Abstract
Recent advances in human stem cell and genome engineering have enabled the generation of genetically-defined human cellular models for brain disorders. These models can be established from a patient’s own cells and can be genetically engineered to generate isogenic, controlled systems for mechanistic studies. Given the challenges of obtaining and working with primary human brain tissue, these models fill a critical gap in our understanding of normal and abnormal human brain development and provide an important complement to animal models. Recently, there has been major progress in modeling the neuropathophysiology of the canonical “mTORopathy” Tuberous Sclerosis Complex (TSC) with such approaches. Studies using two- and three-dimensional cultures of human neurons and glia have provided new insights into how mutations in the TSC1 and TSC2 genes impact human neural development and function. Here we discuss recent progress in human stem cell-based modeling of TSC and highlight challenges and opportunities for further efforts in this area.
Keywords: Tuberous Sclerosis Complex, TSC1, TSC2, mTOR, cortical tuber, human pluripotent stem cells, brain organoids, neurons, astrocytes, disease modeling, CRISPR/Cas9
Introduction
Clinical presentation of TSC
Tuberous Sclerosis Complex (TSC) is a multisystem developmental disorder with a prevalence of about 1 in 6000 births worldwide. TSC causes benign tumors, called hamartomas, in multiple organs including the skin, lungs, kidney, and brain (Crino et al., 2006). Hallmark pathologies of TSC are cortical tubers, which are focal developmental malformations consisting of enlarged and dysplastic neurons, glia and giant cells in the cortex (Mizuguchi and Takashima, 2001). TSC is associated with significant neurological and psychiatric impairments which include epilepsy in about 80% of individuals and variable degrees of intellectual disability (Chu-Shore et al., 2010). TSC patients also have high rates of autism spectrum disorder, attention deficit hyperactivity disorder, and other behavioral and affective disorders (Curatolo et al., 2015). Epilepsy is a major concern in TSC as it can begin in infancy and becomes intractable in about two-thirds of patients (Thiele, 2010). In some cases, surgical resection of the affected brain tissue is required to mitigate seizures.
TSC can be treated with rapamycin (also called sirolimus) and its derivative everolimus, collectively known as rapalogues. Recent clinical trials with these drugs have shown benefit for treating epilepsy in TSC, with approximately 40% seizure reduction in 40% of individuals (French et al., 2016). Rapalogues are also effective at treating the glioneuronal brain tumors that occur in about 5–20% of TSC patients, called subependymal giant cell astrocytomas or SEGAs (Krueger et al., 2010). However, tumors may regrow if treatment is stopped (Miller et al., 2015). Clinical trials with rapalogues focusing on neuropsychological deficits and autistic symptoms in TSC are underway (www.clinicaltrials.gov), although a recent trial did not report significant improvements in neurocognitive functioning with six months of daily everolimus (Krueger et al., 2017). Systemic side-effects associated with chronic rapalogue use are prevalent but generally tolerated and include infections due to immunosuppression and stomatitis (French et al., 2016). Despite some success with rapalogues, there is still an unmet clinical need for TSC treatment and additional therapeutic approaches are required.
Biochemistry and genetics of TSC
TSC is caused by loss-of-function mutations in the genes TSC1 (van Slegtenhorst et al., 1997) or TSC2 (European Chromosome 16 Tuberous Sclerosis, 1993). These two genes encode for the proteins TSC1 and TSC2 that form a multimeric complex together with the protein TBC1D7 (Dibble et al., 2012), which represses mTOR complex 1 (mTORC1) signaling. Biochemically, TSC2 is a GTPase-activating protein (GAP) for the small GTPase Rheb, which prevents its activation of mTOR complex 1 (mTORC1) (Tee et al., 2003). TSC1 is a stabilizer of TSC2, preventing its degradation and enhancing its GAP activity (Tee et al., 2003). As an active signaling node, mTORC1 promotes anabolic cellular processes through a multitude of functions including stimulation of protein and lipid synthesis, cellular metabolic control, and suppression of autophagy, among others (Ben-Sahra and Manning, 2017; Saxton and Sabatini, 2017). Constitutive or deregulated mTORC1 activity, as in the case of TSC1/2 complex loss, causes increased cell growth, altered proteostasis, and transcriptional and translational alterations (Sarbassov et al., 2005). Loss of either TSC1 or TSC2 is sufficient to cause mTORC1 hyperactivity. However, loss-of-function mutations in TSC2 tend to cause greater mTORC1 activation and are associated with more severe epilepsy and cognitive impairment (Jones et al., 1997; Dabora et al., 2001; Zeng et al., 2011; van Eeghen et al., 2012; Blair et al., 2018).
Patients with TSC have germline heterozygous mutations in TSC1 or TSC2; however, pathological lesions including cortical tubers are variable and appear stochastically. A leading hypothesis to explain cortical tuber formation is the “two-hit” model (Crino, 2016). This model proposes that loss of heterozygosity due to a somatic second-hit mutation in TSC1 or TSC2 in a small population of neural progenitor cells (NPCs) causes altered differentiation, development and neuronal migration resulting in a focal malformation. In support of this model, second-hit mutations in TSC1 or TSC2 are frequently identified in TSC-related tumors and have been detected in some cortical tubers (Henske et al., 1996; Sepp et al., 1996; Smolarek et al., 1998; Au et al., 1999; Chan et al., 2004; Crino et al., 2010; Qin et al., 2010; Martin et al., 2017). In addition, studies in mouse and human cellular models have shown that complete loss of TSC1/2 function is required for the formation of enlarged, dysplastic neurons and glia, which do not develop from a heterozygous state (Meikle et al., 2007; Goto et al., 2011; Blair et al., 2018). That said, this idea has been controversial in the field as second-hit mutations have been identified in only a minority of tubers tested (Henske et al., 1996; Crino et al., 2010; Qin et al., 2010; Martin et al., 2017). This may reflect low allelic frequency of the somatic mutation (D’Gama et al., 2017), dilution of the second-hit cells by infiltrating glia, mutations in regulatory regions which are not assessed in exome sequencing studies (Tyburczy et al., 2015), or epigenetic changes.
Other mTORopathies
TSC is representative of a larger class of disorders called “mTORopathies”, caused by mutations in mTOR pathway regulators that result in constitutive activation of mTORC1 signaling. In addition to TSC1 and TSC2, these regulators include PTEN, PI3K, AKT3, DEPDC5, STRADA, Rheb and others (Crino, 2015; Reijnders et al., 2017). Recently, it was shown that in addition to germline mutations, mTORopathies can arise through somatic brain mutations, including gain-of-function mutations in mTOR itself (Lee et al., 2012; D’Gama et al., 2015; Lim et al., 2015; Nakashima et al., 2015; Mirzaa et al., 2016; Moller et al., 2016; D’Gama et al., 2017). These somatic mTORC1-activating mutations have been identified in brain tissue from patients with focal cortical dysplasia and hemimegalencephaly. Notably, an mTOR-activating mutation detected in fewer than 10% of brain cells is sufficient to cause disease (D’Gama et al., 2017). These types of somatic mutations are more challenging to model than inherited mutations as they require a mechanism to induce them in a sparse population of cells and early in brain development. Given potentially similar underlying neuropathophysiology, disease mechanisms discovered in models of TSC are likely to provide insights into this greater class of mTOR-related disorders.
