Skip to main content
Physiology logoLink to Physiology
. 2019 Aug 7;34(5):365–375. doi: 10.1152/physiol.00005.2019

Brain Organoids as Tools for Modeling Human Neurodevelopmental Disorders

Jason W Adams 1,2,3, Fernanda R Cugola 1,2, Alysson R Muotri 1,2
PMCID: PMC6863377  PMID: 31389776

Abstract

Brain organoids recapitulate in vitro the specific stages of in vivo human brain development, thus offering an innovative tool by which to model human neurodevelopmental disease. We review here how brain organoids have been used to study neurodevelopmental disease and consider their potential for both technological advancement and therapeutic development.

Keywords: stem cells, organoids, neurodevelopmental disease, disease modeling

Introduction

Investigating neurodevelopmental disease pathogenesis presents considerable challenge due to limited accessibility of human central nervous system (CNS) tissue and poor recapitulation of animal models. The considerable variability in neuroanatomy and connectivity that exists between individuals because of disparate genetic background and environmental exposure (40) introduces another barrier to understanding disease progression. Our knowledge of cellular phenotypes in neuropathological conditions has historically derived primarily from postmortem analysis of CNS tissue, but tissue represents only a single disease time point and may not be well preserved. Functional neuroimaging and animal models have provided noninvasive alternatives for modeling human neurological diseases, but these models are limited by age, sex, pathological heterogeneity, and inconsistent translatability between species (84) (FIGURE 1).

FIGURE 1.

FIGURE 1.

Advantages and disadvantages of methods of neurodevelopmental disease analysis Neurodevelopmental disease mechanisms can be assessed using various modalities, each of which has unique and complementary strengths, with selection dependent on desired readout.

Recent advancement of stem cell technology has introduced a new model by which to study the human brain. Human embryonic stem cells (hESCs) can be induced into neural stem cells and further differentiated into neurons and glia. Due to their ethically controversial origin, however, hESCs have been only loosely adopted. Cellular reprogramming—a technique that reverts patient-specific somatic cells to a pluripotent state, the induced pluripotent stem cells (iPSCs) (122)—offers a way around this problem. iPSCs carry the genotype of the patient donor and, like hESCs, can be differentiated into many different cell types, including neurons and glia. iPSCs thus enable direct in vitro manipulation of relevant cellular phenotypes affected in nervous system diseases.

Initial efforts to model neurological diseases in vitro consisted of neuronal culturing in monolayer. Neurons and glia can be obtained by differentiating neural progenitor cells (NPCs) that are, in turn, directed from iPSCs or ESCs by dual SMAD inhibition (20). NPCs self-organize into rosettes resembling the embryonic neural tube—thereby mimicking in vivo neocortical development—and are thus a predominant method in several differentiation protocols (93). Alternatively, neurons can be induced directly from fibroblasts via forcible expression of several transcription factors—Brn2, Ascl1, and MytL1—or even a single transcription factor, Ngn2 (17, 90, 140). Regardless, culturing neural cells in a 2D environment limits the opportunity for cells to create the structure and organized network connectivity observed in vivo (56). The need for improved in vitro models that more accurately recapitulate human brain complexity and overcome the limitations of 2D models led to the development of 3D brain organoids (FIGURE 1).

The founding discovery of what would come to be called an “organoid” was made by the Sasai laboratory, a pioneering stem cell research group upon whose shoulders all subsequent organoid research stands. They observed that ESCs and iPSCs could self-organize and aggregate in a manner remarkably recapitulative of in vivo development (32, 85). Indeed, although the Sasai group’s initial monographs detailed the self-formation of optic tissue (32, 85), organoids of other neural fate likewise display highly similar organization, gene expression profiling, topographical induction, and temporal development to that seen during fetal neural development (18, 19, 61, 68, 134). Currently, brain organoids can be generated in two ways: non-patterned or patterned. Non-patterned organoids, generally grown embedded in an extracellular matrix, self-organize into different brain regions via endogenous patterning cues (61, 101). Patterned organoids, in contrast, are differentiated into specific brain regions—forebrain or midbrain, for example—by adding external growth factors (74, 92, 98).

Brain organoids, patterned or otherwise, confer investigative ability unavailable in vivo. Evaluation of normal cellular migration and organoid maturation with immunofluorescence and live imaging can be used to examine tissue stratification and to detect early signs of pathology (11, 18). Genomic engineering tools can modify patient-derived stem cells to investigate gene-function relation (114), regionally patterned organoids can be fused to investigate complex interregional dynamics (11, 114, 135), and patch-clamp electrophysiology and optogenetics can assess functional integration (70) (FIGURE 2). Furthermore, recent detection of spontaneous network activity with regular oscillatory waves, similar to that observed in preterm human electroencephalography, demonstrated the capacity of multi-electrode array to dynamically assess network activity in long-term mature organoids (127). The accessibility of iPSCs and brain organoids has thus introduced unprecedented possibility for in vitro neurodevelopmental disease modeling.

FIGURE 2.

FIGURE 2.

Brain organoid disease modeling and analytic techniques Brain organoids can model congenital structural deficits or be subjected to environmental insult. In addition, genetic engineering and multi-organoid fusion enable the assessment of a broader array of disease mechanisms, such as abnormal interregional development. Various methods can be used to evaluate the developmental changes that underlie the disparate phenotypes observed between normal and diseased organoids.

Applications of Brain Organoids as Neurodevelopmental Disease Models

Perhaps the greatest application of brain organoid technology thus far, in vitro modeling of neurodevelopmental disease enables observation of disease progression throughout neurodevelopment and—in conjunction with novel genetic techniques—the opportunity to interrogate underlying pathological mechanisms with previously precluded precision. The versatility of brain organoids permits modeling diseases of either intrinsic (i.e., genetic) or extrinsic (i.e., environmentally mediated) etiology (FIGURE 2). However, despite recent characterization of functional network development (127), developmental disorders in which gross structural abnormalities predominate remain the more accessible for in vitro modeling.

Autosomal Recessive Primary Microcephaly

Autosomal recessive primary microcephaly (MCPH)—a genetic form of microcephaly, itself a clinical entity of heterogeneous etiology—has been linked to genetic mutations in neurodevelopmental pathways (82). Individuals with MCPH clinically portray nonprogressive intellectual disability and neuropathologically exhibit microcephalic brains, with reduction concentrated in the cerebral cortex (132). MCPH has been modeled with organoids generated from patient-derived iPSCs carrying mutation(s) in either ASPM, the gene that codes for a protein involved with mitotic spindle function and that accounts for a plurality of MCPH cases (36), or CDK5RAP2, a gene whose product localizes to the mitotic spindle pole during neurogenesis (61, 63, 132). Those iPSCs in which ASPM expression was downregulated, predicted to impede neural progenitor proliferation, yielded hypoplastic organoids with fewer proliferative cells, decreased neocortex-like morphology, and diminished neuroepithelial structural integrity (63). Functional analysis revealed calcium activity in fewer cells than the controls—implicating neuronal maturation impediment—and decreased synchrony (63). CDK5RAP2-mutant organoids likewise portrayed hypoplasticity with sparse progenitor and neuroepithelial regions (61). Coincident findings of premature neural differentiation and increased neuron quantity were supported by observation of increased neuronal differentiation upon CDK5RAP2 RNAi-knockdown (61). Successful phenotypic rescue upon electroporated expression of CDK5RAP2 protein confirmed viable in vitro recapitulation of MCPH (61).

Microcephaly in Seckel Syndrome

Seckel syndrome—a disorder argued to be on a clinical spectrum with primary microcephaly and whose clinical features include severe pre- and postnatal growth restriction, microcephaly, and intellectual disability (33, 71)—is observed in individuals with mutated centrosomal-P4.1-associated protein (CPAP). Concordant with other microcephalies, organoids of Seckel syndrome patient-derivation featured diminished size with reduced neuroepithelium and disordered progenitor regions (38). In argument for organoids’ biologically intrinsic mechanistic value, Gabriel et al. (38) identified a ciliary role in NPC sustenance and a regulatory role of ciliary length by CPAP. Indeed, detailed interrogation of Seckel organoids revealed more numerous and longer apical progenitor cilia compared with controls (38). Their work thus demonstrated that, in addition to modeling structural deficits, brain organoids enable investigation—in vitro—of molecular mechanisms underlying neurodevelopmental disease.

