リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「ZEB2 and MEIS1 independently contribute to hematopoiesis via early hematopoietic enhancer activation」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

ZEB2 and MEIS1 independently contribute to hematopoiesis via early hematopoietic enhancer activation

Kitagawa, Yohko Ikenaka, Akihiro Sugimura, Ryohichi Niwa, Akira Saito, Megumu K. 京都大学 DOI:10.1016/j.isci.2023.107893

2023.10.20

概要

Cell differentiation is achieved by acquiring a cell type-specific transcriptional program and epigenetic landscape. While the cell type-specific patterning of enhancers has been shown to precede cell fate decisions, it remains unclear how regulators of these enhancers are induced to initiate cell specification and how they appropriately restrict cells that differentiate. Here, using embryonic stem cell–derived hematopoietic cell differentiation cultures, we show the activation of some hematopoietic enhancers during arterialization of hemogenic endothelium, a prerequisite for hematopoiesis. We further reveal that ZEB2, a factor involved in the transcriptional regulation of arterial endothelial cells, and a hematopoietic regulator MEIS1 are independently required for activating these enhancers. Concomitantly, ZEB2 or MEIS1 deficiency impaired hematopoietic cell development. These results suggest that multiple regulators expressed from an earlier developmental stage non-redundantly contribute to the establishment of hematopoietic enhancer landscape, thereby restricting cell differentiation despite the unrestricted expression of these regulators to hematopoietic cells.

この論文で使われている画像

参考文献

1. Bulger, M., and Groudine, M. (2011).

Functional and mechanistic diversity of distal

transcription enhancers. Cell 144, 327–339.

https://doi.org/10.1016/j.cell.2011.01.024.

2. Hnisz, D., Abraham, B.J., Lee, T.I., Lau, A.,

Saint-Andre´, V., Sigova, A.A., Hoke, H.A., and

Young, R.A. (2013). Super-Enhancers in the

Control of Cell Identity and Disease. Cell 155,

934–947. https://doi.org/10.1016/j.cell.2013.

09.053.

3. Heinz, S., Romanoski, C.E., Benner, C., and

Glass, C.K. (2015). The selection and function

of cell type-specific enhancers. Nat. Rev. Mol.

Cell Biol. 16, 144–154. https://doi.org/10.

1038/nrm3949.

4. Spitz, F., and Furlong, E.E.M. (2012).

Transcription factors: from enhancer binding

to developmental control. Nat. Rev. Genet.

13, 613–626. https://doi.org/10.1038/

nrg3207.

5. Davis, R.L., Weintraub, H., and Lassar, A.B.

(1987). Expression of a single transfected

cDNA converts fibroblasts to myoblasts. Cell

51, 987–1000. https://doi.org/10.1016/00928674(87)90585-X.

6. Kim, H.S., Tan, Y., Ma, W., Merkurjev, D.,

Destici, E., Ma, Q., Suter, T., Ohgi, K.,

Friedman, M., Skowronska-Krawczyk, D., and

Rosenfeld, M.G. (2018). Pluripotency factors

functionally premark cell-type-restricted

enhancers in ES cells. Nature 556, 510–514.

https://doi.org/10.1038/s41586-018-0048-8.

7. Medvinsky, A., and Dzierzak, E. (1996).

Definitive Hematopoiesis Is Autonomously

Initiated by the AGM Region. Cell 86,

897–906. https://doi.org/10.1016/S00928674(00)80165-8.

8. de Bruijn, M.F., Speck, N.A., Peeters, M.C.,

and Dzierzak, E. (2000). Definitive

hematopoietic stem cells first develop within

the major arterial regions of the mouse

embryo. EMBO J. 19, 2465–2474. https://doi.

org/10.1093/emboj/19.11.2465.

9. Clements, W.K., and Traver, D. (2013).

Signalling pathways that control vertebrate

haematopoietic stem cell specification. Nat.

Rev. Immunol. 13, 336–348. https://doi.org/

10.1038/nri3443.

10. Nakagawa, M., Ichikawa, M., Kumano, K.,

Goyama, S., Kawazu, M., Asai, T., Ogawa, S.,

Kurokawa, M., and Chiba, S. (2006). AML1/

Runx1 rescues Notch1-null mutation-induced

deficiency of para-aortic splanchnopleural

hematopoiesis. Blood 108, 3329–3334.

https://doi.org/10.1182/blood-2006-04019570.

11. Robert-Moreno, A., Guiu, J., Ruiz-Herguido,

C., Lo´pez, M.E., Ingle´s-Esteve, J., Riera, L.,

Tipping, A., Enver, T., Dzierzak, E., Gridley, T.,

et al. (2008). Impaired embryonic

haematopoiesis yet normal arterial

development in the absence of the Notch

ligand Jagged1. EMBO J. 27, 1886–1895.

https://doi.org/10.1038/emboj.2008.113.

