Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Cloche is a bHLH-PAS transcription factor that drives haemato-vascular specification

A Corrigendum to this article was published on 22 March 2018

This article has been updated

Abstract

Vascular and haematopoietic cells organize into specialized tissues during early embryogenesis to supply essential nutrients to all organs and thus play critical roles in development and disease. At the top of the haemato-vascular specification cascade lies cloche, a gene that when mutated in zebrafish leads to the striking phenotype of loss of most endothelial and haematopoietic cells1,2,3,4 and a significant increase in cardiomyocyte numbers5. Although this mutant has been analysed extensively to investigate mesoderm diversification and differentiation1,2,3,4,5,6,7 and continues to be broadly used as a unique avascular model, the isolation of the cloche gene has been challenging due to its telomeric location. Here we used a deletion allele of cloche to identify several new cloche candidate genes within this genomic region, and systematically genome-edited each candidate. Through this comprehensive interrogation, we succeeded in isolating the cloche gene and discovered that it encodes a PAS-domain-containing bHLH transcription factor, and that it is expressed in a highly specific spatiotemporal pattern starting during late gastrulation. Gain-of-function experiments show that it can potently induce endothelial gene expression. Epistasis experiments reveal that it functions upstream of etv2 and tal1, the earliest expressed endothelial and haematopoietic transcription factor genes identified to date. A mammalian cloche orthologue can also rescue blood vessel formation in zebrafish cloche mutants, indicating a highly conserved role in vertebrate vasculogenesis and haematopoiesis. The identification of this master regulator of endothelial and haematopoietic fate enhances our understanding of early mesoderm diversification and may lead to improved protocols for the generation of endothelial and haematopoietic cells in vivo and in vitro.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Identification and analysis of candidate genes from the cloche locus.
Figure 2: npas4l is cloche.
Figure 3: cloche/npas4l is expressed in endothelial and haematopoietic precursors, and this expression precedes that of the earliest endothelial and haematopoietic markers etv2 and tal1.
Figure 4: Cloche/Npas4l is a master regulator of the endothelial differentiation cascade.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

GenBank/EMBL/DDBJ

Gene Expression Omnibus

Change history

  • 21 March 2018

    Please see accompanying Corrigendum (http://doi.org/10.1038/nature25991). In the Author Information section, the accession for the PacBio whole-genome sequencing data was wrongly stated as LT571435 from the SRA, instead of PRJEB13442 from the European Nucleotide Archive (ENA). The original Letter has been corrected online.

References

  1. Stainier, D. Y., Weinstein, B. M., Detrich, H. W., III, Zon, L. I. & Fishman, M. C. cloche, an early acting zebrafish gene, is required by both the endothelial and hematopoietic lineages. Development 121, 3141–3150 (1995)

    CAS  PubMed  Google Scholar 

  2. Liao, W. et al. The zebrafish gene cloche acts upstream of a flk-1 homologue to regulate endothelial cell differentiation. Development 124, 381–389 (1997)

    CAS  PubMed  Google Scholar 

  3. Liao, E. C. et al. SCL/Tal-1 transcription factor acts downstream of cloche to specify hematopoietic and vascular progenitors in zebrafish. Genes Dev. 12, 621–626 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liao, W., Ho, C. Y., Yan, Y. L., Postlethwait, J. & Stainier, D. Y. Hhex and Scl function in parallel to regulate early endothelial and blood differentiation in zebrafish. Development 127, 4303–4313 (2000)

    CAS  PubMed  Google Scholar 

  5. Schoenebeck, J. J., Keegan, B. R. & Yelon, D. Vessel and blood specification override cardiac potential in anterior mesoderm. Dev. Cell 13, 254–267 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sumanas, S., Jorniak, T. & Lin, S. Identification of novel vascular endothelial-specific genes by the microarray analysis of the zebrafish cloche mutants. Blood 106, 534–541 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Liu, F., Walmsley, M., Rodaway, A. & Patient, R. Fli1 acts at the top of the transcriptional network driving blood and endothelial development. Curr. Biol. 18, 1234–1240 (2008)

