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:

7q11.23 dosage-dependent dysregulation in human pluripotent stem cells affects transcriptional programs in disease-relevant lineages

This article has been updated

Abstract

Cell reprogramming promises to make characterization of the impact of human genetic variation on health and disease experimentally tractable by enabling the bridging of genotypes to phenotypes in developmentally relevant human cell lineages. Here we apply this paradigm to two disorders caused by symmetrical copy number variations of 7q11.23, which display a striking combination of shared and symmetrically opposite phenotypes—Williams-Beuren syndrome and 7q-microduplication syndrome. Through analysis of transgene-free patient-derived induced pluripotent stem cells and their differentiated derivatives, we find that 7q11.23 dosage imbalance disrupts transcriptional circuits in disease-relevant pathways beginning in the pluripotent state. These alterations are then selectively amplified upon differentiation of the pluripotent cells into disease-relevant lineages. A considerable proportion of this transcriptional dysregulation is specifically caused by dosage imbalances in GTF2I, which encodes a key transcription factor at 7q11.23 that is associated with the LSD1 repressive chromatin complex and silences its dosage-sensitive targets.

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: Patient cohort and expression of WBS chromosome region (WBSCR) genes according to genotype.
Figure 2: 7q11.23 CNV-dependent and GTF2I-specific transcriptional dysregulation.
Figure 3: GTF2I protein complex and genome-wide occupancy.
Figure 4: GTF2I represses BEND4 in a dosage- and LSD1-dependent manner.
Figure 5: Transcriptional dysregulation in iPSC-derived disease-relevant lineages.
Figure 6: Lineage-specific retention of iPSC transcriptional dysregulation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

Change history

  • 09 January 2015

    In the version of this article initially published online, GTF1I knockdown was incorrectly referred to in the legend for Figure 4c. GTF2I is the correct shRNA target in this experiment. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Cahan, P. & Daley, G.Q. Origins and implications of pluripotent stem cell variability and heterogeneity. Nat. Rev. Mol. Cell Biol. 14, 357–368 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sanders, S.J. et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron 70, 863–885 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pober, B.R. Williams-Beuren syndrome. N. Engl. J. Med. 362, 239–252 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Somerville, M.J. et al. Severe expressive-language delay related to duplication of the Williams-Beuren locus. N. Engl. J. Med. 353, 1694–1701 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Merla, G., Brunetti-Pierri, N., Micale, L. & Fusco, C. Copy number variants at Williams-Beuren syndrome 7q11.23 region. Hum. Genet. 128, 3–26 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Van der Aa, N. et al. Fourteen new cases contribute to the characterization of the 7q11.23 microduplication syndrome. Eur. J. Med. Genet. 52, 94–100 (2009).

    Article  PubMed  Google Scholar 

  7. Osborne, L.R. Animal models of Williams syndrome. Am. J. Med. Genet. C. Semin. Med. Genet. 154C, 209–219 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mervis, C.B. et al. Duplication of GTF2I results in separation anxiety in mice and humans. Am. J. Hum. Genet. 90, 1064–1070 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. O'Leary, J. & Osborne, L.R. Global analysis of gene expression in the developing brain of Gtf2ird1 knockout mice. PLoS ONE 6, e23868 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Campuzano, V. et al. Reduction of NADPH-oxidase activity ameliorates the cardiovascular phenotype in a mouse model of Williams-Beuren syndrome. PLoS Genet. 8, e1002458 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fusco, C. et al. Smaller and larger deletions of the Williams Beuren syndrome region implicate genes involved in mild facial phenotype, epilepsy and autistic traits. Eur. J. Hum. Genet. 22, 64–70 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Prontera, P. et al. Brief report: functional MRI of a patient with 7q11.23 duplication syndrome and autism spectrum disorder. J. Autism Dev. Disord. 44, 2608–2613 (2014).