Rodent models of TSC
Rodent models have provided key insights into the consequences of Tsc1/2 loss on brain development and function. Germline knock-out mouse models demonstrated that complete loss of Tsc1 or Tsc2 is embryonic lethal (Onda et al., 1999) (Kwiatkowski et al., 2002), and subsequent conditional knockout models of Tsc1 (Kwiatkowski et al., 2002) and Tsc2 (Hernandez et al., 2007) have been developed. Different applications of Cre-recombinase using viral injections, in utero electroporation, or Cre-expressing mouse lines, have illuminated the effects of Tsc1 or Tsc2 loss on multiple neuronal types. Somatic mTOR-activating mutations have also been modeled in mice by sparse expression of mutant MTOR, constitutively active Rheb, or CRISPR/Cas9 constructs targeting Tsc1 or Tsc2 (Lin et al., 2016; Lim et al., 2017; Park et al., 2018; Nguyen et al., 2019). The Eker rat model of TSC, carrying a spontaneous loss-of-function mutation in Tsc2 was shown to develop sporadic cytomegalic neurons, glia, and SEGAs in aged or irradiated young animals to induce a “second-hit” (Eker et al., 1981; Takahashi et al., 2004; Wenzel et al., 2004).
Collectively, these rodent studies have shown that loss of Tsc1/2 function impacts multiple processes happening at different developmental time points. These include altered neuronal differentiation, survival, migration, morphology, excitability, synaptic plasticity, glial function, and behavior [for relevant reviews see (Tsai and Sahin, 2011; Costa-Mattioli and Monteggia, 2013; Crino, 2013; Magri and Galli, 2013; Lipton and Sahin, 2014)]. In general, complete loss of Tsc1 or Tsc2 is required to observe strong disease-related phenotypes. That said, heterozygous animals do exhibit some changes in synaptic function, neuronal excitability, and behavior (Ehninger et al., 2008; Auerbach et al., 2011; Sato et al., 2012; Tsai et al., 2012). While these rodent models have been and will continue to be powerful research tools, it is important to note that bona fide cortical tuber regions are not readily observed in animal models, suggesting that this pathology may result from unique aspects of human brain development.
Opportunities for human stem cell-based models of TSC
While the rodent and human brain exhibit overall similar developmental patterns and trajectories, there are unique aspects of human brain development that cannot be captured in animal models. In particular, the human cortex develops over a significantly protracted time period compared to mice and exhibits unique cell types and proliferative zones (Miller et al., 2019). For example, the dramatically increased complexity and size of the human cortex is thought to be due to a specific progenitor cell type called outer radial glia that forms the outer subventricular zone, which is not present in rodents (Hansen et al., 2010; Pollen et al., 2015). Notably, outer radial glia have been shown to exhibit high levels of mTORC1 activity and unique expression of mTOR-pathway components (Pollen et al., 2019), indicating an important role for mTOR signaling in human cortical development. The human brain comprises about 85 billion neurons and at the peak of neurogenesis 100,000 new neurons are generated each minute (D’Gama and Walsh, 2018). This massive cell proliferation also results in increased liability for somatic mutations, whose contribution to both normal and abnormal human brain development is becoming increasingly appreciated (D’Gama and Walsh, 2018).
To capture these unique aspects of human brain development and understand how alterations in TSC-mTOR signaling affect these features, human cellular models are needed. Recent advances in somatic cell reprogramming have allowed the derivation of human induced pluripotent stem cells (hiPSCs) from skin or blood cells from patients, which have the advantage of preserving patient-specific genetic information (Okano and Yamanaka, 2014). With the advent of site-specific nucleases as gene-editing tools, most notably CRISPR/Cas9, human pluripotent stem cells (either hiPSCs or human embryonic stem cells, hESCs) can be genetically engineered with good efficiency to generate disease models with targeted mutations (Jinek et al., 2013; Blair et al., 2016; Hockemeyer and Jaenisch, 2016). Combining these approaches, it is also feasible to genetically engineer patient-derived cells to either correct the mutation or introduce an additional mutation, thereby establishing a genetically controlled, isogenic disease model using a patient’s own cells (Soldner et al., 2011). These stem cells can subsequently be differentiated into numerous somatic cell types including neurons and glia for modeling brain disorders (Soldner and Jaenisch, 2018).
Depending on the differentiation protocol employed, two- or three-dimensional cultures comprising different lineages of neurons and glia can be established. Cortical excitatory neurons and astrocytes of the telencephalic lineage are key cell types of relevance for TSC as the dysplastic cells in tubers are positive for glutamatergic and astrocytic markers (Crino, 2013). These cell types can be generated through manipulation of endogenous neuro-ectodermal differentiation pathways either via inhibition of the dual-SMAD pathway (Chambers et al., 2009) or overexpression of transcription factors (Zhang et al., 2013). Studies with cell type-specific conditional knock-out mice have also highlighted cerebellar Purkinje cells as relevant to TSC pathophysiology, particularly the behavioral symptoms of autism (Tsai et al., 2012; Stoodley et al., 2017; Tsai et al., 2018). A human cellular differentiation protocol based on the addition of specific growth factors has recently been established for cerebellar Purkinje cells and specifically applied to disease modeling in TSC (Sundberg et al., 2018), as discussed below.
While differentiation has traditionally been done in two-dimensional (2D) monolayer cultures, protocols have recently been adapted for three-dimensional (3D) differentiation to generate brain spheroids or organoids (collectively called brain organoids here) (Kadoshima et al., 2013; Lancaster et al., 2013; Pasca et al., 2015; Qian et al., 2016). As discussed by Chen, Song & Ming in this special issue, 3D brain organoid models have advantages over 2D models including more complex cytoarchitecture and cellular niches that preserve cell-cell and cell-matrix interactions (Chen et al., 2019).
The approach of differentiating neurons and glia from human pluripotent stem cells generally operates on a human developmental timescale. For example, by transcriptional profiling, a 2–3 month old human brain organoid is roughly equivalent to a 16–19 post-conception week human brain (Pasca et al., 2015). This enables the observation and manipulation of human neural development in approximately real-time. For this reason, neurodevelopmental disorders such as TSC are particularly well-suited to this disease modeling approach. In the next section we will describe published work to date using human stem cell derived-neurons and brain organoids to investigate disease mechanisms in TSC.
Human neuron and brain organoid models of TSC
Forebrain excitatory neurons and glia in 2D cultures
Alterations in differentiation, signaling, and gene expression
Initial work in developing human neuronal models of TSC has focused on the differentiation of genetically engineered hESCs (Costa et al., 2016; Grabole et al., 2016; Blair et al., 2018), TSC patient-derived iPSCs (Li et al., 2017; Zucco et al., 2018; Nadadhur et al., 2019) or gene edited TSC iPSCs (Ebrahimi-Fakhari et al., 2016) into two-dimensional forebrain cultures. These studies were undertaken using a variety of neuronal differentiation methods, investigating the effects of TSC1 or 2 reduction on neural precursors, neurons, astrocytes and, in one case, oligodendrocytes (Nadadhur et al., 2019).
Differentiation into neural precursors proceeded normally in each study with only minor differences observed such as increased neural rosette size in TSC2−/− cultures (Costa et al., 2016) and increased proliferation rate in TSC2+/− cultures (Li et al., 2017), although this was not observed in other studies (Zucco et al., 2018). In contrast to this normal early neural differentiation, terminal differentiation into neurons proved highly problematic for cells with complete loss of TSC1/2 complex function. Specifically, TSC2−/− cultures produced significantly lower numbers of cells expressing the neuronal markers HuC/D (Costa et al., 2016). Notably, loss of one copy of TSC1 or TSC2 was much less deleterious with cultures exhibiting either a minor decrease in HuC/D-positive cells (Costa et al., 2016; Zucco et al., 2018), or no decrease at all (Nadadhur et al., 2019). The differentiation defects in cells with loss of TSC1/2 may be due to a combination of increased neuronal death (Costa et al., 2016), delayed neuronal differentiation (Zucco et al., 2018), or a shift towards astro-glial fate (Costa et al., 2016; Grabole et al., 2016). Dissecting the potential mechanisms of altered differentiation will be an interesting avenue for future investigation with these models.
The expected hyperactivation of mTORC1, as indicated by increased phosphorylation of downstream targets including ribosomal protein S6, was observed in all studies. However the strong effects seen at every developmental stage in TSC2−/− cultures (Costa et al., 2016), were not consistently seen at the NPC stage in TSC2+/− cultures (Blair et al., 2018; Zucco et al., 2018). Transcriptome analysis through RNA sequencing of patient iPSC-derived heterozygous NPCs found 513 differentially expressed transcripts compared to a sibling control line. Gene ontology analysis indicated that these transcripts were primarily involved in neuron migration and development (Zucco et al., 2018). Independent RNA sequencing of isogenic, gene-edited TSC2 heterozygous and homozygous cultures found very few differences between TSC2+/− and TSC2+/+ cells (10 transcripts) but large differences between TSC2−/− and TSC2+/+ with over 2000 transcripts differentially expressed (Grabole et al., 2016). It is possible that some of the differences between the patient iPSC and control line in the study by Zucco et al could be driven by genetic differences independent of the TSC2 mutation. Analysis of additional TSC patient and control cell lines would be helpful to resolve this. In the TSC2−/− cultures in the Grabole et al study, groups of transcripts involved in astrogliosis, inflammation, and glycolysis were all upregulated, which corresponds to observations of poor mitochondrial function in gene edited iPSC-derived TSC2−/− neurons (Ebrahimi-Fakhari et al., 2016). The transcriptome of TSC2−/− cultures also closely corresponded with previous microarray studies of cortical tubers and SEGAs (Boer et al., 2008; Tyburczy et al., 2010).
Given the key involvement of mTORC1 signaling in mRNA translation, translational profiling may reveal further differences in TSC neural cultures that may occur independently of transcriptional changes. This will be an interesting avenue for future exploration. Related to this, a recent study showed that mTORC1 signaling and translation of the translational machinery is high in human pluripotent stem cells but is suppressed during neural differentiation (Blair et al., 2017). In addition, numerous changes in mRNA translation without a corresponding change in mRNA levels were observed across human neuronal development, highlighting the importance of translational control for developing neurons (Blair et al., 2017).
Impact on neuronal morphology and physiology
The most dramatic morphological differences were observed in homozygous TSC2 knockout cells. TSC2−/− NPCs, neurons, and glia exhibited somatic hypertrophy and neurons displayed increased dendritic arborization (Costa et al., 2016; Blair et al., 2018). The effects of heterozygous TSC1 or 2 loss were less clear for these cultures, with either no change in neuronal morphology (Costa et al., 2016; Zucco et al., 2018), minor increases in dendritic branching and no change in soma size (Nadadhur et al., 2019) or increases in both (Li et al., 2017). One note is that the study by Li and colleagues was based on a single cell line from one TSC patient compared to an iPSC line from an unrelated individual. It therefore remains to be determined whether phenotypic differences between these cell lines are due to the TSC2 mutation or a result of cell line variability or genetic background.
Electrophysiological phenotypes were probed in a subset of studies with either whole cell electrophysiology, multi-electrode arrays (MEA), or calcium imaging (Costa et al., 2016; Nadadhur et al., 2019). Whole cell recordings showed a strong decrease in the frequency of spontaneous (sEPSCs) and miniature (mEPSCs) excitatory post-synaptic currents in both TSC2+/− and TSC2−/− neurons in a gene dose-dependent manner (Costa et al., 2016). However, mEPSC amplitude was increased in TSC2−/− neurons, suggestive of increased synaptic strength. TSC2−/−, but not TSC2+/− neurons also had significantly reduced intrinsic excitability, consistent with their morphological alterations and changes in passive membrane properties (Costa et al., 2016). This decreased intrinsic and synaptic excitability in developing TSC2−/− neurons suggests that other circuit components, e.g. inhibitory neurons, may be required to generate hyperexcitability at the network level following loss of TSC1/2 function (Talos et al., 2012; Bateup et al., 2013). By contrast, MEA recordings of heterozygous cultures from patient TSC iPSCs did show increased spontaneous network activity, which was also reflected by the increased frequency, but not amplitude, of calcium transients in these cultures (Nadadhur et al., 2019). Discrepancies between these findings may reflect gene dose-dependent effects, cell line and culture variability (which could have significant effects on network activity levels and development), or inhibitory and excitatory neuron composition of the cultures, which was not explored in these studies.
Treatment with rapalogues and other mTOR inhibitors such as AZD-8055 reversed many of the phenotypes of TSC1 or 2 loss in forebrain neural cultures including altered electrophysiology (Costa et al., 2016; Nadadhur et al., 2019), aberrant morphology (Costa et al., 2016; Nadadhur et al., 2019), hyperactive mTORC1 signaling (Costa et al., 2016; Li et al., 2017; Zucco et al., 2018) and altered mRNA translation (Grabole et al., 2016).
Cerebellar Purkinje cells
While forebrain excitatory cultures deficient in TSC1 or 2 have been the primary focus of most studies thus far because of their potential to develop into cortical tuber-like cells, cerebellar tubers can also form in some TSC patients (Jay et al., 1998; Vaughn et al., 2013). In addition, mouse studies have demonstrated the importance of Tsc1/2 function in cerebellar Purkinje cells for autism-related behaviors (Tsai et al., 2012; Stoodley et al., 2017; Tsai et al., 2018). To generate a cerebellar model for TSC, a new human Purkinje cell differentiation protocol was developed and hiPSC lines from three individuals with TSC were generated, using cells from the parents or unaffected individuals as controls (Sundberg et al., 2018). In addition, this study made use of an established TSC2 heterozygous patient iPSC line, which had been further genetically engineered to create a TSC2−/− cell line together with a repaired TSC2+/+ control cell line (Ebrahimi-Fakhari et al., 2016). This strategy has significant advantages over the use of control iPSC lines from unrelated individuals as it provides an isogenic system in which cells have the same genetic background and differ only in the disease gene.
In this model, many of the same phenotypes as in forebrain cultures were observed including increased rates of NPC proliferation, upregulated expression of astroglial markers, increased cell death, increased cell size, hyperactivation of mTORC1 activity and decreased excitability of differentiated neurons (Sundberg et al., 2018). These properties were observed in both heterozygous and homozygous cultures with more severe deficiencies in TSC2−/− cells. Transcriptomic analysis again revealed more differential gene expression between homozygotes and controls than heterozygotes and controls, with similar differentially expressed transcripts as in forebrain cultures including altered mitochondria and autophagy genes (Grabole et al., 2016; Sundberg et al., 2018). Interestingly, in cerebellar cultures there was also decreased expression of mRNA processing genes, including many genes which are targets of FMRP, the protein disrupted in the neurodevelopmental disorder Fragile X Syndrome. Finally, treatment with mTOR inhibitors reversed all the observed phenotypic effects of complete TSC2 loss (Sundberg et al., 2018).
3D brain organoid models of TSC
Recent developments in 3D differentiation techniques to generate human stem cell-derived brain organoids provide a new platform to investigate neurodevelopmental disorders in a physiologically relevant setting that can be maintained for long periods of time (Kadoshima et al., 2013; Lancaster et al., 2013; Pasca et al., 2015; Qian et al., 2016). Specifically, these models demonstrate cortical patterning, including the presence of human specific cellular niches (Pollen et al., 2019) and neuronal migration (Bershteyn et al., 2017). These features may be particularly relevant for TSC as cortical tubers are developmental malformations that reflect not only altered differentiation but also defective migration and patterning.
A recent study combined 3D neuronal differentiation with CRISPR/Cas9 genome-editing to investigate the “two-hit” hypothesis of cortical tuber development in human brain organoids (Blair et al., 2018). In addition to establishing a panel of hESC lines with constitutive loss-of-function mutations in TSC1 or TSC2, the authors used CRISPR/Cas9 to create an hESC line with a constitutive mutation in one allele of TSC2 and a conditional mutation in the other (TSC2c/−). To generate a second-hit, Cre recombinase was added to TSC2c/− brain organoids to cause biallelic inactivation of TSC2 and expression of a red fluorescent protein at a defined point during development. Applying sub-saturating amounts of Cre recombinase at a stage when NPCs were proliferating resulted in populations of TSC2−/− cells that developed alongside TSC2c/− cells (which are effectively heterozygous), analogous to what is hypothesized to happen in the developing cortex of TSC patients.
Many of the developmental differences seen in 2D neuronal culture were also observed in this model including a strong bias towards an astro-glial cell fate, altered cell morphology, cytomegaly, and activation of mTORC1 signaling (Blair et al., 2018). These phenotypes became more apparent over developmental time, with relatively minor alterations at the neural precursor stage and greater abnormalities following terminal neuronal or astrocyte differentiation. This may be because mTOR signaling is normally high in stem cells (hPSCs and NPCs) and becomes strongly suppressed during neurogenesis. Failure to suppress mTOR signaling during neuronal differentiation due to loss of the TSC1/2 complex may alter proteostasis and interfere with transcriptional and translational programs. Within the organoids, the second-hit mutation resulted in the cell autonomous generation of multiple cell types found in cortical tubers including dysmorphic neurons, dysplastic astrocytes, and giant cells. Comparisons between wild-type controls and cells heterozygous or homozygous knock-out for TSC1 or TSC2 revealed differentiation defects and mTORC1 hyperactivation only in organoids with homozygous deletion of TSC1 or TSC2, supporting the two-hit model. In addition, phenotypes tended to be more severe in organoids with complete TSC2 loss-of-function compared to TSC1.
Chronic treatment of brain organoids with rapamycin prevented mTORC1 hyperactivation and cytomegaly of TSC2−/− cells (Blair et al., 2018). In addition, rapamycin treatment biased differentiation towards neuronal fates, indicating that mTOR signaling can bi-directionally control cell fate and thus have a major impact on nervous system development. In addition, while early treatment with rapamycin shifted cell fate, later treatment did not, demonstrating a critical window for mTOR to regulate cell fate decisions in developing brain organoids. Removal of rapamycin after early treatment caused the return of mTORC1 hyperactivity, indicating the potential necessity of chronic rapalogue use to fully treat TSC (Blair et al., 2018).
Summary of human neural models of TSC
Taken together, the studies described above demonstrate the power of human stem cell-based disease modeling for neurodevelopmental disorders such as TSC. In particular, human cell studies have led to the robust generation of tuber-like cells, including giant cells, in vitro, enabling future studies into the molecular mechanisms for this developmental abnormality in the context of human-specific neural development. Collectively, these studies have revealed profound impairments in neuronal differentiation and development due to TSC1/2 mutations together with increased production of astro-glial cells. These findings are consistent with patient histology literature describing the altered cell morphology and molecular expression patterns of cortical tuber cells (Crino, 2013). In addition, findings in human neural models of TSC have supported the idea that alterations in astrocyte differentiation and function are important for TSC neuropathophysiology (Wong and Crino, 2012). Thus, these initial studies have established and validated new models for TSC that can be employed to perform mechanistic investigations into how TSC-mTOR signaling contributes to early human brain development in both normal and pathological states.
In addition to allowing the study of human-specific cell biology, a key advantage of human cellular systems over rodent models is the ability to observe and manipulate the very earliest stages of neural development. This is technically challenging in mice as germline loss of Tsc1 or Tsc2 causes embryonic lethality prior to brain development. To circumvent this, brain-specific deletion of Tsc1 or 2 can be utilized; however, this requires expression of Cre recombinase, which may not be expressed early enough in development to induce the full panoply of abnormalities that occur in utero in TSC. This could be a factor limiting the development of bona fide cortical tubers in rodent models. The ability to generate long-lived human neural cultures that develop from a stem cell state therefore provides a unique platform to perform early manipulations and to analyze their long-term consequences on neuron and astrocyte development.
Perspectives, challenges, and future directions
While human stem cell-based models for the neurological aspects of TSC have many compelling features, there are important caveats and considerations to this approach. In particular, while the ability to differentiate neurons from a stem cell state provides an opportunity to model the earliest stages of human brain development, it is important to note that these cells generally develop on a human time scale. Specifically, 3D brain organoids that are cultured for 2–3 months are transcriptionally similar to first or second trimester human fetal brain (Pasca et al., 2015). Therefore, the neurons and astrocytes in these models are reflective of an immature, fetal state. This is less of a problem, and indeed may be a useful feature, for studying abnormalities of brain development such as cortical tubers and related focal dysplasias. However, it can present a challenge when seeking to study aspects of TSC that may emerge later in development. Related to this, immaturity also applies to the functional properties of the neurons and astrocytes generated. For example, in 2D cortical cultures, neurons do not robustly fire action potentials until 7–10 weeks post-differentiation (Johnson et al., 2007). The maturation process can be accelerated by direct conversion of stem cells into functional neurons via forced expression of neurogenic transcription factors (Zhang et al., 2013). However, this type of protocol bypasses normal developmental stages that may be important to preserve in certain disease models. In brain organoids, mature electrophysiological activity is detected around 14 weeks and later (Yoon et al., 2019) and it may take >6 months post-differentiation for coordinated network activity to develop (Quadrato et al., 2017). In terms of astrocyte function, a recent single cell transcriptomics analysis of human cortical spheroids across development noted a shift in astrocyte gene signatures from fetal to post-natal around nine months in culture, with further maturation and development extending to spheroids grown for almost two years (Sloan et al., 2017). Therefore, if these more mature functional properties are required before pathophysiology emerges, human neural cultures may need to be maintained for very long periods of time.
Another consideration is that while undirected brain organoid models that generate multiple cell types exist (Lancaster et al., 2013), the variability and stochasticity with which different cell types are generated are a potential impediment to reproducibility required for disease modeling (Quadrato et al., 2017). As a result, the majority of differentiation protocols are directed, in that they are designed to generate particular neural cell type(s) arising from a specific developmental lineage. One therefore needs to consider what cell types are most pertinent to the disease and it may not be possible to have all relevant cell types present in a given model. This may be important in TSC, in which changes in several different cell types and non-cell autonomous effects may converge to produce disease phenotypes. For example, in mice, a growth factor secreted by Tsc1 KO neurons impairs oligodendrocyte development, which in turn negatively affects the myelination of cortical neurons (Ercan et al., 2017). In addition, in a Tsc1 conditional knock-out mouse model, changes in inhibitory synapses onto excitatory neurons are what drive hyperactivity of the hippocampal network (Bateup et al., 2013). Neuronal differentiation approaches tend to produce mainly glutamatergic or GABAergic neurons but generally not both as these arise from different lineages. Excitatory/inhibitory circuits have been modeled in 2D cultures by mixing separately derived excitatory and inhibitory neurons together (Yang et al., 2017), or in 3D cultures by fusing pallium (excitatory glutamatergic lineage) and subpallium (inhibitory GABA-ergic lineage) organoids together into an “assembloid” (Birey et al., 2017).
A particularly exciting aspect of human stem cell-based disease modeling is the option to develop patient-specific models. These have the major advantage of preserving patient-specific genetic information. The disadvantage is the challenge in finding an appropriate control as differences between cell lines generated from different individuals could reflect cell line variability or differences in genetic background unrelated to disease state. Gene editing approaches can be employed to generate isogenic cell lines with defined mutations. This approach can be done in the context of patient cells to capitalize on the advantages of both technologies, namely generating an isogenic disease model with patient-specific cells (Ebrahimi-Fakhari et al., 2016; Blair et al., 2018). Advances in gene editing have also facilitated more sophisticated genetic models in which conditional mutations can be generated that were previously only feasible in animal models (Pak et al., 2015; Blair et al., 2018). These conditional strategies may be particularly relevant to mTORopathies as somatic mutations in the mTOR pathway can lead to disease (D’Gama et al., 2017). In this case, using patient-derived cells to establish a model is not possible, as the somatic mutation would not necessarily be present in the fibroblasts or blood cells used for somatic cell reprogramming. Advanced gene editing can also be used for the expression of additional tools such as genetically encoded calcium indicators to monitor neuronal activity, optogenetics proteins for neuronal activation or silencing, or fluorescent cellular organelle reporters (Roberts et al., 2017).
In summary, the concurrent technical developments of neuronal differentiation from hPSCs, iPSC creation from human somatic cells, and genome engineering have facilitated the modeling of neurodevelopmental disorders in a laboratory setting. In particular, by applying these techniques to TSC, new human cellular models have been developed to not only answer questions about the developmental origins of the disorder but also give biological insights into treatment. We anticipate that the continued use and refinement of these models in conjunction with animal models will eventually lead to more effective and less invasive treatment for TSC and related mTORopathies.
Acknowledgements
We thank our collaborator Dirk Hockemeyer for sharing his expertise in human stem cell biology and gene editing. John Blair’s work was supported by Predoctoral Awards from the American Epilepsy Society and Canadian Institutes for Health Research (DFS-147661). This work was supported by R01NS097823, a Hellman Fellows Award, a Brain Research Foundation Seed Grant, an Alfred P. Sloan Foundation fellowship, and a Shurl and Kay Curci Foundation grant to Helen Bateup. Helen Bateup is a Chan Zuckerberg Biohub Investigator.
References
- Au KS, Hebert AA, Roach ES, Northrup H. 1999. Complete inactivation of the TSC2 gene leads to formation of hamartomas. Am J Hum Genet 65:1790–1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Auerbach BD, Osterweil EK, Bear MF. 2011. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 480:63–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bateup HS, Johnson CA, Denefrio CL, Saulnier JL, Kornacker K, Sabatini BL. 2013. Excitatory/Inhibitory synaptic imbalance leads to hippocampal hyperexcitability in mouse models of tuberous sclerosis. Neuron 78:510–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben-Sahra I, Manning BD. 2017. mTORC1 signaling and the metabolic control of cell growth. Curr Opin Cell Biol 45:72–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bershteyn M, Nowakowski TJ, Pollen AA, Di Lullo E, Nene A, Wynshaw-Boris A, Kriegstein AR. 2017. Human iPSC-Derived Cerebral Organoids Model Cellular Features of Lissencephaly and Reveal Prolonged Mitosis of Outer Radial Glia. Cell Stem Cell 20:435–449 e434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, Pasca SP. 2017. Assembly of functionally integrated human forebrain spheroids. Nature 545:54–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blair JD, Bateup HS, Hockemeyer DF. 2016. Establishment of Genome-edited Human Pluripotent Stem Cell Lines: From Targeting to Isolation. J Vis Exp:e53583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blair JD, Hockemeyer D, Bateup HS. 2018. Genetically engineered human cortical spheroid models of tuberous sclerosis. Nat Med 24:1568–1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blair JD, Hockemeyer D, Doudna JA, Bateup HS, Floor SN. 2017. Widespread Translational Remodeling during Human Neuronal Differentiation. Cell Rep 21:2005–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boer K, Jansen F, Nellist M, Redeker S, van den Ouweland AM, Spliet WG, van Nieuwenhuizen O, Troost D, Crino PB, Aronica E. 2008. Inflammatory processes in cortical tubers and subependymal giant cell tumors of tuberous sclerosis complex. Epilepsy Res 78:7–21. [DOI] [PubMed] [Google Scholar]
- 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 27:275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan JA, Zhang H, Roberts PS, Jozwiak S, Wieslawa G, Lewin-Kowalik J, Kotulska K, Kwiatkowski DJ. 2004. Pathogenesis of tuberous sclerosis subependymal giant cell astrocytomas: biallelic inactivation of TSC1 or TSC2 leads to mTOR activation. J Neuropathol Exp Neurol 63:1236–1242. [DOI] [PubMed] [Google Scholar]
- Chen HI, Song H, Ming GL. 2019. Applications of Human Brain Organoids to Clinical Problems. Dev Dyn 248:53–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu-Shore CJ, Major P, Camposano S, Muzykewicz D, Thiele EA. 2010. The natural history of epilepsy in tuberous sclerosis complex. Epilepsia 51:1236–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa-Mattioli M, Monteggia LM. 2013. mTOR complexes in neurodevelopmental and neuropsychiatric disorders. Nat Neurosci 16:1537–1543. [DOI] [PubMed] [Google Scholar]
- Costa V, Aigner S, Vukcevic M, Sauter E, Behr K, Ebeling M, Dunkley T, Friedlein A, Zoffmann S, Meyer CA, Knoflach F, Lugert S, Patsch C, Fjeldskaar F, Chicha-Gaudimier L, Kiialainen A, Piraino P, Bedoucha M, Graf M, Jessberger S, Ghosh A, Bischofberger J, Jagasia R. 2016. mTORC1 Inhibition Corrects Neurodevelopmental and Synaptic Alterations in a Human Stem Cell Model of Tuberous Sclerosis. Cell Rep 15:86–95. [DOI] [PubMed] [Google Scholar]
- Crino PB. 2013. Evolving neurobiology of tuberous sclerosis complex. Acta Neuropathol 125:317–332. [DOI] [PubMed] [Google Scholar]
- Crino PB. 2015. mTOR signaling in epilepsy: insights from malformations of cortical development. Cold Spring Harb Perspect Med 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crino PB. 2016. The mTOR signalling cascade: paving new roads to cure neurological disease. Nat Rev Neurol 12:379–392. [DOI] [PubMed] [Google Scholar]
- Crino PB, Aronica E, Baltuch G, Nathanson KL. 2010. Biallelic TSC gene inactivation in tuberous sclerosis complex. Neurology 74:1716–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crino PB, Nathanson KL, Henske EP. 2006. The tuberous sclerosis complex. N Engl J Med 355:1345–1356. [DOI] [PubMed] [Google Scholar]
- Curatolo P, Moavero R, de Vries PJ. 2015. Neurological and neuropsychiatric aspects of tuberous sclerosis complex. Lancet Neurol 14:733–745. [DOI] [PubMed] [Google Scholar]
- D’Gama AM, Geng Y, Couto JA, Martin B, Boyle EA, LaCoursiere CM, Hossain A, Hatem NE, Barry BJ, Kwiatkowski DJ, Vinters HV, Barkovich AJ, Shendure J, Mathern GW, Walsh CA, Poduri A. 2015. Mammalian target of rapamycin pathway mutations cause hemimegalencephaly and focal cortical dysplasia. Ann Neurol 77:720–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Gama AM, Walsh CA. 2018. Somatic mosaicism and neurodevelopmental disease. Nat Neurosci 21:1504–1514. [DOI] [PubMed] [Google Scholar]
- D’Gama AM, Woodworth MB, Hossain AA, Bizzotto S, Hatem NE, LaCoursiere CM, Najm I, Ying Z, Yang E, Barkovich AJ, Kwiatkowski DJ, Vinters HV, Madsen JR, Mathern GW, Blumcke I, Poduri A, Walsh CA. 2017. Somatic Mutations Activating the mTOR Pathway in Dorsal Telencephalic Progenitors Cause a Continuum of Cortical Dysplasias. Cell Rep 21:3754–3766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dabora SL, Jozwiak S, Franz DN, Roberts PS, Nieto A, Chung J, Choy YS, Reeve MP, Thiele E, Egelhoff JC, Kasprzyk-Obara J, Domanska-Pakiela D, Kwiatkowski DJ. 2001. Mutational analysis in a cohort of 224 tuberous sclerosis patients indicates increased severity of TSC2, compared with TSC1, disease in multiple organs. Am J Hum Genet 68:64–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dibble CC, Elis W, Menon S, Qin W, Klekota J, Asara JM, Finan PM, Kwiatkowski DJ, Murphy LO, Manning BD. 2012. TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1. Mol Cell 47:535–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ebrahimi-Fakhari D, Saffari A, Wahlster L, DiNardo A, Turner D, Lewis TL Jr, Conrad C, Rothberg JM, Lipton JO, Kolker S, Hoffmann GF, Han MJ, Polleux F, Sahin M. 2016. Impaired Mitochondrial Dynamics And Mitophagy In Neuronal Models Of Tuberous Sclerosis Complex. Cell Rep 17:2162. [DOI] [PubMed] [Google Scholar]
- Ehninger D, Han S, Shilyansky C, Zhou Y, Li W, Kwiatkowski DJ, Ramesh V, Silva AJ. 2008. Reversal of learning deficits in a Tsc2+/− mouse model of tuberous sclerosis. Nat Med 14:843–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eker R, Mossige J, Johannessen JV, Aars H. 1981. Hereditary renal adenomas and adenocarcinomas in rats. Diagn Histopathol 4:99–110. [PubMed] [Google Scholar]
- Ercan E, Han JM, Di Nardo A, Winden K, Han MJ, Hoyo L, Saffari A, Leask A, Geschwind DH, Sahin M. 2017. Neuronal CTGF/CCN2 negatively regulates myelination in a mouse model of tuberous sclerosis complex. J Exp Med 214:681–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- European Chromosome 16 Tuberous Sclerosis C. 1993. Identification and characterization of the tuberous sclerosis gene on chromosome 16. Cell 75:1305–1315. [DOI] [PubMed] [Google Scholar]
- French JA, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, Curatolo P, de Vries PJ, Dlugos DJ, Berkowitz N, Voi M, Peyrard S, Pelov D, Franz DN. 2016. Adjunctive everolimus therapy for treatment-resistant focal-onset seizures associated with tuberous sclerosis (EXIST-3): a phase 3, randomised, double-blind, placebo-controlled study. Lancet 388:2153–2163. [DOI] [PubMed] [Google Scholar]
- Goto J, Talos DM, Klein P, Qin W, Chekaluk YI, Anderl S, Malinowska IA, Di Nardo A, Bronson RT, Chan JA, Vinters HV, Kernie SG, Jensen FE, Sahin M, Kwiatkowski DJ. 2011. Regulable neural progenitor-specific Tsc1 loss yields giant cells with organellar dysfunction in a model of tuberous sclerosis complex. Proc Natl Acad Sci U S A 108:E1070–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grabole N, Zhang JD, Aigner S, Ruderisch N, Costa V, Weber FC, Theron M, Berntenis N, Spleiss O, Ebeling M, Yeo GW, Jagasia R, Kiialainen A. 2016. Genomic analysis of the molecular neuropathology of tuberous sclerosis using a human stem cell model. Genome Med 8:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen DV, Lui JH, Parker PR, Kriegstein AR. 2010. Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature 464:554–561. [DOI] [PubMed] [Google Scholar]
- Henske EP, Scheithauer BW, Short MP, Wollmann R, Nahmias J, Hornigold N, van Slegtenhorst M, Welsh CT, Kwiatkowski DJ. 1996. Allelic loss is frequent in tuberous sclerosis kidney lesions but rare in brain lesions. Am J Hum Genet 59:400–406. [PMC free article] [PubMed] [Google Scholar]
- Hernandez O, Way S, McKenna J 3rd, Gambello MJ. 2007. Generation of a conditional disruption of the Tsc2 gene. Genesis 45:101–106. [DOI] [PubMed] [Google Scholar]
- Hockemeyer D, Jaenisch R. 2016. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 18:573–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jay V, Edwards V, Musharbash A, Rutka JT. 1998. Cerebellar pathology in tuberous sclerosis. Ultrastruct Pathol 22:331–339. [DOI] [PubMed] [Google Scholar]
- Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. 2013. RNA-programmed genome editing in human cells. Elife 2:e00471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson MA, Weick JP, Pearce RA, Zhang SC. 2007. Functional neural development from human embryonic stem cells: accelerated synaptic activity via astrocyte coculture. J Neurosci 27:3069–3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones AC, Daniells CE, Snell RG, Tachataki M, Idziaszczyk SA, Krawczak M, Sampson JR, Cheadle JP. 1997. Molecular genetic and phenotypic analysis reveals differences between TSC1 and TSC2 associated familial and sporadic tuberous sclerosis. Hum Mol Genet 6:2155–2161. [DOI] [PubMed] [Google Scholar]
- Kadoshima T, Sakaguchi H, Nakano T, Soen M, Ando S, Eiraku M, Sasai Y. 2013. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proc Natl Acad Sci U S A 110:20284–20289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P, Wilson KA, Byars A, Sahmoud T, Franz DN. 2010. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med 363:1801–1811. [DOI] [PubMed] [Google Scholar]
- Krueger DA, Sadhwani A, Byars AW, de Vries PJ, Franz DN, Whittemore VH, Filip-Dhima R, Murray D, Kapur K, Sahin M. 2017. Everolimus for treatment of tuberous sclerosis complex-associated neuropsychiatric disorders. Ann Clin Transl Neurol 4:877–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwiatkowski DJ, Zhang H, Bandura JL, Heiberger KM, Glogauer M, el-Hashemite N, Onda H. 2002. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum Mol Genet 11:525–534. [DOI] [PubMed] [Google Scholar]
- Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA. 2013. Cerebral organoids model human brain development and microcephaly. Nature 501:373–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Huynh M, Silhavy JL, Kim S, Dixon-Salazar T, Heiberg A, Scott E, Bafna V, Hill KJ, Collazo A, Funari V, Russ C, Gabriel SB, Mathern GW, Gleeson JG. 2012. De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause hemimegalencephaly. Nat Genet 44:941–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Cao J, Chen M, Li J, Sun Y, Zhang Y, Zhu Y, Wang L, Zhang C. 2017. Abnormal Neural Progenitor Cells Differentiated from Induced Pluripotent Stem Cells Partially Mimicked Development of TSC2 Neurological Abnormalities. Stem Cell Reports 8:883–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim JS, Gopalappa R, Kim SH, Ramakrishna S, Lee M, Kim WI, Kim J, Park SM, Lee J, Oh JH, Kim HD, Park CH, Lee JS, Kim S, Kim DS, Han JM, Kang HC, Kim HH, Lee JH. 2017. Somatic Mutations in TSC1 and TSC2 Cause Focal Cortical Dysplasia. Am J Hum Genet 100:454–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim JS, Kim WI, Kang HC, Kim SH, Park AH, Park EK, Cho YW, Kim S, Kim HM, Kim JA, Kim J, Rhee H, Kang SG, Kim HD, Kim D, Kim DS, Lee JH. 2015. Brain somatic mutations in MTOR cause focal cortical dysplasia type II leading to intractable epilepsy. Nat Med 21:395–400. [DOI] [PubMed] [Google Scholar]
- Lin TV, Hsieh L, Kimura T, Malone TJ, Bordey A. 2016. Normalizing translation through 4E-BP prevents mTOR-driven cortical mislamination and ameliorates aberrant neuron integration. Proc Natl Acad Sci U S A 113:11330–11335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipton JO, Sahin M. 2014. The neurology of mTOR. Neuron 84:275–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magri L, Galli R. 2013. mTOR signaling in neural stem cells: from basic biology to disease. Cell Mol Life Sci 70:2887–2898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 2017. The genomic landscape of tuberous sclerosis complex. Nat Commun 8:15816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meikle L, Talos DM, Onda H, Pollizzi K, Rotenberg A, Sahin M, Jensen FE, Kwiatkowski DJ. 2007. A mouse model of tuberous sclerosis: neuronal loss of Tsc1 causes dysplastic and ectopic neurons, reduced myelination, seizure activity, and limited survival. J Neurosci 27:5546–5558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller DJ, Bhaduri A, Sestan N, Kriegstein A. 2019. Shared and derived features of cellular diversity in the human cerebral cortex. Curr Opin Neurobiol 56:117–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller JM, Wachsman A, Haker K, Majlessipour F, Danielpour M, Puliyanda D. 2015. The effects of everolimus on tuberous sclerosis-associated lesions can be dramatic but may be impermanent. Pediatr Nephrol 30:173–177. [DOI] [PubMed] [Google Scholar]
- Mirzaa GM, Campbell CD, Solovieff N, Goold C, Jansen LA, Menon S, Timms AE, Conti V, Biag JD, Adams C, Boyle EA, Collins S, Ishak G, Poliachik S, Girisha KM, Yeung KS, Chung BHY, Rahikkala E, Gunter SA, McDaniel SS, Macmurdo CF, Bernstein JA, Martin B, Leary R, Mahan S, Liu S, Weaver M, Doerschner M, Jhangiani S, Muzny DM, Boerwinkle E, Gibbs RA, Lupski JR, Shendure J, Saneto RP, Novotny EJ, Wilson CJ, Sellers WR, Morrissey M, Hevner RF, Ojemann JG, Guerrini R, Murphy LO, Winckler W, Dobyns WB. 2016. Association of MTOR Mutations With Developmental Brain Disorders, Including Megalencephaly, Focal Cortical Dysplasia, and Pigmentary Mosaicism. JAMA Neurol 73:836–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mizuguchi M, Takashima S. 2001. Neuropathology of tuberous sclerosis. Brain Dev 23:508–515. [DOI] [PubMed] [Google Scholar]
- Moller RS, Weckhuysen S, Chipaux M, Marsan E, Taly V, Bebin EM, Hiatt SM, Prokop JW, Bowling KM, Mei D, Conti V, de la Grange P, Ferrand-Sorbets S, Dorfmuller G, Lambrecq V, Larsen LH, Leguern E, Guerrini R, Rubboli G, Cooper GM, Baulac S. 2016. Germline and somatic mutations in the MTOR gene in focal cortical dysplasia and epilepsy. Neurol Genet 2:e118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nadadhur AG, Alsaqati M, Gasparotto L, Cornelissen-Steijger P, van Hugte E, Dooves S, Harwood AJ, Heine VM. 2019. Neuron-Glia Interactions Increase Neuronal Phenotypes in Tuberous Sclerosis Complex Patient iPSC-Derived Models. Stem Cell Reports 12:42–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakashima M, Saitsu H, Takei N, Tohyama J, Kato M, Kitaura H, Shiina M, Shirozu H, Masuda H, Watanabe K, Ohba C, Tsurusaki Y, Miyake N, Zheng Y, Sato T, Takebayashi H, Ogata K, Kameyama S, Kakita A, Matsumoto N. 2015. Somatic Mutations in the MTOR gene cause focal cortical dysplasia type IIb. Ann Neurol 78:375–386. [DOI] [PubMed] [Google Scholar]
- Nguyen LH, Mahadeo T, Bordey A. 2019. mTOR Hyperactivity Levels Influence the Severity of Epilepsy and Associated Neuropathology in an Experimental Model of Tuberous Sclerosis Complex and Focal Cortical Dysplasia. J Neurosci. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okano H, Yamanaka S. 2014. iPS cell technologies: significance and applications to CNS regeneration and disease. Mol Brain 7:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onda H, Lueck A, Marks PW, Warren HB, Kwiatkowski DJ. 1999. Tsc2(+/−) mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background. J Clin Invest 104:687–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pak C, Danko T, Zhang Y, Aoto J, Anderson G, Maxeiner S, Yi F, Wernig M, Sudhof TC. 2015. Human Neuropsychiatric Disease Modeling using Conditional Deletion Reveals Synaptic Transmission Defects Caused by Heterozygous Mutations in NRXN1. Cell Stem Cell 17:316–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park SM, Lim JS, Ramakrishina S, Kim SH, Kim WK, Lee J, Kang HC, Reiter JF, Kim DS, Kim HH, Lee JH. 2018. Brain Somatic Mutations in MTOR Disrupt Neuronal Ciliogenesis, Leading to Focal Cortical Dyslamination. Neuron 99:83–97 e87. [DOI] [PubMed] [Google Scholar]
- Pasca 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, Pasca SP. 2015. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat Methods 12:671–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollen AA, Bhaduri A, Andrews MG, Nowakowski TJ, Meyerson OS, Mostajo-Radji MA, Di Lullo E, Alvarado B, Bedolli M, Dougherty ML, Fiddes IT, Kronenberg ZN, Shuga J, Leyrat AA, West JA, Bershteyn M, Lowe CB, Pavlovic BJ, Salama SR, Haussler D, Eichler EE, Kriegstein AR. 2019. Establishing Cerebral Organoids as Models of Human-Specific Brain Evolution. Cell 176:743–756 e717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollen AA, Nowakowski TJ, Chen J, Retallack H, Sandoval-Espinosa C, Nicholas CR, Shuga J, Liu SJ, Oldham MC, Diaz A, Lim DA, Leyrat AA, West JA, Kriegstein AR. 2015. Molecular identity of human outer radial glia during cortical development. Cell 163:55–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 2016. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 165:1238–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin W, Chan JA, Vinters HV, Mathern GW, Franz DN, Taillon BE, Bouffard P, Kwiatkowski DJ. 2010. Analysis of TSC cortical tubers by deep sequencing of TSC1, TSC2 and KRAS demonstrates that small second-hit mutations in these genes are rare events. Brain Pathol 20:1096–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quadrato G, Nguyen T, Macosko EZ, Sherwood JL, Min Yang S, Berger DR, Maria N, Scholvin J, Goldman M, Kinney JP, Boyden ES, Lichtman JW, Williams ZM, McCarroll SA, Arlotta P. 2017. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545:48–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reijnders MRF, Kousi M, van Woerden GM, Klein M, Bralten J, Mancini GMS, van Essen T, Proietti-Onori M, Smeets EEJ, van Gastel M, Stegmann APA, Stevens SJC, Lelieveld SH, Gilissen C, Pfundt R, Tan PL, Kleefstra T, Franke B, Elgersma Y, Katsanis N, Brunner HG. 2017. Variation in a range of mTOR-related genes associates with intracranial volume and intellectual disability. Nat Commun 8:1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roberts B, Haupt A, Tucker A, Grancharova T, Arakaki J, Fuqua MA, Nelson A, Hookway C, Ludmann SA, Mueller IA, Yang R, Horwitz R, Rafelski SM, Gunawardane RN. 2017. Systematic gene tagging using CRISPR/Cas9 in human stem cells to illuminate cell organization. Mol Biol Cell 28:2854–2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarbassov DD, Ali SM, Sabatini DM. 2005. Growing roles for the mTOR pathway. Curr Opin Cell Biol 17:596–603. [DOI] [PubMed] [Google Scholar]
- Sato A, Kasai S, Kobayashi T, Takamatsu Y, Hino O, Ikeda K, Mizuguchi M. 2012. Rapamycin reverses impaired social interaction in mouse models of tuberous sclerosis complex. Nat Commun 3:1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saxton RA, Sabatini DM. 2017. mTOR Signaling in Growth, Metabolism, and Disease. Cell 168:960–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sepp T, Yates JR, Green AJ. 1996. Loss of heterozygosity in tuberous sclerosis hamartomas. J Med Genet 33:962–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sloan SA, Darmanis S, Huber N, Khan TA, Birey F, Caneda C, Reimer R, Quake SR, Barres BA, Pasca SP. 2017. Human Astrocyte Maturation Captured in 3D Cerebral Cortical Spheroids Derived from Pluripotent Stem Cells. Neuron 95:779–790 e776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smolarek TA, Wessner LL, McCormack FX, Mylet JC, Menon AG, Henske EP. 1998. Evidence that lymphangiomyomatosis is caused by TSC2 mutations: chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes from women with lymphangiomyomatosis. Am J Hum Genet 62:810–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Jaenisch R. 2018. Stem Cells, Genome Editing, and the Path to Translational Medicine. Cell 175:615–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Laganiere J, Cheng AW, Hockemeyer D, Gao Q, Alagappan R, Khurana V, Golbe LI, Myers RH, Lindquist S, Zhang L, Guschin D, Fong LK, Vu BJ, Meng X, Urnov FD, Rebar EJ, Gregory PD, Zhang HS, Jaenisch R. 2011. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell 146:318–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stoodley CJ, D’Mello AM, Ellegood J, Jakkamsetti V, Liu P, Nebel MB, Gibson JM, Kelly E, Meng F, Cano CA, Pascual JM, Mostofsky SH, Lerch JP, Tsai PT. 2017. Altered cerebellar connectivity in autism and cerebellar-mediated rescue of autism-related behaviors in mice. Nat Neurosci 20:1744–1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sundberg M, Tochitsky I, Buchholz DE, Winden K, Kujala V, Kapur K, Cataltepe D, Turner D, Han MJ, Woolf CJ, Hatten ME, Sahin M. 2018. Purkinje cells derived from TSC patients display hypoexcitability and synaptic deficits associated with reduced FMRP levels and reversed by rapamycin. Mol Psychiatry 23:2167–2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi DK, Dinday MT, Barbaro NM, Baraban SC. 2004. Abnormal Cortical Cells and Astrocytomas in the Eker Rat Model of Tuberous Sclerosis Complex. Epilepsia 45:1525–1530. [DOI] [PubMed] [Google Scholar]
- Talos DM, Sun H, Kosaras B, Joseph A, Folkerth RD, Poduri A, Madsen JR, Black PM, Jensen FE. 2012. Altered inhibition in tuberous sclerosis and type IIb cortical dysplasia. Ann Neurol 71:539–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J. 2003. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr Biol 13:1259–1268. [DOI] [PubMed] [Google Scholar]
- Thiele EA. 2010. Managing and understanding epilepsy in tuberous sclerosis complex. Epilepsia 51 Suppl 1:90–91. [DOI] [PubMed] [Google Scholar]
- Tsai P, Sahin M. 2011. Mechanisms of neurocognitive dysfunction and therapeutic considerations in tuberous sclerosis complex. Curr Opin Neurol 24:106–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai PT, Hull C, Chu Y, Greene-Colozzi E, Sadowski AR, Leech JM, Steinberg J, Crawley JN, Regehr WG, Sahin M. 2012. Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature 488:647–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai PT, Rudolph S, Guo C, Ellegood J, Gibson JM, Schaeffer SM, Mogavero J, Lerch JP, Regehr W, Sahin M. 2018. Sensitive Periods for Cerebellar-Mediated Autistic-like Behaviors. Cell Rep 25:357–367 e354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyburczy ME, Dies KA, Glass J, Camposano S, Chekaluk Y, Thorner AR, Lin L, Krueger D, Franz DN, Thiele EA, Sahin M, Kwiatkowski DJ. 2015. Mosaic and Intronic Mutations in TSC1/TSC2 Explain the Majority of TSC Patients with No Mutation Identified by Conventional Testing. PLoS Genet 11:e1005637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyburczy ME, Kotulska K, Pokarowski P, Mieczkowski J, Kucharska J, Grajkowska W, Roszkowski M, Jozwiak S, Kaminska B. 2010. Novel proteins regulated by mTOR in subependymal giant cell astrocytomas of patients with tuberous sclerosis complex and new therapeutic implications. Am J Pathol 176:1878–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Eeghen AM, Black ME, Pulsifer MB, Kwiatkowski DJ, Thiele EA. 2012. Genotype and cognitive phenotype of patients with tuberous sclerosis complex. Eur J Hum Genet 20:510–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Slegtenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, Verhoef S, Lindhout D, van den Ouweland A, Halley D, Young J, Burley M, Jeremiah S, Woodward K, Nahmias J, Fox M, Ekong R, Osborne J, Wolfe J, Povey S, Snell RG, Cheadle JP, Jones AC, Tachataki M, Ravine D, Sampson JR, Reeve MP, Richardson P, Wilmer F, Munro C, Hawkins TL, Sepp T, Ali JB, Ward S, Green AJ, Yates JR, Kwiatkowska J, Henske EP, Short MP, Haines JH, Jozwiak S, Kwiatkowski DJ. 1997. Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science 277:805–808. [DOI] [PubMed] [Google Scholar]
- Vaughn J, Hagiwara M, Katz J, Roth J, Devinsky O, Weiner H, Milla S. 2013. MRI characterization and longitudinal study of focal cerebellar lesions in a young tuberous sclerosis cohort. AJNR Am J Neuroradiol 34:655–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wenzel HJ, Patel LS, Robbins CA, Emmi A, Yeung RS, Schwartzkroin PA. 2004. Morphology of cerebral lesions in the Eker rat model of tuberous sclerosis. Acta Neuropathol 108:97–108. [DOI] [PubMed] [Google Scholar]
- Wong M, Crino PB. 2012. Tuberous sclerosis and epilepsy: role of astrocytes. Glia 60:1244–1250. [DOI] [PubMed] [Google Scholar]
- Yang N, Chanda S, Marro S, Ng YH, Janas JA, Haag D, Ang CE, Tang Y, Flores Q, Mall M, Wapinski O, Li M, Ahlenius H, Rubenstein JL, Chang HY, Buylla AA, Sudhof TC, Wernig M. 2017. Generation of pure GABAergic neurons by transcription factor programming. Nat Methods 14:621–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoon SJ, Elahi LS, Pasca AM, Marton RM, Gordon A, Revah O, Miura Y, Walczak EM, Holdgate GM, Fan HC, Huguenard JR, Geschwind DH, Pasca SP. 2019. Reliability of human cortical organoid generation. Nat Methods 16:75–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng LH, Rensing NR, Zhang B, Gutmann DH, Gambello MJ, Wong M. 2011. Tsc2 gene inactivation causes a more severe epilepsy phenotype than Tsc1 inactivation in a mouse model of tuberous sclerosis complex. Hum Mol Genet 20:445–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, Sudhof TC. 2013. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78:785–798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zucco AJ, Pozzo VD, Afinogenova A, Hart RP, Devinsky O, D’Arcangelo G. 2018. Neural progenitors derived from Tuberous Sclerosis Complex patients exhibit attenuated PI3K/AKT signaling and delayed neuronal differentiation. Mol Cell Neurosci 92:149–163. [DOI] [PMC free article] [PubMed] [Google Scholar]