Macrocephaly

Just as some mutations confer a microcephalic phenotype, others—PTEN loss-of-function, for example—have macrocephalic consequence (15). Cortical organoids generated from PTEN-knockout hESCs exhibited increased neuroepithelial outgrowth, surface area, and volume (64). Most striking, however, was the enhanced gyrification evident in PTEN-mutant organoids. At the cellular level, PTEN-mutants displayed more proliferative cells throughout development, an increase reflected in expanded neural progenitor pools in the ventricular and subventricular zones. It was over these pools of increased proliferative cellularity that the neuroepithelial curvature was more pronounced, an observation later supported biophysically (55). Viral reintroduction of PTEN offered successful phenotypic rescue, with normal neural differentiation and proliferation and the generation of smooth organoids. Indeed, manipulating AKT signaling effected a phenotypic dose response: less AKT signaling yielded smaller, smoother cortical organoids, and increased AKT signaling generated larger organoids with increased fold density (64).

Congenital Lissencephaly

Lissencephaly is a clinical entity characterized by decreased or absent gyration resulting from deficient neuronal migration during development (49). Heterozygous chromosome 17p13.3 deletion results in Miller-Dieker syndrome (MDS), a neurodevelopmental lissencephalic disorder whose clinical characteristics include intellectual disability and seizures (13). This deletion often involves LIS1 and YWHAE, the gene products of which are components of a multiprotein complex that has regulatory function of cytoskeletal protein dynamics (133). Because the LIS1 multiprotein complex is involved with the mitotic spindle and radial glia proliferation, protein defects within the complex yield structural defects during development (137). Organoids developed from MDS patient-derived iPSCs exhibited smaller size and slower expansion compared with controls, and likewise featured structural aberrancies in neuroepithelial loops, including reduced apical and basal membrane length, loop diameter, and overall size of a ventricle-like space (50). Interrogation of MDS organoids at finer resolution revealed disrupted radial glia cytoskeletal networks with diminished projection of truncated microtubules toward the basal membrane (50). Moreover, mitotic spindle cleavage in MDS organoids exhibited a non-random planar switch (from vertical to horizontal and asymmetric) and disrupted adhesion molecules in the ventricular zone. Reduced β-catenin expression in organoids and influential N-cadherin signaling in MDS patient rosettes implicated Wnt pathway disruption in MDS; pharmacological β-catenin activation via CHIR99021 inhibition of GSK3β rescued division plane orientation, cortical loop structure, and apical membrane length (50). Notably, expressing LIS1 or 14-3-3ε (the protein product of YWHAE) in MDS patient cells offered partial restoration of a control phenotype (50).

Individuals with MDS exhibit pronouncedly disrupted cortical cellular distribution: immature neurons appear in the deepest cortical layer, and cell proliferation markers imply disruption of neural progenitor proliferation (112). Organoids from MDS patients—developed to investigate neural progenitor defects in vitro—revealed a disrupted ventricular zone surface, disproportionately frequent horizontal cleavage angles, and outer radial glia with longer mitotic delay before cytokinesis (9). Despite intact migratory processes, MDS organoids exhibited impaired neural migration such that migration was initiated but not sustained and was observed to be slower and less linear. Remarkably, restoration of the LIS1 and 14-3-3ε proteins completely rescued phenotypic aberrancy to wild-type (9).

Karzbrun et al. (55) sought to define the physical forces that regulate cortical folding and then apply that understanding to a lissencephalic organoid model. Physically, organoid surface wrinkling appeared to be subject to the mechanical instability instigated by compression, such that wrinkling starkly emerged when the nuclear density reached a threshold (55). Application to a heterozygous LIS1 mutant organoid model revealed a wider range of cortical thicknesses than control organoids and the longer wavelengths (i.e., distance from one wrinkle crest to another) and decreased curvature expected of lissencephaly (55). LIS1 mutants exhibited decreased expression of cytoskeletal genes, and assessment of ESCs and NPCs with atomic force microscopy indicated the LIS1 mutant cells were twice as elastic as their control counterparts (55). Increased elasticity would be expected to confer resistance against a compressive force inducing folding and might partially explain the decreased gyrification of lissencephaly. Such a model would likewise be consistent with a separate group’s observation of increased neuroepithelial curvature in areas with more proliferating cells (64).

Sandhoff Disease

Sandhoff disease is an autosomal recessive lysosomal storage disorder in which mutant-HEXB insufficiency of β-hexosaminidase causes lysosomal accumulation of GM2 ganglioside (104, 105). The GM2 gangliosidoses clinically feature developmental delay or regression, seizures, macrocephaly, hypotonia, and progressive decline in motor and cognitive functioning (3, 124). Allende et al. (3) developed organoids from a Sandhoff disease patient and from CRISPR/Cas9-corrected HEXB mutant isogenic iPSCs. Compared with HEXB-corrected organoids, Sandhoff disease patient organoids appeared macrocephalic, with increased proliferation and dysfunctional differentiation (3). By week 4 of growth, immunostaining revealed GM2 ganglioside accumulation and even inclusion body development in Sandhoff disease organoids; HEXB-corrected organoids accumulated significantly less GM2 ganglioside (3). In evidence of phenotypic rescue, restoration of β-hexosaminidase via AAV-HEXA/B injection decreased Sandhoff disease organoid size and lessened GM2 accumulation (3).

Tuberous Sclerosis Complex

Tuberous sclerosis complex (TSC) is a multisystem developmental disorder in which heterozygous germline mutations in TSC1 or TSC2—encoding hamartin and tuberin, respectively—cause constitutive activation of mechanistic target of rapamycin complex 1 (mTORC1) (95). The wide-ranging clinical features of TSC include seizures, autism, and intellectual disability; facial angiofibromas; cardiac rhabomyomas; and, most characteristically, cortical tubers (28). Blair et al. (12) sought to investigate the underlying genetic mechanisms of cortical tuber development—the proposed of which include “second hit” loss of heterozygosity, abnormal retrotransposition, and haploinsufficiency (52, 75)—using cortical spheroids generated from CRISPR/Cas9-mutated (both hetero and homozygously) TSC1 or TSC2 hESCs. Homozygous TSC2-knockout spheroids exhibited persistently upregulated mTORC1 during neuronal differentiation, a period customarily of quiescent mTORC1 signaling (12). Homozygous TSC1 or TSC2 knockouts additionally featured enhanced mTOR-dependent, gliogenic JAK-STAT signaling: treatment with the mTOR inhibitor rapamycin reduced pathway activation, decreased astrocytic GFAP expression, and increased expression of neuronal markers (12). Blair et al. (12) then engineered hESCs with a Cre-inducible TSC2 mutation to assess the verity of the second-hit hypothesis. Homozygous, but not heterozygous, spheroids exhibited enlarged, misshapen neurons, increased glial cells, and hypertrophic, filamentous “giant cells” resembling those characteristic of cortical tubers. Spheroids developed from a heterozygous TSC patient, reprogrammed from fibroblasts and engineered to receive a second hit, subsequently confirmed the necessity of biallelic inactivation to develop the dysmorphic cells characteristic of TSC. Just as rapamycin earlier reduced JAK-STAT signaling to combat mTORC1 overactivation, so, too, could it reduce cellular enlargement and recover neuronal—rather than glial—differentiation. A developmental time dependency constrained treatment efficacy, however, such that rapamycin administered too late offered insufficient phenotypic rescue (12).

Rett Syndrome

Rett syndrome is a neurodevelopmental disorder almost universally caused by X-linked mutations in MECP2, the gene encoding methyl-CpG binding protein 2. Clinically, females with Rett syndrome typically experience normally appearing development for 12–18 mo followed by regression—in domains such as hand movement and language—and the onset of gait abnormalities (51, 86); males experience severe congenital encephalopathy and early death (108). Rett patient-derived cells exhibit structural and connectivity defects (73), and a suppressive role of MeCP2 has been implicated in posttranscriptional micro-RNA processing essential for neural development (22). Brain organoids generated from Rett syndrome patients revealed upregulated miR-199 and miR-214, two micro-RNAs that are involved in ERK and AKT signaling pathways for neurogenesis and neural differentiation (78). Moreover, Rett mutant organoids displayed increased ventricular area with decreased ventricular wall thickness, consistent with increased neural progenitors and proliferation with concomitantly impaired neurogenesis (78).

Autism Spectrum Disorder

The term autism spectrum disorder (ASD) describes a set of clinically heterogeneous disorders whose core characteristics include behavioral stereotypies and impairments in social interaction and language (1). ASD is thought to be of both genetic and environmental etiological derivation (100), and, despite the wide-ranging concordance estimates observed for twins, the estimated heritability of ASD is 0.7–0.8 (26, 118). Perhaps underlying the broad clinical heterogeneity, ASD is associated with a broad array of genetic profiles and aberrancies—from widely varying allelic frequencies (e.g., common, rare, etc.) to disparate inheritance patterns (e.g., dominant, X-linked, etc.) to variant type (e.g., insertion/deletion, copy number variant, etc.) (100). Despite this genetic heterogeneity, autism may represent a convergence phenotype of several common pathways (39); in vitro cellular modeling may offer a versatile modality by which to interrogate these various underlying mechanisms (43, 67, 72).