12. Burns, C.E., Galloway, J.L., Smith, A.C.H.,

Keefe, M.D., Cashman, T.J., Paik, E.J.,

Mayhall, E.A., Amsterdam, A.H., and Zon, L.I.

(2009). A genetic screen in zebrafish defines a

hierarchical network of pathways required for

hematopoietic stem cell emergence. Blood

113, 5776–5782. https://doi.org/10.1182/

blood-2008-12-193607.

13. Monteiro, R., Pinheiro, P., Joseph, N.,

Peterkin, T., Koth, J., Repapi, E., Bonkhofer,

F., Kirmizitas, A., and Patient, R. (2016).

Transforming Growth Factor b Drives

Hemogenic Endothelium Programming and

the Transition to Hematopoietic Stem Cells.

Dev. Cell 38, 358–370. https://doi.org/10.

1016/j.devcel.2016.06.024.

14. Jung, H.S., Uenishi, G., Park, M.A., Liu, P.,

Suknuntha, K., Raymond, M., Choi, Y.J.,

Thomson, J.A., Ong, I.M., and Slukvin, I.I.

(2021). SOX17 integrates HOXA and arterial

programs in hemogenic endothelium to drive

definitive lympho-myeloid hematopoiesis.

Cell Rep. 34, 108758. https://doi.org/10.

1016/j.celrep.2021.108758.

15. Zeng, Y., He, J., Bai, Z., Li, Z., Gong, Y., Liu, C.,

Ni, Y., Du, J., Ma, C., Bian, L., et al. (2019).

Tracing the first hematopoietic stem cell

generation in human embryo by single-cell

RNA sequencing. Cell Res. 29, 881–894.

https://doi.org/10.1038/s41422-019-0228-6.

16. Niwa, A., Heike, T., Umeda, K., Oshima, K.,

Kato, I., Sakai, H., Suemori, H., Nakahata, T.,

and Saito, M.K. (2011). A novel serum-free

monolayer culture for orderly hematopoietic

differentiation of human pluripotent cells via

mesodermal progenitors. PLoS One 6,

e22261. https://doi.org/10.1371/journal.

pone.0022261.

17. Ohta, R., Sugimura, R., Niwa, A., and Saito,

M.K. (2019). Hemogenic Endothelium

Differentiation from Human Pluripotent Stem

Cells in A Feeder- and Xeno-free Defined

Condition. J. Vis. Exp. https://doi.org/10.

3791/59823.

18. Choi, K.-D., Vodyanik, M.A., Togarrati, P.P.,

Suknuntha, K., Kumar, A., Samarjeet, F.,

Probasco, M.D., Tian, S., Stewart, R.,

Thomson, J.A., and Slukvin, I.I. (2012).

Identification of the hemogenic endothelial

progenitor and its direct precursor in human

pluripotent stem cell differentiation cultures.

Cell Rep. 2, 553–567. https://doi.org/10.1016/

j.celrep.2012.08.002.

19. Ditadi, A., Sturgeon, C.M., Tober, J., Awong,

G., Kennedy, M., Yzaguirre, A.D., Azzola, L.,

Ng, E.S., Stanley, E.G., French, D.L., et al.

(2015). HUMAN DEFINITIVE HAEMOGENIC

ENDOTHELIUM AND ARTERIAL VASCULAR

ENDOTHELIUM REPRESENT DISTINCT

LINEAGES. Nat. Cell Biol. 17, 580–591.

https://doi.org/10.1038/ncb3161.

20. Uenishi, G.I., Jung, H.S., Kumar, A., Park,

M.A., Hadland, B.K., McLeod, E., Raymond,

M., Moskvin, O., Zimmerman, C.E., Theisen,

D.J., et al. (2018). NOTCH signaling specifies

21.

22.

23.

24.

25.

26.

27.

28.

29.

arterial-type definitive hemogenic

endothelium from human pluripotent stem

cells. Nat. Commun. 9, 1828. https://doi.org/

10.1038/s41467-018-04134-7.

Nottingham, W.T., Jarratt, A., Burgess, M.,

Speck, C.L., Cheng, J.-F., Prabhakar, S.,

Rubin, E.M., Li, P.-S., Sloane-Stanley, J.,

Kong-a-San, J., and de Bruijn, M.F.T.R. (2007).

Runx1-mediated hematopoietic stem-cell

emergence is controlled by a Gata/Ets/SCLregulated enhancer. Blood 110, 4188–4197.

https://doi.org/10.1182/blood-2007-07100883.

Xiang, P., Wei, W., Lo, C., Rosten, P., Hou, J.,

Hoodless, P.A., Bilenky, M., Bonifer, C.,

Cockerill, P.N., Kirkpatrick, A., et al. (2014).

Delineating MEIS1 cis-regulatory elements

active in hematopoietic cells. Leukemia 28,

433–436. https://doi.org/10.1038/leu.

