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.

  • Article
  • Published:

The noncoding RNA IPW regulates the imprinted DLK1-DIO3 locus in an induced pluripotent stem cell model of Prader-Willi syndrome

Abstract

Parental imprinting is a form of epigenetic regulation that results in parent-of-origin differential gene expression. To study Prader-Willi syndrome (PWS), a developmental imprinting disorder, we generated case-derived induced pluripotent stem cells (iPSCs) harboring distinct aberrations in the affected region on chromosome 15. In studying PWS-iPSCs and human parthenogenetic iPSCs, we unexpectedly found substantial upregulation of virtually all maternally expressed genes (MEGs) in the imprinted DLK1-DIO3 locus on chromosome 14. Subsequently, we determined that IPW, a long noncoding RNA in the critical region of the PWS locus, is a regulator of the DLK1-DIO3 region, as its overexpression in PWS and parthenogenetic iPSCs resulted in downregulation of MEGs in this locus. We further show that gene expression changes in the DLK1-DIO3 region coincide with chromatin modifications rather than DNA methylation levels. Our results suggest that a subset of PWS phenotypes may arise from dysregulation of an imprinted locus distinct from the PWS region.

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: Genome-wide gene expression analysis in PWS-iPSCs.
Figure 2: Characterization of the DLK1-DIO3 locus in PWS.
Figure 3: IPW regulates gene expression at the DLK1-DIO3 locus.
Figure 4: IPW affects chromatin modifications at the DLK1-DIO3 locus.
Figure 5: Model summarizing the effect of IPW depletion on the expression of MEGs in the DLK1-DIO3 region during early development.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Ferguson-Smith, A.C. Genomic imprinting: the emergence of an epigenetic paradigm. Nat. Rev. Genet. 12, 565–575 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Cassidy, S.B., Schwartz, S., Miller, J.L. & Driscoll, D.J. Prader-Willi syndrome. Genet. Med. 14, 10–26 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Kishore, S. & Stamm, S. The snoRNA HBII-52 regulates alternative splicing of the serotonin receptor 2C. Science 311, 230–232 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Yin, Q.F. et al. Long noncoding RNAs with snoRNA ends. Mol. Cell 48, 219–230 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Pick, M. et al. Clone- and gene-specific aberrations of parental imprinting in human induced pluripotent stem cells. Stem Cells 27, 2686–2690 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Stelzer, Y., Yanuka, O. & Benvenisty, N. Global analysis of parental imprinting in human parthenogenetic induced pluripotent stem cells. Nat. Struct. Mol. Biol. 18, 735–741 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Stelzer, Y. et al. Identification of novel imprinted differentially methylated regions by global analysis of human-parthenogenetic-induced pluripotent stem cells. Stem Cell Reports 1, 79–89 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Urbach, A., Bar-Nur, O., Daley, G.Q. & Benvenisty, N. Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell 6, 407–411 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Stelzer, Y., Sagi, I. & Benvenisty, N. Involvement of parental imprinting in the antisense regulation of onco-miR-372-373. Nat. Commun. 4, 2724 (2013).

    Article  PubMed  Google Scholar 

  11. Seitz, H. et al. Imprinted microRNA genes transcribed antisense to a reciprocally imprinted retrotransposon-like gene. Nat. Genet. 34, 261–262 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Vassena, R. et al. Accumulation of instability in serial differentiation and reprogramming of parthenogenetic human cells. Hum. Mol. Genet. 21, 3366–3373 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. de Smith, A.J. et al. A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism. Hum. Mol. Genet. 18, 3257–3265 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Duker, A.L. et al. Paternally inherited microdeletion at 15q11.2 confirms a significant role for the SNORD116 C/D box snoRNA cluster in Prader-Willi syndrome. Eur. J. Hum. Genet. 18, 1196–1201 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sahoo, T. et al. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat. Genet. 40, 719–721 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wevrick, R., Kerns, J.A. & Francke, U. Identification of a novel paternally expressed gene in the Prader-Willi syndrome region. Hum. Mol. Genet. 3, 1877–1882 (1994).

