Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that affects an individual’s reciprocal social interaction and communication ability. Numerous genetic and environmental conditions are associated with ASD, including tuberous sclerosis complex, phosphatase and tensin homolog hamartoma tumor syndrome, fragile X syndrome, and neurofibromatosis 1. The pathogenic molecular mechanisms of these diseases are integrated into the hyperactivation of mTORC1 (mechanistic target of rapamycin complex 1). Rodent models of these diseases have shown high mTORC1 activity in the brain and ASD-related behavioral deficits, which were reversed by the mTORC1 inhibitor rapamycin. Environmental stress can also affect this signaling pathway. In utero exposure to valproate caused ASD in offspring and enhanced mTORC1 activity in the brain, which was sensitive to mTORC1 inhibition. mTORC1 is a signaling hub for diverse cellular functions, including protein synthesis, through the phosphorylation of its targets, such as ribosomal protein S6 kinases. Metabotropic glutamate receptor 5–mediated synaptic function is also affected by the dysregulation of mTORC1 activity, such as in fragile X syndrome and tuberous sclerosis complex. Reversing these downstream changes that are associated with mTORC1 activation normalizes behavioral defects in rodents. Despite abundant preclinical evidence, few clinical studies have investigated the treatment of ASD and cognitive deficits. Therapeutics other than mTORC1 inhibitors failed to show efficacy in fragile X syndrome and neurofibromatosis 1. mTORC1 inhibitors have been tested mainly in tuberous sclerosis complex, and their effects on ASD and neuropsychological deficits are promising. mTORC1 is a promising target for the pharmacological treatment of ASD associated with mTORC1 activation.
Keywords: Animal models, Autism spectrum disorder, Autophagy, Mechanistic target of rapamycin, Metabotropic glutamate receptor, Protein synthesis
Autism Spectrum Disorder and Mechanistic Target of Rapamycin
Autism spectrum disorder (ASD) is a major neurodevelopmental disorder that affects around 1% of the general population. ASD symptoms are categorized into 1) social communication deficits and 2) reciprocal and repetitive interests/behaviors (1). Various genetic deficits are identified in individuals with ASD, including single-gene disorders (2), copy number variations in specific loci, and chromosomal alterations (3). Environmental stress, such as premature birth and drug exposure in utero, can also give rise to ASD in offspring (4,5). Despite accumulating knowledge of the etiology of ASD, relevant knowledge that can contribute to the development of effective treatments for ASD is still sparse.
One mechanism of ASD that may reveal new therapeutic strategies is dysregulation of the mTOR (mechanistic target of rapamycin) complex (mTORC). The central component of the complex is mTOR, a protein with serine-threonine kinase activity. mTOR forms complexes with several proteins to form mTORC1 and mTORC2 (6). The most important difference between mTORC1 and mTORC2 is that mTORC1 consists of raptor (regulatory associated protein of mTOR), whereas mTORC2 consists of rictor (rapamycin-insensitive companion of mTOR), making it insensitive to rapamycin (7,8). mTORC1 activity is under the regulation of environmental signals, such as growth factors, hypoxia, and energy levels (9). Some inputs activate mTORC1, such as the PI3K/Akt (phosphoinositide 3-kinase/protein kinase B) pathway (10) and Ras/MEK/ERK (rat sarcoma/mitogen-activated protein kinase kinase/extracellular signal–regulated kinase) pathway (11), whereas others inhibit mTORC1, such as the hypoxia/AMPK (5′-adenosine monophosphate–activated protein kinase) pathway (12). These signals are transmitted to TSC1/2 (tuberous sclerosis complex 1/2), which is suppressed by inputs from most pathways while activated by the AMPK pathway. The activation of TSC1/2 suppresses downstream Rheb (Ras homolog enriched in brain) by converting the active GTP (guanosine-5′-triphosphate)–bound form to the inactive GDP (guanosine diphosphate)–bound form. Rheb directly stimulates mTORC1, and TSC1/2 activity results in a reduction of mTORC1 activity, and vice versa (13). mTORC1 phosphorylates its targets and modulates their functions, including S6Ks (ribosomal protein S6 kinases, which control global protein synthesis) (14), eIF4E (eukaryotic translation initiation factor 4E)–binding proteins (4E-BPs; which enhance the initiation of cap-dependent translation) (15), and ULK1 (unc51-like autophagy-activating kinase 1, which suppresses the initiation of macroautophagy) (16).