ASD may be attributable to heterogeneous intercellular transcriptomic changes with convergent cellular and molecular pathways (99), one of which may be excitatory/inhibitory imbalance (103, 123). Cortical organoids using iPSCs drawn from macrocephalic ASD individuals revealed, compared with unaffected family members, transcriptomic differences in cell fate and proliferation; cytoskeletal regulation of dynamic cytologic growth, guidance, and maintenance; synaptic assembly and channel functioning, such as potassium ion channel upregulation and ligand-receptor interactions; and upregulated GABAergic enzyme synthesis (74). Despite the low number of individuals tested, the similar findings observed in an independent cohort of ASD individuals increase confidence in the data (72). Cytologic analysis exhibited increased VGAT+ inhibitory synapse formation with concomitantly increased GABAergic neural fate, a finding subsequently confirmed electrophysiologically (74). Transcriptomic upregulation correlated with autism symptom severity, and one of the principal genes upregulated throughout development was FOXG1, the product of which is a developmental transcription factor for the telencephalon and mutations in which confer neurodevelopmental dysfunction (4, 15, 53, 74, 79, 97). Remarkably, interference with FOXG1 expression could revert the GABAergic neural production in ASD organoids to normal (74). Evaluation of another gene frequently involved in autism, the chromatin-remodeling factor CHD8 (chromodomain helicase DNA-binding protein 8), likewise revealed upregulation of transcripts used for GABAergic interneuronal development (130). Although ASD is associated with an excitatory/inhibitory imbalance, the concordant findings of GABAergic increase in these models contrasts with evidence indicating GABAergic reduction in ASD (21, 34).

Microglia in Organoids

Concomitantly with imbalanced excitation and inhibition, individuals with ASD and other neuropsychiatric disorders are proposed to have aberrant synaptic structure and connectivity (7, 48, 73, 94, 126). Microglia function in synaptic remodeling during development (77, 107, 117, 120) and may be involved in ASD and other neurodevelopmental disorders (35, 59, 91, 106). For example, ASD patients exhibit increased microglia density and microglial morphology indicative of activation (81, 121, 125), and wild-type microglial engraftment could impede disease progression and improve phenotype in a Rett syndrome mouse model (31). Indeed, rodents have, to date, been the primary model for studying microglia (10, 117); however, murine and human microglia are thought to differ markedly (111a). Recent derivation of microglia from hiPSCs offers a novel opportunity by which to study microglial function in neurological disease (2, 83). Two groups separately derived microglia-like cells from human iPS cells that could, in organoid co-culture, integrate and respond to either laser- or needle-induced traumatic injury (2, 83), and a third group induced an inflammatory response by exposing neurospheres co-cultured with human microglia to bacterial lipopolysaccharide and flaviviral infection (80). Whereas it had been thought that microglia—of primitive myeloid progenitor yolk-sac derivation (42)—must be introduced to neuroectodermally derived organoids, recent modification of an undirected-differentiation brain organoid protocol, in which all three primitive germ layers are present in early organoidogenesis, revealed intrinsic development of Iba-1-positive microglia from mesodermal precursors (89). Such inclusion of microglia in organoids promises expanded opportunities for disease modeling.

Teratogens Modeling

In addition to genetically based neurodevelopmental disease modeling, the in vitro accessibility and manipulability of brain organoids has also facilitated their use in studying CNS teratogens. Alcohol consumption during pregnancy, perhaps the foremost CNS teratogen, is a major public health concern (111). Clinically, fetal alcohol spectrum disorder features developmental delay, behavioral impairment, and cognitive dysfunction (102). The neuropathological consequences of prenatal alcohol exposure include decreased brain volume and thinning of the corpus callosum (reviewed in Ref. 62). Exposure of brain organoids to alcohol impaired neural maturation, reduced neurite outgrowth, and increased cell death (141). Nicotine is another common teratogen, fetal exposure to which can disrupt development and increase long-lasting health risks for the offspring even into adulthood (14). Brain organoids exposed to nicotine showed impaired neuronal migration and differentiation (131).

As with toxin exposure, viral insult also presents an external mechanism by which neural development can be impaired (6). Exposure to Zika virus in utero has been associated with an increased frequency of congenital microcephaly (88). A causal mechanism underlying this association was established by infecting brain organoids and a mouse model with a Brazilian strain of Zika virus (29). Consistent with the microcephaly observed in mice, the virus depleted the neural progenitor cell pool of organoids by preferentially infecting NPCs, causing cell death, and disrupting the cortical plate (29).

Fusion, Genome Engineering, and the Next Generation of Organoid Technology

Modeling neurological disorders in the past has often required animal or other model systems, and neurodevelopmental disorders present a particular challenge due to limited accessibility. The enhanced breadth and depth of organoid technology using human cells, however, has facilitated the modeling of an ever-increasing array of disorders with both unprecedented recapitulative accuracy and genetic manipulative capacity. Multi-organoid fusion, genomic engineering, and methods of vascularization promise further advancement in disease modeling and pharmacological development.

Multi-Organoid Fusion

Development of the nervous system is a complex yet synchronous dance of multi-modal events, including neuronal migration and the establishment of synaptic interconnectivity. Although organoid modeling methods to date recapitulate neural development remarkably well (61, 98), the disrupted neuronal migration and regional interconnectivity underlying some neurological disorders argues for methods that can model these actions. Fusing together organoids of different pre-specified brain regions offers a modality by which to model such interregional interaction (5, 11, 134, 135). Birey et al. (11) pre-patterned and fused pallium and subpallium spheroids using patient-derived cells to identify GABAergic interneuronal migration deficits present in Timothy syndrome. Timothy syndrome—a multisystem neurodevelopmental disorder resulting from defective L-type calcium channels (LTCC) secondary to mutations in their encoding gene, CACNA1C (CaV1.2)—clinically includes features such as cognitive impairment and autism, cardiac defects, syndactyly, and immune deficiency (119). Assessment of interneuronal migration in fused spheroids revealed increased neuronal saltation frequency but decreased length and speed (11). Because Timothy syndrome results from gain-of-function mutation, adding the LTCC blocker nimodipine to patient-derived cells rescued the defective saltatory phenotype (11). Multi-organoid fusion thus offers a viable means by which to model interregional developmental dynamics as well as phenotypic defects associated with disease.

Genome Engineering

Novel techniques in genomic engineering—including zinc-finger nucleases, transcription activator-like effector nucleases (TALEN), and the CRISPR/Cas9 system (23, 45, 46, 54, 69, 113a)—have enhanced our ability to manipulate the human cellular genome with unprecedented precision (16, 58). The considerable genetic heterogeneity underlying neurological disease demands techniques by which to more efficiently define the effects of genetic variants on neurodevelopmental pathways and phenotypic change (76). These new techniques have enhanced the utility of in vitro disease modeling by enabling pathogenic mutations to be introduced into wild-type stem cells or mutations to be corrected in patient-derived stem cells (113, 128). Despite concern of off-target effects and inefficiency (37, 65, 69, 139), whole-genome sequencing suggests these effects may be less prevalent in iPSCs than feared (116, 129), and updated genome editing strategies—such as target selection and guide protein modification—have sought to minimize these effects (24, 25, 47, 57, 69, 113a, 130, 139). In vitro organoid modeling of several of the diseases featured in this review has only been successful because of genomic manipulability, and further genomic innovation promises disease modeling with even greater complexity—sporadic diseases, for example (44)—and portends enhanced clinical translatability (109).

Transplantation, Vascularization, and Pharmaceutical Development

One of the primary factors limiting further development of organoid technology has been size restriction imposed by insufficient nutrient delivery to the organoid interior due to the absence of vascularization (41, 136). One solution is to appropriate in vivo nutrient distribution machinery (30, 70). Brain organoids engrafted into the retrosplenial cortex of immunodeficient mice successfully vascularized, yielding coincidently increasing graft surface area with minimal or absent apoptosis (70). The engrafted organoids retained differentiability, underwent astrocytic and oligodendrocytic gliogenesis, and functionally integrated (70). Another solution—separate differentiation of patient-derived iPSCs into cortical organoids and endothelial cells with subsequent co-culture—likewise exhibited organoid vascularization (96).