2013.287.

Shlyueva, D., Stampfel, G., and Stark, A.

(2014). Transcriptional enhancers: from

properties to genome-wide predictions. Nat.

Rev. Genet. 15, 272–286. https://doi.org/10.

1038/nrg3682.

Verschueren, K., Remacle, J.E., Collart, C.,

Kraft, H., Baker, B.S., Tylzanowski, P., Nelles,

L., Wuytens, G., Su, M.-T., Bodmer, R., et al.

(1999). SIP1, a Novel Zinc Finger/

Homeodomain Repressor, Interacts with

Smad Proteins and Binds to 50 -CACCT

Sequences in Candidate Target Genes.

J. Biol. Chem. 274, 20489–20498. https://doi.

org/10.1074/jbc.274.29.20489.

Challen, G.A., and Goodell, M.A. (2010).

Runx1 isoforms show differential expression

patterns during hematopoietic development

but have similar functional effects in adult

hematopoietic stem cells. Exp. Hematol. 38,

403–416. https://doi.org/10.1016/j.exphem.

2010.02.011.

Wang, H., Liu, C., Liu, X., Wang, M., Wu, D.,

Gao, J., Su, P., Nakahata, T., Zhou, W., Xu, Y.,

et al. (2018). MEIS1 Regulates Hemogenic

Endothelial Generation, Megakaryopoiesis,

and Thrombopoiesis in Human Pluripotent

Stem Cells by Targeting TAL1 and FLI1. Stem

Cell Rep. 10, 447–460. https://doi.org/10.

1016/j.stemcr.2017.12.017.

Arner, E., Daub, C.O., Vitting-Seerup, K.,

Andersson, R., Lilje, B., Drabløs, F.,

Lennartsson, A., Ro¨nnerblad, M.,

Hrydziuszko, O., Vitezic, M., et al. (2015).

Transcribed enhancers lead waves of

coordinated transcription in transitioning

mammalian cells. Science 347, 1010–1014.

https://doi.org/10.1126/science.1259418.

Kim, Y.W., Lee, S., Yun, J., and Kim, A. (2015).

Chromatin looping and eRNA transcription

precede the transcriptional activation of gene

in the b-globin locus. Biosci. Rep. 35, e00179.

https://doi.org/10.1042/BSR20140126.

Menendez, P., Navarro-Montero, O., RamosMejia, V., Ayllo´n, V., Stanley, E., Elefanty, A.,

and Real, P. (2016). RUNX1C regulates

iScience 26, 107893, October 20, 2023

15

iScience

ll

Article

OPEN ACCESS

30.

31.

32.

33.

34.

35.

36.

16

hematopoietic specification of human

embryonic stem cells. Exp. Hematol. 44, S89.

https://doi.org/10.1016/j.exphem.2016.

06.186.

Navarro-Montero, O., Ayllon, V., Lamolda,

M., Lo´pez-Onieva, L., Montes, R., Bueno, C.,

Ng, E., Guerrero-Carreno, X., Romero, T.,

Romero-Moya, D., et al. (2017). RUNX1c

Regulates Hematopoietic Differentiation of

Human Pluripotent Stem Cells Possibly in

Cooperation with Proinflammatory Signaling.

Stem Cell. 35, 2253–2266. https://doi.org/10.

1002/stem.2700.

Lichtinger, M., Ingram, R., Hannah, R., Mu¨ller,

D., Clarke, D., Assi, S.A., Lie-A-Ling, M.,

Noailles, L., Vijayabaskar, M.S., Wu, M., et al.

(2012). RUNX1 reshapes the epigenetic

landscape at the onset of haematopoiesis.

EMBO J. 31, 4318–4333. https://doi.org/10.

1038/emboj.2012.275.

Park, M.A., Kumar, A., Jung, H.S., Uenishi, G.,

Moskvin, O.V., Thomson, J.A., and Slukvin, I.I.

(2018). Activation of Arterial Program Drives

Development of Definitive Hemogenic

Endothelium with Lymphoid Potential. Cell

Rep. 23, 2467–2481. https://doi.org/10.1016/

j.celrep.2018.04.092.

Goossens, S., Janzen, V., Bartunkova, S.,

Yokomizo, T., Drogat, B., Crisan, M., Haigh,

K., Seuntjens, E., Umans, L., Riedt, T., et al.

(2011). The EMT regulator Zeb2/Sip1 is

essential for murine embryonic

hematopoietic stem/progenitor cell

differentiation and mobilization. Blood 117,

5620–5630. https://doi.org/10.1182/blood2010-08-300236.

Wang, L., Menendez, P., Shojaei, F., Li, L.,

Mazurier, F., Dick, J.E., Cerdan, C., Levac, K.,

and Bhatia, M. (2005). Generation of

hematopoietic repopulating cells from

human embryonic stem cells independent of

ectopic HOXB4 expression. J. Exp. Med. 201,

1603–1614. https://doi.org/10.1084/jem.

