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.

  • Perspective
  • Published:

Modeling the complex genetic architectures of brain disease

Abstract

The genetic architecture of each individual comprises common and rare variants that, acting alone and in combination, confer risk of disease. The cell-type-specific and/or context-dependent functional consequences of the risk variants linked to brain disease must be resolved. Coupling human induced pluripotent stem cell (hiPSC)-based technology with CRISPR-based genome engineering facilitates precise isogenic comparisons of variants across genetic backgrounds. Although functional-validation studies are typically performed on one variant in isolation and in one cell type at a time, complex genetic diseases require multiplexed gene perturbations to interrogate combinations of genes and resolve physiologically relevant disease biology. Our aim is to discuss advances at the intersection of genomics, hiPSCs and CRISPR. A better understanding of the molecular mechanisms underlying disease risk will improve genetic diagnosis, drive phenotypic drug discovery and pave the way toward precision medicine.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: 2D versus 3D culture systems to resolve cell-type effects and interactions.
Fig. 2: Unidirectional and bidirectional network perturbations.
Fig. 3: Coupling hiPSC and CRISPR platforms to accelerate functional validations of brain disease-risk loci.

Similar content being viewed by others

References

  1. Sullivan, P. F. & Geschwind, D. H. Defining the genetic, genomic, cellular, and diagnostic architectures of psychiatric disorders. Cell 177, 162–183 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Flint, J. & Ideker, T. The great hairball gambit. PLoS Genet. 15, e1008519 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Skene, N. G. et al. Genetic identification of brain cell types underlying schizophrenia. Nat. Genet. 50, 825–833 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Sims, R. et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat. Genet. 49, 1373–1384 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Brennand, K. et al. Phenotypic differences in hiPSC NPCs derived from patients with schizophrenia. Mol. Psychiatry 20, 361–368 (2015).

    Article  PubMed  CAS  Google Scholar 

  7. Mariani, J. et al. Modeling human cortical development in vitro using induced pluripotent stem cells. Proc. Natl Acad. Sci. USA 109, 12770–12775 (2012).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  8. Paşca, A. M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671–678 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Qian, X. et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165, 1238–1254 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Nicholas, C. R. et al. Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. Cell Stem Cell 12, 573–586 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Rajarajan, P., Flaherty, E., Akbarian, S. & Brennand, K.J. CRISPR-based functional evaluation of schizophrenia risk variants. Schizophr. Res. https://doi.org/10.1016/j.schres.2019.06.017 (2019).

  12. Brennand, K. J. Personalized medicine in a dish: the growing possibility of neuropsychiatric disease drug discovery tailored to patient genetic variants using stem cells. Stem Cell Investig. 4, 91 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Haggarty, S. J., Silva, M. C., Cross, A., Brandon, N. J. & Perlis, R. H. Advancing drug discovery for neuropsychiatric disorders using patient-specific stem cell models. Mol. Cell. Neurosci. 73, 104–115 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Yang, N. et al. Generation of pure GABAergic neurons by transcription factor programming. Nat. Methods 14, 621–628 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Theka, I. et al. Rapid generation of functional dopaminergic neurons from human induced pluripotent stem cells through a single-step procedure using cell lineage transcription factors. Stem Cells Transl. Med. 2, 473–479 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Vadodaria, K. C. et al. Generation of functional human serotonergic neurons from fibroblasts. Mol. Psychiatry 21, 49–61 (2016).

    Article  PubMed  CAS  Google Scholar 

  18. Goto, K. et al. Simple derivation of spinal motor neurons from ESCs/iPSCs using Sendai virus vectors. Mol. Ther. Methods Clin. Dev. 4, 115–125 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Canals, I. et al. Rapid and efficient induction of functional astrocytes from human pluripotent stem cells. Nat. Methods 15, 693–696 (2018).

    Article  PubMed  CAS  Google Scholar 

  20. Ehrlich, M. et al. Rapid and efficient generation of oligodendrocytes from human induced pluripotent stem cells using transcription factors. Proc. Natl Acad. Sci. USA 114, E2243–E2252 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  21. Kuijlaars, J. et al. Sustained synchronized neuronal network activity in a human astrocyte co-culture system. Sci. Rep. 6, 36529 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Marro, S. G. et al. Neuroligin-4 regulates excitatory synaptic transmission in human neurons. Neuron 103, 617–626.e6 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  23. Kawaguchi, K., Kageyama, R. & Sano, M. Topological defects control collective dynamics in neural progenitor cell cultures. Nature 545, 327–331 (2017).

    Article  PubMed  CAS  Google Scholar 

  24. Dezonne, R. S. et al. Derivation of functional human astrocytes from cerebral organoids. Sci. Rep. 7, 45091 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Marton, R. M. et al. Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat. Neurosci. 22, 484–491 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Mansour, A. A. et al. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 36, 432–441 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Cakir, B. et al. Engineering of human brain organoids with a functional vascular-like system. Nat. Methods 16, 1169–1175 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  28. Abud, E. M. et al. iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94, 278–293.e9 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Birey, F. et al. Assembly of functionally integrated human forebrain spheroids. Nature 545, 54–59 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Xiang, Y. et al. Fusion of regionally specified hPSC-derived organoids models human brain development and interneuron migration. Cell Stem Cell 21, 383–398.e7 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Bagley, J. A., Reumann, D., Bian, S., Lévi-Strauss, J. & Knoblich, J. A. Fused cerebral organoids model interactions between brain regions. Nat. Methods 14, 743–751 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Xiang, Y. et al. hESC-derived thalamic organoids form reciprocal projections when fused with cortical organoids. Cell Stem Cell 24, 487–497.e7 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Paşca, S. P. Assembling human brain organoids. Science 363, 126–127 (2019).

    Article  PubMed  CAS  Google Scholar 

  34. Quadrato, G. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48–53 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Velasco, S. et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 570, 523–527 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Yoon, S. J. et al. Reliability of human cortical organoid generation. Nat. Methods 16, 75–78 (2019).

    Article  PubMed  CAS  Google Scholar 

  37. Adli, M. The CRISPR tool kit for genome editing and beyond. Nat. Commun. 9, 1911 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Rajarajan, P., Gil, S. E., Brennand, K. J. & Akbarian, S. Spatial genome organization and cognition. Nat. Rev. Neurosci. 17, 681–691 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Benner, C. et al. FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics 32, 1493–1501 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Dobbyn, A. et al. Landscape of conditional eQTL in dorsolateral prefrontal cortex and co-localization with schizophrenia GWAS. Am. J. Hum. Genet. 102, 1169–1184 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Huckins, L. M. et al. Gene expression imputation across multiple brain regions provides insights into schizophrenia risk. Nat. Genet. 51, 659–674 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Zetsche, B. et al. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat. Biotechnol. 35, 31–34 (2017).

    Article  PubMed  CAS  Google Scholar 

  45. Kleinstiver, B. P. et al. Engineered CRISPR–Cas12a variants with increased activities and improved targeting ranges for gene, epigenetic and base editing. Nat. Biotechnol. 37, 276–282 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Liu, J.-J. et al. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 566, 218–223 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Grünewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Thakore, P. I. et al. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nat. Methods 12, 1143–1149 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Chavez, A. et al. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods 12, 326–328 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Schrode, N. et al. Synergistic effects of common schizophrenia risk variants. Nat. Genet. 51, 1475–1485 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Zhang, X. et al. Multiplex gene regulation by CRISPR-ddCpf1. Cell Discov. 3, 17018 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Konermann, S. et al. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell 173, 665–676.e14 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Lu, C. et al. Overexpression of NEUROG2 and NEUROG1 in human embryonic stem cells produces a network of excitatory and inhibitory neurons. FASEB J. 33, 5287–5299 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Liu, Y. et al. CRISPR activation screens systematically identify factors that drive neuronal fate and reprogramming. Cell Stem Cell 23, 758–771.e8 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Tian, R. et al. CRISPR interference-based platform for multimodal genetic screens in human iPSC-derived neurons. Neuron 104, 239–255.e12 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  59. Dixit, A. et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866.e17 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882.e21 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Mimitou, E. P. et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat. Methods 16, 409–412 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Inoue, F., Kreimer, A., Ashuach, T., Ahituv, N. & Yosef, N. Identification and massively parallel characterization of regulatory elements driving neural induction. Cell Stem Cell 25, 713–727.e10 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Rowe, R. G. & Daley, G. Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 20, 377–388 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  65. Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Topol, A. et al. Dysregulation of miRNA-9 in a subset of schizophrenia patient-derived neural progenitor cells. Cell Rep. 15, 1024–1036 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Brennand, K. J. et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 473, 221–225 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Marchetto, M. C. et al. Altered proliferation and networks in neural cells derived from idiopathic autistic individuals. Mol. Psychiatry 22, 820–835 (2017).

    Article  PubMed  CAS  Google Scholar 

  69. Choi, S. H. et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274–278 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Readhead, B. et al. Expression-based drug screening of neural progenitor cells from individuals with schizophrenia. Nat. Commun. 9, 4412 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Mertens, J. et al. Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder. Nature 527, 95–99 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Stern, S., Linker, S., Vadodaria, K. C., Marchetto, M. C. & Gage, F. H. Prediction of response to drug therapy in psychiatric disorders. Open Biol. 8, 180031 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Hou, L. et al. Genetic variants associated with response to lithium treatment in bipolar disorder: a genome-wide association study. Lancet 387, 1085–1093 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Jiang, X. et al. Sodium valproate rescues expression of TRANK1 in iPSC-derived neural cells that carry a genetic variant associated with serious mental illness. Mol. Psychiatry 24, 613–624 (2019).

    Article  PubMed  CAS  Google Scholar 

  75. Moffat, J. G., Vincent, F., Lee, J. A., Eder, J. & Prunotto, M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat. Rev. Drug Discov. 16, 531–543 (2017).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Poon, A. et al. Modeling neurodegenerative diseases with patient-derived induced pluripotent cells: possibilities and challenges. N. Biotechnol. 39, 190–198 (2017).

    Article  PubMed  CAS  Google Scholar 

  78. Zhou, M. et al. Seamless genetic conversion of SMN2 to SMN1 via CRISPR/Cpf1 and single-stranded oligodeoxynucleotides in spinal muscular atrophy patient-specific induced pluripotent stem cells. Hum. Gene Ther. 29, 1252–1263 (2018).

    Article  PubMed  CAS  Google Scholar 

  79. Ho, S. M. et al. Evaluating synthetic activation and repression of neuropsychiatric-related genes in hiPSC-derived NPCs, neurons, and astrocytes. Stem Cell Reports 9, 615–628 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Nihongaki, Y. et al. CRISPR–Cas9-based photoactivatable transcription systems to induce neuronal differentiation. Nat. Methods 14, 963–966 (2017).

    Article  PubMed  CAS  Google Scholar 

  82. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  PubMed  CAS  Google Scholar 

  83. Tak, Y. E. et al. Inducible and multiplex gene regulation using CRISPR–Cpf1-based transcription factors. Nat. Methods 14, 1163–1166 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Klann, T. S. et al. CRISPR–Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat. Biotechnol. 35, 561–568 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Williams, R. M. et al. Genome and epigenome engineering CRISPR toolkit for in vivo modulation of cis-regulatory interactions and gene expression in the chicken embryo. Development 145, dev160333 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247.e17 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Ziller, M. J. et al. Dissecting the functional consequences of de novo DNA methylation dynamics in human motor neuron differentiation and physiology. Cell Stem Cell 22, 559–574.e9 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Liu, X. S. et al. Rescue of fragile X syndrome neurons by DNA methylation editing of the FMR1 gene. Cell 172, 979–992.e6 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Abudayyeh, O. O. et al. RNA targeting with CRISPR–Cas13. Nature 550, 280–284 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Cox, D. B. T. et al. RNA editing with CRISPR-Cas13. Science 358, 1019–1027 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was partially supported by US National Institutes of Health (NIH) grants R56 MH101454 (K.J.B.), R01 MH106056 (K.J.B.) and R01 MH109897 (K.J.B.).

Author information

Authors and Affiliations

Authors

Contributions

M.B.F., T.A. and K.J.B. wrote the manuscript.

Corresponding author

Correspondence to Kristen J. Brennand.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fernando, M.B., Ahfeldt, T. & Brennand, K.J. Modeling the complex genetic architectures of brain disease. Nat Genet 52, 363–369 (2020). https://doi.org/10.1038/s41588-020-0596-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-020-0596-3

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