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.

  • Technical Report
  • Published:

Engineering microdeletions and microduplications by targeting segmental duplications with CRISPR

Abstract

Recurrent, reciprocal genomic disorders resulting from non-allelic homologous recombination (NAHR) between near-identical segmental duplications (SDs) are a major cause of human disease, often producing phenotypically distinct syndromes. The genomic architecture of flanking SDs presents a challenge for modeling these syndromes; however, the capability to efficiently generate reciprocal copy number variants (CNVs) that mimic NAHR would represent a valuable modeling tool. We describe here a CRISPR/Cas9 genome engineering method, single-guide CRISPR/Cas targeting of repetitive elements (SCORE), to model reciprocal genomic disorders and demonstrate its capabilities by generating reciprocal CNVs of 16p11.2 and 15q13.3, including alteration of one copy-equivalent of the SDs that mediate NAHR in vivo. The method is reproducible, and RNA sequencing reliably clusters transcriptional signatures from human subjects with in vivo CNVs and their corresponding in vitro models. This new approach will provide broad applicability for the study of genomic disorders and, with further development, may also permit efficient correction of these defects.

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: Generation of 16p11.2 rMDS in human iPSC by CRISPR/Cas9.
Figure 2: Gene expression characterization of putative CNV lines.
Figure 3: Generation of 15q13.3 rMDS in human iPSC by CRISPR/Cas9.

Similar content being viewed by others

References

  1. Stankiewicz, P. & Lupski, J.R. Genome architecture, rearrangements and genomic disorders. Trends Genet. 18, 74–82 (2002).

    Article  CAS  Google Scholar 

  2. Tucker, T. et al. Prevalence of selected genomic deletions and duplications in a French-Canadian population-based sample of newborns. Mol. Genet. Genomic Med. 1, 87–97 (2013).

    Article  CAS  Google Scholar 

  3. Konialis, C. et al. Uncovering recurrent microdeletion syndromes and subtelomeric deletions/duplications through non-selective application of a MLPA-based extended prenatal panel in routine prenatal diagnosis. Prenat. Diagn. 31, 571–577 (2011).

    Article  CAS  Google Scholar 

  4. Maillard, A.M. et al. 16p11.2 European Consortium. The 16p11.2 locus modulates brain structures common to autism, schizophrenia and obesity. Mol. Psychiatry 20, 140–147 (2015).

    Article  CAS  Google Scholar 

  5. Weiss, L.A. et al. Autism Consortium. Association between microdeletion and microduplication at 16p11.2 and autism. N. Engl. J. Med. 358, 667–675 (2008).

    Article  CAS  Google Scholar 

  6. Flicek, P. et al. Ensembl 2013. Nucleic Acids Res. 41, D48–D55 (2013).

    Article  CAS  Google Scholar 

  7. Walters, R.G. et al. A new highly penetrant form of obesity due to deletions on chromosome 16p11.2. Nature 463, 671–675 (2010).

    Article  CAS  Google Scholar 

  8. Golzio, C. et al. KCTD13 is a major driver of mirrored neuroanatomical phenotypes of the 16p11.2 copy number variant. Nature 485, 363–367 (2012).

    Article  CAS  Google Scholar 

  9. Blumenthal, I. et al. Transcriptional consequences of 16p11.2 deletion and duplication in mouse cortex and multiplex autism families. Am. J. Hum. Genet. 94, 870–883 (2014).

    Article  CAS  Google Scholar 

  10. Gasiunas, G., Barrangou, R., Horvath, P. & Siksnys, V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl. Acad. Sci. USA 109, E2579–E2586 (2012).

    Article  CAS  Google Scholar 

  11. Sander, J.D. & Joung, J.K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    Article  CAS  Google Scholar 

  12. Miller, J.C. et al. An improved zinc-finger nuclease architecture for highly specific genome editing. Nat. Biotechnol. 25, 778–785 (2007).

    Article  CAS  Google Scholar 

  13. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    Article  CAS  Google Scholar 

  14. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  Google Scholar 

  15. Makarova, K.S. et al. Evolution and classification of the CRISPR-Cas systems. Nat. Rev. Microbiol. 9, 467–477 (2011).

    Article  CAS  Google Scholar 

  16. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

  17. Platt, R.J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  Google Scholar 

  18. Mandal, P.K. et al. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell 15, 643–652 (2014).

    Article  CAS  Google Scholar 

  19. Canver, M.C. et al. Characterization of genomic deletion efficiency mediated by clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 nuclease system in mammalian cells. J. Biol. Chem. 289, 21312–21324 (2014).

    Article  Google Scholar 

  20. Xiao, A. et al. Chromosomal deletions and inversions mediated by TALENs and CRISPR/Cas in zebrafish. Nucleic Acids Res. 41, e141 (2013).

    Article  CAS  Google Scholar 

  21. Veres, A. et al. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell 15, 27–30 (2014).

    Article  CAS  Google Scholar 

  22. Sheridan, S.D. et al. Epigenetic characterization of the FMR1 gene and aberrant neurodevelopment in human induced pluripotent stem cell models of fragile X syndrome. PLoS One 6, e26203 (2011).