    Article  PubMed  Google Scholar 

  13. Warren, L. et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618–630 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  15. Pasi, C.E. et al. Genomic instability in induced stem cells. Cell Death Differ. 18, 745–753 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Leung, A. et al. Induced pluripotent stem cell modeling of multisystemic, hereditary transthyretin amyloidosis. Stem Cell Reports 1, 451–463 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Abyzov, A. et al. Somatic copy number mosaicism in human skin revealed by induced pluripotent stem cells. Nature 492, 438–442 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ma, H. et al. Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature 511, 177–183 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Antonell, A. et al. Partial 7q11.23 deletions further implicate GTF2I and GTF2IRD1 as the main genes responsible for the Williams-Beuren syndrome neurocognitive profile. J. Med. Genet. 47, 312–320 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Edelmann, L. et al. An atypical deletion of the Williams-Beuren syndrome interval implicates genes associated with defective visuospatial processing and autism. J. Med. Genet. 44, 136–143 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Hirota, H. et al. Williams syndrome deficits in visual spatial processing linked to GTF2IRD1 and GTF2I on chromosome 7q11.23. Genet. Med. 5, 311–321 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Lazebnik, M.B., Tussie-Luna, M.I., Hinds, P.W. & Roy, A.L. Williams-Beuren syndrome–associated transcription factor TFII-I regulates osteogenic marker genes. J. Biol. Chem. 284, 36234–36239 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Malenfant, P. et al. Association of GTF2i in the Williams-Beuren syndrome critical region with autism spectrum disorders. J. Autism Dev. Disord. 42, 1459–1469 (2012).

    Article  PubMed  Google Scholar 

  24. Sakurai, T. et al. Haploinsufficiency of Gtf2i, a gene deleted in Williams Syndrome, leads to increases in social interactions. Autism Res. 4, 28–39 (2011).

    Article  PubMed  Google Scholar 

  25. Kruse, K., Pankau, R., Gosch, A. & Wohlfahrt, K. Calcium metabolism in Williams-Beuren syndrome. J. Pediatr. 121, 902–907 (1992).

    Article  CAS  PubMed  Google Scholar 

  26. Gothelf, D., Farber, N., Raveh, E., Apter, A. & Attias, J. Hyperacusis in Williams syndrome: characteristics and associated neuroaudiologic abnormalities. Neurology 66, 390–395 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Kern, J.K. et al. The pattern of sensory processing abnormalities in autism. Autism 10, 480–494 (2006).

    Article  PubMed  Google Scholar 

  28. Pankau, R., Partsch, C.J., Winter, M., Gosch, A. & Wessel, A. Incidence and spectrum of renal abnormalities in Williams-Beuren syndrome. Am. J. Med. Genet. 63, 301–304 (1996).

    Article  CAS  PubMed  Google Scholar 

  29. Wang, K.S., Liu, X., Zhang, Q., Aragam, N. & Pan, Y. Parent-of-origin effects of FAS and PDLIM1 in attention-deficit/hyperactivity disorder. J. Psychiatry Neurosci. 37, 46–52 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ohno, K., Kato, H., Funahashi, S., Hasegawa, T. & Sato, K. Characterization of CLP36/Elfin/PDLIM1 in the nervous system. J. Neurochem. 111, 790–800 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Donaudy, F. et al. Nonmuscle myosin heavy-chain gene MYH14 is expressed in cochlea and mutated in patients affected by autosomal dominant hearing impairment (DFNA4). Am. J. Hum. Genet. 74, 770–776 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Antonell, A., Vilardell, M. & Perez Jurado, L.A. Transcriptome profile in Williams-Beuren syndrome lymphoblast cells reveals gene pathways implicated in glucose intolerance and visuospatial construction deficits. Hum. Genet. 128, 27–37 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Ferrero, G.B. et al. An atypical 7q11.23 deletion in a normal IQ Williams-Beuren syndrome patient. Eur. J. Hum. Genet. 18, 33–38 (2010).