20041888.

Azcoitia, V., Aracil, M., Martı´nez-A, C., and

Torres, M. (2005). The homeodomain protein

Meis1 is essential for definitive

hematopoiesis and vascular patterning in the

mouse embryo. Dev. Biol. 280, 307–320.

https://doi.org/10.1016/j.ydbio.2005.01.004.

Cvejic, A., Serbanovic-Canic, J., Stemple,

D.L., and Ouwehand, W.H. (2011). The role of

meis1 in primitive and definitive

iScience 26, 107893, October 20, 2023

37.

38.

39.

40.

41.

42.

43.

44.

hematopoiesis during zebrafish

development. Haematologica 96, 190–198.

https://doi.org/10.3324/haematol.2010.

027698.

Doss, M.X., and Sachinidis, A. (2019). Current

Challenges of iPSC-Based Disease Modeling

and Therapeutic Implications. Cells 8, 403.

https://doi.org/10.3390/cells8050403.

Chen, B., Teng, J., Liu, H., Pan, X., Zhou, Y.,

Huang, S., Lai, M., Bian, G., Mao, B., Sun, W.,

et al. (2017). Inducible overexpression of

RUNX1b/c in human embryonic stem cells

blocks early hematopoiesis from mesoderm.

J. Mol. Cell Biol. 9, 262–273. https://doi.org/

10.1093/jmcb/mjx032.

Riddell, J., Gazit, R., Garrison, B.S., Guo, G.,

Saadatpour, A., Mandal, P.K., Ebina, W.,

Volchkov, P., Yuan, G.-C., Orkin, S.H., and

Rossi, D.J. (2014). Reprogramming

committed murine blood cells to induced

hematopoietic stem cells with defined

factors. Cell 157, 549–564. https://doi.org/10.

1016/j.cell.2014.04.006.

Yanagimachi, M.D., Niwa, A., Tanaka, T.,

Honda-Ozaki, F., Nishimoto, S., Murata, Y.,

Yasumi, T., Ito, J., Tomida, S., Oshima, K.,

et al. (2013). Robust and highly-efficient

differentiation of functional monocytic cells

from human pluripotent stem cells under

serum- and feeder cell-free conditions. PLoS

One 8, e59243. https://doi.org/10.1371/

journal.pone.0059243.

Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto,

R., Habib, N., Hsu, P.D., Wu, X., Jiang, W.,

Marraffini, L.A., and Zhang, F. (2013).

Multiplex Genome Engineering Using

CRISPR/Cas Systems. Science 339, 819–823.

https://doi.org/10.1126/science.1231143.

Martin, M. (2011). Cutadapt removes adapter

sequences from high-throughput sequencing

reads. EMBnet. J. 17, 10–12. https://doi.org/

10.14806/ej.17.1.200.

Kim, D., Langmead, B., and Salzberg, S.L.

(2015). HISAT: a fast spliced aligner with low

memory requirements. Nat. Methods 12,

357–360. https://doi.org/10.1038/

nmeth.3317.

Liao, Y., Smyth, G.K., and Shi, W. (2014).

featureCounts: an efficient general purpose

program for assigning sequence reads to

genomic features. Bioinformatics 30,

923–930. https://doi.org/10.1093/

bioinformatics/btt656.

45. Vera Alvarez, R., Pongor, L.S., Marin˜oRamı´rez, L., and Landsman, D. (2019).

TPMCalculator: one-step software to quantify

mRNA abundance of genomic features.

Bioinformatics 35, 1960–1962. https://doi.

org/10.1093/bioinformatics/bty896.

46. Gu, Z., Eils, R., and Schlesner, M. (2016).

Complex heatmaps reveal patterns and

correlations in multidimensional genomic

data. Bioinformatics 32, 2847–2849. https://

doi.org/10.1093/bioinformatics/btw313.

47. Langmead, B., and Salzberg, S.L. (2012). Fast

gapped-read alignment with Bowtie 2. Nat.

Methods 9, 357–359. https://doi.org/10.

1038/nmeth.1923.

48. Zhang, Y., Liu, T., Meyer, C.A., Eeckhoute, J.,

Johnson, D.S., Bernstein, B.E., Nusbaum, C.,

Myers, R.M., Brown, M., Li, W., and Liu, X.S.

(2008). Model-based Analysis of ChIP-Seq

(MACS). Genome Biol. 9, R137. https://doi.

org/10.1186/gb-2008-9-9-r137.

49. Heinz, S., Benner, C., Spann, N., Bertolino, E.,

Lin, Y.C., Laslo, P., Cheng, J.X., Murre, C.,

Singh, H., and Glass, C.K. (2010). Simple

combinations of lineage-determining

transcription factors prime cis-regulatory

elements required for macrophage and B cell

identities. Mol. Cell 38, 576–589. https://doi.

org/10.1016/j.molcel.2010.05.004.