    Article  CAS  PubMed  Google Scholar 

  8. Rossant, J. & Howard, L. Signaling pathways in vascular development. Annu. Rev. Cell Dev. Biol. 18, 541–573 (2002)

    Article  CAS  PubMed  Google Scholar 

  9. Adams, R. H. & Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nature Rev. Mol. Cell Biol. 8, 464–478 (2007)

    Article  CAS  Google Scholar 

  10. Herbert, S. P. & Stainier, D. Y. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat. Rev. Mol. Cell Biol. 12, 551–564 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hirschi, K. K. Hemogenic endothelium during development and beyond. Blood 119, 4823–4827 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. De Val, S. & Black, B. L. Transcriptional control of endothelial cell development. Dev. Cell 16, 180–195 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Begley, C. G. et al. The gene SCL is expressed during early hematopoiesis and encodes a differentiation-related DNA-binding motif. Proc. Natl Acad. Sci. USA 86, 10128–10132 (1989)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Endoh, M., Ogawa, M., Orkin, S. & Nishikawa, S. SCL/tal-1-dependent process determines a competence to select the definitive hematopoietic lineage prior to endothelial differentiation. EMBO J. 21, 6700–6708 (2002)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Van Handel, B. et al. Scl represses cardiomyogenesis in prospective hemogenic endothelium and endocardium. Cell 150, 590–605 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lacaud, G., Robertson, S., Palis, J., Kennedy, M. & Keller, G. Regulation of hemangioblast development. Ann. NY Acad. Sci. 938, 96–108 (2001)

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Huber, T. L., Kouskoff, V., Fehling, H. J., Palis, J. & Keller, G. Haemangioblast commitment is initiated in the primitive streak of the mouse embryo. Nature 432, 625–630 (2004)

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Vogeli, K. M., Jin, S. W., Martin, G. R. & Stainier, D. Y. A common progenitor for haematopoietic and endothelial lineages in the zebrafish gastrula. Nature 443, 337–339 (2006)

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Yang, L. et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 453, 524–528 (2008)

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Xiong, J. W., Yu, Q., Zhang, J. & Mably, J. D. An acyltransferase controls the generation of hematopoietic and endothelial lineages in zebrafish. Circ. Res. 102, 1057–1064 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ooe, N., Saito, K., Mikami, N., Nakatuka, I. & Kaneko, H. Identification of a novel basic helix-loop-helix-PAS factor, NXF, reveals a Sim2 competitive, positive regulatory role in dendritic-cytoskeleton modulator Drebrin gene expression. Mol. Cell. Biol. 24, 608–616 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lin, Y. et al. Activity-dependent regulation of inhibitory synapse development by Npas4. Nature 455, 1198–1204 (2008)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Smith, B. W. et al. The aryl hydrocarbon receptor directs hematopoietic progenitor cell expansion and differentiation. Blood 122, 376–385 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Iyer, N. V. et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev. 12, 149–162 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Peng, J., Zhang, L., Drysdale, L. & Fong, G. H. The transcription factor EPAS-1/hypoxia-inducible factor 2α plays an important role in vascular remodeling. Proc. Natl Acad. Sci. USA 97, 8386–8391 (2000)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Field, H. A., Dong, P. D., Beis, D. & Stainier, D. Y. Formation of the digestive system in zebrafish. II. Pancreas morphogenesis. Dev. Biol. 261, 197–208 (2003)

    Article  CAS  PubMed  Google Scholar 

  27. Pauli, A. et al. Systematic identification of long noncoding RNAs expressed during zebrafish embryogenesis. Genome Res. 22, 577–591 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Parker, L. & Stainier, D. Y. Cell-autonomous and non-autonomous requirements for the zebrafish gene cloche in hematopoiesis. Development 126, 2643–2651 (1999)

    CAS  PubMed  Google Scholar 

  29. Ramírez-Bergeron, D. L., Runge, A., Adelman, D. M., Gohil, M. & Simon, M. C. HIF-dependent hematopoietic factors regulate the development of the embryonic vasculature. Dev. Cell 11, 81–92 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Han, Y., Kuang, S. Z., Gomer, A. & Ramirez-Bergeron, D. L. Hypoxia influences the vascular expansion and differentiation of embryonic stem cell cultures through the temporal expression of vascular endothelial growth factor receptors in an ARNT-dependent manner. Stem Cells 28, 799–809 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jin, S. W., Beis, D., Mitchell, T., Chen, J. N. & Stainier, D. Y. Cellular and molecular analyses of vascular tube and lumen formation in zebrafish. Development 132, 5199–5209 (2005)

    Article  CAS  PubMed  Google Scholar 

  32. Yi, H. et al. Duplex-specific nuclease efficiently removes rRNA for prokaryotic RNA-seq. Nucleic Acids Res. 39, e140 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhao, W. et al. Comparison of RNA-seq by poly (A) capture, ribosomal RNA depletion, and DNA microarray for expression profiling. BMC Genomics 15, 419 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nature Methods 9, 357–359 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Roberts, A., Pimentel, H., Trapnell, C. & Pachter, L. Identification of novel transcripts in annotated genomes using RNA-seq. Bioinformatics 27, 2325–2329 (2011)