    Article  CAS  Google Scholar 

  23. Shen, Y. et al. Intra-family phenotypic heterogeneity of 16p11.2 deletion carriers in a three-generation Chinese family. Am. J. Med. Genet. B Neuropsychiatr. Genet. 156, 225–232 (2011).

    Article  Google Scholar 

  24. Kumar, R.A. et al. Recurrent 16p11.2 microdeletions in autism. Hum. Mol. Genet. 17, 628–638 (2008).

    Article  CAS  Google Scholar 

  25. Sharp, A.J. et al. A recurrent 15q13.3 microdeletion syndrome associated with mental retardation and seizures. Nat. Genet. 40, 322–328 (2008).

    Article  CAS  Google Scholar 

  26. van Bon, B.W. et al. Further delineation of the 15q13 microdeletion and duplication syndromes: a clinical spectrum varying from non-pathogenic to a severe outcome. J. Med. Genet. 46, 511–523 (2009).

    Article  CAS  Google Scholar 

  27. Stefansson, H. et al. GROUP. Large recurrent microdeletions associated with schizophrenia. Nature 455, 232–236 (2008).

    Article  CAS  Google Scholar 

  28. Antonacci, F. et al. Palindromic GOLGA8 core duplicons promote chromosome 15q13.3 microdeletion and evolutionary instability. Nat. Genet. 46, 1293–1302 (2014).

    Article  CAS  Google Scholar 

  29. Li, J. et al. Efficient inversions and duplications of mammalian regulatory DNA elements and gene clusters by CRISPR/Cas9. J. Mol. Cell Biol. 7, 284–298 (2015).

    Article  CAS  Google Scholar 

  30. Vassos, E. et al. Penetrance for copy number variants associated with schizophrenia. Hum. Mol. Genet. 19, 3477–3481 (2010).

    Article  CAS  Google Scholar 

  31. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  32. Marçais, G. & Kingsford, C. A fast, lock-free approach for efficient parallel counting of occurrences of k-mers. Bioinformatics 27, 764–770 (2011).

    Article  Google Scholar 

  33. Pliatsika, V. & Rigoutsos, I. “Off-Spotter”: very fast and exhaustive enumeration of genomic lookalikes for designing CRISPR/Cas guide RNAs. Biol. Direct 10, 4 (2015).

    Article  Google Scholar 

  34. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  Google Scholar 

  35. McClive, P.J. & Sinclair, A.H. Rapid DNA extraction and PCR-sexing of mouse embryos. Mol. Reprod. Dev. 60, 225–226 (2001).

    Article  CAS  Google Scholar 

  36. Yaffe, M.B. et al. A motif-based profile scanning approach for genome-wide prediction of signaling pathways. Nat. Biotechnol. 19, 348–353 (2001).

    Article  CAS  Google Scholar 

  37. Jiang, L. et al. Synthetic spike-in standards for RNA-seq experiments. Genome Res. 21, 1543–1551 (2011).

    Article  CAS  Google Scholar 

  38. Wu, T.D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    Article  CAS  Google Scholar 

  39. DeLuca, D.S. et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics 28, 1530–1532 (2012).

    Article  CAS  Google Scholar 

  40. Wang, L., Wang, S. & Li, W. RSeQC: quality control of RNA-seq experiments. Bioinformatics 28, 2184–2185 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  42. Verbovšek, U. et al. Expression analysis of all protease genes reveals cathepsin K to be overexpressed in glioblastoma. PLoS One 9, e111819 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Haggarty and S. Sheridan (Center for Human Genetic Research, Massachusetts General Hospital) for generously providing the control iPSC line. These studies were supported by funding from the Simons Foundation for Autism Research (SFARI 328656 and 346042 (M.E.T.) and SFARI 308955 (J.F.G.)), the Nancy Lurie Marks Family Foundation (J.F.G. and M.E.T.), the US National Institutes of Health (R01NS093200 (J.F.G. and M.E.T.), R00MH095867 (M.E.T.), P01GM061354 (J.F.G. and M.E.T.)), the March of Dimes (M.E.T.), NARSAD (M.E.T.) and Autism Speaks (J.F.G.). Research reported in this publication was also partially supported by US National Cancer Institute award P30CA034196 (C.L.) and National Human Genome Research Institute award U41HG007497 (C.L.). We gratefully acknowledge the resources provided by the AGRE consortium and the participating AGRE families. AGRE is a program of Autism Speaks and is supported, in part, by grant 1U24MH081810 from the US National Institute of Mental Health to C.M. Lajonchere. C.L. is supported by an Ewha Womans University Distinguished Professorship. D.J.C.T. is a recipient of the Postdoctoral Research Abroad Program sponsored by Ministry of Science and Technology of Taiwan (102-2917-I-564-012).

Author information

Authors and Affiliations

Authors

Contributions

M.E.T., J.F.G., D.J.C.T., A.S. and I.B. conceived and designed the studies. D.J.C.T., P.M., C.M.S. and A.S. performed molecular studies. A.R., R.L.C. and S.E. performed computational and statistical analyses. X.C., Y.S. and M.S. obtained 16p11.2 family tissue samples. C.Z. and C.L. designed and performed microarray studies. D.J.C.T., J.F.G. and M.E.T. wrote the manuscript.