50. Yu, G., Wang, L.-G., Han, Y., and He, Q.-Y.

(2012). clusterProfiler: an R Package for

Comparing Biological Themes Among Gene

Clusters. OMICS A J. Integr. Biol. 16,

284–287. https://doi.org/10.1089/omi.

2011.0118.

51. Guo, J., Ma, D., Huang, R., Ming, J., Ye, M.,

Kee, K., Xie, Z., and Na, J. (2017). An inducible

CRISPR-ON system for controllable gene

activation in human pluripotent stem cells.

Protein Cell 8, 379–393. https://doi.org/10.

1007/s13238-016-0360-8.

52. Butler, A., Hoffman, P., Smibert, P., Papalexi,

E., and Satija, R. (2018). Integrating single-cell

transcriptomic data across different

conditions, technologies, and species. Nat.

Biotechnol. 36, 411–420. https://doi.org/10.

1038/nbt.4096.

53. Binns, D., Dimmer, E., Huntley, R., Barrell, D.,

O’Donovan, C., and Apweiler, R. (2009).

QuickGO: a web-based tool for Gene

Ontology searching. Bioinformatics 25, 3045–

3046. https://doi.org/10.1093/

bioinformatics/btp536.

iScience

ll

Article

OPEN ACCESS

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE

SOURCE

IDENTIFIER

Antibodies

Anti-H3K27ac

GeneTex

GTX60815; RRID: AB_2888004

Anti-H3K4me1

ActiveMotif

39297; RRID: AB_2615075

Anti-H3K27me3

Millipore

07-449; RRID: AB_310624

Anti-SIP1 (ZEB2)

Bethyl Laboratories

A302474A; RRID: AB_1944271

Anti-MEIS1

Abcam

ab19867; RRID: AB_776272

Anti-NOTCH1

Cell Signaling Technology

3608S; RRID: AB_2153354

BV421 anti-CD34

BD Biosicneces

744904; RRID: AB_2742572

APC anti-CD309

BIOLEGEND

359916; RRID: AB_2565928

PECy7 anti-CD45

BIOLEGEND

304016; RRID: AB_314404

FITC anti-CD73

BIOLEGEND

344015; RRID: AB_2561808

PE anti-DLL4

Miltenyi Biotec

130-096-567; RRID: AB_10831209

APC anti-CD184 (CXCR4)

BD Biosicneces

555976; RRID: AB_398616

Anti-b-Actin

Cell Signaling Technology

5125S; RRID: AB_1903890

Anti-rabbit-HRP

Cell Signaling Technology

7074S; RRID: AB_2099233

BMP-8

R&D

314-BP-010

VEGF

R&D

293-VE-010

Chemicals, peptides, and recombinant proteins

CHIR99021

Wako

038-23101

SCF

R&D

255-SC/CF

bFGF

Wako

064-05381

SB431542

Wako

031-24291

TPO

R&D

288-TPN

FLT3L

R&D

308-GMP

FP6

R&D

8954-SR

iMatrix-511

Matrixome

892 012

iMatrix-511 silk

Matrixome

892 021

RetroNectin

TAKARA

T100B

mTeSR1

STEMCELL Technologies

ST-85850

Essential 8

Thermo Fisher Scientific

A1517001

Essential 6

Thermo Fisher Scientific

A1516401

StemLine II

Sigma-Aldrich

S0192

TrypLE Select

Gibco

A1217702

Puromycin

InvivoGen

ant-pr-1

G418

Nacalai Tesque

09380-44

Critical commercial assays

SMART-seq v4 Ultra Low Input RNA Kit for Sequencing

Takara

Z4891N

SMARTer ThruPLEX DNA-seq 48S Kit

Takara

RB4427

NextSeq 500/550 High Output Kit v2.5 (75 Cycles)

Illumina

20024906

CD34 MicroBead Kit, human

Miltenyi Biotec

130-046-702

ChIP DNA Clean & Concentrator

Zymo Research

D5205

(Continued on next page)

iScience 26, 107893, October 20, 2023

17

iScience

ll

Article

OPEN ACCESS

Continued

REAGENT or RESOURCE

SOURCE

IDENTIFIER

This study

PRJNA783033

This study

N/A

pX330-U6-Chimeric_BB-CBh-hSpCas9

Cong et al.41

Addgene #42230

AAVS1-idCas9-vpr

Guo et al.51

Addgene #89985

Cutadapt (v1.15)

EMBnet.Journal42

https://cutadapt.readthedocs.io/en/stable/index.html

Bowtie2 (v2.2.5)

Langmead and Salzberg47

http://bowtie-bio.sourceforge.net/bowtie2/index.shtml

Hisat2 (v2.1.0)