Several human genetic disorders are known to be associated with mTORC1 activation and ASD (Figure 1). Rapamycin does not directly bind to mTOR but instead binds to FKBP12 (FK506-binding protein 12) to form the FKBP12-rapamycin complex. This complex can then bind to mTOR in mTORC1 and suppress mTORC1 activity, whereas mTORC2 does not interact with the FKBP12 complex because of the presence of rictor (17). mTORC1 inhibitors are currently widely used to treat tumors and drug-resistant epilepsy in TSC (18) and malignancies in different organs (19). Rapamycin and its analogs are expected to reverse mTORC1 hyperactivity and related neuropsychiatric deficits, such as ASD.
Figure 1.
Signaling pathways that involve mTORC1. Physiological inputs to these pathways include growth factors through RTK, the elevation of calcium ion concentrations through CaMKKb, and hypoxia and glucose deprivation through LKB1. These and other signals that stem from FMRP converge in TSC1/TSC2. The activation of TSC1/TSC2 results in mTORC1 inhibition and decreases protein synthesis (ribosomal protein S6), macroautophagy initiation (ULK1), and cap-dependent translation initiation (EIF4E). The net result of the defect that is discussed in the text (highlighted in orange) is mTORC1 activation. A number of drugs that have been evaluated in humans (highlighted in yellow) inhibit each target molecule. Arrows indicate activation. Lack of arrows indicates inhibition. 4E-BP, eukaryotic translation initiation factor 4E-binding protein; AKT, protein kinase B; AMPK, adenosine monophosphate–activated protein kinase; CaMKKb, calcium/calmodulin-dependent protein kinase kinase β; CYFIP1, cytoplasmic fragile X mental retardation protein-interacting protein 1; eIF4E, eukaryotic translation initiation factor 4E; ERK, extracellular signal–regulated kinase; FMRP, fragile X mental retardation protein; GSK-3, glycogen synthase kinase 3; LKB1, liver kinase B1 (serine/threonine protein kinase 11); mTORC, mechanistic target of rapamycin complex; NF1, neurofibromatosis 1; PI3K, phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; raptor, regulatory associated protein of mTOR; RAS, Rat sarcoma; Rheb, Ras homolog enriched in brain; rictor, rapamycin-insensitive companion of mTOR; RTK, receptor tyrosine kinase; S6, ribosomal protein S6; S6K, S6 kinase; TSC, tuberous sclerosis complex; ULK1, unc51-like autophagy-activating kinase 1; VPA, valproic acid.
ASD and Human Diseases
Human genetic disorders and environmental factors have provided insights into the critical role of mTORC1 dysregulation in ASD. Although not every disorder is found in all individuals with ASD, these disorders provide insights into the ways in which ASD is caused and how it can be reversed pharmacologically in a molecular manner (Table 1).
Table 1.
ASD-Related Behavioral Deficits in Rodent Models of mTORC1-Associated ASD
| Mutation/Exposure | Findings | Treatment | Reference |
|---|---|---|---|
| Tsc1+/- | (F) ↓ social interaction, ↓ nest building | – | (38) |
| (M, F) ↓ social interaction, ↑ rearings | 5 mg/kg rapamycin, 2 days | (41) | |
| Tsc2+/- | (M, F) ↓ social interaction, ↑ rearings | 5 mg/kg rapamycin, 2 days | (41) |
| (M) ↓ sociability, ↓ social novelty preference | 3 mg/kg rapamycin, 1 week | (117) | |
| (M) ↓ reversal learning | 30 mg/kg MPEP, 2 days | (120) | |
| Tsc2+/−, Rats | (M) ↓ social interaction, ↓ rearings | – | (42) |
| (M) ↓ social interaction, ↓ social discrimination | 1 mg/kg everolimus, 3 times per week | (43) | |
| L7Cre;Tsc1flox/flox | (M) ↓ sociability, ↓ social novelty preference, ↑ self-grooming, ↑ ultrasonic vocalizations |