    Article  CAS  PubMed  Google Scholar 

  17. Guttman, M. et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458, 223–227 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Guttman, M. et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477, 295–300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rinn, J.L. et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Khalil, A.M. et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. USA 106, 11667–11672 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, X. et al. A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints. Dev. Cell 15, 547–557 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Collins, R.E. et al. In vitro and in vivo analyses of a Phe/Tyr switch controlling product specificity of histone lysine methyltransferases. J. Biol. Chem. 280, 5563–5570 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Nishikawa, S., Goldstein, R.A. & Nierras, C.R. The promise of human induced pluripotent stem cells for research and therapy. Nat. Rev. Mol. Cell Biol. 9, 725–729 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Soldner, F. & Jaenisch, R. Medicine. iPSC disease modeling. Science 338, 1155–1156 (2012).

    Article  PubMed  Google Scholar 

  25. Chamberlain, S.J. et al. Induced pluripotent stem cell models of the genomic imprinting disorders Angelman and Prader-Willi syndromes. Proc. Natl. Acad. Sci. USA 107, 17668–17673 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yang, J. et al. Induced pluripotent stem cells can be used to model the genomic imprinting disorder Prader-Willi syndrome. J. Biol. Chem. 285, 40303–40311 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cabili, M.N. et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25, 1915–1927 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Mann, M.R. & Bartolomei, M.S. Towards a molecular understanding of Prader-Willi and Angelman syndromes. Hum. Mol. Genet. 8, 1867–1873 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Falk, M.J., Curtis, C.A., Bass, N.E., Zinn, A.B. & Schwartz, S. Maternal uniparental disomy chromosome 14: case report and literature review. Pediatr. Neurol. 32, 116–120 (2005).

    Article  PubMed  Google Scholar 

  30. Hordijk, R. et al. Maternal uniparental disomy for chromosome 14 in a boy with a normal karyotype. J. Med. Genet. 36, 782–785 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hosoki, K. et al. Maternal uniparental disomy 14 syndrome demonstrates Prader-Willi syndrome–like phenotype. J. Pediatr. 155, 900–903 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Reik, W. & Walter, J. Evolution of imprinting mechanisms: the battle of the sexes begins in the zygote. Nat. Genet. 27, 255–256 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Gupta, R.A. et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464, 1071–1076 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Epsztejn-Litman, S. et al. De novo DNA methylation promoted by G9a prevents reprogramming of embryonically silenced genes. Nat. Struct. Mol. Biol. 15, 1176–1183 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Fibroblasts obtained from an individual with PWS harboring complete mUPD of chromosome 15 were kindly donated by V. Gross-Tsur (Multidisciplinary Prader-Willi Syndrome Clinic, Child Neurology Unit, Shaare Zedek Medical Center). This research was partially funded by the Israel Science Foundation–Morasha Foundation (grant 1252/12) and by the Israel Ministry of Science and Technology Infrastructure (grant 3-9693).

Author information

Authors and Affiliations

Authors

Contributions

Y.S. contributed to the conception and design of the study, the collection and assembly of data, data analysis and interpretation, and manuscript writing. I.S. contributed to the collection and assembly of data and graphic design. O.Y. and R.E. contributed to the collection and assembly of data. N.B. contributed to the conception and design of the study, financial support, data analysis and interpretation, and manuscript writing.

Corresponding author

Correspondence to Nissim Benvenisty.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of PWS-iPSCs.

(a) G-band karyotyping of representative PWS-iPSC lines, demonstrating a diploid genome, excluding chromosome 15 aberrations. (b) Representative alkaline phosphatase staining for PWS-iPSC lines. Scale bar, 200 μm. (c) Mean expression levels ± s.d. of representative pluripotency markers in four PWS-iPSC lines (PWS-iPSC-1-A and B and PWS-iPSC-2-A and B) compared with five control (WT) PSCs (two iPSC and three ESC lines). (d) Gene expression analysis of 20-d-old embryoid bodies (EBs) from the two PWS-iPSC lines, demonstrating differentiation in vitro into all three embryonic germ layers. DES, NCAM1 and AFP represent markers for mesoderm, ectoderm and endoderm, respectively (mean expression levels ± s.d.; two biological replicates). (e) Mean expression levels ± s.d. of representative potential targets of PEGs in the PWS locus, in PWS and control (WT) fibroblasts.

