Abstract
Approximately 50% of patients with the genetic disease tuberous sclerosis complex (TSC) present with autism spectrum disorder. Although a number of studies have investigated the link between autism and TSC, the etiology of autism spectrum disorder in these patients remains unclear. Abnormal cerebellar function during critical phases of development could disrupt functional processes in the brain, leading to development of autistic features. Accordingly, we review the potential role of cerebellar dysfunction in the pathogenesis of autism spectrum disorder in TSC. We also introduce conditional knockout mouse models of Tsc1 and Tsc2 that link cerebellar circuitry to the development of autistic-like features. Taken together, these preclinical and clinical investigations indicate the cerebellum has a profound regulatory role during development of social communication and repetitive behaviors.
Keywords: magnetic resonance imaging, mouse models, mTOR, Purkinje cell, cerebellum
TSC and Autism Spectrum Disorder
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by significant neurologic disability.1 TSC results from a mutation of either the TSC1 or TSC2 gene, whose protein products, hamartin and tuberin, form a heterodimeric complex that negatively regulates mechanistic target of rapamycin (mTOR) signaling. The mTOR pathway is a critical signaling network that regulates protein synthesis, cell proliferation, cell metabolism, and cell growth.2,3 More than 80% of TSC patients have cortical tubers (hamartomas), which contain large glial cells and dysplastic neurons and have loss of normal architecture in cerebral cortex.4
Some studies have shown that tuber formation affects the severity of neurological symptoms in TSC, although patients without tubers may also have significant developmental deficits, including intellectual disability and autism.5–10 In addition, seizures that emerge during critical developmental periods profoundly impact the severity of the neurological, cognitive, and psychological symptoms of TSC patients.10–12 Several studies have demonstrated that TSC2 mutations are associated with a more severe TSC phenotype compared with mutations in TSC1.13,14 In addition, several studies indicate that disruption of the TSC pathway contributes to autism spectrum disorder, and approximately 50% of TSC patients fulfill criteria for autism spectrum disorder.15,16 This makes TSC one of the most common monogenetic causes of autism spectrum disorder.
Autism spectrum disorder is highly heterogeneous and can usually be diagnosed as early as 2 years of age. The condition presents with a wide range of deficits, including difficulty with social communication, repetitive behaviors, and restricted interests. In addition, patients with autism spectrum disorder often have intellectual disability and epilepsy. Neuropathological studies have consistently demonstrated a loss of cerebellar Purkinje cells in individuals diagnosed with autism spectrum disorder versus typically developing controls.17–20 Imaging studies have shown that patients diagnosed with autism spectrum disorder have gray and white matter abnormalities in the cerebellum, dating to early childhood.21–25 Significantly, premature infants with isolated cerebellar hemorrhage have a higher incidence of autism spectrum disorder, suggesting that cerebellar dysfunction early in development contributes to the pathogenesis of autism.26,27 These findings have raised the hypothesis that abnormal cerebellar function could disrupt higher-order social cognitive processes and lead to autism spectrum disorder.
In the case of individuals with TSC, several lines of evidence suggest a correlation with cerebellar involvement and presence of autism spectrum disorder symptoms. A relationship between cerebellar tubers and autism was first reported in 2000.28 Imaging studies indicate the presence of abnormal cerebellar findings in TSC patients, with 4 to 13% of these patients displaying cerebellar atrophy.29–31 Interestingly, cerebellar tubers have unique a morphology and specialized structure compared with supratentorial tubers. In one study, 33% of TSC patients had cerebellar lesions, which were described as having a pyramidal/wedge shaped appearance.30 All the cerebellar tubers studied had increased apparent diffusion coefficient values, and most of them had “zebra-like” contrast enhancement. Despite this presentation, these patients did not have any ‘typical’ cerebellar symptoms on neurological examination.30 There also appears to be an association between the presence of cerebellar tubers and occurrence of cerebellar atrophy.29,32 Taken together, these studies indicate that 24% to 33% of TSC patients have cerebellar lesions.30,31,33
Research conducted by Drs. Diane and Harry Chugani and their colleagues demonstrate that cerebellar lesions detected on MRI scans also have functional correlates on PET scans.33 They characterized features of cerebellar lesions in a functional imaging study of 78 TSC patients as part of an epilepsy surgery evaluation. Twenty-seven percent of these patients had cerebellar lesions with increased alpha-[(11)C]methyl-L-tryptophan (AMT) uptake and decreased glucose metabolism. In addition, this study showed that those with right-sided lesions were more likely to have communication problems and autism spectrum disorders.33
Furthermore, it has been previously demonstrated that cerebellar cortical lesions may play a role in the dentatothalamofrontal circuits. Positron emission tomography (PET) scans showed that children with TSC and autism spectrum disorders had increased glucose metabolism in the deep cerebellar nuclei bilaterally. Moreover, glucose hypermetabolism in the deep cerebellar nuclei and occurrence of infantile spasms were both associated with disturbances in behavior and communication skills, and difficulties in social interaction.34 As the cerebellar cortex inhibits the deep cerebellar nuclei, these findings indicate hypofunction of the cerebellar cortex. These findings may not be restricted to autism spectrum disorders associated with TSC. In fact, in a study of a series of individuals with ‘idiopathic’ autism spectrum disorder, PET scans also displayed increased AMT uptake in the right dentate nucleus and decreased AMT uptake in the left cortical and thalamic regions,35 supporting the notion that the role of the cerebellum in autism spectrum disorders may be more generalizable.
In summary, in several cohorts of TSC patients the presence of cerebellar lesions correlates with a diagnosis of autism spectrum disorder.28,33 Although a number of clinical investigations have been performed for TSC patients, the mechanistic link between cerebellar dysfunction and autism spectrum disorder remains to be determined.