6 mg/kg rapamycin, daily from P7 | (44) |
| Pcp2Cre;Tsc2flox/ko | (M, F) ↓ sociability, ↓ social novelty preference, ↑ marble burying |
2 mg/kg rapamycin, 3 times per week | (45) |
| Pten+/− | (M, F) ↓ sociability, ↓ social novelty preference, ↑ brain mass |
– | (58) |
| (F) ↓ sociability, ↓ social novelty preference | 0.3 mg/kg SB 242084 (5-HT2C receptor antagonist), 20 min before testing | (59) | |
| (F) ↓ sociability | 75 mg/kg PF-4708671 (S6K1 inhibitor), daily from P4 to P14 | (60) | |
| (F) ↓ sociability, ↓ social novelty preference (M, F) ↑ brain mass |
– | (144) | |
| L7Cre;Ptenflox/flox | (M) ↓ social interaction, ↓ sociability, ↑ rearings | – | (145) |
| NseCre;Pten+/flox | (M) ↓ sociability, ↓ social novelty preference | – | (146) |
| NseCre;Ptenflox/flox | (M) ↓ social interaction, ↓ sociability, ↓ social novelty preference, ↑ brain mass | – | (61) |
| (M) ↓ social interaction, ↑ brain mass | 10 mg/kg rapamycin, 5 times per week | (62) | |
| GFAPCre;Ptenflox/flox | (NR) ↓ sociability, ↑ marble burying, ↑ hole-poke, ↑ rearings | – | (147) |
| Nestin-CreRET2;Ptenflox/flox | (M) ↓ social interaction, ↓ sociability, ↑ brain mass | – | (148) |
| OxtCre;Ptenflox/flox | (M) ↓ social recognition (F) ↓ social novelty preference |
– | (149) |
| Camk2aCre;Ptenflox/flox | (M, F) ↓ reversal learning, ↓ social novelty preference | Genetic removal of rapamycin-insensitive companion of mTOR | (63) |
| Nkx2.1Cre;Pten+/flox | (M) ↓ social interaction | – | (150) |
| Pten-ΔPDZ | (NR) ↓ sociability, ↑ brain mass | – | (151) |
| Fmr1 KO (C57BL/6 Background) | (M) ↓ reversal learning, ↓ social novelty preference, ↑ marble burying | Genetic removal of S6K1 | (74) |
| Fmr1 KO (C57BL/6J Background) | (M) ↓ social novelty preference | 1.75 mg/kg rapamycin, incorporated into mouse chow | (84) |
| (M) ↑ perseveration | – | (152) | |
| Fmr1 KO (FVB/129P2 Background) | (M) ↑ perseveration | 20 mg/kg MPEP, 30 min before testing | (153) |
| Nf1+/− | (M) ↓ long-term social learning | Genetic removal of Pak1 | (91) |
| GFAPCre;Nf1flox/flox | (M) ↓ ultrasonic vocalizations (M, F) ↓ ultrasonic vocalizations |
– | (93) |
| VPA In Utero Single Dose | (M) ↓ social interaction | 10 mg/kg rapamycin, 2 days | (105) |
| (M) ↓ sociability, ↑ marble burying | 5 mg/kg rapamycin, 5 days | (106) | |
| (M) ↓ sociability | 30 mg/kg BrBzGCp2 (glyoxylate 1 inhibitor), 10 hours before testing | (154) | |
| (M) ↓ sociability, ↓ social novelty preference, ↑ self-grooming, ↑ marble burying | Clonazepam and baclofen infused into mPFC | (155) | |
| VPA In Utero Single Dose, Rats | (M) ↓ sociability, ↓ social novelty preference | 1 mg/kg rapamycin, daily from P23 to P33 5 mg/kg sulindac, daily from P23 to P33 |
(107) |
| S6K1 KO | (M) ↓ social novelty preference, ↑ marble burying | – | (74) |
| 4E-BP2 KO | (M) ↓ sociability, ↓ social interaction, ↑ self-grooming, ↑ marble burying, ↑ ultrasonic vocalizations | Genetic addition of eIF4E | (110) |
| (M) ↓ sociability, ↑ marble burying | 0.3 mg/kg JNJ162596385 (mGlu1 antagonist), 30 min before testing 3 mg/kg fenobam (mGlu5 antagonist), 24 hours before testing |
(111) | |
| eIF4E Transgenic | (M) ↓ sociability, ↓ social interaction, ↑ self-grooming, ↑ marble burying | 4EGI-1 infused intracerebroventricularly | (112) |
| CamKIICre;Atg7flox/flox | (M) ↓ social interaction, ↓ sociability, ↓ social novelty preference | – | (117) |
ASD, autism spectrum disorder; F, female; KO, knockout; M, male; mPFC, medial prefrontal cortex; P, postnatal day; VPA, valproic acid.