Corresponding authors

Correspondence to James F Gusella or Michael E Talkowski.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The pedigrees of families with 16p11.2 microdeletion (family 1, 2) or microduplication.

Two 16p11.2 deletion families and one 16p11.2 duplication family used as controls are shown. Individuals with 16p11.2 rMDS are presented as shaded squares. Subject information of 16p11.2 microdeletion Family 1 was reported in Shen et al23. Probands are indicated by arrows.

Supplementary Figure 2 CRISPR strategy and validation

(a) Strategy for PCR characterization of 16p11.2 rMDS in iPSC lines (575 kb deletion). The ~1 kb PCR product is specific to the iPSC harboring 16p11.2 rMDS. (b) Left: PCR characterization reveals a clear ~1 kb band for the CRISPR generated 16p11.2 rMDS iPSC “+” compared to the control iPSC “C” and the CRISPR treated line that failed to generate an rMDS “−“. Right: Sanger sequencing of the 575 kb rMDS deletion (red dashes = deleted bases; red bases = insertion sequence; blue bases = PAM sequence).

Supplementary Figure 3 Overview of the RNAseq experiment.

RNAseq was performed on CRISPR-treated lines harboring putative CNV (one for 575kb deletion, three for 740 kb deletion, five for 740 kb duplication), CRISPR-treated lines without CNV (Cas9 1-3), untreated 8330-controls (Control 1-3), as well as family lines. Those samples labeled were selected for genome-wide DNA microarray analysis.

Supplementary Figure 4 Representative array comparative genomic hybridization (aCGH).

aCGH results generated at Jackson Laboratories are shown for four representative lines harboring putative CNV, and for the iPSCs from the family lines harboring CNV. Normalized log2 ratios are provided for comparison of the CRISPR treated line to controls for each probe. Probes with log2 ratio less than −0.25 are indicated as red dots for deletion, and probes with ratios greater than 0.25 are indicated as green dots for duplication. (a) CRISPR/Cas9 treated 740Kb microdeletion line and (b) family line harboring deletion. (c) CRISPR/Cas9 740 kb microduplication line and (d) family line harboring duplication.

Supplementary Figure 5 The junction sequences of deletions/duplications in 16p11.2 rMDS iPSC lines created by the SCORE approach.

Schematic illustrating the junction sequences found in the individual iPSC lines harboring 16p11.2 microdeletion and microduplication. (a) In the 16p11.2 microdeletion iPSC lines, three different junction sequences observed in the individual iPSC lines (Del 1-3), two different junction sequences were found in the Del 4 line, a single-nucleotide T insertion was detected in four deletion lines (Del 5-8), and the remaining deletion lines were perfectly homologous with the reference (Del 9-12). (b) In the 16p11.2 microduplication iPSC lines, only one aberrant junction sequence found in four of the five iPSC lines. The ratio of certain junction sequence in all amplicons are shown as percentage of amplicons. (Red dashes = deleted bases; red bases = insertion bases; gray triangle = insertion point; orange arrow = CRISPR/Cas9 cut site; those samples labeled were selected for RNAseq analysis)

Supplementary Figure 6 MAPK3 protein expression level in 16p11.2 rMDS iPSC lines.

Representative gel pattern and statistics for MAPK3 protein level (indicated as arrow) in the iPSC lines harboring 16p11.2 microdeletion (575 kb deletion: n = 1, Del 1; 740 kb deletion: n = 3, Del 2-4), microduplication (n = 5, Dup1-5), and 8330 control iPSC lines. All iPSC lines used in western blot analyses were confirmed by RNAseq. β-actin was used as an internal control. Results from western blot showed that the protein level of MAPK3 (MAPK3/β-actin) was significantly increased in 740kb duplication lines (q = 4.391, p = 0.004) but decreased in 740kb deletion lines (q = 2.93, p = 0.031) compared with 8330 control lines ((F2,8) = 43.548, P<0.001). Experiments are in triplicate. Data are mean ± SEM. p <0.05 and **p<0.01 compared with the control group from One-Way ANOVA. The full, uncropped blots are shown in Supplementary Figure 8.

Supplementary Figure 7 Copy number analysis of SCG5 and TJP1 genes.

Compared with 8330 control, there is no significant dosage difference in SCG5 and TJP1 genes (directly outside the CRISPR target region), suggesting the size of deletion/duplication made by CRISPR in our 15q13.3 rMDS iPSC lines are specific to the predicted region. The relative copy number was determined using relative quantitation method (See Online Method). For each gene, experiments are in triplicate. Data are mean ± SEM.

Supplementary Figure 8 MAPK3 protein expression level in 16p11.2 rMDS iPSC lines.

The full, uncropped blots are shown here.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tai, D., Ragavendran, A., Manavalan, P. et al. Engineering microdeletions and microduplications by targeting segmental duplications with CRISPR. Nat Neurosci 19, 517–522 (2016). https://doi.org/10.1038/nn.4235

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4235

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research