    Article  PubMed  Google Scholar 

  34. Morris, C.A. et al. GTF2I hemizygosity implicated in mental retardation in Williams syndrome: genotype-phenotype analysis of five families with deletions in the Williams syndrome region. Am. J. Med. Genet. A. 123A, 45–59 (2003).

    Article  PubMed  Google Scholar 

  35. Makeyev, A.V. & Bayarsaihan, D. Molecular basis of Williams-Beuren syndrome: TFII-I regulated targets involved in craniofacial development. Cleft Palate Craniofac. J. 48, 109–116 (2011).

    Article  PubMed  Google Scholar 

  36. Gocke, C.B. & Yu, H. ZNF198 stabilizes the LSD1-CoREST-HDAC1 complex on chromatin through its MYM-type zinc fingers. PLoS ONE 3, e3255 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hakimi, M.A., Dong, Y., Lane, W.S., Speicher, D.W. & Shiekhattar, R. A candidate X-linked mental retardation gene is a component of a new family of histone deacetylase–containing complexes. J. Biol. Chem. 278, 7234–7239 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Ming, G.L. et al. Cellular reprogramming: recent advances in modeling neurological diseases. J. Neurosci. 31, 16070–16075 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yang, P. et al. RCOR2 is a subunit of the LSD1 complex that regulates ESC property and substitutes for SOX2 in reprogramming somatic cells to pluripotency. Stem Cells 29, 791–801 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Chimge, N.O., Makeyev, A.V., Ruddle, F.H. & Bayarsaihan, D. Identification of the TFII-I family target genes in the vertebrate genome. Proc. Natl. Acad. Sci. USA 105, 9006–9010 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Makeyev, A.V. et al. Diversity and complexity in chromatin recognition by TFII-I transcription factors in pluripotent embryonic stem cells and embryonic tissues. PLoS ONE 7, e44443 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fan, A.X. et al. Genomic and proteomic analysis of transcription factor TFII-I reveals insight into the response to cellular stress. Nucleic Acids Res. 42, 7625–7641 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sabaratnam, M., Turk, J. & Vroegop, P. Case report: autistic disorder and chromosomal abnormality 46, XX duplication (4) p12-p13. Eur. Child Adolesc. Psychiatry 9, 307–311 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Ramasamy, A. et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 17, 1418–1428 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hawrylycz, M.J. et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature 489, 391–399 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Letra, A. et al. Follow-up association studies of chromosome region 9q and nonsyndromic cleft lip/palate. Am. J. Med. Genet. A. 152A, 1701–1710 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shi, Y., Kirwan, P., Smith, J., Robinson, H.P. & Livesey, F.J. Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat. Neurosci. 15, 477–486 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Shi, Y., Kirwan, P. & Livesey, F.J. Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat. Protoc. 7, 1836–1846 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Menendez, L. et al. Directed differentiation of human pluripotent cells to neural crest stem cells. Nat. Protoc. 8, 203–212 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Bonilla-Claudio, M. et al. Bmp signaling regulates a dose-dependent transcriptional program to control facial skeletal development. Development 139, 709–719 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cobourne, M.T. et al. Sonic hedgehog signalling inhibits palatogenesis and arrests tooth development in a mouse model of the nevoid basal cell carcinoma syndrome. Dev. Biol. 331, 38–49 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Dennis, J.F. et al. Mutations in Hedgehog acyltransferase (Hhat) perturb Hedgehog signaling, resulting in severe acrania-holoprosencephaly-agnathia craniofacial defects. PLoS Genet. 8, e1002927 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dobreva, G. et al. SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell 125, 971–986 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Kurosaka, H., Iulianella, A., Williams, T. & Trainor, P.A. Disrupting hedgehog and WNT signaling interactions promotes cleft lip pathogenesis. J. Clin. Invest. 124, 1660–1671 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Metzis, V. et al. Patched1 is required in neural crest cells for the prevention of orofacial clefts. Hum. Mol. Genet. 22, 5026–5035 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Singh, S., Yin, X., Pisano, M.M. & Greene, R.M. Molecular profiles of mitogen activated protein kinase signaling pathways in orofacial development. Birth Defects Res. A Clin. Mol. Teratol. 79, 35–44 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhao, X. et al. The role of SATB2 in skeletogenesis and human disease. Cytokine Growth Factor Rev. 25, 35–44 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Minoux, M. & Rijli, F.M. Molecular mechanisms of cranial neural crest cell migration and patterning in craniofacial development. Development 137, 2605–2621 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Phillips, H.M. et al. Neural crest cell survival is dependent on Rho kinase and is required for development of the mid face in mouse embryos. PLoS ONE 7, e37685 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ge, X. et al. Modeling supravalvular aortic stenosis syndrome with human induced pluripotent stem cells. Circulation 126, 1695–1704 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Barnett, C. et al. Williams syndrome transcription factor is critical for neural crest cell function in Xenopus laevis. Mech. Dev. 129, 324–338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Burgold, T. et al. The H3K27 demethylase JMJD3 is required for maintenance of the embryonic respiratory neuronal network, neonatal breathing, and survival. Cell Rep. 2, 1244–1258 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Mohn, F. et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol. Cell 30, 755–766 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Burgold, T. et al. The histone H3 lysine 27–specific demethylase Jmjd3 is required for neural commitment. PLoS ONE 3, e3034 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bernstein, B.E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol. 25, 681–686 (2007).