Kim et al.43

http://daehwankimlab.github.io/hisat2/

BEDTools (v.2.27.1)

Bedtools

https://github.com/arq5x/bedtools2

MACS2 (v2.1.1.20160309)

Zhang et al.48

https://github.com/taoliu/MACS

samtools (v1.7)

Samtools

http://www.htslib.org/

featureCounts (v1.6.0)

Liao et al.44

http://subread.sourceforge.net/

R (v3.6.1)

The R Project

https://www.r-project.org/

R: DESeq2 (v1.24.0)

Bioconductor

http://bioconductor.org/packages/release/bioc/html/

R: ClusterProfiler (v3.12.0)

Bioconductor

R: ComplexHeatmap (v2.0.0)

Bioconductor

Deposited data

RNA-seq and ChIP-seq data

Oligonucleotides

gRNA for CRISPR, see method details for

oligonucleotide sequences

Recombinant DNA

Software and algorithms

DESeq2.html

https://bioconductor.org/packages/release/bioc/html/

clusterProfiler.html

https://bioconductor.riken.jp/packages/3.2/bioc/html/

ComplexHeatmap.html

R: ggplot2 (v3.2.1)

CRAN

https://cran.r-project.org/web/packages/ggplot2/index.html

GenomeJack (v3.1)

Mitsubishi Space Software

http://genomejack.net/japanese/index.html

Homer

Heinz et al.49

http://homer.ucsd.edu/homer/

TPMCalculator

Vera Alvarez et al.45

https://github.com/ncbi/TPMCalculator

GraphPad Prism 7

GraphPad Software

https://www.graphpad.com/scientific-software/prism/

FlowJo

BD Biosicneces

https://www.flowjo.com/

CountBright Absolute Counting Beads

Thermo Fisher Scientific

C36950

Dynabeads M-280 Sheep anti-Rabbit IgG

Thermo Fisher Scientific

11203D

Other

RESOURCE AVAILABILITY

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Megumu K. Saito

(msaito@cira.kyoto-u.ac.jp).

Materials availability

All requests for resources and reagents should be directed to the lead contact author. All reagents will be made available on request after

completion of a Materials Transfer Agreement.

Data and code availability

 RNA-seq and ChIP-seq data have been deposited at NCBI SRA and are publicly available as of the date of publication. Accession

numbers are listed in the key resources table.

18

iScience 26, 107893, October 20, 2023

iScience

Article

ll

OPEN ACCESS

 All original code is available in this paper’s supplemental information (Methods S1).

 Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Cell lines

The human ESC line KhES-1 and iPSC line 201B2 were kindly provided by Norio Nakatsuji (Kyoto University, Kyoto, Japan) and Shinya Yamanaka (Kyoto University, Kyoto, Japan), respectively. Both lines were generated from female donors. All cells were regularly karyotyped and

tested for the presence of mycoplasma. The use of human embryonic stem cells (ESCs) in Kyoto University was approved by the Ministry

of Education Culture, Sports, Science and Technology of Japan (MEXT). The study plan for recombinant DNA research was approved by

the recombinant DNA experiments safety committee of Kyoto University.

METHOD DETAILS

Monolayer hematopoietic cell differentiation via HE

PSCs spheroids were formed on an EZSPHERE SP microplate (IWAKI) to tightly control colony numbers and speed up the time required to

obtain colonies of sufficient size. They were transferred to iMatrix-511-coated plates the next day and maintained in mTeSR1 (STEMCELL

Technologies) for three more days. When undifferentiated colonies reached 750–1000 mm in diameter (day 0), hematopoietic cell differentiation was initiated based on previously described protocols.16,17,40 Briefly, the culture media was replaced with Essential-8 medium

(#A1517001, Thermo Fisher Scientific) containing 80 ng/mL BMP4 (#314-BP-010, R&D), 80 ng/mL VEGF 165 (#293-VE-010, R&D), and 4 mM

GSK-3 inhibitor CHIR99021 (#038-23101, Wako). On day 2, the media was changed to Essential-6 medium (#A1516401, Thermo Fisher Scientific) with 80 ng/mL VEGF, 25 ng/mL bFGF (#064-05381, Wako), 2 mM ALK5 inhibitor SB431542 (#031-24291, Wako), and 50 ng/mL SCF (#255SC/CF, R&D). On day 4, CD34+ cells were isolated using a human CD34 Microbead Kit (Miltenyi Biotec) and re-plated on Retronectin-coated

plates at a density of 2.5 x 104 cells/cm2 in hematopoietic cell differentiation medium, which contains Stemline-II medium (#S0192, SigmaAldrich), 50 ng/mL SCF, 50 ng/mL Flt-3 Ligand (#308-GMP, R&D), 5 ng/mL TPO (#288-TPN, R&D) and 20 ng/mL FP6 (#8954-SR, R&D). The

media was thereafter changed every 3 days. 10 mM DAPT or 2 mM SB431542 were added on day 4 and/or 7 for the inhibition of Notch

and TGF-b signaling, respectively, in Figures 3 and S3.