Cerebellar Development
The cerebellum is a highly specialized brain region with carefully detailed architecture and cell layer formations, first described by Cajal. Although the cerebellum has been studied for several decades, our understanding of its functional roles and connectivity to other brain regions remains incomplete. Anatomically the cerebellum can be divided into separate lobes, white matter, and cortical layers. The cerebellum consists of special classes of cerebellar neurons: granule cells, Purkinje cells, and interneurons. We will focus on describing development and differentiation of Purkinje cells, whose dysfunction and reduced number have been previously linked to autism occurrence.17–20
There is a close resemblance in cerebellar development among vertebrates. Mice cerebellar primordium consists of 2 distinct germinal matrices, including the ventral ventricular zone and the dorsal rhombic lip, which are formed between embryonic days 8.5–9.5. This is a birthplace for cerebellar GABAergic neuronal precursors, including the Purkinje cells and glutamatergic neurons.36 The concentration of transcriptional factors, cell position, and migration affect the final fate of the cells in the developing cerebellum.37–39
Purkinje cell precursors in the murine cerebellum develop between embryonic days 11–13. During the differentiation process, Purkinje cells migrate along the radial glial fibers out beyond the mantle of postmitotic precursors. After birth, Purkinje cells continue to migrate along the radial glial system to form a zone (molecular layer) overlying the germinative zone. In addition, the granule neurons migrate via the radial glial pathway through the zone of immature Purkinje cells. This migration process facilitates the formation of another neuronal layer of the cerebellar cortex, the internal granule cell layer.39
The directed migration of interneurons and postmitotic precursors of Purkinje cells results in the formation of the cerebellar cortex. The expansion and development of the cerebellum depends on the proliferation rate of granule cell progenitors related to the ratio of developing Purkinje cells, which in the human cerebellum is around 400 granule cells per one Purkinje cell.37,38 Purkinje cells form connections with basket and satellite cells that provide inhibitory input to other Purkinje cells. Of note, Purkinje cells are the only output neuron of the cerebellar cortex. Purkinje cells’ inhibitory neurotransmission establishes the patterning of the cerebellum during development and has a crucial role in the neuronal activity in the cerebellum.40
Several transcription factors and growth factors affect the development of Purkinje cells and granule cells with canonical manner. For example, isthmic organizer at the midbrain-hindbrain boundary secretes fibroblast growth factor 8 (FGF8), which plays an important role in the earliest stage of cerebellar development.41–44 FGF8 regulates expression of several transcription factors, including region-specific OTX2, EN1/2, PAX2/5/8, and GBX2. Importantly, FGF8 secretion regulates Wnt1 expression at the midbrain-hindbrain boundary, forming a positive feedback loop, which also activates EN2 and PAX2 expression.45–49
The En1 and En2 expression in developing precursor cells affects the formation of the cerebellar region, and together with the bone morphogenic proteins, dorsalizes cells within this region.50,51 In the granule cell layer, the dorsal genes Zic1/2 and Math1, play important roles in the regulation of the granular neurons’ specification and migration.52 In the ventricular zone, Ptf1a- (pancreas transcription factor 1a), which encodes a bHLH transcription factor, and Neph3 (nephrin-like protein 3), also known as KIRREL2, a membrane molecule and a product of a Ptf1a target gene, expression gives rise to all Purkinje cells.53–55 The neuroepithelium in the developing mice cerebellar plate expresses E-cadherin, Neph3, Ptf1a, and Corl2 with a spatiotemporal pattern that overlaps with Purkinje cell generation (embryonic days 12–14).
Purkinje cell precursors in the subventricular zone and postmitotic stage express lineage-specific marker Corl2 (SKOR2).53,55–57 Mature Purkinje cells can be characterized by their expression of Calbindin, L7, and PCP2.55,58,59 Granule cells migrate on glial scaffolds, and this process is mediated by a family of neuron-glial adhesion molecules, known as astrotactins. ACTN2 (astrotactin 2) has been implicated in ASD in at least 2 genetic cohorts,60,61 supporting the role for abnormal cerebellar development contributing to ASD.
Current Knowledge of Cerebellar Dysfunction and Autism
A number of studies have described that the cerebellum regulates cognitive function, and a loss of cerebellar cells has been linked to autism spectrum disorder.19,21,62,63 To establish solid proof that cerebellar deficits link to development of autism spectrum disorder, several research groups have investigated patients with syndromic forms of autism who have cerebellar vermis hypoplasia.64–67 According to these studies, the decreased number of cells in the cerebellum appeared to result from developmental hypoplasia rather than neuronal degeneration during later stages of development.64–67
In addition, postmortem analyses have revealed that patients with autism spectrum disorder had reduced Purkinje cell numbers, reduced number of cells in the cerebellar nuclei, excessive number of Bergmann glia, and activated microglia and cytokine production in cerebellar white matter.17,68,69 Related to this, several diffusion tensor imaging studies to analyze cerebral white matter tracts have been performed in children with autism spectrum disorder, who were then compared with typically developing children.70–72 These studies indicate that children with autism spectrum disorder who have impairment in white matter microstructure, increase in the diffusivity of bilateral superior cerebellar peduncles and reduced connectivity in the corpus callosum and internal capsule.70–72
In addition, behavioral assessment has shown that cerebellum and brainstem dysfunction can lead to saccadic eye movement abnormalities in patients with autism spectrum disorder.73 Growing evidence suggests that the cerebellum has an important regulatory role in language, cognition, and emotions.74–77 The cerebellum also contributes to visual perceptual learning.78 According to this study, patients who had focal damage to the posterior cerebellum displayed strongly diminished learning capacities compared with age- and gender-matched controls, in terms of both rate and amount of improvement over time.78
Animal Models for Describing Cerebellar Dysfunction and Autism Symptoms
Despite pathological, imaging, and genetic studies in patients, all indirectly point to an “upstream” role for the cerebellum in autism; direct mechanistic proof of the involvement of cerebellar neurons in autism spectrum disorder had been lacking. Animal models have started to fill this gap. Several genetic models have been established for studying autism spectrum disorder in mouse models. The genes commonly linked to the development of autism spectrum disorder are: TSC genes TSC1 and TSC2, Fragile X mental retardation gene (Fmr1), neuroligin 3 and 4 (NLGN), SCA1, and methyl-CpG-binding protein type 2 gene (MECP2).79–86 In addition, mutations in Engrailed, PTEN, Reelin, and DLX are linked to development of autism spectrum disorder.87–91 These mouse models have clear behavioral and neurophysiological features linked to autism spectrum disorder and cerebellar development, including aggressive behavior, anxiety, seizures, gastrointestinal problems, motor deficits, abnormal sleep, and sensory problems. Because TSC1/2 mutations cause wide penetration of autism for these patients, we will focus here on Tsc1/2 mice models with cerebellar deficits.
Despite the complex origin of autism spectrum disorders and their heterogeneous cerebellar deficits, recent studies of Tsc1 and Tsc2 mutant mice show clear evidence of these deficits contributing to development of behavioral abnormalities similar to the core features of these disorders.83–85 Our lab has generated a mutant mouse model in which the Tsc1 deletion was restricted to the cerebellum, specifically Purkinje cells. Most notably, the Tsc1-deficient mouse model showed core characteristics of autistic-like behaviors: lack of interest in socializing, repetitive behaviors, restricted interests, and abnormal communication.83 This provides clear evidence that neurodevelopmental deficits restricted to the cerebellum can result in a wide range of typical autistic-like behaviors.
At the cellular level, deletion of Tsc1 only from Purkinje cells during postnatal development resulted in the decreased number of Purkinje cells in mice cerebellum (in the null mutants but not in heterozygous), and the cells demonstrated hypo-excitability (in both null and heterozygous mutants). Together, these results indicate that Purkinje cell hypofunction (not necessarily number) is associated with the behavioral deficits in this model. Interestingly, increased spine density in Purkinje cells was linked to altered behavioral phenotype associated with autism spectrum disorder, and treatment of these mice with rapamycin prevented development of these autistic-like behaviors, providing strong evidence that the TSC/mTOR pathway underlies these defects.
Independent studies by Gambello and colleagues demonstrated that the loss of Tsc2 in mice Purkinje cells caused a progressive increase in cell size and subsequent apoptotic cell death. In Tsc2-deficient mice, Purkinje cell death was associated with motor deficits. Remarkably, oxidative stress and endoplasmic reticulum stress were both increased in Tsc2 null Purkinje cells. In line with the studies of Tsc1 knockout mice models, cell death and endoplasmic reticulum stress phenotypes were rescued by treatment with the mTORC1 inhibitor rapamycin in the Tsc2 knockout model.84,85 Together, these results indicate that Purkinje cells are a crucial part of the neural circuitry underlying autistic-like behavior in mice and alteration of the cerebellar function can result in profound defects in behavior that are typically thought to be related to cerebral cortical dysfunction in autism spectrum disorder.
Because Purkinje cells are the only output of the cerebellar cortex, Purkinje cell abnormalities could influence the rest of the brain via the projections of the deep nuclei to and from specific regions of the thalamus, basal ganglia, and (via polysynaptic connections) the neocortex. In fact, anatomical tracing in non-human primates demonstrates long-range connections between prefrontal cortex and cerebellar hemispheres, consistent with a nonmotor role for the cerebellum.92 Future studies are needed to understand how Purkinje cell dysfunction and/or hypo-excitability lead to autistic-like symptoms in these animal models.
mTOR Pathway and Rapamycin Treatment
mTOR is a conserved serine/threonine kinase and it affects downstream of the PI3K/AKT pathway, where it activates S6K1 and 4EBP1, which are involved in mRNA translation (see review3). mTOR consists of 2 multiprotein complexes: mTORC1 and mTORC2.93–95. The mTORC1 pathway is sensitive to acute rapamycin treatment. Regulation of the mTORC1 pathway occurs via neurotransmitters, nutrients, growth factors, and axon guidance cues. Compared with mTORC1, mTORC2 is generally resistant to rapamycin treatment and not as well-studied in the brain. Several extracellular growth factors affect mTOR pathway activation (e.g., brain-derived neurotrophic factor [BDNF], ciliary neurotrophic factor [CNTF], glutamate, insulin, insulin-like growth factor 1 [IGF1], and vascular endothelial growth factor [VEGF]).96–98 mTOR regulates cell survival, cell growth, proliferation, autophagy, and synaptic function. It is also linked to cellular stress pathways in TSC models.99–104
TSC1 and TSC2 proteins form a heterodimeric complex that negatively regulates mTORC1. The TSC1 gene, located on chromosome 9q34, encodes a transcript that contains 23 exons.105 The TSC2 gene is located on chromosome 16p13, where it encodes a large transcript that contains 41 exons.106 TSC1 and TSC2 function together with TBC1D7 to activate GTPase-activating protein (GAP). This complex inhibits mTOR pathway by converting active Rheb-GTP to inactive Rheb-GDP form.107,108 In the case of tuberous sclerosis, TSC1 or TSC2 are mutated and their protein products are not functional, leading to unregulated activation of the mTOR pathway, and subsequently causing abnormal protein synthesis, cellular growth and proliferation.2,99–104 Thus, inhibition of the mTOR pathway is a potential target for pharmacotherapy treatment of TSC. In addition to rapamycin, there are other drugs that are molecular analogues of rapamycin, called ‘rapalogues’ (e.g, temsirolimus, deforolimus, and everolimus).109 Also, dual PI3K-mTOR inhibitors are being tested for mTOR regulation110 and for TSC treatment.109
Clinical Trials for TSC with Rapalogues
Rapamycin (also known as sirolimus), temsirolimus, deforolimus (also known as ridaforolimus), and everolimus (also known as RAD001) are currently being studied in clinical trials for a variety of indications, including neurocognition, autism, angiofibromas, angiomyolipomas, and multiple cancers occurring with TSC (see study details in www.clinicaltrials.gov). Here we briefly introduce 3 clinical trials with rapamycin and rapalogues that are ongoing for treatment of neurocognitive symptoms and autism in patients with TSC.
We conducted a clinical study for evaluation of the safety of everolimus (RAD001) on TSC patients (www.clinicaltrials.gov; NCT01289912). In 2011, we initiated a Phase II placebo-controlled clinical trial at Boston Children’s Hospital and Cincinnati Children’s Hospital Medical Center for patients with TSC ages 6–21 years old with an IQ of 60 or greater. Primary outcomes of the study were: 1) safety of everolimus compared with placebo in patients with TSC and 2) assessment of the efficacy of everolimus on neurocognition in patients with TSC in comparison with a placebo group of patients, as measured by well-validated, standardized, direct, and indirect neurocognitive tools. This study also included secondary outcomes, which were frequency of seizures, sleep disturbances, autism spectrum disorder features, academic skills, and behavioral problems. This study was completed at the end of 2014, and data analysis is currently in process.
A similar clinical trial has recently been initiated at the Erasmus Medical Center in Rotterdam, Netherlands, to study everolimus efficacy for treatment of autism and neuropsychological deficits in children with TSC (NCT01730209), with expected completion by 2016. The study focuses on TSC patients between 4–15 years of age with an IQ estimated <80 and autism spectrum disorder and/or learning disabilities that require assistance and remedial teaching. The primary outcome of this 12-month study is cognitive ability measured by IQ levels. The secondary outcomes include evaluation of autistic features, social and communicational skills, working memory and attention, visual-motor integration, executive functioning, sleep problems, sensory-related difficulties, overall health, school success, and seizure frequency.
Lastly, a feasibility study of “Rapalogues for Autism Phenotype in TSC” began in 2014 at the Kennedy Krieger Institute, Baltimore (NCT01929642). The aim of this study is to assess the safety and feasibility of administering rapalogues sirolimus or everolimus in 2- to 30-year-old patients with TSC and self-injury and to measure cognitive and behavioral changes, including reduction in autistic symptoms, aggressive behaviors, and self-injury as well as improvements in cognition across multiple domains of cognitive function. Primary outcome measures are assessments of parental stress, caregiver burden, and family compliance to study protocol. Secondary outcomes include evaluation of whether treatment of TSC patients with sirolimus reduces the frequency, severity, or duration of repetitive, self-injurious, and aggressive behavior.
In summary, these clinical trials will provide valuable information on the safety and potential effects of rapalogues on neurocognitive symptoms, learning difficulties, and autism in patients with TSC. Clinical trials such as these will reveal which treatments may be most beneficial for ameliorating symptoms of TSC, at what age treatments should begin, and how long treatment must continue for long-lasting neurocognitive benefit. Moreover, identifying treatments effective for autism and neurocognitive deficits in TSC may be effective more broadly for other individuals with autism or conditions caused by genes in the same or similar gene pathways.
Future Prospects
Based on preclinical studies, rapalogues hold great promise for future treatment of TSC-related behavioral and physiological symptoms. However, effective therapy directed at the cognitive impairments and autism associated with TSC is still in the early stages of clinical research. Although the underlying etiology for development of autism spectrum disorder remains to be defined, continued research towards this goal is ongoing. In the future, the use of new imaging technologies can be utilized for finding novel correlations of network activity with individual symptoms in individual TSC patients and will enable development of targeted therapies for these patients.
Recent progress in studies with Tsc1 and Tsc2 knockout animal models have shown that genetic dysfunctions in physiological contents can provide insights on the developmental regulators, synaptic interactions, neurotransmitters, and novel transcriptional pathways that contribute to autism spectrum disorder susceptibility in the presence of Tsc1/2 dysfunction.83,84 Although the mTORC1 inhibitor rapamycin has been shown to alleviate some of the behavioral deficits in Purkinje cells-specific Tsc1/2 knockout mice, it may not be an ideal therapeutic for the chronic treatment of TSC patients because of its side effects.
We acknowledge that the human patient brain is different structurally and functionally from mice models. Thus, ideally, we need to have access to patient cerebellar tissue to understand the cellular and circuitry abnormalities leading to autistic behaviors as well as for the development of novel therapies for TSC patients. However, nervous system tissues from patients are rarely accessible for these in vitro types of experiments. To overcome this problem, Yamanaka and colleagues have developed cell reprogramming technologies that allow production of human induced pluripotent stem cells (iPSCs) from any patient somatic tissues.111,112 Neural differentiation of human iPSC allows characterization of neurodevelopmental disease phenotypes with in vitro-platform in laboratories.113–115 Thus, in the future researchers can use TSC patient-derived iPSCs for identification of novel molecular targets to rescue TSC-deficient neural cells. By targeting cerebellar cells and especially Purkinje cells, novel therapeutic strategies for TSC can be established. In the future, development of the iPSC-derived Purkinje cells in combination with studies of Tsc1/2 knockout mouse models with clear autistic-like phenotypes will provide a powerful system to develop new treatments for TSC and autism.
Acknowledgments
The authors thank the families who participated in the research studies reviewed in this article. We wish to also thank Rachel Friedman and Kira Dies for critically reviewing the manuscript, and Melanie Fridl Ross, MSJ, ELS, for editing a draft of this paper. Due to limited space we have not quoted all literature in the field, and we apologize to those whose articles are not referenced.
Declaration of Conflicting Interests
Research in the Sahin lab is supported in part by sponsored research agreements with Novartis and Shire.
Funding
Research in MS’s laboratory is supported by the National Institutes of Health (U01 NS082320, P20 NS080199, P30 HD018655), Department of Defense, Tuberous Sclerosis Alliance, Autism Speaks, Nancy Lurie Marks Family Foundation, Simons Foundation, Harvard Stem Cell Institute, Boston Children’s Hospital Translational Research Program, and Novartis, and Shire. The Developmental Synaptopathies Consortium (U54NS092090) is a part of the National Center for Advancing Translational Sciences (NCATS) Rare Diseases Clinical Research Network (RDCRN). RDCRN is an initiative of the Office of Rare Diseases Research (ORDR), NCATS, and funded through collaboration between NCATS, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, and National Institute of Child Health and Human Development.
Footnotes
This work was presented in part at the 2014 Neurobiology of Disease in Children Symposium: Autism, held in conjunction with the 43rd Annual Meeting of the Child Neurology Society, Columbus, Ohio, October 21-23, 2014.
Author Contributions
Drs. Sundberg and Sahin wrote the article together and are sole authors.
References
- 1.Kwiatkowski DJ, Whittemore VH, Thiele EA, editors. Tuberous Sclerosis Complex: Genes, Clinical Features and Therapeutics. 1st ed. Weinheim: Wiley-VCH; 2010. p. 409. [Google Scholar]
- 2.Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis complex. N Engl J Med. 2006;355:1345–1356. doi: 10.1056/NEJMra055323. [DOI] [PubMed] [Google Scholar]
- 3.Lipton JO, Sahin M. The neurology of mTOR. Neuron. 2014;84:275–291. doi: 10.1016/j.neuron.2014.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mizuguchi M, Takashima S. Neuropathology of tuberous sclerosis. Brain Dev. 2001;23:508–515. doi: 10.1016/s0387-7604(01)00304-7. [DOI] [PubMed] [Google Scholar]
- 5.Goodman M, Lamm SH, Engel A, et al. Cortical tuber count: a biomarker indicating neurologic severity of tuberous sclerosis complex. J Child Neurol. 1997;12:85–90. doi: 10.1177/088307389701200203. [DOI] [PubMed] [Google Scholar]
- 6.Major P, Rakowski S, Simon MV, et al. Are cortical tubers epileptogenic? Evidence from electrocorticography. Epilepsia. 2009;50:147–154. doi: 10.1111/j.1528-1167.2008.01814.x. [DOI] [PubMed] [Google Scholar]
- 7.Wong V, Khong PL. Tuberous sclerosis complex: correlation of magnetic resonance imaging (MRI) findings with comorbidities. J Child Neurol. 2006;21:99–105. doi: 10.1177/08830738060210020901. [DOI] [PubMed] [Google Scholar]
- 8.Numis AL, Major P, Montenegro MA, et al. Identification of risk factors for autism spectrum disorders in tuberous sclerosis complex. Neurology. 2011;76:981–987. doi: 10.1212/WNL.0b013e3182104347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bolton PF, Park RJ, Higgins JN, et al. Neuro-epileptic determinants of autism spectrum disorders in tuberous sclerosis complex. Brain. 2002;125(Pt 6):1247–1255. doi: 10.1093/brain/awf124. [DOI] [PubMed] [Google Scholar]
- 10.Kaczorowska M, Jurkiewicz E, Domanska-Pakiela D, et al. Cerebral tuber count and its impact on mental outcome of patients with tuberous sclerosis complex. Epilepsia. 2011;52:22–27. doi: 10.1111/j.1528-1167.2010.02892.x. [DOI] [PubMed] [Google Scholar]
- 11.Wang S, Fallah A. Optimal management of seizures associated with tuberous sclerosis complex: current and emerging options. Neuropsychiatr Dis Treat. 2014:2021–2030. doi: 10.2147/NDT.S51789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mohamed AR, Bailey CA, Freeman JL, et al. Intrinsic epileptogenicity of cortical tubers revealed by intracranial EEG monitoring. Neurology. 2012;79:2249–2257. doi: 10.1212/WNL.0b013e3182768923. [DOI] [PubMed] [Google Scholar]
- 13.Kothare SV, Singh K, Chalifoux JR, et al. Severity of manifestations in tuberous sclerosis complex in relation to genotype. Epilepsia. 2014;55:1025–1029. doi: 10.1111/epi.12680. [DOI] [PubMed] [Google Scholar]
- 14.Dabora SL, Jozwiak S, Franz DN, et al. 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. 2001;68:64–80. doi: 10.1086/316951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jeste SS, Sahin M, Bolton P, et al. Characterization of autism in young children with tuberous sclerosis complex. J Child Neurol. 2008;23:520–525. doi: 10.1177/0883073807309788. [DOI] [PubMed] [Google Scholar]
- 16.Wiznitzer M. Autism and tuberous sclerosis. J Child Neurol. 2004;19:675–679. doi: 10.1177/08830738040190090701. [DOI] [PubMed] [Google Scholar]
- 17.Bauman ML, Kemper TL. Neuroanatomic observations of the brain in autism: a review and future directions. Int J Dev Neurosci. 2005;23:183–187. doi: 10.1016/j.ijdevneu.2004.09.006. [DOI] [PubMed] [Google Scholar]
- 18.Casanova MF. The neuropathology of autism. Brain Pathol. 2007;17:422–433. doi: 10.1111/j.1750-3639.2007.00100.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fatemi SH, Aldinger KA, Ashwood P, et al. Consensus paper: pathological role of the cerebellum in autism. Cerebellum. 2012;11:777–807. doi: 10.1007/s12311-012-0355-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Whitney ER, Kemper TL, Bauman ML, et al. Cerebellar Purkinje cells are reduced in a subpopulation of autistic brains: a stereological experiment using calbindin-D28k. Cerebellum. 2008;7:406–416. doi: 10.1007/s12311-008-0043-y. [DOI] [PubMed] [Google Scholar]
- 21.Courchesne E, Yeung-Courchesne R, Press GA, et al. Hypoplasia of cerebellar vermal lobules VI and VII in autism. N Engl J Med. 1988;318:1349–1354. doi: 10.1056/NEJM198805263182102. [DOI] [PubMed] [Google Scholar]
- 22.Amaral DG, Schumann CM, Nordahl CW. Neuroanatomy of autism. Trends Neurosci. 2008;31:137–145. doi: 10.1016/j.tins.2007.12.005. [DOI] [PubMed] [Google Scholar]
- 23.Kates WR, Burnette CP, Eliez S, et al. Neuroanatomic variation in monozygotic twin pairs discordant for the narrow phenotype for autism. Am J Psychiatry. 2004;161:539–546. doi: 10.1176/appi.ajp.161.3.539. [DOI] [PubMed] [Google Scholar]
- 24.Akshoomoff N, Lord C, Lincoln AJ, et al. Outcome classification of preschool children with autism spectrum disorders using MRI brain measures. J Am Acad Child Adolesc Psychiatry. 2004;43:349–357. doi: 10.1097/00004583-200403000-00018. [DOI] [PubMed] [Google Scholar]
- 25.Allen G, Courchesne E. Differential effects of developmental cerebellar abnormality on cognitive and motor functions in the cerebellum: an fMRI study of autism. Am J Psychiatry. 2003;160:262–273. doi: 10.1176/appi.ajp.160.2.262. [DOI] [PubMed] [Google Scholar]
- 26.Limperopoulos C, Bassan H, Gauvreau K, et al. Does cerebellar injury in premature infants contribute to the high prevalence of long-term cognitive, learning, and behavioral disability in survivors? Pediatrics. 2007;120:584–593. doi: 10.1542/peds.2007-1041. [DOI] [PubMed] [Google Scholar]
- 27.Limperopoulos C, Chilingaryan G, Guizard N, et al. Cerebellar injury in the premature infant is associated with impaired growth of specific cerebral regions. Pediatric Res. 2010;68:145–150. doi: 10.1203/PDR.0b013e3181e1d032. [DOI] [PubMed] [Google Scholar]
- 28.Weber AM, Egelhoff JC, McKellop JM, et al. Autism and the cerebellum: evidence from tuberous sclerosis. J Autism Dev Disord. 2000;30:511–517. doi: 10.1023/a:1005679108529. [DOI] [PubMed] [Google Scholar]
- 29.Marti-Bonmati L, Menor F, Dosda R. Tuberous sclerosis: differences between cerebral and cerebellar cortical tubers in a pediatric population. AJNR Am J Neuroradiol. 2000;21:557–560. [PMC free article] [PubMed] [Google Scholar]
- 30.Ertan G, Arulrajah S, Tekes A, et al. Cerebellar abnormality in children and young adults with tuberous sclerosis complex: MR and diffusion weighted imaging findings. J Neuroradiol. 2010;37:231–238. doi: 10.1016/j.neurad.2009.12.006. [DOI] [PubMed] [Google Scholar]
- 31.Menor F, Marti-Bonmati L, Mulas F, et al. Neuroimaging in tuberous sclerosis: a clinicoradiological evaluation in pediatric patients. Pediatr Radiol. 1992;22:485–489. doi: 10.1007/BF02012989. [DOI] [PubMed] [Google Scholar]
- 32.Vaughn J, Hagiwara M, Katz J, et al. MRI characterization and longitudinal study of focal cerebellar lesions in a young tuberous sclerosis cohort. AJNR Am J Neuroradiol. 2013;34:655–659. doi: 10.3174/ajnr.A3260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Eluvathingal TJ, Behen ME, Chugani HT, et al. Cerebellar lesions in tuberous sclerosis complex: neurobehavioral and neuroimaging correlates. J Child Neurol. 2006;21:846–851. doi: 10.1177/08830738060210100301. [DOI] [PubMed] [Google Scholar]
- 34.Asano E, Chugani DC, Muzik O, et al. Autism in tuberous sclerosis complex is related to both cortical and subcortical dysfunction. Neurology. 2001;57:1269–1277. doi: 10.1212/wnl.57.7.1269. [DOI] [PubMed] [Google Scholar]
- 35.Chugani DC, Muzik O, Rothermel R, et al. Altered serotonin synthesis in the dentatothalamocortical pathway in autistic boys. Ann Neurol. 1997;42:666–669. doi: 10.1002/ana.410420420. [DOI] [PubMed] [Google Scholar]
- 36.Sillitoe RV, Joyner AL. Morphology, molecular codes, and circuitry produce the three-dimensional complexity of the cerebellum. Annu Rev Cell Dev Biol. 2007;23:549–577. doi: 10.1146/annurev.cellbio.23.090506.123237. [DOI] [PubMed] [Google Scholar]
- 37.Hatten ME, Heintz N. Mechanisms of neural patterning and specification in the developing cerebellum. Annu Rev Neurosci. 1995;18:385–408. doi: 10.1146/annurev.ne.18.030195.002125. [DOI] [PubMed] [Google Scholar]
- 38.Hatten ME. Central nervous system neuronal migration. Annu Rev Neurosci. 1999;22:511–539. doi: 10.1146/annurev.neuro.22.1.511. [DOI] [PubMed] [Google Scholar]
- 39.Altman J, Bayer SA. Embryonic development of the rat cerebellum. II. Translocation and regional distribution of the deep neurons. J Comp Neurol. 1985;231:27–41. doi: 10.1002/cne.902310104. [DOI] [PubMed] [Google Scholar]
- 40.White JJ, Arancillo M, Stay TL, et al. Cerebellar zonal patterning relies on Purkinje cell neurotransmission. J Neurosci. 2014;34:8231–8245. doi: 10.1523/JNEUROSCI.0122-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Martinez S, Crossley PH, Cobos I, et al. FGF8 induces formation of an ectopic isthmic organizer and isthmocerebellar development via a repressive effect on Otx2 expression. Development. 1999;126:1189–1200. doi: 10.1242/dev.126.6.1189. [DOI] [PubMed] [Google Scholar]
- 42.Yu T, Meiners LC, Danielsen K, et al. Deregulated FGF and homeotic gene expression underlies cerebellar vermis hypoplasia in CHARGE syndrome. Elife. 2013;2:e01305. doi: 10.7554/eLife.01305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Crossley PH, Martinez S, Martin GR. Midbrain development induced by FGF8 in the chick embryo. Nature. 1996;380:66–68. doi: 10.1038/380066a0. [DOI] [PubMed] [Google Scholar]
- 44.Joyner AL. Engrailed, Wnt and Pax genes regulate midbrain--hindbrain development. Trends Genet. 1996;12:15–20. doi: 10.1016/0168-9525(96)81383-7. [DOI] [PubMed] [Google Scholar]
- 45.Thomas KR, Capecchi MR. Targeted disruption of the murine int-1 proto-oncogene resulting in severe abnormalities in midbrain and cerebellar development. Nature. 1990;346:847–850. doi: 10.1038/346847a0. [DOI] [PubMed] [Google Scholar]
- 46.McMahon AP, Gavin BJ, Parr B, et al. The Wnt family of cell signalling molecules in postimplantation development of the mouse. Ciba Found Symp. 1992:165199–165212. doi: 10.1002/9780470514221.ch12. discussion 212–218. [DOI] [PubMed] [Google Scholar]
- 47.Bally-Cuif L, Alvarado-Mallart RM, Darnell DK, et al. Relationship between Wnt-1 and En-2 expression domains during early development of normal and ectopic met-mesencephalon. Development. 1992;115:999–1009. doi: 10.1242/dev.115.4.999. [DOI] [PubMed] [Google Scholar]
- 48.Millen KJ, Hui CC, Joyner AL. A role for En-2 and other murine homologues of Drosophila segment polarity genes in regulating positional information in the developing cerebellum. Development. 1995;121:3935–3945. doi: 10.1242/dev.121.12.3935. [DOI] [PubMed] [Google Scholar]
- 49.Simeone A. Positioning the isthmic organizer where Otx2 and Gbx2meet. Trends Genet. 2000;16:237–240. doi: 10.1016/s0168-9525(00)02000-x. [DOI] [PubMed] [Google Scholar]
- 50.Wilson SL, Kalinovsky A, Orvis GD, et al. Spatially restricted and developmentally dynamic expression of engrailed genes in multiple cerebellar cell types. Cerebellum. 2011;10:356–372. doi: 10.1007/s12311-011-0254-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sillitoe RV, Vogel MW, Joyner AL. Engrailed homeobox genes regulate establishment of the cerebellar afferent circuit map. J Neurosci. 2010;30:10015–10024. doi: 10.1523/JNEUROSCI.0653-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Aruga J, Inoue T, Hoshino J, et al. Zic2 controls cerebellar development in cooperation with Zic1. J Neurosci. 2002;22:218–225. doi: 10.1523/JNEUROSCI.22-01-00218.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Mizuhara E, Minaki Y, Nakatani T, et al. Purkinje cells originate from cerebellar ventricular zone progenitors positive for Neph3 and E-cadherin. Dev Biol. 2010;338:202–214. doi: 10.1016/j.ydbio.2009.11.032. [DOI] [PubMed] [Google Scholar]
- 54.Hoshino M, Nakamura S, Mori K, et al. Ptf1a, a bHLH transcriptional gene, defines GABAergic neuronal fates in cerebellum. Neuron. 2005;47:201–213. doi: 10.1016/j.neuron.2005.06.007. [DOI] [PubMed] [Google Scholar]
- 55.Muguruma K, Nishiyama A, Ono Y, et al. Ontogeny-recapitulating generation and tissue integration of ES cell-derived Purkinje cells. Nat Neurosci. 2010;13:1171–1180. doi: 10.1038/nn.2638. [DOI] [PubMed] [Google Scholar]
- 56.Nakatani T, Minaki Y, Kumai M, et al. The c-Ski family member and transcriptional regulator Corl2/Skor2 promotes early differentiation of cerebellar Purkinje cells. Dev Biol. 2014;388:68–80. doi: 10.1016/j.ydbio.2014.01.016. [DOI] [PubMed] [Google Scholar]
- 57.Minaki Y, Nakatani T, Mizuhara E, et al. Identification of a novel transcriptional corepressor, Corl2, as a cerebellar Purkinje cell-selective marker. Gene Expr Patterns. 2008;8:418–423. doi: 10.1016/j.gep.2008.04.004. [DOI] [PubMed] [Google Scholar]
- 58.Sillitoe RV, Gopal N, Joyner AL. Embryonic origins of ZebrinII parasagittal stripes and establishment of topographic Purkinje cell projections. Neuroscience. 2009;162:574–588. doi: 10.1016/j.neuroscience.2008.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Whitney ER, Kemper TL, Rosene DL, et al. Calbindin-D28k is a more reliable marker of human Purkinje cells than standard Nissl stains: a stereological experiment. J Neurosci Methods. 2008;168:42–47. doi: 10.1016/j.jneumeth.2007.09.009. [DOI] [PubMed] [Google Scholar]
- 60.Lionel AC, Tammimies K, Vaags AK, et al. Disruption of the ASTN2/TRIM32 locus at 9q33.1 is a risk factor in males for autism spectrum disorders, ADHD and other neurodevelopmental phenotypes. Hum Mol Genet. 2014;23:2752–2768. doi: 10.1093/hmg/ddt669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Glessner JT, Wang K, Cai G, et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature. 2009;459:569–573. doi: 10.1038/nature07953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Palmen SJ, van Engeland H. Review on structural neuroimaging findings in autism. J Neural Transm. 2004;111:903–929. doi: 10.1007/s00702-003-0068-9. [DOI] [PubMed] [Google Scholar]
- 63.DiCicco-Bloom E, Lord C, Zwaigenbaum L, et al. The developmental neurobiology of autism spectrum disorder. J Neurosci. 2006;26:6897–6906. doi: 10.1523/JNEUROSCI.1712-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Webb SJ, Sparks BF, Friedman SD, et al. Cerebellar vermal volumes and behavioral correlates in children with autism spectrum disorder. Psychiatry Res. 2009;172:61–67. doi: 10.1016/j.pscychresns.2008.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Becker R, Stiemer B, Neumann L, et al. Mild ventriculomegaly, mild cerebellar hypoplasia and dysplastic choroid plexus as early prenatal signs of CHARGE association. Fetal Diagn Ther. 2001;16:280–283. doi: 10.1159/000053928. [DOI] [PubMed] [Google Scholar]
- 66.Sanlaville D, Verloes A. CHARGE syndrome: an update. Eur J Hum Genet. 2007;15:389–399. doi: 10.1038/sj.ejhg.5201778. [DOI] [PubMed] [Google Scholar]
- 67.Aldinger KA, Kogan J, Kimonis V, et al. Cerebellar and posterior fossa malformations in patients with autism-associated chromosome 22q13 terminal deletion. Am J Med Genet A. 2013;161:131–136. doi: 10.1002/ajmg.a.35700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bailey A, Luthert P, Dean A, et al. A clinicopathological study of autism. Brain. 1998;121(Pt 5):889–905. doi: 10.1093/brain/121.5.889. [DOI] [PubMed] [Google Scholar]
- 69.Vargas DL, Nascimbene C, Krishnan C, et al. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:67–81. doi: 10.1002/ana.20315. [DOI] [PubMed] [Google Scholar]
- 70.Brito AR, Vasconcelos MM, Domingues RC, et al. Diffusion tensor imaging findings in school-aged autistic children. J Neuroimaging. 2009;19:337–343. doi: 10.1111/j.1552-6569.2009.00366.x. [DOI] [PubMed] [Google Scholar]
- 71.Sivaswamy L, Kumar A, Rajan D, et al. A diffusion tensor imaging study of the cerebellar pathways in children with autism spectrum disorder. J Child Neurol. 2010;25:1223–1231. doi: 10.1177/0883073809358765. [DOI] [PubMed] [Google Scholar]
- 72.Aoki Y, Abe O, Nippashi Y, et al. Comparison of white matter integrity between autism spectrum disorder subjects and typically developing individuals: a meta-analysis of diffusion tensor imaging tractography studies. Mol Autism. 2013;4:25. doi: 10.1186/2040-2392-4-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Schmitt LM, Cook EH, Sweeney JA, et al. Saccadic eye movement abnormalities in autism spectrum disorder indicate dysfunctions in cerebellum and brainstem. Mol Autism. 2014;5:47. doi: 10.1186/2040-2392-5-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Schmahmann JD, Weilburg JB, Sherman JC. The neuropsychiatry of the cerebellum - insights from the clinic. Cerebellum. 2007;6:254–267. doi: 10.1080/14734220701490995. [DOI] [PubMed] [Google Scholar]
- 75.Stoodley CJ, Schmahmann JD. Evidence for topographic organization in the cerebellum of motor control versus cognitive and affective processing. Cortex. 2010;46:831–844. doi: 10.1016/j.cortex.2009.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Koziol LF, Budding D, Andreasen N, et al. Consensus paper: the cerebellum’s role in movement and cognition. Cerebellum. 2014;13:151–177. doi: 10.1007/s12311-013-0511-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Marien P, Ackermann H, Adamaszek M, et al. Consensus paper: Language and the cerebellum: an ongoing enigma. Cerebellum. 2014;13:386–410. doi: 10.1007/s12311-013-0540-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Deluca C, Golzar A, Santandrea E, et al. The cerebellum and visual perceptual learning: evidence from a motion extrapolation task. Cortex. 2014;58:52–71. doi: 10.1016/j.cortex.2014.04.017. [DOI] [PubMed] [Google Scholar]
- 79.Bernardet M, Crusio WE. Fmr1 KO mice as a possible model of autistic features. Scientific World Journal. 2006;6:1164–1176. doi: 10.1100/tsw.2006.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hays SA, Huber KM, Gibson JR. Altered neocortical rhythmic activity states in Fmr1 KO mice are due to enhanced mGluR5 signaling and involve changes in excitatory circuitry. J Neurosci. 2011;31:14223–14234. doi: 10.1523/JNEUROSCI.3157-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Grossman AW, Aldridge GM, Lee KJ, et al. Developmental characteristics of dendritic spines in the dentate gyrus of Fmr1 knockout mice. Brain Res. 2010;1355:221–227. doi: 10.1016/j.brainres.2010.07.090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Shahbazian MD, Orr HT, Zoghbi HY. Reduction of Purkinje cell pathology in SCA1 transgenic mice by p53 deletion. Neurobiol Dis. 2001;8:974–981. doi: 10.1006/nbdi.2001.0444. [DOI] [PubMed] [Google Scholar]
- 83.Tsai PT, Hull C, Chu Y, et al. Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature. 2012;488:647–651. doi: 10.1038/nature11310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Reith RM, Way S, McKenna J3rd, et al. Loss of the tuberous sclerosis complex protein tuberin causes Purkinje cell degeneration. Neurobiol Dis. 2011;43:113–122. doi: 10.1016/j.nbd.2011.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Reith RM, McKenna J, Wu H, et al. Loss of Tsc2 in Purkinje cells is associated with autistic-like behavior in a mouse model of tuberous sclerosis complex. Neurobiol Dis. 2013;51:93–103. doi: 10.1016/j.nbd.2012.10.014. [DOI] [PubMed] [Google Scholar]
- 86.Baudouin SJ, Gaudias J, Gerharz S, et al. Shared synaptic pathophysiology in syndromic and nonsyndromic rodent models of autism. Science. 2012;338:128–132. doi: 10.1126/science.1224159. [DOI] [PubMed] [Google Scholar]
- 87.Han S, Tai C, Westenbroek RE, et al. Autistic-like behaviour in Scn1a+/− mice and rescue by enhanced GABA-mediated neurotransmission. Nature. 2012;489:385–390. doi: 10.1038/nature11356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hadj-Sahraoui N, Frederic F, Delhaye-Bouchaud N, et al. Gender effect on Purkinje cell loss in the cerebellum of the heterozygous reeler mouse. J Neurogenet. 1996;11:45–58. doi: 10.3109/01677069609107062. [DOI] [PubMed] [Google Scholar]
- 89.Zhou J, Blundell J, Ogawa S, et al. Pharmacological inhibition of mTORC1 suppresses anatomical, cellular, and behavioral abnormalities in neural-specific Pten knock-out mice. J Neurosci. 2009;29:1773–1783. doi: 10.1523/JNEUROSCI.5685-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Argyropoulos A, Gilby KL, Hill-Yardin EL. Studying autism in rodent models: reconciling endophenotypes with comorbidities. Front Hum Neurosci. 2013;7:417. doi: 10.3389/fnhum.2013.00417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gharani N, Benayed R, Mancuso V, et al. Association of the homeobox transcription factor, ENGRAILED 2, 3, with autism spectrum disorder. Mol Psychiatry. 2004;9:474–484. doi: 10.1038/sj.mp.4001498. [DOI] [PubMed] [Google Scholar]
- 92.Strick PL, Dum RP, Fiez JA. Cerebellum and nonmotor function. Annu Rev Neurosci. 2009;32:413–434. doi: 10.1146/annurev.neuro.31.060407.125606. [DOI] [PubMed] [Google Scholar]
- 93.Dibble CC, Manning BD. Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat Cell Biol. 2013;15:555–564. doi: 10.1038/ncb2763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Zoncu R, Bar-Peled L, Efeyan A, et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science. 2011;334:678–683. doi: 10.1126/science.1207056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274–293. doi: 10.1016/j.cell.2012.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Quevedo C, Salinas M, Alcazar A. Regulation of cap-dependent translation by insulin-like growth factor-1 in neuronal cells. Biochem Biophys Res Commun. 2002;291:560–566. doi: 10.1006/bbrc.2002.6479. [DOI] [PubMed] [Google Scholar]
- 97.Takei N, Inamura N, Kawamura M, et al. Brain-derived neurotrophic factor induces mammalian target of rapamycin-dependent local activation of translation machinery and protein synthesis in neuronal dendrites. J Neurosci. 2004;24:9760–9769. doi: 10.1523/JNEUROSCI.1427-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Takei N, Kawamura M, Hara K, et al. Brain-derived neurotrophic factor enhances neuronal translation by activating multiple initiation processes: comparison with the effects of insulin. J Biol Chem. 2001;276:42818–42825. doi: 10.1074/jbc.M103237200. [DOI] [PubMed] [Google Scholar]
- 99.Di Nardo A, Wertz MH, Kwiatkowski E, et al. Neuronal Tsc1/2 complex controls autophagy through AMPK-dependent regulation of ULK1. Hum Mol Genet. 2014;23:3865–3874. doi: 10.1093/hmg/ddu101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Di Nardo A, Kramvis I, Cho N, et al. Tuberous sclerosis complex activity is required to control neuronal stress responses in an mTOR-dependent manner. J Neurosci. 2009;29:5926–5937. doi: 10.1523/JNEUROSCI.0778-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Goto J, Talos DM, Klein P, et al. 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. 2011;108:E1070–E1079. doi: 10.1073/pnas.1106454108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Choi YJ, Di Nardo A, Kramvis I, et al. Tuberous sclerosis complex proteins control axon formation. Genes Dev. 2008;22:2485–2495. doi: 10.1101/gad.1685008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zhang J, Kim J, Alexander A, et al. A tuberous sclerosis complex signalling node at the peroxisome regulates mTORC1 and autophagy in response to ROS. Nat Cell Biol. 2013;15:1186–1196. doi: 10.1038/ncb2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yuan E, Tsai PT, Greene-Colozzi E, et al. Graded loss of tuberin in an allelic series of brain models of TSC correlates with survival, and biochemical, histological and behavioral features. Hum Mol Genet. 2012;21:4286–4300. doi: 10.1093/hmg/dds262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.van Slegtenhorst M, de Hoogt R, Hermans C, et al. Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science. 1997:277, 805–808. doi: 10.1126/science.277.5327.805. [DOI] [PubMed] [Google Scholar]
- 106.Xu L, Sterner C, Maheshwar MM, et al. Alternative splicing of the tuberous sclerosis 2 (TSC2) gene in human and mouse tissues. Genomics. 1995;27:475–480. doi: 10.1006/geno.1995.1079. [DOI] [PubMed] [Google Scholar]
- 107.Li Y, Inoki K, Guan KL. Biochemical and functional characterizations of small GTPase Rheb and TSC2 GAP activity. Mol Cell. Biol. 2004;24:7965–7975. doi: 10.1128/MCB.24.18.7965-7975.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Inoki K, Li Y, Xu T, et al. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 2003;17:1829–1834. doi: 10.1101/gad.1110003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Owonikoko TK, Khuri FR. Targeting the PI3K/AKT/mTOR pathway: biomarkers of success and tribulation. Am Soc Clin Oncol Educ Book. 2013 doi: 10.1200/EdBook_AM.2013.33.e395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yuan J, Mehta PP, Yin MJ, et al. PF-04691502, a potent and selective oral inhibitor of PI3K and mTOR kinases with antitumor activity. Mol Cancer Ther. 2011;10(11):2189–99. doi: 10.1158/1535-7163.MCT-11-0185. [DOI] [PubMed] [Google Scholar]
- 111.Takahashi K, Okita K, Nakagawa M, et al. Induction of pluripotent stem cells from fibroblast cultures. Nat Protoc. 2007;2:3081–3089. doi: 10.1038/nprot.2007.418. [DOI] [PubMed] [Google Scholar]
- 112.Sivapatham R, Zeng X. Generation and characterization of patient-specific induced pluripotent stem cell for disease modeling. Methods Mol Biol. 2014 Dec 18; doi: 10.1007/7651_2014_157. [ePub ahead of print] [DOI] [PubMed] [Google Scholar]
- 113.Prilutsky D, Palmer NP, Smedemark-Margulies N, et al. iPSC-derived neurons as a higher-throughput readout for autism: promises and pitfalls. Trends Mol Med. 2014;20:91–104. doi: 10.1016/j.molmed.2013.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Merkle FT, Eggan K. Modeling human disease with pluripotent stem cells: from genome association to function. Cell Stem Cell. 2013;12:656–668. doi: 10.1016/j.stem.2013.05.016. [DOI] [PubMed] [Google Scholar]
- 115.Chamberlain SJ, Chen PF, Ng KY, et al. Induced pluripotent stem cell models of the genomic imprinting disorders Angelman and Prader-Willi syndromes. Proc Natl Acad Sci U S A. 2010;107:17668–17673. doi: 10.1073/pnas.1004487107. [DOI] [PMC free article] [PubMed] [Google Scholar]