    Article  CAS  PubMed  Google Scholar 

  67. Blecher-Gonen, R. et al. High-throughput chromatin immunoprecipitation for genome-wide mapping of in vivo protein-DNA interactions and epigenomic states. Nat. Protoc. 8, 539–554 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Adamo, A. et al. LSD1 regulates the balance between self-renewal and differentiation in human embryonic stem cells. Nat. Cell Biol. 13, 652–659 (2011).

    Article  CAS  PubMed  Google Scholar 

  70. Ortega Muñoz, A. et al. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors. International Bureau of the World Intellectual Property Organization patent number WO2013057322 (2013).

Download references

Acknowledgements

We thank the AFSW (Associazione Famiglie Sindrome di Williams) and AISW (Associazione Italiana Sindrome di Williams) for agreeing to participate and making this study possible and the Genomic and Genetic Disorder Biobank, Galliera Genetic Bank and members of the Telethon Network of Genetic Biobanks (project numbers GTB12001G and GTB12001A), along with the EuroBioBank network, for providing us with specimens. We also thank scientists at the Drug Discovery Unit, Drug Development Program (DDU-DDP) of the European Institute of Oncology (IEO) for sharing with us the two LSD1 inhibitors used in this study; A. Bachi, A. Cattaneo and P. Soffiantini from the Mass Spectrometry service of the FIRC (Fondazione Italiana per la Ricerca sul Cancro) Institute of Molecular Oncology (IFOM); F. Pisati for processing of the teratomas; P. Andrews (University of Sheffield) for sharing two control iPSC lines (CTL2-C1 and CTL2-C2; reprogrammed from CRL-2429 fibroblasts); G. Mostoslavsky and the Center for Regenerative Medicine of Boston University for sharing the BU1Cr3-1 line; G. Barbagiovanni for help with FACS profiling and analysis; and L. Marelli along with all other members of the Testa laboratory for discussion. This work was funded by the European Research Council (consolidator grant number 616441-DISEASEAVATARS to G.T.), the Italian Ministry of Health (Ricerca Corrente to G.T. and G.M. and Bando Giovani Ricercatori 2008 and 2009 to G.T.), the EPIGEN Flagship Project of the Italian National Research Council (G.T.), the Jerome-Lejeune Foundation (G.T. and G.M.), the ERA-NET Neuron Program (G.T.), the Umberto Veronesi Foundation (S.A. and G.D.) and the Federation of European Biochemical Societies (FEBS; fellowship awarded to A.A. to work in the laboratory of G.T.).