    Article  CAS  PubMed  Google Scholar 

  37. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bedell, V. M. et al. In vivo genome editing using a high-efficiency TALEN system. Nature 491, 114–118 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jao, L. E., Wente, S. R. & Chen, W. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc. Natl Acad. Sci. USA 110, 13904–13909 (2013)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  41. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at http://arxiv.org/abs/1303.3997v2 (2013)

  42. Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. Preprint at http://arxiv.org/abs/1207.3907 (2012)

  43. Thisse, C. & Thisse, B. High-resolution in situ hybridization to whole-mount zebrafish embryos. Nature Protocols 3, 59–69 (2008)

    Article  CAS  PubMed  Google Scholar 

  44. Marshall, O. J. PerlPrimer: cross-platform, graphical primer design for standard, bisulphite and real-time PCR. Bioinformatics 20, 2471–2472 (2004)

    Article  CAS  PubMed  Google Scholar 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013)

    Article  CAS  PubMed  Google Scholar 

  46. Trapnell, C. et al. Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Dereeper, A. et al. Phylogeny.fr: robust phylogenetic analysis for the non-specialist. Nucleic Acids Res. 36, W465–W469 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Piskacek, S. et al. Nine-amino-acid transactivation domain: establishment and prediction utilities. Genomics 89, 756–768 (2007)

    Article  CAS  PubMed  Google Scholar 

  49. Dooley, K. A., Davidson, A. J. & Zon, L. I. Zebrafish scl functions independently in hematopoietic and endothelial development. Dev. Biol. 277, 522–536 (2005)

    Article  CAS  PubMed  Google Scholar 

  50. Sumanas, S. & Lin, S. Ets1-related protein is a key regulator of vasculogenesis in zebrafish. PLoS Biol. 4, e10 (2006)

    Article  CAS  PubMed  Google Scholar 

  51. Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nature Methods 8, 424–429 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kattman, S. J. et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8, 228–240 (2011)

    Article  CAS  PubMed  Google Scholar 

  53. Lin, B. et al. High-purity enrichment of functional cardiovascular cells from human iPS cells. Cardiovasc. Res. 95, 327–335 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all laboratory members who have worked on cloche over the years starting with W. Liao and H. Sawyer (UCSF), as well as J. Collins (Sanger Institute) for help in expanding the GRCz10 assembly, W. Coppieters (Liège), Z. Wang (JGI), X. Chen (BGI), H. Yuan (BGI) for their hard work in trying to resolve the cloche locus, C. Helker, M. Higuchi and C. Gerri for reagents, discussions and reading of the manuscript, A. Borchers (Marburg) for help with Xenopus experiments, and funding from the DFG (S.R.), AHA (S.R., S.-W.J., N.C.), NIH (N.C., A.J.G., D.Y.R.S.), the Ragnar Söderberg Foundation and Swedish Research Council (O.A.), and the Packard Foundation and Max Planck Society (D.Y.R.S.).

Author information

Authors and Affiliations

Authors

Contributions

S.R., O.S., A.V. and D.Y.R.S. designed and performed experiments identifying cloche and its function. N.F., Mi.M, S.M.A. and S.M. performed reverse genetics and downstream analysis. N.C., S.-W.J., T.K., W.-S.C., S.S., R.L. and J.A. worked on the meiotic mapping. S.L., Ma.M., M.T.L., I.F. and D.T. contributed to sequencing and bioinformatics. Human embryonic stem cell assays were handled by A.W. Supervision by D.Y.R.S., N.C., O.A., G.P.S., A.J.G. and B.N. All authors contributed to data analysis and manuscript preparation.

Corresponding author

Correspondence to Didier Y. R. Stainier.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Zebrafish npas4l sequences have been deposited to GenBank (KX066018, KX066019), RNA-seq data deposited to GEO (GSE76690), and PacBio whole-genome sequencing data to ENA (PRJEB13442).

Reviewer Information

Nature thanks K. Alitalo and K. Poss and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Illustration of the predicted mutant proteins.