Much of the immediate clinical utility expected of the cortical organoid system is its pharmaceutical potential; the true efficacy of such a model, however, demands vasculature and a blood-brain barrier (BBB). To that end, a vascularized 3D model has been used for multi-disciplinary interrogation of neurotoxicity (110), and dynamic BBB spheroids have been employed to model drug transport and neurotoxicity (24, 87). Following co-culture of endothelial cells, astrocytes, and pericytes, the spheroid exhibited an interior with preponderant astrocytes and an exterior surface of pericytes and endothelial cells (8). Critically, this external surface appeared dynamic, with regulated permeability, expression and activity of a P-glycoprotein efflux pump, and peptide receptor-mediated transcytosis (24). Successful development of these early BBB models suggests organoid technology—irrespective of BBB inclusion—may relieve dependence on imperfect animal models for pharmaceutical development and toxin interrogation, and may well confer future opportunity for personalized therapeutics (66, 110).

Future Challenges

Brain organoid technology has greatly enhanced neurodevelopmental disease research, but, despite its potential, many challenges and technical limitations remain. Organoid protocols may exhibit significant batch variability and can give rise to different compositions of brain regions (60); however, improvements aiming to reduce organoid heterogeneity and improve reproducibility are being developed (98, 114, 128, 138). Organoids are also limited by their minimal or lack of relevant cellular subtypes (e.g., microglia and endothelial cells), which restricts their resemblance of in vivo development to only early gestation (92, 98). Later neocortical development requires vascularization for nutrient diffusion, the absence of which results in interior necrosis (60). As was previously mentioned, however, novel vascularization techniques are initial steps to resolving this challenge (70, 96). Moreover, because most differentiation protocols favor ectodermal fate, microglia, of mesodermal origination, are usually absent. Improved methods of co-culturing or introducing these cells into brain organoids is necessary for accurate disease modeling. Last, to advance the translation of pathophysiological findings from organoids to the human brain, it is imperative to understand whether brain organoids create the neural circuitry observed in the human brain and address any ensuing ethical concerns.

Acknowledgments

This work was supported by grants from the California Institute for Regenerative Medicine (CIRM) DISC2-09649; the National Institutes of Health through 5T32 GM-007198 (to J.W.A.), R01 MH-108528, R01 MH-094753, R01 MH-109885, R01 MH-100175, and R56 MH-109587; SFARI Grant No. 345469; and a NARSAD Independent Investigator Grant to A.R.M.

A.R.M. is a co-founder and has equity interest in TISMOO, a company dedicated to genetic analysis and brain organoid modeling focusing on therapeutic applications customized for autism spectrum disorder and other neurological disorders with genetic origins. The terms of this arrangement have been reviewed and approved by the University of California San Diego in accordance with its conflict-of-interest policies.

J.W.A., F.R.C., and A.R.M. prepared figures; J.W.A. and F.R.C. drafted manuscript; J.W.A., F.R.C., and A.R.M. edited and revised manuscript; J.W.A., F.R.C., and A.R.M. approved final version of manuscript.

References

  • 1.Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet 9: 341–355, 2008. doi: 10.1038/nrg2346 . A corrigendum for this article is available at http://dx.doi.org/10.1038/nrg2861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH, Newman SA, Yeromin AV, Scarfone VM, Marsh SE, Fimbres C, Caraway CA, Fote GM, Madany AM, Agrawal A, Kayed R, Gylys KH, Cahalan MD, Cummings BJ, Antel JP, Mortazavi A, Carson MJ, Poon WW, Blurton-Jones M. iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94: 278–293.e9, 2017. doi: 10.1016/j.neuron.2017.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Allende ML, Cook EK, Larman BC, Nugent A, Brady JM, Golebiowski D, Sena-Esteves M, Tifft CJ, Proia RL. Cerebral organoids derived from Sandhoff disease-induced pluripotent stem cells exhibit impaired neurodifferentiation. J Lipid Res 59: 550–563, 2018. doi: 10.1194/jlr.M081323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ariani F, Hayek G, Rondinella D, Artuso R, Mencarelli MA, Spanhol-Rosseto A, Pollazzon M, Buoni S, Spiga O, Ricciardi S, Meloni I, Longo I, Mari F, Broccoli V, Zappella M, Renieri A. FOXG1 is responsible for the congenital variant of Rett syndrome. Am J Hum Genet 83: 89–93, 2008. doi: 10.1016/j.ajhg.2008.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bagley JA, Reumann D, Bian S, Lévi-Strauss J, Knoblich JA. Fused cerebral organoids model interactions between brain regions. Nat Methods 14: 743–751, 2017. doi: 10.1038/nmeth.4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bale JF., Jr Fetal infections and brain development. Clin Perinatol 36: 639–653, 2009. doi: 10.1016/j.clp.2009.06.005. [DOI] [PubMed] [Google Scholar]
  • 7.Belmonte MK, Allen G, Beckel-Mitchener A, Boulanger LM, Carper RA, Webb SJ. Autism and abnormal development of brain connectivity. J Neurosci 24: 9228–9231, 2004. doi: 10.1523/JNEUROSCI.3340-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bergmann S, Lawler SE, Qu Y, Fadzen CM, Wolfe JM, Regan MS, Pentelute BL, Agar NYR, Cho CF. Blood-brain-barrier organoids for investigating the permeability of CNS therapeutics. Nat Protoc 13: 2827–2843, 2018. doi: 10.1038/s41596-018-0066-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bershteyn M, Nowakowski TJ, Pollen AA, Di Lullo E, Nene A, Wynshaw-Boris A, Kriegstein AR. Human iPSC-derived cerebral organoids model cellular features of lissencephaly and reveal prolonged mitosis of outer radial glia. Cell Stem Cell 20: 435–449.e4, 2017. doi: 10.1016/j.stem.2016.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bilimoria PM, Stevens B. Microglia function during brain development: new insights from animal models. Brain Res 1617: 7–17, 2015. doi: 10.1016/j.brainres.2014.11.032. [DOI] [PubMed] [Google Scholar]
  • 11.Birey F, Andersen J, Makinson CD, Islam S, Wei W, Huber N, Fan HC, Metzler KRC, Panagiotakos G, Thom N, O’Rourke NA, Steinmetz LM, Bernstein JA, Hallmayer J, Huguenard JR, Paşca SP. Assembly of functionally integrated human forebrain spheroids. Nature 545: 54–59, 2017. doi: 10.1038/nature22330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Blair JD, Hockemeyer D, Bateup HS. Genetically engineered human cortical spheroid models of tuberous sclerosis. Nat Med 24: 1568–1578, 2018. doi: 10.1038/s41591-018-0139-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Blazejewski SM, Bennison SA, Smith TH, Toyo-Oka K. Neurodevelopmental genetic diseases associated with microdeletions and microduplications of chromosome 17p13.3. Front Genet 9: 80, 2018. doi: 10.3389/fgene.2018.00080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bruin JE, Gerstein HC, Holloway AC. Long-term consequences of fetal and neonatal nicotine exposure: a critical review. Toxicol Sci 116: 364–374, 2010. doi: 10.1093/toxsci/kfq103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Butler MG, Dasouki MJ, Zhou XP, Talebizadeh Z, Brown M, Takahashi TN, Miles JH, Wang CH, Stratton R, Pilarski R, Eng C. Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet 42: 318–321, 2005. doi: 10.1136/jmg.2004.024646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Byrne SM, Mali P, Church GM. Genome editing in human stem cells. Methods Enzymol 546: 119–138, 2014. doi: 10.1016/B978-0-12-801185-0.00006-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Caiazzo M, Dell’Anno MT, Dvoretskova E, Lazarevic D, Taverna S, Leo D, Sotnikova TD, Menegon A, Roncaglia P, Colciago G, Russo G, Carninci P, Pezzoli G, Gainetdinov RR, Gustincich S, Dityatev A, Broccoli V. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476: 224–227, 2011. doi: 10.1038/nature10284. [DOI] [PubMed] [Google Scholar]
  • 18.Camp JG, Badsha F, Florio M, Kanton S, Gerber T, Wilsch-Bräuninger M, Lewitus E, Sykes A, Hevers W, Lancaster M, Knoblich JA, Lachmann R, Pääbo S, Huttner WB, Treutlein B. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci USA 112: 15672–15677, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cederquist GY, Asciolla JJ, Tchieu J, Walsh RM, Cornacchia D, Resh MD, Studer L. Specification of positional identity in forebrain organoids. Nat Biotechnol 37: 436–444, 2019. doi: 10.1038/s41587-019-0085-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 27: 275–280, 2009. doi: 10.1038/nbt.1529 A corrigendum for this article is available at . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chao HT, Chen H, Samaco RC, Xue M, Chahrour M, Yoo J, Neul JL, Gong S, Lu HC, Heintz N, Ekker M, Rubenstein JLR, Noebels JL, Rosenmund C, Zoghbi HY. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 468: 263–269, 2010. doi: 10.1038/nature09582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cheng T-L, Wang Z, Liao Q, Zhu Y, Zhou W-H, Xu W, Qiu Z. MeCP2 suppresses nuclear microRNA processing and dendritic growth by regulating the DGCR8/Drosha complex. Dev Cell 28: 547–560, 2014. doi: 10.1016/j.devcel.2014.01.032. [DOI] [PubMed] [Google Scholar]
  • 23.Chiba K, Hockemeyer D. Genome Editing in Human Pluripotent Stem Cells Using Site-Specific Nucleases. New York, NY: Humana Press, 2015, p. 267–280. doi: 10.1007/978-1-4939-1862-1_15. [DOI] [PubMed] [Google Scholar]
  • 24.Cho CF, Wolfe JM, Fadzen CM, Calligaris D, Hornburg K, Chiocca EA, Agar NYR, Pentelute BL, Lawler SE. Blood-brain-barrier spheroids as an in vitro screening platform for brain-penetrating agents. Nat Commun 8: 15623, 2017. doi: 10.1038/ncomms15623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae S, Kim JS. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res 24: 132–141, 2014. doi: 10.1101/gr.162339.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Colvert E, Tick B, McEwen F, Stewart C, Curran SR, Woodhouse E, Gillan N, Hallett V, Lietz S, Garnett T, Ronald A, Plomin R, Rijsdijk F, Happé F, Bolton P. Heritability of autism spectrum disorder in a UK population-based twin sample. JAMA Psychiatry 72: 415–423, 2015. doi: 10.1001/jamapsychiatry.2014.3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis complex. N Engl J Med 355: 1345–1356, 2006. doi: 10.1056/NEJMra055323. [DOI] [PubMed] [Google Scholar]
  • 29.Cugola FR, Fernandes IR, Russo FB, Freitas BC, Dias JLM, Guimarães KP, Benazzato C, Almeida N, Pignatari GC, Romero S, Polonio CM, Cunha I, Freitas CL, Brandão WN, Rossato C, Andrade DG, Faria DP, Garcez AT, Buchpigel CA, Braconi CT, Mendes E, Sall AA, Zanotto PMDA, Peron JPS, Muotri AR, Beltrão-Braga PCBB. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534: 267–271, 2016. doi: 10.1038/nature18296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Daviaud N, Friedel RH, Zou H. Vascularization and engraftment of transplanted human cerebral organoids in mouse cortex. eNeuro 5: 1–18, 2018. doi: 10.1523/ENEURO.0219-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Derecki NC, Cronk JC, Lu Z, Xu E, Abbott SBG, Guyenet PG, Kipnis J. Wild-type microglia arrest pathology in a mouse model of Rett syndrome. Nature 484: 105–109, 2012. doi: 10.1038/nature10907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, Okuda S, Sekiguchi K, Adachi T, Sasai Y. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472: 51–56, 2011. doi: 10.1038/nature09941. [DOI] [PubMed] [Google Scholar]
  • 33.Majewski F, Goecke T, Opitz JM; F. M . Studies of microcephalic primordial dwarfism I: approach to a delineation of the Seckel syndrome. Am J Med Genet 12: 7–21, 1982. doi: 10.1002/ajmg.1320120103. [DOI] [PubMed] [Google Scholar]
  • 34.Fatemi SH, Reutiman TJ, Folsom TD, Rooney RJ, Patel DH, Thuras PD. mRNA and protein levels for GABAAalpha4, α5, β1 and GABABR1 receptors are altered in brains from subjects with autism. J Autism Dev Disord 40: 743–750, 2010. doi: 10.1007/s10803-009-0924-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, Erreni M, Markicevic M, Starvaggi-Cucuzza C, Otero K, Piccio L, Cignarella F, Perrucci F, Tamborini M, Genua M, Rajendran L, Menna E, Vetrano S, Fahnestock M, Paolicelli RC, Matteoli M. The microglial innate immune receptor TREM2 is required for synapse elimination and normal brain connectivity. Immunity 48: 979–991.e8, 2018. doi: 10.1016/j.immuni.2018.04.016. [DOI] [PubMed] [Google Scholar]
  • 36.Francis F, Meyer G, Fallet-Bianco C, Moreno S, Kappeler C, Socorro AC, Tuy FPD, Beldjord C, Chelly J. Human disorders of cortical development: from past to present. Eur J Neurosci 23: 877–893, 2006. doi: 10.1111/j.1460-9568.2006.04649.x. [DOI] [PubMed] [Google Scholar]
  • 37.Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, Sander JD. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol 31: 822–826, 2013. doi: 10.1038/nbt.2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gabriel E, Wason A, Ramani A, Gooi LM, Keller P, Pozniakovsky A, Poser I, Noack F, Telugu NS, Calegari F, Šarić T, Hescheler J, Hyman AA, Gottardo M, Callaini G, Alkuraya FS, Gopalakrishnan J. CPAP promotes timely cilium disassembly to maintain neural progenitor pool. EMBO J 35: 803–819, 2016. doi: 10.15252/embj.201593679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Geschwind DH. Autism: many genes, common pathways? Cell 135: 391–395, 2008. doi: 10.1016/j.cell.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Geschwind DH, Flint J. Genetics and genomics of psychiatric disease. Science 349: 1489–1494, 2015. doi: 10.1126/science.aaa8954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Giandomenico SL, Lancaster MA. Probing human brain evolution and development in organoids. Curr Opin Cell Biol 44: 36–43, 2017. doi: 10.1016/j.ceb.2017.01.001. [DOI] [PubMed] [Google Scholar]
  • 42.Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330: 841–845, 2010. doi: 10.1126/science.1194637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Griesi-Oliveira K, Acab A, Gupta AR, Sunaga DY, Chailangkarn T, Nicol X, Nunez Y, Walker MF, Murdoch JD, Sanders SJ, Fernandez TV, Ji W, Lifton RP, Vadasz E, Dietrich A, Pradhan D, Song H, Ming GL, Gu X, Haddad G, Marchetto MCN, Spitzer N, Passos-Bueno MR, State MW, Muotri AR. Modeling non-syndromic autism and the impact of TRPC6 disruption in human neurons. Mol Psychiatry 20: 1350–1365, 2015. doi: 10.1038/mp.2014.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hockemeyer D, Jaenisch R. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 18: 573–586, 2016. doi: 10.1016/j.stem.2016.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hockemeyer D, Soldner F, Beard C, Gao Q, Mitalipova M, DeKelver RC, Katibah GE, Amora R, Boydston EA, Zeitler B, Meng X, Miller JC, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Jaenisch R. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol 27: 851–857, 2009. doi: 10.1038/nbt.1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hockemeyer D, Wang H, Kiani S, Lai CS, Gao Q, Cassady JP, Cost GJ, Zhang L, Santiago Y, Miller JC, Zeitler B, Cherone JM, Meng X, Hinkley SJ, Rebar EJ, Gregory PD, Urnov FD, Jaenisch R. Genetic engineering of human pluripotent cells using TALE nucleases. Nat Biotechnol 29: 731–734, 2011. doi: 10.1038/nbt.1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O, Cradick TJ, Marraffini LA, Bao G, Zhang F. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 31: 827–832, 2013. doi: 10.1038/nbt.2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hutsler JJ, Zhang H. Increased dendritic spine densities on cortical projection neurons in autism spectrum disorders. Brain Res 1309: 83–94, 2010. doi: 10.1016/j.brainres.2009.09.120. [DOI] [PubMed] [Google Scholar]
  • 49.Icenogle DA, Kaplan AM. A review of congenital neurologic malformations. Clin Pediatr (Phila) 20: 565–576, 1981. doi: 10.1177/000992288102000903. [DOI] [PubMed] [Google Scholar]
  • 50.Iefremova V, Manikakis G, Krefft O, Jabali A, Weynans K, Wilkens R, Marsoner F, Brändl B, Müller FJ, Koch P, Ladewig J. An organoid-based model of cortical development identifies non-cell-autonomous defects in Wnt signaling contributing to Miller-Dieker Syndrome. Cell Reports 19: 50–59, 2017. doi: 10.1016/j.celrep.2017.03.047. [DOI] [PubMed] [Google Scholar]
  • 51.Ip JPK, Mellios N, Sur M. Rett syndrome: insights into genetic, molecular and circuit mechanisms. Nat Rev Neurosci 19: 368–382, 2018. doi: 10.1038/s41583-018-0006-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jacob-Hirsch J, Eyal E, Knisbacher BA, Roth J, Cesarkas K, Dor C, Farage-Barhom S, Kunik V, Simon AJ, Gal M, Yalon M, Moshitch-Moshkovitz S, Tearle R, Constantini S, Levanon EY, Amariglio N, Rechavi G. Whole-genome sequencing reveals principles of brain retrotransposition in neurodevelopmental disorders. Cell Res 28: 187–203, 2018. doi: 10.1038/cr.2018.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jacob FD, Ramaswamy V, Andersen J, Bolduc FV. Atypical Rett syndrome with selective FOXG1 deletion detected by comparative genomic hybridization: case report and review of literature. Eur J Hum Genet 17: 1577–1581, 2009. doi: 10.1038/ejhg.2009.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol 14: 49–55, 2013. doi: 10.1038/nrm3486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Karzbrun E, Kshirsagar A, Cohen SR, Hanna JH, Reiner O. Human brain organoids on a chip reveal the physics of folding. Nat Phys 14: 515–522, 2018. doi: 10.1038/s41567-018-0046-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kirwan P, Turner-Bridger B, Peter M, Momoh A, Arambepola D, Robinson HPC, Livesey FJ. Development and function of human cerebral cortex neural networks from pluripotent stem cells in vitro. Development 142: 3178–3187, 2015. doi: 10.1242/dev.123851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, Joung JK. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529: 490–495, 2016. doi: 10.1038/nature16526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Komor AC, Badran AH, Liu DR. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168: 20–36, 2017. doi: 10.1016/j.cell.2016.10.044 A correction for this article is available at . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Koyama R, Ikegaya Y. Microglia in the pathogenesis of autism spectrum disorders. Neurosci Res 100: 1–5, 2015. doi: 10.1016/j.neures.2015.06.005. [DOI] [PubMed] [Google Scholar]
  • 60.Lancaster MA, Knoblich JA. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 345: 1247125, 2014. doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
  • 61.Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA. Cerebral organoids model human brain development and microcephaly. Nature 501: 373–379, 2013. doi: 10.1038/nature12517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lebel C, Roussotte F, Sowell ER. Imaging the impact of prenatal alcohol exposure on the structure of the developing human brain. Neuropsychol Rev 21: 102–118, 2011. doi: 10.1007/s11065-011-9163-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li R, Sun L, Fang A, Li P, Wu Q, Wang X. Recapitulating cortical development with organoid culture in vitro and modeling abnormal spindle-like (ASPM related primary) microcephaly disease. Protein Cell 8: 823–833, 2017. doi: 10.1007/s13238-017-0479-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Li Y, Muffat J, Omer A, Bosch I, Lancaster MA, Sur M, Gehrke L, Knoblich JA, Jaenisch R. Induction of expansion and folding in human cerebral organoids. Cell Stem Cell 20: 385–396.e3, 2017. doi: 10.1016/j.stem.2016.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lin Y, Cradick TJ, Brown MT, Deshmukh H, Ranjan P, Sarode N, Wile BM, Vertino PM, Stewart FJ, Bao G. CRISPR/Cas9 systems have off-target activity with insertions or deletions between target DNA and guide RNA sequences. Nucleic Acids Res 42: 7473–7485, 2014. doi: 10.1093/nar/gku402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lou YR, Leung AW. Next generation organoids for biomedical research and applications. Biotechnol Adv 36: 132–149, 2018. doi: 10.1016/j.biotechadv.2017.10.005 A corrigendum for this article is available at . [DOI] [PubMed] [Google Scholar]
  • 67.Di Lullo E, Kriegstein AR. The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci 18: 573–584, 2017. doi: 10.1038/nrn.2017.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Luo C, Lancaster MA, Castanon R, Nery JR, Knoblich JA, Ecker JR. Cerebral organoids recapitulate epigenomic signatures of the human fetal brain. Cell Reports 17: 3369–3384, 2016. doi: 10.1016/j.celrep.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, Yang L, Church GM. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol 31: 833–838, 2013. doi: 10.1038/nbt.2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mansour AA, Gonçalves JT, Bloyd CW, Li H, Fernandes S, Quang D, Johnston S, Parylak SL, Jin X, Gage FH. An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol 36: 432–441, 2018. doi: 10.1038/nbt.4127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Marakhonov AV, Konovalov FA, Makaov AK, Vasilyeva TA, Kadyshev VV, Galkina VA, Dadali EL, Kutsev SI, Zinchenko RA. Primary microcephaly case from the Karachay-Cherkess Republic poses an additional support for microcephaly and Seckel syndrome spectrum disorders. BMC Med Genomics 11, Suppl 1: 8, 2018. doi: 10.1186/s12920-018-0326-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Marchetto MC, Belinson H, Tian Y, Freitas BC, Fu C, Vadodaria K, Beltrao-Braga P, Trujillo CA, Mendes APD, Padmanabhan K, Nunez Y, Ou J, Ghosh H, Wright R, Brennand K, Pierce K, Eichenfield L, Pramparo T, Eyler L, Barnes CC, Courchesne E, Geschwind DH, Gage FH, Wynshaw-Boris A, Muotri AR. Altered proliferation and networks in neural cells derived from idiopathic autistic individuals. Mol Psychiatry 22: 820–835, 2017. doi: 10.1038/mp.2016.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Marchetto MCN, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y, Chen G, Gage FH, Muotri AR. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143: 527–539, 2010. doi: 10.1016/j.cell.2010.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mariani J, Coppola G, Zhang P, Abyzov A, Provini L, Tomasini L, Amenduni M, Szekely A, Palejev D, Wilson M, Gerstein M, Grigorenko EL, Chawarska K, Pelphrey KA, Howe JR, Vaccarino FM. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell 162: 375–390, 2015. doi: 10.1016/j.cell.2015.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Martin KR, Zhou W, Bowman MJ, Shih J, Au KS, Dittenhafer-Reed KE, Sisson KA, Koeman J, Weisenberger DJ, Cottingham SL, DeRoos ST, Devinsky O, Winn ME, Cherniack AD, Shen H, Northrup H, Krueger DA, MacKeigan JP. The genomic landscape of tuberous sclerosis complex. Nat Commun 8: 15816, 2017. doi: 10.1038/ncomms15816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.McClellan J, King MC. Genetic heterogeneity in human disease. Cell 141: 210–217, 2010. doi: 10.1016/j.cell.2010.03.032. [DOI] [PubMed] [Google Scholar]