Author information

Authors and Affiliations

Authors

Contributions

S.A. initiated this project and set up human iPSC reprogramming and culture, including mRNA-based reprogramming. S.A. and A.A. reprogrammed the lines presented in this study. A.A., S.A., G.D. and M.Z. cultured and characterized iPSC lines and profiled transcriptomes. A.A. performed the biochemical characterization of the GTF2I complex and GTF2I and LSD1 ChIP-seq. A.A. performed the Nanostring experiment. A.A. and V.A. generated the GTF2I RNAi lines. S.A. established human iPSC differentiation into the cortical neural and neural crest lineages. S.A. and A.A. differentiated human iPSCs into cortical neural progenitors. S.A. analyzed NPCs and NCSCs by microarray. S.A. and M.Z. differentiated human iPSCs into NCSCs and MSCs. P.-L.G. performed the computational analysis for the microarray, Nanostring, RNA-seq and ChIP-seq data sets. P.-L.G. created the WikiWilliams-7qGeneBase web platform. G.M. organized the recruitment of patients, including molecular diagnostics and derivation of fibroblast cultures (with L.M., C.F. and B.A.). O.P., M.C. and G.M. performed aCGH analysis. G.P. performed histopathological analysis of teratomas. A.S. diagnosed and recruited patient AtWBS1, E.B. diagnosed and recruited patient WBS4, and P.P. and E.D. diagnosed and recruited patient 7dupASD1. R.M. and J.C. performed RNA-seq on a subset of samples. C.U. provided two control iPSC lines. B.H. provided mRNA reprogramming kits and expertise. P.-L.G., S.A., A.A. and G.T. wrote the manuscript. G.T. conceived, designed and supervised the study.

Corresponding author

Correspondence to Giuseppe Testa.

Ethics declarations

Competing interests

B.H. is the director of research and development for Stemgent and Asterand. All other authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 iPSC line derivation.

(a) Schematic representation of mRNA-mediated reprogramming. The scale bar represents 400 μm. (b) GFP tracking of the mesenchymal-to-epithelial transition during reprogramming. The scale bar represents 50 μm. (c) For each genetic condition, expression of ALP by immunohistochemistry, immunofluorescence for pluripotency markers and staining of three iPSC-derived teratomas expressing markers specific for the three germ layers (right) are shown. The scale bar represents 400 μm. (d) Nanostring measurements for pluripotency markers. (e) Principal-component analysis of the published data comparing iPSCs, IVF-derived hESCs and somatic cell nuclear transfer–derived hESCs (SCNT-hESCs)18. Plotted are the first components able to distinguish, in the published data (after trimmed mean of M values normalization with ours), between iPSCs and SCNT-hESCs. Although our lines span the spectrum of variation on these components, most of them side with SCNT-hESCs and IVF-hESCs.

ALP, alkaline phosphatase; H&E, hematoxylin and eosin; DES, desmin; CK, cytokeratin.

Supplementary Figure 2 Expression of genes of the WBSCR in iPSCs.

(a) Expression of genes included in and directly flanking the WBSCR rearrangement as measured by RNA-seq (see Nanostring validation in Fig. 1c). The order of genes reflects their relative chromosomal position, and the horizontal colored bars indicate which genes are included in the CNVs. (bd) Protein blot (b) and densitometry analyses (c,d) of BAZ1B and GTF2I protein levels in a representative subset of iPSC lines. Changes in GTF2I protein levels are statistically significant according to a two-tailed t test (*p<0.05, **P < 0.01); differences in BAZ1B protein levels, although showing a clear trend, are not statistically significant in this assay.

Supplementary Figure 3 Transcriptional profiling of patient-derived and control iPSCs.

(a–c) Top most-specific GO biological processes enriched among DEGs between control versus 7dupASD (a), control versus WBS (b), and WBS versus 7dupASD (c). (d) Top most-specific enrichments for GO biological processes among the union of DEGs when excluding the external control lines from the analysis.

Supplementary Figure 4 Genes differentially expressed in a symmetrical manner in WBS and 7dupASD iPSCs.