Flow cytometric analysis and cell sorting

For the isolation of cells from day 0 and 2, adherent cells were treated with 50% TrypLE Select (Gibco) in PBS supplemented with 0.5 mM EDTA

(Gibco) and harvested by pipetting. Viability of cells was ensured to be > 90% for the RNA-seq and ChIP-seq experiments. On days 4 and 7, the

supernatant was removed, and adherent cells were collected with 50% TrypLE Select. After staining with DAPI and antibodies, CD34+KDR+

cells were sorted or analyzed using BD FACSAriaII (BD Biosciences). To analyze both CD34+CD45– and CD34+CD45+ cells on day 10, the supernatant and adherent cells were collected. Dead cells were removed by DAPI staining, and the gating was set so that the purity of the sorted

cells was over 90%. The following antibodies were used: BV421 anti-CD34 (744904, BD Biosciences), APC anti-CD309 (3559916, Biolegend),

PECy7 anti-CD45 (304016, Biolegend), FITC anti-CD73 (344015, Biolegend), PE anti-DLL4 (130-096-567, Miltenyi Biotec), and APC anti-CD184

(555976, BD Biosciences). Flow cytometric data were analyzed using FlowJo (version 10.6.1, BD Biosciences).

Generation of ZEB2-deficient and MEIS1-deficient PSCs

A pX330-U6-Chimeric_BB-CBh-hSpCas9 plasmid41 (Addgene #42230) was modified to replace Cas9 with Cas9-T2A-puromycin. This plasmid

was digested by BbsI, and the following oligonucleotides were annealed and inserted.

ZEB2: (forward) CACCGATCCAGACCGCAATTAACAA

(reverse) AAACTTGTTAATTGCGGTCTGGATC

MEIS1: (forward) CACCG TACTTGTACCCCCCGCGAGC

(reverse) AAACGCTCGCGGGGGGTACAAGTAC

The plasmids were transfected into human ESCs or human iPSCs using a NEPA21 electroporator (NEPAGENE) and cuvettes of 2-mm gap.

The transfected cells were transiently selected by adding 1 mg/mL puromycin from 24 to 48 hours after the transfection. Single colonies were

harvested for genotyping by Sanger sequencing. At least two knockout colonies were expanded for further analysis. To generate ZEB2/MEIS1

double-deficient ESCs, ZEB2-deficient ESCs were transfected with the pX330-U6-Chimeric_BB-CBh-hSpCas9 plasmid containing MEIS1-targeting gRNA.

Loss of the target protein was confirmed by western blotting. Briefly, CD34+ cells on day 4 were isolated using the human CD34 Microbead

Kit (Miltenyi Biotec) and lysed with RIPA buffer (Wako) for 30 minutes on ice. After removing cell debris by centrifugation, the lysate was mixed

with 23 Laemmli Sample Buffer (Bio-Rad Laboratories) containing 5% total volume 2-mercaptoethanol (Nacalai tesque) and boiled for 5 minutes at 95 C. Polyacrylamide gel electrophoresis was performed on SDS-polyacrylamide gels, and proteins were transferred to a nitrocellulose membrane (Merck Millipore). The membrane was then incubated with 5% BSA in Tris-buffered saline with tween 20 (Santa Cruz Biotechnology, lnc.) for blocking. The primary antibody reaction was performed at 4 C overnight. The secondary antibody incubation was performed

for 90 minutes at room temperature. The target protein was detected using ECL chemiluminescence reagents (Thermo Fisher Scientific). An

iScience 26, 107893, October 20, 2023

19

ll

OPEN ACCESS

iScience

Article

antibody against b-Actin was reacted for 60 minutes at room temperature. The following primary and secondary antibodies were used: antiZEB2 (1:1000, 61095, Active Motif), anti-MEIS1 (1:1000, abcam ab19867), anti-b-Actin (1:5000, #5125S, Cell Signaling Technology), and antirabbit-HRP (1:2500, #7074S, Cell Signaling Technology).

RNA-seq

FACS-sorted cells were lysed in RLT buffer (Qiagen), and RNA was extracted using RNAClean XP beads (Beckman Coulter). Reverse transcription was performed using a SMART-Seq v4 Ultra Low Input RNA Kit for Sequencing (Takara). cDNA was then fragmented using a Covaris

Focused-ultrasonicator M220 (M&S Instruments Inc.). The library was constructed using a SMARTer ThruPLEX DNA-seq 48S Kit (Takara)

and sequenced on a NextSeq 500 System (Illumina) with 75-bp single-end reads. No technical replicates were generated.