Schematic representations show the wild-type protein (top) and the corresponding mutant protein (bottom) for each of the candidate genes in the cloche region. Functional protein domains are annotated, and the information about the molecular lesion(s) is indicated next to the protein’s name. Red numbers indicate the last native amino acid before the induced frameshift mutation. For the two genes that were targeted at more than one position, the extension of the longer mutant protein is indicated by transparency. Except for the npas4l mutation, none of the other 18 mutations causes an obvious morphological phenotype at 48 hpf. For proteins with incomplete sequence information (Inpp5a, Ptk7b, Srfb), primary structure was extrapolated from orthologues from other teleost species.

Extended Data Figure 2 5′ and 3′ RACE PCR for npas4l, and physical map of the cloche/npas4l locus in relation to klhdc3.

a, Primer design was based on a 513 bp fragment from GRCz10 that is deleted in clom39 mutants. b, c, Agarose gels showing the products of 5′ (b) and 3′ (c) RACE PCRs using cDNAs generated from 24 hpf or tailbud (TB) mRNAs. Multiple fragments were cloned for sequencing and bands that extended the npas4l sequence to the 5′ (b) and 3′ (c) are highlighted in blue boxes. d, Physical assembly of the cloche locus based on the extension of KN150101.1 by whole-genome sequencing. KN150101.1 (ex)/LT571435 is shown in reverse-complementary orientation with the blue region marking newly obtained sequence information. Alignment of the klhdc3 expressed sequence tag (EST) BC068372.1 establishes a physical connection between KN150101.1 (ex) and Chr. 13. The tightly linked SSLP marker z709 lies between klhdc3 and cloche/npas4l.

Extended Data Figure 3 bHLH-PAS protein relatedness and phylogeny of Cloche/Npas4l.

ac, Individual clustering diagrams based on the amino acid sequence of the bHLH domain (a), the PAS-A domain (b), and the PAS-B domain (c) of mouse class I (NPAS1, NPAS3, NPAS4, HIF1A-HIF3A, AHR, SIM1/2) and class II (ARNT, ARNT2) bHLH-PAS proteins, together with the protein encoded by zebrafish cloche/npas4l. Analysis using the respective zebrafish proteins leads to similar results; the similarity between full-length zebrafish Npas4l and Npas4a is 39.5%, and between Npas4l and Npas4b it is 33%. d, GenBank accession numbers of proteins used in this analysis. e, Unrooted phylogenetic tree of Npas4 and Npas4l proteins across different phyla (using the sequence from the N terminus of the bHLH domain to the C terminus of the PAS-B domain). The cloche/npas4l gene is clearly present from lampreys to birds, but appears to be missing in mammals. f, Detailed information on the NPAS4 and NPAS4L protein sequences as well as synteny information of the corresponding genes. Predicted proteins were manually corrected when necessary using gene prediction methods (Genscan; AUGUSTUS) and BLAST tools.

Extended Data Figure 4 Additional phenotypic analysis of npas4lbns59 mutant.

af, Whole-mount in situ hybridization for kdrl (a, b), gata1a (c, d) and hbae3 (e, f) expression at the 15-somite stage in wild-type and npas4lbns59/bns59 embryos. g, h, Ventral views of Tg(kdrl:EGFP) sibling (g) and npas4lbns59 mutant (h) hearts at 48–hpf; a, atrium; v, ventricle. i, Quantification of the number of atrial endocardial cells in homozygous wild-type siblings (n = 9) and npas4lbns59 mutants (n = 9) at 60 hpf. Values represent means ± s.e.m. ***P < 0.001 by t-test. j, Penetrance of the vascular phenotype based on Tg(kdrl:EGFP) expression in 48 hpf npas4lbns59/bns59 embryos. kv, Side-by-side comparison of endothelial and haematopoietic marker gene expression between different cloche alleles. Whole-mount in situ hybridization for etv2 (kn), tal1 (or) and gata1a (sv) expression at the 15-somite stage in wild-types and clom39, clos5 and npas4lbns59 mutants. x/y, number of embryos showing representative phenotype (x), number of embryos examined (y); m39 and s5 mutants were not confirmed by genotyping, hence the approximately 1/4 ratio in those panels. Scale bars, 200 μm (except for g and h, 100 μm).

Extended Data Figure 5 Morpholino knockdown of npas4l, etv2 and tal1.