  • 77.McKenzie AT, Wang M, Hauberg ME, Fullard JF, Kozlenkov A, Keenan A, Hurd YL, Dracheva S, Casaccia P, Roussos P, Zhang B. Synaptic pruning by microglia is necessary for normal brain development [Online]. Sci Rep 8: 8868, 2018. doi: 10.1038/s41598-018-27293-5. http://www.nature.com/articles/s41598-018-27293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mellios N, Feldman DA, Sheridan SD, Ip JPK, Kwok S, Amoah SK, Rosen B, Rodriguez BA, Crawford B, Swaminathan R, Chou S, Li Y, Ziats M, Ernst C, Jaenisch R, Haggarty SJ, Sur M. MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol Psychiatry 23: 1051–1065, 2018. doi: 10.1038/mp.2017.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mitter D, Pringsheim M, Kaulisch M, Plümacher KS, Schröder S, Warthemann R, Abou Jamra R, Baethmann M, Bast T, Büttel HM, Cohen JS, Conover E, Courage C, Eger A, Fatemi A, Grebe TA, Hauser NS, Heinritz W, Helbig KL, Heruth M, Huhle D, Höft K, Karch S, Kluger G, Korenke GC, Lemke JR, Lutz RE, Patzer S, Prehl I, Hoertnagel K, Ramsey K, Rating T, Rieß A, Rohena L, Schimmel M, Westman R, Zech FM, Zoll B, Malzahn D, Zirn B, Brockmann K. FOXG1 syndrome: genotype-phenotype association in 83 patients with FOXG1 variants. Genet Med 20: 98–108, 2018. doi: 10.1038/gim.2017.75. [DOI] [PubMed] [Google Scholar]
  • 80.Abreu CM, Gama L, Krasemann S, Chesnut M, Odwin-Dacosta S, Hogberg HT, Hartung T, Pamies D. Microglia increase inflammatory responses in iPSC-derived human brainspheres. Front Microbiol 9: 2766, 2018. doi: 10.3389/fmicb.2018.02766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, Courchesne E, Everall IP. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry 68: 368–376, 2010. doi: 10.1016/j.biopsych.2010.05.024. [DOI] [PubMed] [Google Scholar]
  • 82.Morris-Rosendahl DJ, Kaindl AM. What next-generation sequencing (NGS) technology has enabled us to learn about primary autosomal recessive microcephaly (MCPH). Mol Cell Probes 29: 271–281, 2015. doi: 10.1016/j.mcp.2015.05.015. [DOI] [PubMed] [Google Scholar]
  • 83.Muffat J, Li Y, Yuan B, Mitalipova M, Omer A, Corcoran S, Bakiasi G, Tsai LH, Aubourg P, Ransohoff RM, Jaenisch R. Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med 22: 1358–1367, 2016. doi: 10.1038/nm.4189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Muotri AR. The human model: changing focus on autism research. Biol Psychiatry 79: 642–649, 2016. doi: 10.1016/j.biopsych.2015.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Nakano T, Ando S, Takata N, Kawada M, Muguruma K, Sekiguchi K, Saito K, Yonemura S, Eiraku M, Sasai Y. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell 10: 771–785, 2012. doi: 10.1016/j.stem.2012.05.009. [DOI] [PubMed] [Google Scholar]
  • 86.Neul JL, Kaufmann WE, Glaze DG, Christodoulou J, Clarke AJ, Bahi-Buisson N, Leonard H, Bailey MES, Schanen NC, Zappella M, Renieri A, Huppke P, Percy AK; RettSearch Consortium . Rett syndrome: revised diagnostic criteria and nomenclature. Ann Neurol 68: 944–950, 2010. doi: 10.1002/ana.22124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Nzou G, Wicks RT, Wicks EE, Seale SA, Sane CH, Chen A, Murphy SV, Jackson JD, Atala AJ. Human cortex spheroid with a functional blood brain barrier for high-throughput neurotoxicity screening and disease modeling. Sci Rep 8: 7413, 2018. doi: 10.1038/s41598-018-25603-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Oliveira WK De, Cortez-escalante J, Tenório W, Holanda G, Madeleine G. Increase in reported prevalence of microcephaly in infants born to women living in areas with confirmed zika virus transmission during the first trimester of pregnancy—Brazil, 2015. MMWR Morb Mortal Wkly Rep 65: 242–247, 2016. doi: 10.15585/mmwr.mm6509e2. [DOI] [PubMed] [Google Scholar]
  • 89.Ormel PR, Vieira de Sá R, van Bodegraven EJ, Karst H, Harschnitz O, Sneeboer MAM, Johansen LE, van Dijk RE, Scheefhals N, Berdenis van Berlekom A, Ribes Martínez E, Kling S, MacGillavry HD, van den Berg LH, Kahn RS, Hol EM, de Witte LD, Pasterkamp RJ. Microglia innately develop within cerebral organoids. Nat Commun 9: 4167, 2018. doi: 10.1038/s41467-018-06684-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, Yang TQ, Citri A, Sebastiano V, Marro S, Südhof TC, Wernig M. Induction of human neuronal cells by defined transcription factors. Nature 476: 220–223, 2011. doi: 10.1038/nature10202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Pardo CA, Vargas DL, Zimmerman AW. Immunity, neuroglia and neuroinflammation in autism. Int Rev Psychiatry 17: 485–495, 2005. doi: 10.1080/02646830500381930. [DOI] [PubMed] [Google Scholar]
  • 92.Paşca AM, Sloan SA, Clarke LE, Tian Y, Makinson CD, Huber N, Kim CH, Park JY, O’Rourke NA, Nguyen KD, Smith SJ, Huguenard JR, Geschwind DH, Barres BA, Paşca SP. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat Methods 12: 671–678, 2015. doi: 10.1038/nmeth.3415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Paşca SP, Panagiotakos G, Dolmetsch RE. Generating human neurons in vitro and using them to understand neuropsychiatric disease. Annu Rev Neurosci 37: 479–501, 2014. doi: 10.1146/annurev-neuro-062012-170328. [DOI] [PubMed] [Google Scholar]
  • 94.Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci 14: 285–293, 2011. doi: 10.1038/nn.2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Peron A, Northrup H. Tuberous sclerosis complex. Am J Med Genet C Semin Med Genet 178: 274–277, 2018. doi: 10.1002/ajmg.c.31657. [DOI] [PubMed] [Google Scholar]
  • 96.Pham MT, Pollock KM, Rose MD, Cary WA, Stewart HR, Zhou P, Nolta JA, Waldau B. Generation of human vascularized brain organoids. Neuroreport 29: 588–593, 2018. doi: 10.1097/WNR.0000000000001014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hanashima C, Li SC, Shen L, Lai E, Fishell G. Foxg1 suppresses early cortical cell fate. Science 303: 56–59, 2004. doi: 10.1126/science.1090674. [DOI] [PubMed] [Google Scholar]
  • 98.Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C, Yao B, Hamersky GR, Jacob F, Zhong C, Yoon KJ, Jeang W, Lin L, Li Y, Thakor J, Berg DA, Zhang C, Kang E, Chickering M, Nauen D, Ho CY, Wen Z, Christian KM, Shi PY, Maher BJ, Wu H, Jin P, Tang H, Song H, Ming GL. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165: 1238–1254, 2016. doi: 10.1016/j.cell.2016.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Quesnel-Vallières M, Weatheritt RJ, Cordes SP, Blencowe BJ. Autism spectrum disorder: insights into convergent mechanisms from transcriptomics. Nat Rev Genet 20: 51–63, 2019. doi: 10.1038/s41576-018-0066-2. [DOI] [PubMed] [Google Scholar]
  • 100.Ramaswami G, Geschwind DH. Genetics of autism spectrum disorder. Handb Clin Neurol 147: 321–329, 2018. doi: 10.1016/B978-0-444-63233-3.00021-X. [DOI] [PubMed] [Google Scholar]
  • 101.Renner M, Lancaster MA, Bian S, Choi H, Ku T, Peer A, Chung K, Knoblich JA. Self-organized developmental patterning and differentiation in cerebral organoids. EMBO J 36: 1316–1329, 2017. doi: 10.15252/embj.201694700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Riley EP, Infante MA, Warren KR. Fetal alcohol spectrum disorders: an overview. Neuropsychol Rev 21: 73–80, 2011. doi: 10.1007/s11065-011-9166-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rubenstein JLR. Three hypotheses for developmental defects that may underlie some forms of autism spectrum disorder. Curr Opin Neurol 23: 118–123, 2010. doi: 10.1097/WCO.0b013e328336eb13. [DOI] [PubMed] [Google Scholar]
  • 104.Sandhoff K. Variation of beta-N-acetylhexosaminidase-pattern in Tay-Sachs disease. FEBS Lett 4: 351–354, 1969. doi: 10.1016/0014-5793(69)80274-7. [DOI] [PubMed] [Google Scholar]
  • 105.Sandhoff K, Harzer K, Wässle W, Jatzkewitz H. Enzyme alterations and lipid storage in three variants of Tay-Sachs disease. J Neurochem 18: 2469–2489, 1971. doi: 10.1111/j.1471-4159.1971.tb00204.x. [DOI] [PubMed] [Google Scholar]