Reads were trimmed by Cutadapt (version 1.15)42 and mapped to the human genome hg19 by Hisat2 (version 2.1.0).43 Tags were counted

by featureCounts (version 1.6.0)44 and normalized using the DESeq2 package (version 1.24.0) in R. Differentially expressed genes were defined

as those with FDR < 0.01 and log2 fold-change > 2. The expression of RUNX1 splice variants was determined by TPMCalculator (version

0.0.3).45 Heatmaps showing gene expression patterns were generated using the ComplexHeatmap package (version 2.0.0)46 in R. Data

from individual samples were treated independently in all PCA plots and heatmaps.

ChIP-seq

The following number of cells were used for the ChIP-seq: 1 x 105 for H3K27ac, 2 x 105 for H3K4me1 and H3K27me3, and 1-5 x 106 for transcription factor ChIP-seq. The antibodies used were anti-H3K27ac (GeneTex, GTX60815), anti-H3K4me1 (Active Motif, 39297), anti-H3K27me3

(Millipore, 07-449), anti-ZEB2 (Bethyl Laboratories, A302474A), anti-MEIS1 (Abcam, ab19867), anti-Notch1 (Cell Signaling Technology, 3608S),

anti-SOX6 (Abcam, ab30455), anti-SOX17 (R&D Systems, AF1924-SP), and anti-SPI1 (Cell Signaling Technology, 2266). Cells were cross-linked

in 1% (w/v) formaldehyde solution for 5 min (histone ChIP-seq) or 30 min (transcription factor ChIP-seq) and lysed. Cross-linked DNA was fragmented by sonication using a Digital Sonifier (Branson) and incubated overnight at 4 C with 50 mL DynaBeads IgG magnetic beads (Thermo

Fisher) conjugated with 2.5 mg antibodies in the presence of 1% FBS and 10-30 mg salmon sperm DNA. Samples were then washed, eluted,

reverse cross-linked at 65 C overnight, and purified using ChIP DNA Clean & Concentrator (Zymo Research). For transcription factor ChIPseq, purified ChIP DNA was fragmented using a Covaris Focused-ultrasonicator M220 (M&S Instruments Inc.). The library was prepared using

a SMARTer ThruPLEX DNA-seq 48S Kit (Takara) and sequenced on a NextSeq 500 System (Illumina) with 75-bp single-end reads.

ChIP-seq reads were trimmed by Cutadapt and mapped by bowtie247 to hg19 after removing the reads mapped to salmon. For visualization, ChIP peaks were called and normalized by the number of mapped reads (-SPMR option) using MACS248 with input reads as a control

and are presented in GenomeJACK Browser (version 3.1, Mitsubishi Space Software). The y-axis of a ChIP-seq track indicates the count per

million (CPM). To identify ChIP peaks, the findPeaks program in the Homer package49 was used. For H3K27ac ChIP-seq, the program was

used with -region option and 20-fold enrichment over input as a cutoff. For transcription factor ChIP-seq, both the peak size and minimum

distance of peaks were set to 500 bp, the local fold change cutoff was disabled, and the default settings were applied for all other parameters.

ChIP-seq tag counts at the target regions were determined by featureCounts (version 1.6.0) and normalized by the number of uniquely mapped reads (tag counts per region O uniquely mapped reads 3 1,000,000). Heatmaps showing relative signal intensities were generated using

the ComplexHeatmap package (version 2.0.0) in R.

Definition of hematopoietic enhancers

First, regions with H3K27ac marks in day 10 CD34+CD45+ cells were identified by the findPeaks program in the Homer package. Regions overlapping promoter regions (defined as up to 2.5 kb upstream of the transcription start site indicated in NCBI RefSeq) were removed. Regions

with H3K27ac marks were similarly identified in day 0 and day 2 cells and H3K27ac-marked regions in day 10 CD34+CD45+ cells which overlap

with H3K27ac-marked regions in day 0 and day 2 cells were also removed to define hematopoietic enhancers.

Gene ontology analysis

A gene ontology analysis of defined gene sets was conducted using ClusterProfiler (version 3.12.0)50 in R. All gene sets in the Gene Ontology

database were used, except for the data in Figure 1B, where the gene sets whose name contain ‘development’ or ‘differentiation’ were

selected.

Motif analysis

A motif analysis was performed using the findMotifsGenome program in the Homer package with default settings. For the background

sequence, proximal enhancers [ENCFF036NSJ] and distal enhancers [ENCFF535MKS] from candidate cis-Regulatory Elements predicted

in the ENCODE project (https://www.encodeproject.org/) were combined, and the genome coordinates were converted from hg38 to

hg19 using UCSC LiftOver. Results with q-value < 0.0001, log P-value < -30 and the ratio of % target sequences with motifs to % background

sequences with motifs > 1.5 in at least one cluster of hematopoietic enhancers were selected for the data presentation.

20

iS ...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る