Supplementary Figure 2 Genome-wide gene expression analysis in PWS-iPSCs derived from a patient harboring a maternal UPD of chromosome 15.

(a) Establishment of PWS-iPSCs with maternal UPD in the PWS region. (b,c) Analysis of gene expression by moving average plot along chromosome 15q (b, logarithmic scale) and 14q (c) in PWS-iPSC-3 relative to normal (WT) PSCs. Vertical dashed lines mark the genomic regions of the PWS (b) and DLK1-DIO3 (c) loci.

Supplementary Figure 3 Analysis of known imprinted genes in PWS-iPSCs.

(a) RTL1 expression ratio (in RPKM, logarithmic scale, two biological replicates), demonstrating its marked downregulation in Pg-iPSCs relative to normal PSCs. (b) qRT-PCR of the mean relative fold change ± s.d. (three biological replicates each) of RTL1, MEG3, miR-370 and miR-409 in normal PSCs before and after knockdown of RTL1 (+siRNA). (c) qRT-PCR of the mean relative fold change ± s.d. of miR-371-3 in normal PSCs, Pg-iPSCs and PWS-iPSCs (two biological replicates each), demonstrating comparable expression levels of miR-371-3 in PWS-iPSCs and normal PSCs. (d) Mean expression levels ± s.d. of representative known PEGs in distinct loci. Analyses were conducted on three PWS-iPSC lines (PWS-iPSC-1-A and B and PWS-iPSC-2-A), three Pg-iPSC lines (Pg-iPSC-A-11, 20 and 26) and five control PSCs (two iPSC and three ESC lines). WT-PSCs, normal PSCs.

Supplementary Figure 4 Gene expression analyses in PWS cell lines.

(a) Analysis of gene expression by moving average plot along chromosome 14q in PWS parental fibroblasts relative to control fibroblasts. Vertical dashed lines mark the genomic boundaries of the DLK1-DIO3 region. (b) qRT-PCR of the mean relative fold change ± s.d. (three biological replicates each) in MEG3 in mature PWS cells derived from three affected individuals, showing negligible expression levels in patient-derived B cells. N.E., not expressed. (c) Expression analysis of all MEGs represented in the expression arrays in the parental fibroblasts (left panel) and undifferentiated iPSCs (right panel). WT, control.

Supplementary Figure 5 Proper monoallelic expression of imprinted genes in the DLK1-DIO3 locus in PWS-iPSCs.

Direct sequencing of the transcribed region of MEG3 and RTL1 in genomic DNA (gDNA) and complementary DNA (cDNA) in PWS-iPSCs derived from three patients. Note that both MEG3 and RTL1 are expressed in a monoallelic fashion.

Supplementary Figure 6 Chromatin regulation in PWS-iPSCs.

(a) qRT-PCR of the mean relative fold change ± s.d. (three biological replicates each) in HOTAIR in PWS-iPSCs before and after transfection with an overexpression plasmid. (b) qRT-PCR of the mean relative fold change ± s.d. (three biological replicates each) in MEG3 in PWS-iPSCs before and after transfection with the HOTAIR overexpression plasmid, relative to control (WT) PSCs. (c) Allelic sequence analysis following ChIP for H3K9me3 reveals the association of this histone modification with both methylated and unmethylated alleles of the IG-DMR in normal PSCs. (d) qRT-PCR of the mean relative fold change ± s.d. in ZFP57 in Pg-iPSCs (three biological replicates each) and PWS-iPSCs (three biological replicates each) following introduction of IPW, relative to control (WT) iPSCs.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1–3 (PDF 1164 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stelzer, Y., Sagi, I., Yanuka, O. et al. The noncoding RNA IPW regulates the imprinted DLK1-DIO3 locus in an induced pluripotent stem cell model of Prader-Willi syndrome. Nat Genet 46, 551–557 (2014). https://doi.org/10.1038/ng.2968

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2968

This article is cited by

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