Knockdown of Npas4l was achieved by co-injecting a translation blocking morpholino targeting the npas4l translational start site and a splice morpholino targeting the intron-exon splice site I2E3. a, Schematic representation of the npas4l genomic locus including the I2E3 splice morpholino (red) and the oligos used to test for splicing defects by RT–PCR (black). b, Analysis of npas4l mRNA splicing by RT–PCR shows a significant reduction in wild-type npas4l mRNA at the tailbud stage following injection of 6 ng of the I2E3 morpholino at the one-cell stage (3 different experiments, sMO1-sMO3, are shown). uc, uninjected control; sMO, splice morpholino injected; H2O, water control; wild-type product, 296 bp. c, d, Knockdown of Etv2 was achieved by injecting two previously published ATG morpholinos50. Fluorescence micrographs of Tg(kdrl:EGFP) control (ctrl) and etv2 MO1 + MO2 injected embryos. df, Knockdown of Tal1 was achieved by injecting a previously published splice morpholino49. d, e, Confocal projections of Tg(kdrl:EGFP) ctrl and tal1 MO injected embryos. g, Analysis of tal1 mRNA splicing by RT–PCR shows a significant reduction in wild-type tal1 mRNA at the two-somite stage after injecting 6.5 ng of tal1 MO at the one-cell stage. Three different experiments, sMO1–sMO3, are shown). uc, uninjected control; sMO, splice morpholino injected; H2O, water control; wild-type product, 401 bp. Scale bars, 200 μm.

Extended Data Figure 6 npas4l rescues clos5/s5 embryos.

af, Confocal projections of the anterior (ac) and posterior (df) vasculature of 54 hpf Tg(kdrl:EGFP) sibling, clos5/s5 and clos5/s5 embryos injected with 25 pg npas4l mRNA at the one-cell stage. Scale bars, 200 μm.

Extended Data Figure 7 Expression levels of npas4l, etv2, tal1 and gata1a during development.

ac, RT–qPCR for npas4l, etv2 and tal1 expression; 20–40 embryos from single pair matings were pooled at each of the indicated stages; expression is normalized to the corresponding 24 hpf sample. Error bars represent s.e.m. from three biological replicates. d, Data extracted from ref. 27. npas4l expression clearly precedes etv2, tal1 and gata1a expression; it appears to be minimal until 6 hpf, peaks around the end of gastrulation/early somitogenesis (in 10 hpf sample from this data set), and is clearly down by 28 hpf, the stage when the other transcripts peak. Expression at each developmental stage represents a single RNA-seq experiment on 1,000 pooled embryos, and bars indicate the 95% confidence interval (Cufflinks). e, RT–qPCR for gata1a expression, performed and analysed as in ac. Whole-mount in situ hybridization for gata1a expression in wild-type embryos at the 2-somite (f) and 18-somite (g) stages. Scale bars, 200 μm.

Extended Data Figure 8 Potential Npas4l targets.

RT–qPCR analysis of etv2, fli1a, fli1b, lmo2, sox7, sox18 and tal1 expression in shield stage embryos injected with npas4l mRNA at the one-cell stage, normalized to uninjected controls. Error bars represent s.e.m. from three biological replicates of 20–40 pooled embryos per condition.

Extended Data Figure 9 NPAS4 expression during human embryonic stem cell derived vascular endothelial differentiation.

a, Scheme of the protocol used to differentiate human embryonic stem cells towards the vascular endothelial (VE) lineage highlighting the (1) mesoderm induction stage and (2) the VE lineage specification and differentiation stage. b, Representative flow cytometric analysis for day 5 KDR+ PDGFRA VE progenitors and day 10 CD31+ KDR+ VE cells. c, RT–qPCR analysis of temporal expression of NPAS4 compared to OCT4, T, MESP1, ETV2, AHR, FLI1, CDH5 and PECAM1 over the course of human embryonic stem cell derived VE differentiation. Values are relative to the housekeeping gene TBP and depicted as a fold change in which average peak expression of the individual gene is set to 1. Raw data can be found in Figshare (http://dx.doi.org/10.6084/m9.figshare.3383572). Error bars represent s.e.m. from three biological replicates.

Extended Data Figure 10 AHR overexpression induces expression of early endothelial and haematopoietic markers in P19 mouse embryonic carcinoma cells.

a, RT–qPCR analysis of Etv2, Tal1, Lmo2 and Sox7 expression following overexpression of human AHR in P19 cells. Error bars represent s.e.m. from three biological replicates derived from independent transfections (*P < 0.05 and **P < 0.01 by t-test).

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-4. (PDF 261 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reischauer, S., Stone, O., Villasenor, A. et al. Cloche is a bHLH-PAS transcription factor that drives haemato-vascular specification. Nature 535, 294–298 (2016). https://doi.org/10.1038/nature18614

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18614

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing