1932

Abstract

Induced pluripotent stem cell (iPSC) technology holds promise for modeling neurodegenerative diseases. Traditional approaches for disease modeling using animal and cellular models require knowledge of disease mutations. However, many patients with neurodegenerative diseases do not have a known genetic cause. iPSCs offer a way to generate patient-specific models and study pathways of dysfunction in an in vitro setting in order to understand the causes and subtypes of neurodegeneration. Furthermore, iPSC-based models can be used to search for candidate therapeutics using high-throughput screening. Here we review how iPSC-based models are currently being used to further our understanding of neurodegenerative diseases, as well as discuss their challenges and future directions.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-biodatasci-092820-025214
2021-07-20
2024-05-05
Loading full text...

Full text loading...

/deliver/fulltext/biodatasci/4/1/annurev-biodatasci-092820-025214.html?itemId=/content/journals/10.1146/annurev-biodatasci-092820-025214&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Petrov D, Mansfield C, Moussy A, Hermine O 2017. ALS clinical trials review: 20 years of failure. Are we any closer to registering a new treatment?. Front. Aging Neurosci. 9:68
    [Google Scholar]
  2. 2. 
    Mehta D, Jackson R, Paul G, Shi J, Sabbagh M. 2017. Why do trials for Alzheimer's disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin. Investig. Drugs 26:6735–39
    [Google Scholar]
  3. 3. 
    Gan L, Cookson MR, Petrucelli L, La Spada AR 2018. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat. Neurosci. 21:101300–9
    [Google Scholar]
  4. 4. 
    Taylor JP, Brown RH, Cleveland DW. 2016. Decoding ALS: from genes to mechanism. Nature 539:7628197–206
    [Google Scholar]
  5. 5. 
    Hodge RD, Bakken TE, Miller JA, Smith KA, Barkan ER et al. 2019. Conserved cell types with divergent features in human versus mouse cortex. Nature 573:777261–68
    [Google Scholar]
  6. 6. 
    Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ et al. 1998. Embryonic stem cell lines derived from human blastocysts. Science 282:53911145–47
    [Google Scholar]
  7. 7. 
    Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:4663–76
    [Google Scholar]
  8. 8. 
    Okita K, Ichisaka T, Yamanaka S. 2007. Generation of germline-competent induced pluripotent stem cells. Nature 448:7151313–17
    [Google Scholar]
  9. 9. 
    Wernig M, Meissner A, Foreman R, Brambrink T, Ku M et al. 2007. In vitro reprogramming of fibro-blasts into a pluripotent ES-cell-like state. Nature 448:7151318–24
    [Google Scholar]
  10. 10. 
    Maherali N, Sridharan R, Xie W, Utikal J, Eminli S et al. 2007. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell 1:155–70
    [Google Scholar]
  11. 11. 
    Guenther MG, Frampton GM, Soldner F, Hockemeyer D, Mitalipova M et al. 2010. Chromatin structure and gene expression programs of human embryonic and induced pluripotent stem cells. Cell Stem Cell 7:2249–57
    [Google Scholar]
  12. 12. 
    Gutierrez-Aranda I, Ramos-Mejia V, Bueno C, Munoz-Lopez M, Real PJ et al. 2010. Human induced pluripotent stem cells develop teratoma more efficiently and faster than human embryonic stem cells regardless the site of injection. Stem Cells 28:91568–70
    [Google Scholar]
  13. 13. 
    Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. 2009. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 27:3275–80
    [Google Scholar]
  14. 14. 
    Hong YJ, Do JT. 2019. Neural lineage differentiation from pluripotent stem cells to mimic human brain tissues. Front. Bioeng. Biotechnol. 7:400
    [Google Scholar]
  15. 15. 
    Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH et al. 2017. iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94:2278–93.e9
    [Google Scholar]
  16. 16. 
    Zhao J, Davis MD, Martens YA, Shinohara M, Graff-Radford NR et al. 2017. APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum. Mol. Genet. 26:142690–700
    [Google Scholar]
  17. 17. 
    Ilieva H, Polymenidou M, Cleveland DW. 2009. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 187:6761–72
    [Google Scholar]
  18. 18. 
    Choi SH, Veeraraghavalu K, Lazarov O, Marler S, Ransohoff RM et al. 2008. Non-cell-autonomous effects of presenilin 1 variants on enrichment-mediated hippocampal progenitor cell proliferation and differentiation. Neuron 59:4568–80
    [Google Scholar]
  19. 19. 
    Lancaster MA, Renner M, Martin C-A, Wenzel D, Bicknell LS et al. 2013. Cerebral organoids model human brain development and microcephaly. Nature 501:7467373–79
    [Google Scholar]
  20. 20. 
    Zhang X, Hu D, Shang Y, Qi X. 2020. Using induced pluripotent stem cell neuronal models to study neurodegenerative diseases. Biochim. Biophys. Acta Mol. Basis Dis. 1866:4165431
    [Google Scholar]
  21. 21. 
    Gonzalez C, Armijo E, Bravo-Alegria J, Becerra-Calixto A, Mays CE, Soto C. 2018. Modeling amyloid beta and tau pathology in human cerebral organoids. Mol. Psychiatry 23:122363–74
    [Google Scholar]
  22. 22. 
    Klimmt J, Dannert A, Paquet D. 2020. Neurodegeneration in a dish: advancing human stem-cell-based models of Alzheimer's disease. Curr. Opin. Neurobiol. 61:96–104
    [Google Scholar]
  23. 23. 
    Wray S. 2021. Modelling neurodegenerative disease using brain organoids. Semin. Cell Dev. Biol. 111:6066
    [Google Scholar]
  24. 24. 
    Kuijlaars J, Oyelami T, Diels A, Rohrbacher J, Versweyveld S et al. 2016. Sustained synchronized neuronal network activity in a human astrocyte co-culture system. Sci. Rep. 6:36529
    [Google Scholar]
  25. 25. 
    Canfield SG, Stebbins MJ, Faubion MG, Gastfriend BD, Palecek SP, Shusta EV. 2019. An isogenic neurovascular unit model comprised of human induced pluripotent stem cell-derived brain microvascular endothelial cells, pericytes, astrocytes, and neurons. Fluids Barriers CNS 16:25
    [Google Scholar]
  26. 26. 
    Cheng X-Y, Biswas S, Li J, Mao C-J, Chechneva O et al. 2020. Human iPSCs derived astrocytes rescue rotenone-induced mitochondrial dysfunction and dopaminergic neurodegeneration in vitro by donating functional mitochondria. Transl. Neurodegener. 9:13
    [Google Scholar]
  27. 27. 
    Lin C-Y, Yoshida M, Li L-T, Ikenaka A, Oshima S et al. 2019. iPSC-derived functional human neuromuscular junctions model the pathophysiology of neuromuscular diseases. JCI Insight 4:18e124299
    [Google Scholar]
  28. 28. 
    Osaki T, Uzel SGM, Kamm RD. 2018. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci. Adv. 4:10eaat5847
    [Google Scholar]
  29. 29. 
    Soto C, Pritzkow S. 2018. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat. Neurosci. 21:101332–40
    [Google Scholar]
  30. 30. 
    Kouroupi G, Taoufik E, Vlachos IS, Tsioras K, Antoniou N et al. 2017. Defective synaptic connectivity and axonal neuropathology in a human iPSC-based model of familial Parkinson's disease. PNAS 114:18E3679–88
    [Google Scholar]
  31. 31. 
    Fujimori K, Ishikawa M, Otomo A, Atsuta N, Nakamura R et al. 2018. Modeling sporadic ALS in iPSC-derived motor neurons identifies a potential therapeutic agent. Nat. Med. 24:101579–89
    [Google Scholar]
  32. 32. 
    Ren Y, Jiang H, Hu Z, Fan K, Wang J et al. 2015. Parkin mutations reduce the complexity of neuronal processes in iPSC-derived human neurons. Stem Cells 33:168–78
    [Google Scholar]
  33. 33. 
    Mehta SR, Tom CM, Wang Y, Bresee C, Rushton D et al. 2018. Human Huntington's disease iPSC-derived cortical neurons display altered transcriptomics, morphology, and maturation. Cell Rep 25:41081–96.e6
    [Google Scholar]
  34. 34. 
    An MC, Zhang N, Scott G, Montoro D, Wittkop T et al. 2012. Genetic correction of Huntington's disease phenotypes in induced pluripotent stem cells. Cell Stem Cell 11:2253–63
    [Google Scholar]
  35. 35. 
    HD iPSC Consort 2012. Induced pluripotent stem cells from patients with Huntington's disease show CAG-repeat-expansion-associated phenotypes. Cell Stem Cell 11:2264–78
    [Google Scholar]
  36. 36. 
    Franz D, Olsen HL, Klink O, Gimsa J. 2017. Automated and manual patch clamp data of human induced pluripotent stem cell-derived dopaminergic neurons. Sci. Data 4:170056
    [Google Scholar]
  37. 37. 
    Chang C-Y, Ting H-C, Liu C-A, Su H-L, Chiou T-W et al. 2020. Induced pluripotent stem cell (iPSC)-based neurodegenerative disease models for phenotype recapitulation and drug screening. Molecules 25:82000
    [Google Scholar]
  38. 38. 
    Hollingsworth EW, Vaughn JE, Orack JC, Skinner C, Khouri J et al. 2017. iPhemap: an atlas of phenotype to genotype relationships of human iPSC models of neurological diseases. EMBO Mol. Med. 9:121742–62
    [Google Scholar]
  39. 39. 
    Xu X, Lei Y, Luo J, Wang J, Zhang S et al. 2013. Prevention of β-amyloid induced toxicity in human iPS cell-derived neurons by inhibition of Cyclin-dependent kinases and associated cell cycle events. Stem Cell Res 10:2213–27
    [Google Scholar]
  40. 40. 
    Malik N, Efthymiou AG, Mather K, Chester N, Wang X et al. 2014. Compounds with species and cell type specific toxicity identified in a 2000 compound drug screen of neural stem cells and rat mixed cortical neurons. NeuroToxicology 45:192–200
    [Google Scholar]
  41. 41. 
    Höing S, Rudhard Y, Reinhardt P, Glatza M, Stehling M et al. 2012. Discovery of inhibitors of microglial neurotoxicity acting through multiple mechanisms using a stem-cell-based phenotypic assay. Cell Stem Cell 11:5620–32
    [Google Scholar]
  42. 42. 
    Burkhardt MF, Martinez FJ, Wright S, Ramos C, Volfson D et al. 2013. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol. Cell Neurosci. 56:355–64
    [Google Scholar]
  43. 43. 
    Brownjohn PW, Smith J, Portelius E, Serneels L, Kvartsberg H et al. 2017. Phenotypic screening identifies modulators of amyloid precursor protein processing in human stem cell models of Alzheimer's disease. Stem Cell Rep 8:4870–82
    [Google Scholar]
  44. 44. 
    Kondo T, Imamura K, Funayama M, Tsukita K, Miyake M et al. 2017. iPSC-based compound screening and in vitro trials identify a synergistic anti-amyloid β combination for Alzheimer's disease. Cell Rep 21:82304–12
    [Google Scholar]
  45. 45. 
    Imamura K, Izumi Y, Watanabe A, Tsukita K, Woltjen K et al. 2017. The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci. Transl. Med. 9:391eaaf3962
    [Google Scholar]
  46. 46. 
    Morimoto S, Takahashi S, Fukushima K, Saya H, Suzuki N et al. 2019. Ropinirole hydrochloride remedy for amyotrophic lateral sclerosis—protocol for a randomized, double-blind, placebo-controlled, single-center, and open-label continuation phase I/IIa clinical trial (ROPALS trial). Regen. Ther. 11:143–66
    [Google Scholar]
  47. 47. 
    Linsley JW, Linsley DA, Lamstein J, Ryan G, Shah K et al. 2020. Super-human cell death detection with biomarker-optimized neural networks. bioRxiv 2020.08.04.237032. https://doi.org/10.1101/2020.08.04.237032
    [Crossref]
  48. 48. 
    Scheeder C, Heigwer F, Boutros M. 2018. Machine learning and image-based profiling in drug discovery. Curr. Opin. Syst. Biol. 10:43–52
    [Google Scholar]
  49. 49. 
    Little D, Ketteler R, Gissen P, Devine MJ. 2019. Using stem cell-derived neurons in drug screening for neurological diseases. Neurobiol. Aging 78:130–41
    [Google Scholar]
  50. 50. 
    Kwart D, Gregg A, Scheckel C, Murphy EA, Paquet D et al. 2019. A large panel of isogenic APP and PSEN1 mutant human iPSC neurons reveals shared endosomal abnormalities mediated by APP β-CTFs, not Aβ. Neuron 104:2256–70.e5
    [Google Scholar]
  51. 51. 
    Lin Y-T, Seo J, Gao F, Feldman HM, Wen H-L et al. 2018. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer's disease phenotypes in human iPSC-derived brain cell types. Neuron 98:61141–54.e7
    [Google Scholar]
  52. 52. 
    Heman-Ackah SM, Manzano R, Hoozemans JJ, Scheper W, Flynn R et al. 2017. Alpha-synuclein induces the unfolded protein response in Parkinson's disease SNCA triplication iPSC-derived neurons. Hum. Mol. Genet. 26:224441–50
    [Google Scholar]
  53. 53. 
    Kim H, Park HJ, Choi H, Chang Y, Park H et al. 2019. Modeling G2019S-LRRK2 sporadic Parkinson's disease in 3D midbrain organoids. Stem Cell Rep 12:3518–31
    [Google Scholar]
  54. 54. 
    Bhinge A, Namboori SC, Zhang X, VanDongen AMJ, Stanton LW. 2017. Genetic correction of SOD1 mutant iPSCs reveals ERK and JNK activated AP1 as a driver of neurodegeneration in amyotrophic lateral sclerosis. Stem Cell Rep 8:4856–69
    [Google Scholar]
  55. 55. 
    Kiskinis E, Sandoe J, Williams LA, Boulting GL, Moccia R et al. 2014. Pathways disrupted in human ALS motor neurons identified through genetic correction of mutant SOD1. Cell Stem Cell 14:6781–95
    [Google Scholar]
  56. 56. 
    Abo-Rady M, Kalmbach N, Pal A, Schludi C, Janosch A et al. 2020. Knocking out C9ORF72 exacerbates axonal trafficking defects associated with hexanucleotide repeat expansion and reduces levels of heat shock proteins. Stem Cell Rep 14:3390–405
    [Google Scholar]
  57. 57. 
    De Santis R, Santini L, Colantoni A, Peruzzi G, de Turris V et al. 2017. FUS mutant human motoneurons display altered transcriptome and microRNA pathways with implications for ALS pathogenesis. Stem Cell Rep 9:51450–62
    [Google Scholar]
  58. 58. 
    Fernández-Santiago R, Carballo-Carbajal I, Castellano G, Torrent R, Richaud Y et al. 2015. Aberrant epigenome in iPSC-derived dopaminergic neurons from Parkinson's disease patients. EMBO Mol. Med. 7:121529–46
    [Google Scholar]
  59. 59. 
    Sackmann C, Hallbeck M. 2020. Oligomeric amyloid-β induces early and widespread changes to the proteome in human iPSC-derived neurons. Sci. Rep. 10:6538
    [Google Scholar]
  60. 60. 
    Markmiller S, Soltanieh S, Server KL, Mak R, Jin W et al. 2018. Context-dependent and disease-specific diversity in protein interactions within stress granules. Cell 172:3590–604.e13
    [Google Scholar]
  61. 61. 
    Conlon EG, Manley JL. 2017. RNA-binding proteins in neurodegeneration: mechanisms in aggregate. Genes Dev 31:151509–28
    [Google Scholar]
  62. 62. 
    Kapeli K, Pratt GA, Vu AQ, Hutt KR, Martinez FJ et al. 2016. Distinct and shared functions of ALS-associated proteins TDP-43, FUS and TAF15 revealed by multisystem analyses. Nat. Commun. 7:12143
    [Google Scholar]
  63. 63. 
    Tank EM, Figueroa-Romero C, Hinder LM, Bedi K, Archbold HC et al. 2018. Abnormal RNA stability in amyotrophic lateral sclerosis. Nat. Commun. 9:2845
    [Google Scholar]
  64. 64. 
    De Santis R, Alfano V, de Turris V, Colantoni A, Santini L et al. 2019. Mutant FUS and ELAVL4 (HuD) aberrant crosstalk in amyotrophic lateral sclerosis. Cell Rep 27:133818–31.e5
    [Google Scholar]
  65. 65. 
    Nijssen J, Aguila J, Hoogstraaten R, Kee N, Hedlund E 2018. Axon-Seq decodes the motor axon transcriptome and its modulation in response to ALS. Stem Cell Rep 11:61565–78
    [Google Scholar]
  66. 66. 
    Volpato V, Smith J, Sandor C, Ried JS, Baud A et al. 2018. Reproducibility of molecular phenotypes after long-term differentiation to human iPSC-derived neurons: a multi-site omics study. Stem Cell Rep 11:4897–911
    [Google Scholar]
  67. 67. 
    Germain P-L, Testa G. 2017. Taming human genetic variability: transcriptomic meta-analysis guides the experimental design and interpretation of iPSC-based disease modeling. Stem Cell Rep 8:61784–96
    [Google Scholar]
  68. 68. 
    Volpato V, Webber C. 2020. Addressing variability in iPSC-derived models of human disease: guidelines to promote reproducibility. Dis. Model. Mech. 13:dmm042317
    [Google Scholar]
  69. 69. 
    Engle SJ, Blaha L, Kleiman RJ. 2018. Best practices for translational disease modeling using human iPSC-derived neurons. Neuron 100:4783–97
    [Google Scholar]
  70. 70. 
    Soneson C, Robinson MD. 2018. Bias, robustness and scalability in single-cell differential expression analysis. Nat. Methods 15:4255–61
    [Google Scholar]
  71. 71. 
    Lang C, Campbell KR, Ryan BJ, Carling P, Attar M et al. 2019. Single-cell sequencing of iPSC-dopamine neurons reconstructs disease progression and identifies HDAC4 as a regulator of Parkinson cell phenotypes. Cell Stem Cell 24:193–106.e6
    [Google Scholar]
  72. 72. 
    Cuomo ASE, Seaton DD, McCarthy DJ, Martinez I, Bonder MJ et al. 2020. Single-cell RNA-sequencing of differentiating iPS cells reveals dynamic genetic effects on gene expression. Nat. Commun. 11:810
    [Google Scholar]
  73. 73. 
    Wang X, Park J, Susztak K, Zhang NR, Li M. 2019. Bulk tissue cell type deconvolution with multi-subject single-cell expression reference. Nat. Commun. 10:380
    [Google Scholar]
  74. 74. 
    Zaitsev K, Bambouskova M, Swain A, Artyomov MN. 2019. Complete deconvolution of cellular mixtures based on linearity of transcriptional signatures. Nat. Commun. 10:2209
    [Google Scholar]
  75. 75. 
    Burke EE, Chenoweth JG, Shin JH, Collado-Torres L, Kim S-K et al. 2020. Dissecting transcriptomic signatures of neuronal differentiation and maturation using iPSCs. Nat. Commun. 11:462
    [Google Scholar]
  76. 76. 
    Mertens J, Reid D, Lau S, Kim Y, Gage FH. 2018. Aging in a dish: iPSC-derived and directly induced neurons for studying brain aging and age-related neurodegenerative diseases. Annu. Rev. Genet. 52:271–93
    [Google Scholar]
  77. 77. 
    Meissner A, Mikkelsen TS, Gu H, Wernig M, Hanna J et al. 2008. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454:7205766–70
    [Google Scholar]
  78. 78. 
    Horvath S. 2013. DNA methylation age of human tissues and cell types. Genome Biol 14:103156
    [Google Scholar]
  79. 79. 
    Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A et al. 2011. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev 25:212248–53
    [Google Scholar]
  80. 80. 
    Studer L, Vera E, Cornacchia D. 2015. Programming and reprogramming cellular age in the era of induced pluripotency. Cell Stem Cell 16:6591–600
    [Google Scholar]
  81. 81. 
    Barnat M, Capizzi M, Aparicio E, Boluda S, Wennagel D et al. 2020. Huntington's disease alters human neurodevelopment. Science 369:6505787–93
    [Google Scholar]
  82. 82. 
    Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y et al. 2011. Modeling familial Alzheimer's disease with induced pluripotent stem cells. Hum. Mol. Genet. 20:234530–39
    [Google Scholar]
  83. 83. 
    Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB et al. 2015. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525:756756–61
    [Google Scholar]
  84. 84. 
    Coyne AN, Zaepfel BL, Hayes L, Fitchman B, Salzberg Y et al. 2020. G4C2 repeat RNA initiates a POM121-mediated reduction in specific nucleoporins in C9orf72 ALS/FTD. Neuron 107:61124–40.e11
    [Google Scholar]
  85. 85. 
    Ho R, Workman MJ, Mathkar P, Wu K, Kim KJ et al. 2020. Single-cell RNA-seq analysis of human iPSC-derived motor neurons resolves early and predictive ALS signatures. bioRxiv 2020.04.27.064584. https://doi.org/10.1101/2020.04.27.064584
    [Crossref]
  86. 86. 
    Vera E, Bosco N, Studer L. 2016. Generating late-onset human iPSC-based disease models by inducing neuronal age-related phenotypes through telomerase manipulation. Cell Rep 17:41184–92
    [Google Scholar]
  87. 87. 
    Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B et al. 2013. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 13:6691–705
    [Google Scholar]
  88. 88. 
    Tang Y, Liu M-L, Zang T, Zhang C-L. 2017. Direct reprogramming rather than iPSC-based reprogramming maintains aging hallmarks in human motor neurons. Front. Mol. Neurosci. 10:359
    [Google Scholar]
  89. 89. 
    Ziff OJ, Patani R. 2019. Harnessing cellular aging in human stem cell models of amyotrophic lateral sclerosis. Aging Cell 18:1e12862
    [Google Scholar]
  90. 90. 
    Liguori I, Russo G, Curcio F, Bulli G, Aran L et al. 2018. Oxidative stress, aging, and diseases. Clin. Interv. Aging 13:757–72
    [Google Scholar]
  91. 91. 
    Saez I, Vilchez D. 2014. The mechanistic links between proteasome activity, aging and age-related diseases. Curr. Genom. 15:138–51
    [Google Scholar]
  92. 92. 
    Nekrasov ED, Vigont VA, Klyushnikov SA, Lebedeva OS, Vassina EM et al. 2016. Manifestation of Huntington's disease pathology in human induced pluripotent stem cell-derived neurons. Mol. Neurodegener. 11:27
    [Google Scholar]
  93. 93. 
    Schwartzentruber J, Foskolou S, Kilpinen H, Rodrigues J, Alasoo K et al. 2018. Molecular and functional variation in iPSC-derived sensory neurons. Nat. Genet. 50:54–61
    [Google Scholar]
  94. 94. 
    Young AM, Kumasaka N, Calvert F, Hammond TR, Knights A et al. 2019. A map of transcriptional heterogeneity and regulatory variation in human microglia. bioRxiv 2019.12.20.874099. https://doi.org/10.1101/2019.12.20.874099
    [Crossref]
  95. 95. 
    Jerber J, Seaton DD, Cuomo ASE, Kumasaka N, Haldane J et al. 2020. Population-scale single-cell RNA-seq profiling across dopaminergic neuron differentiation. bioRxiv 2020.05.21.103820. https://doi.org/10.1101/2020.05.21.103820
    [Crossref]
  96. 96. 
    Griesi-Oliveira K, Fogo MS, Pinto BGG, Alves AY, Suzuki AM et al. 2020. Transcriptome of iPSC-derived neuronal cells reveals a module of co-expressed genes consistently associated with autism spectrum disorder. Mol. Psychiatry. https://doi.org/10.1038/s41380-020-0669-9
    [Crossref] [Google Scholar]
  97. 97. 
    Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S et al. 2019. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:D1D607–13
    [Google Scholar]
  98. 98. 
    HD iPSC Consort 2020. Bioenergetic deficits in Huntington's disease iPSC-derived neural cells and rescue with glycolytic metabolites. Hum. Mol. Genet. 29:111757–71
    [Google Scholar]
  99. 99. 
    Wu Z, Pan S, Chen F, Long G, Zhang C, Yu PS. 2020. A comprehensive survey on graph neural networks. IEEE Trans. Neural Netw. Learn. Syst. 32:424
    [Google Scholar]
  100. 100. 
    Tian R, Gachechiladze MA, Ludwig CH, Laurie MT, Hong JY et al. 2019. CRISPR interference-based platform for multimodal genetic screens in human iPSC-derived neurons. Neuron 104:2239–55.e12
    [Google Scholar]
  101. 101. 
    Glymour C, Zhang K, Spirtes P. 2019. Review of causal discovery methods based on graphical models. Front. Genet 10:524
    [Google Scholar]
/content/journals/10.1146/annurev-biodatasci-092820-025214
Loading
/content/journals/10.1146/annurev-biodatasci-092820-025214
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error