Supplementary Figure 5 Comparison of different antibodies assessed by ChIP-seq and immunoprecipitation assays.

(a) Protein blot validation of the immunoprecipitation efficiency of two different GTF2I antibodies in a control iPSC line. (b) Enrichment plot showing the distribution of reads across the genome; the samples using the Bethyl antibodies have a distinctively greater degree of enrichment compared with the other samples. (c) Most gene targets identified with the Bethyl antibody are also identified with the other antibodies. Importantly, nearly all core targets (i.e., genes with a high-confidence peak across all control and 7dupASD samples) are identified by all three antibodies. (d) ChIP-qPCR validation of GTF2I targets shown as enrichment over 0.05% total input. EOMES and SNAP25 promoters have been used as negative controls.

Supplementary Figure 6 Characterization of NCSC and MSC lines derived from WBS, AtWBS, 7dupASD and control iPSC lines.

NCSC (a) and MSC (b) phase-contrast microscopy shows a similar morphology across the four genotypes. The scale bar represents 400 μm. (c) Immunofluorescence analysis indicates positivity for two NCSC markers (HNK1 and NGFR) in a representative iPSC-derived NCSC line. The scale bar represents 50 μm. (d,e) Flow cytometry analysis indicates a high percentage of HNK1-NGFR and CD73-CD44 double-positive cells in, respectively, NCSC (d) and MSC (e) lines. (f) Plot of RNA-seq expression levels of genes included in the WBSCR at the MSC stage. For better visualization, genes were separated into low/medium (left) and high (right) expression.

Supplementary Figure 7 Characterization of DEGs found in both iPSCs and MSCs.

(a) MSC DEGs that are also DEGs in iPSCs have higher expression. (b) The proportion of overlapping DEGs in MSCs does not correlate with expression levels in iPSCs. (c) The vast majority of DEGs in iPSCs are downregulated in differentiated MSCs, and the overlap between iPSC and MSC DEGs increases with greater fold changes from iPSCs to MSCs.

Supplementary Figure 8 Grafical representation of the core results of the study and their integration into the WikiWilliams/7q11GB database.

(a) Graphical representation of the lineage-specific retention of DEGs. (b) Schematic representation of the data gathered in the open-access WikiWilliams/7qGB.

Supplementary Figure 9 The WikiWilliams/7q11GB web platform.

Representative screenshot of the WikiWilliams/7q11GB database as it appears to users searching for a specific gene of interest. All transcriptomic and genomic data presented in this paper as well as previously published data sets can be easily interrogated in a multilayered format integrated with several biological databases.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1 and 10 (PDF 3864 kb)

Supplementary Table 2

Summary of the copy number variations (CNVs) identified through aCGH. (XLS 34 kb)

Supplementary Table 3

GO biological processes enriched among linear DEGs, defined as mean (control) within a 20–80% range between mean (WBS) and mean (7dupASD), and abs(Pearson correlation) > 0.5 with WBS copy number. (XLSX 117 kb)

Supplementary Table 4

Proportion of DEGs, in each comparison between genotypes, attributable to GTF2I. (XLSX 10 kb)

Supplementary Table 5

GTF2I interactors identified through mass spectrometry analysis. (XLSX 9 kb)

Supplementary Table 6

GTF2I target classification according to ChIP analysis. (XLSX 48 kb)

Supplementary Table 7

GO biological processes enriched among the union of NCSC DEGs. (XLSX 164 kb)

Supplementary Table 8

Comparison of GO biological processes enriched among MSC DEGs and in MSC shuffling. (XLSX 124 kb)

Supplementary Table 9

List of performed experiments (Nat. Biotechnol. 25, 681–686, 2007). (XLSX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Adamo, A., Atashpaz, S., Germain, PL. et al. 7q11.23 dosage-dependent dysregulation in human pluripotent stem cells affects transcriptional programs in disease-relevant lineages. Nat Genet 47, 132–141 (2015). https://doi.org/10.1038/ng.3169

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3169

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