mapping and identifying mutations
from genetic screens. Worm 2, e25081 (2013). Medline doi:10.4161/worm.25081
35. J. Vandamme, G. Lettier, S. Sidoli, E. Di Schiavi, O. Nørregaard Jensen, A. E. Salcini, The
C. elegans H3K27 demethylase UTX-1 is essential for normal development, independent
of its enzymatic activity. PLOS Genet. 8, e1002647 (2012). Medline
doi:10.1371/journal.pgen.1002647
36. T. Sengoku, S. Yokoyama, Structural basis for histone H3 Lys 27 demethylation by
UTX/KDM6A. Genes Dev. 25, 2266–2277 (2011). Medline doi:10.1101/gad.172296.111
37. A. G. Fraser, R. S. Kamath, P. Zipperlen, M. Martinez-Campos, M. Sohrmann, J. Ahringer,
Functional genomic analysis of C. elegans chromosome I by systematic RNA
interference. Nature 408, 325–330 (2000). Medline doi:10.1038/35042517
38. K. Fisher, S. M. Southall, J. R. Wilson, G. B. Poulin, Methylation and demethylation
activities of a C. elegans MLL-like complex attenuate RAS signalling. Dev. Biol. 341,
142–153 (2010). Medline doi:10.1016/j.ydbio.2010.02.023
39. D. J. Katz, T. M. Edwards, V. Reinke, W. G. Kelly, A C. elegans LSD1 demethylase
contributes to germline immortality by reprogramming epigenetic memory. Cell 137,
308–320 (2009). Medline doi:10.1016/j.cell.2009.02.015

3

40. J. Christensen, K. Agger, P. A. Cloos, D. Pasini, S. Rose, L. Sennels, J. Rappsilber, K. H.
Hansen, A. E. Salcini, K. Helin, RBP2 belongs to a family of demethylases, specific for
tri-and dimethylated lysine 4 on histone 3. Cell 128, 1063–1076 (2007). Medline
doi:10.1016/j.cell.2007.02.003
41. M. G. Lee, C. Wynder, N. Cooch, R. Shiekhattar, An essential role for CoREST in
nucleosomal histone 3 lysine 4 demethylation. Nature 437, 432–435 (2005). Medline
42. T. Simonet, R. Dulermo, S. Schott, F. Palladino, Antagonistic functions of SET-2/SET1 and
HPL/HP1 proteins in C. elegans development. Dev. Biol. 312, 367–383 (2007). Medline
doi:10.1016/j.ydbio.2007.09.035

4


Aperçu du document TRANSDIFFERENCIATION 2.pdf - page 1/32

 
TRANSDIFFERENCIATION 2.pdf - page 2/32
TRANSDIFFERENCIATION 2.pdf - page 3/32
TRANSDIFFERENCIATION 2.pdf - page 4/32
TRANSDIFFERENCIATION 2.pdf - page 5/32
TRANSDIFFERENCIATION 2.pdf - page 6/32
 







Télécharger le fichier (PDF)





Documents récents du même auteur


HENRI   19 juillet 2016
IDEE   19 juillet 2016
CAFE   19 juillet 2016
PEAU   19 juillet 2016
NOVLANGUE   19 juillet 2016
AVC   19 juillet 2016
DAMIEN   19 juillet 2016
JET LAG   19 juillet 2016
A Putative Human Pheromon1   19 juillet 2016
JORDI LLORENS   19 juillet 2016
reactive and proactive aggression   17 juillet 2016
Holy Warriors Exploring the Psychological Processe   17 juillet 2016
baillement orl (1)   6 juillet 2016
Bienfait de la randonnée   6 juin 2016
pleine lune et sommeil   21 avril 2016
Past experience   18 avril 2016
GENERATION AND TRANSPLANTATION NEURONES 3D   25 mars 2016
Musiq&Parf LaCelle 05-03-2016   6 mars 2016
HFEA approval for new   27 février 2016
CRISP CHINE 2   27 février 2016

Sur le même sujet..







Ce fichier a été mis en ligne par un utilisateur du site Fichier PDF. Identifiant unique du document: 00260662.
⚠️  Signaler un contenu illicite
Pour plus d'informations sur notre politique de lutte contre la diffusion illicite de contenus protégés par droit d'auteur, consultez notre page dédiée.