  • 106.Schafer DP, Heller CT, Gunner G, Heller M, Gordon C, Hammond T, Wolf Y, Jung S, Stevens B. Microglia contribute to circuit defects in Mecp2 null mice independent of microglia-specific loss of Mecp2 expression. eLife 5: e15224, 2016. doi: 10.7554/eLife.15224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74: 691–705, 2012. doi: 10.1016/j.neuron.2012.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Schüle B, Armstrong DD, Vogel H, Oviedo A, Francke U. Severe congenital encephalopathy caused by MECP2 null mutations in males: central hypoxia and reduced neuronal dendritic structure. Clin Genet 74: 116–126, 2008. doi: 10.1111/j.1399-0004.2008.01005.x. [DOI] [PubMed] [Google Scholar]
  • 109.Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, Sasaki N, Boymans S, Cuppen E, van der Ent CK, Nieuwenhuis EES, Beekman JM, Clevers H. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13: 653–658, 2013. doi: 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 110.Schwartz MP, Hou Z, Propson NE, Zhang J, Engstrom CJ, Santos Costa V, Jiang P, Nguyen BK, Bolin JM, Daly W, Wang Y, Stewart R, Page CD, Murphy WL, Thomson JA. Human pluripotent stem cell-derived neural constructs for predicting neural toxicity. Proc Natl Acad Sci USA 112: 12516–12521, 2015. doi: 10.1073/pnas.1516645112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sebastiani G, Borr C, Casanova MA, Tutusaus MP, Ferrero S. The effects of alcohol and drugs of abuse on maternal nutritional profile during pregnancy. Nutrients 10: E1008, 2018. doi: 10.3390/nu10081008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111a.SHAME of the slums. Am J Public Health Nations Health 43: 621–623, 1953. doi: 10.2105/AJPH.43.5_Pt_1.621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sheen VL, Ferland RJ, Neal J, Harney M, Hill RS, Banham A, Brown P, Chenn A, Corbo J, Hecht J, Folkerth R, Walsh CA. Neocortical neuronal arrangement in Miller Dieker syndrome. Acta Neuropathol 111: 489–496, 2006. doi: 10.1007/s00401-005-0010-3. [DOI] [PubMed] [Google Scholar]
  • 113.Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov 16: 115–130, 2017. doi: 10.1038/nrd.2016.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113a.Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. Rationally engineered Cas9 nucleases with improved specificity. Science 351: 84–88, 2016. doi: 10.1126/science.aad5227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sloan SA, Andersen J, Pașca AM, Birey F, Pașca SP. Generation and assembly of human brain region-specific three-dimensional cultures. Nat Protoc 13: 2062–2085, 2018. doi: 10.1038/s41596-018-0032-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Smith C, Gore A, Yan W, Abalde-Atristain L, Li Z, He C, Wang Y, Brodsky RA, Zhang K, Cheng L, Ye Z. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell 15: 12–13, 2014. doi: 10.1016/j.stem.2014.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sominsky L, De Luca S, Spencer SJ. Microglia: Key players in neurodevelopment and neuronal plasticity. Int J Biochem Cell Biol 94: 56–60, 2018. doi: 10.1016/j.biocel.2017.11.012. [DOI] [PubMed] [Google Scholar]
  • 118.Spence SJ. The genetics of autism. Semin Pediatr Neurol 11: 196–204, 2004. doi: 10.1016/j.spen.2004.07.003. [DOI] [PubMed] [Google Scholar]
  • 119.Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, Priori SG, Sanguinetti MC, Keating MT. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell 119: 19–31, 2004. doi: 10.1016/j.cell.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 120.Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke AMM, Lambris JD, Smith SJ, John SWM, Barres BA. The classical complement cascade mediates CNS synapse elimination. Cell 131: 1164–1178, 2007. doi: 10.1016/j.cell.2007.10.036. [DOI] [PubMed] [Google Scholar]
  • 121.Suzuki K, Sugihara G, Ouchi Y, Nakamura K, Futatsubashi M, Takebayashi K, Yoshihara Y, Omata K, Matsumoto K, Tsuchiya KJ, Iwata Y, Tsujii M, Sugiyama T, Mori N. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiatry 70: 49–58, 2013. doi: 10.1001/jamapsychiatry.2013.272. [DOI] [PubMed] [Google Scholar]
  • 122.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861–872, 2007. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 123.Tang X, Kim J, Zhou L, Wengert E, Zhang L, Wu Z, Carromeu C, Muotri AR, Marchetto MCN, Gage FH, Chen G. KCC2 rescues functional deficits in human neurons derived from patients with Rett syndrome. Proc Natl Acad Sci USA 113: 751–756, 2016. doi: 10.1073/pnas.1524013113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Tavasoli AR, Parvaneh N, Ashrafi MR, Rezaei Z, Zschocke J. Clinical presentation and outcome in infantile Sandhoff disease: a case series of 25 patients from Iranian neurometabolic bioregistry with five novel mutations. Orphanet J Rare Dis 13: 130, 2018. doi: 10.1186/s13023-018-0876-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tetreault NA, Hakeem AY, Jiang S, Williams BA, Allman E, Wold BJ, Allman JM. Microglia in the cerebral cortex in autism. J Autism Dev Disord 42: 2569–2584, 2012. doi: 10.1007/s10803-012-1513-0. [DOI] [PubMed] [Google Scholar]
  • 126.Toro R, Konyukh M, Delorme R, Leblond C, Chaste P, Fauchereau F, Coleman M, Leboyer M, Gillberg C, Bourgeron T. Key role for gene dosage and synaptic homeostasis in autism spectrum disorders. Trends Genet 26: 363–372, 2010. doi: 10.1016/j.tig.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 127.Trujillo CA, Gao R, Negraes PD, Chaim IA, Domissy A, Vandenberghe M, Devor A, Yeo GW, Voytek B, Muotri AR. Nested oscillatory dynamics in cortical organoids model early human brain network development. bioRxiv: 358622, 2018. doi: 10.1101/358622. [DOI] [Google Scholar]
  • 128.Trujillo CA, Muotri AR. Brain organoids and the study of neurodevelopment. Trends Mol Med 24: 982–990, 2018. doi: 10.1016/j.molmed.2018.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Cowan CA, Talkowski ME, Musunuru K. 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. doi: 10.1016/j.stem.2014.04.020. An erratum for this article is available at http://dx.doi.org/10.1016/j.stem.2014.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wang P, Mokhtari R, Pedrosa E, Kirschenbaum M, Bayrak C, Zheng D, Lachman HM. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol Autism 8: 11, 2017. doi: 10.1186/s13229-017-0124-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wang Y, Wang L, Zhu Y, Qin J. Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab Chip 18: 851–860, 2018. doi: 10.1039/C7LC01084B. [DOI] [PubMed] [Google Scholar]
  • 132.Woods CG, Bond J, Enard W. Autosomal recessive primary microcephaly (MCPH): a review of clinical, molecular, and evolutionary findings. Am J Hum Genet 76: 717–728, 2005. doi: 10.1086/429930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wynshaw-Boris A. Lissencephaly and LIS1: insights into the molecular mechanisms of neuronal migration and development. Clin Genet 72: 296–304, 2007. doi: 10.1111/j.1399-0004.2007.00888.x. [DOI] [PubMed] [Google Scholar]
  • 134.Xiang Y, Tanaka Y, Cakir B, Patterson B, Kim KY, Sun P, Kang YJ, Zhong M, Liu X, Patra P, Lee SH, Weissman SM, Park IH. hESC-derived thalamic organoids form reciprocal projections when fused with cortical organoids. Cell Stem Cell 24: 487–497.e7, 2019. doi: 10.1016/j.stem.2018.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Xiang Y, Tanaka Y, Patterson B, Kang YJ, Govindaiah G, Roselaar N, Cakir B, Kim KY, Lombroso AP, Hwang SM, Zhong M, Stanley EG, Elefanty AG, Naegele JR, Lee SH, Weissman SM, Park IH. Fusion of regionally specified hPSC-derived organoids models human brain development and interneuron migration. Cell Stem Cell 21: 383–398.e7, 2017. doi: 10.1016/j.stem.2017.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yin X, Mead BE, Safaee H, Langer R, Karp JM, Levy O. Engineering stem cell organoids. Cell Stem Cell 18: 25–38, 2016. doi: 10.1016/j.stem.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yingling J, Youn YH, Darling D, Toyo-Oka K, Pramparo T, Hirotsune S, Wynshaw-Boris A. Neuroepithelial stem cell proliferation requires LIS1 for precise spindle orientation and symmetric division. Cell 132: 474–486, 2008. doi: 10.1016/j.cell.2008.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Yoon SJ, Elahi LS, Pașca AM, Marton RM, Gordon A, Revah O, Miura Y, Walczak EM, Holdgate GM, Fan HC, Huguenard JR, Geschwind DH, Pașca SP. Reliability of human cortical organoid generation. Nat Methods 16: 75–78, 2019. doi: 10.1038/s41592-018-0255-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zhang XH, Tee LY, Wang XG, Huang QS, Yang SH. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids 4: e264, 2015. doi: 10.1038/mtna.2015.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhang Y, Pak C, Han Y, Ahlenius H, Zhang Z, Chanda S, Marro S, Patzke C, Acuna C, Covy J, Xu W, Yang N, Danko T, Chen L, Wernig M, Südhof TC. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78: 785–798, 2013. doi: 10.1016/j.neuron.2013.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zhu Y, Wang L, Yin F, Yu Y, Wang Y. Probing impaired neurogenesis in human brain organoids exposed to alcohol. Integr Biol (Camb) 9: 968–978, 2017. doi: 10.1039/C7IB00105C. [DOI] [PubMed] [Google Scholar]

Articles from Physiology are provided here courtesy of American Physiological Society

RESOURCES