Tuberous Sclerosis Complex
Historically, TSC (Mendelian Inheritance in Man [MIM] #191100, #613254) has been defined as a neurocutaneous syndrome (genetic disorder that affects the skin and brain) and presents with facial angiofibroma, epilepsy, and intellectual disability (20,21). The identification of two genes that cause TSC, TSC1 (22) and TSC2 (23), expanded knowledge of phenotypic variations of TSC, such as those that are represented in current diagnostic criteria. Major manifestations of TSC include hamartomatous lesions in different organs and in different age groups (24). TSC is estimated to occur in 1 in 6000 live births, with no regional, ethnic, or sexual differences (25,26). Compared with other neurocutaneous syndromes, TSC is more likely to be associated with neurologic complications, such as characteristic brain tumors (e.g., subependymal giant cell astrocytoma) and drug-resistant epilepsy (27). Neurodevelopmental disorders and psychiatric problems, such as anxiety and depression, are also often found in patients with TSC. These disorders are collectively referred to as TSC-associated neuropsychiatric disorders (28). Up to half of patients with TSC are diagnosed with ASD, equally divided among males and female patients (2,29). This lack of sex differences contrasts with the observation that males are approximately three times more vulnerable to ASD than females (30). This suggests that a diagnosis of TSC may be an independent risk factor for ASD, regardless of sex.
The TSC1 and TSC2 genes encode hamartin and tuberin, respectively (22,23). TSC1 and TSC2 form a heterodimer that receives signaling inputs (Figure 1). The activated heterodimer suppresses Rheb, resulting in mTORC1 inhibition. The haploinsufficiency of either TSC1 or TSC2 weakens the suppression of Rheb and renders mTORC1 uninhibited. Activated mTORC1 is considered a central pathomechanism of TSC (Figure 1) (13). Individuals with mutations of TSC1 or TSC2 generally present the same phenotype, whereas individuals with TSC2 mutations are more severely affected in certain aspects, such as more severe skin and kidney involvement (31,32), more severe intellectual disability, heavier seizure burden (32,33), and ASD (33,34). Despite these findings, the mechanisms by which these phenotypes arise are not well understood, especially how TSC2 mutations cause more severe manifestations.
Soon after the identification of TSC2, the Eker rat model of hereditary renal carcinoma was found to have a germline mutation of the Tsc2 gene (35), followed by the establishment of a mouse model of TSC with the germline haploinsufficiency of Tsc1 and Tsc2 (36,37). These rodents, however, lack characteristic brain lesions, such as cortical tubers, that are found in most human patients with TSC (38,39). Similar to patients with TSC, Tsc1+/− and Tsc2+/− heterozygous knockout mice exhibited ASD-related deficits in social interaction in both males and females and cognitive deficits (38,40,41). Eker rats also exhibited ASD-related social impairments, regardless of concomitant status epilepticus (42,43). This body of evidence suggests that epilepsy and brain lesions are not essential for developing ASD or cognitive deficits in TSC.
The influence of the complete deletion of Tsc1 and Tsc2 on the brain and behavior has been investigated to explain the gap between humans who harbor obvious brain lesions and mice that have no morphological changes in the brain. The loss of Tsc1 and Tsc2 in cerebellar Purkinje cells resulted in progressive Purkinje cell loss and ataxia and ASD-related social deficits (44,45). This phenotype was much more severe when Tsc1 and Tsc2 were deleted primarily in glial cells. These knockout mice exhibited epileptic seizures, progressive macrocephaly, and early mortality. Histological alterations included diffuse glial cell proliferation and the dispersion of hippocampal pyramidal cells (46,47). Intriguingly, these histological and neurologic changes and the elevation of mTORC1 activation were more severe in Tsc2 knockout mice (47), consistent with more severe manifestations in human patients with TSC.
These rodent models also provide preclinical data on the potential therapeutic usefulness of mTORC1 inhibitors for neurologic manifestations of TSC. Short-term rapamycin treatment reversed ASD-related behavior (41,43) and cognitive deficits (40) through mTORC1 inhibition. Chronic rapamycin treatment prevented seizures, premature death (47), ASD-related behavioral deficits (44), and related histological abnormalities without serious adverse effects. These findings strongly suggest that hyperactivated mTORC1 is sufficient to cause ASD and other cognitive deficits in TSC.
PTEN Hamartoma Tumor Syndrome
The PTEN gene was originally discussed with regard to genetic predispositions to various tumors, such as Bannayan-Riley-Ruvalcaba syndrome (MIM #153480; e.g., macrocephaly, intellectual disability, and multiple intestinal hamartomas), Cowden syndrome (MIM #158350; e.g., macrocephaly, mucocutaneous lesions, intestinal polyps, and a higher risk of malignancies) (48,49), and Lhermitte-Duclos syndrome (MIM #158350; e.g., dysplastic gangliocytoma of the cerebellum, ataxia, and increase in intracranial pressure) (50). These disorders share macrocephaly as a common presentation and developmental problems, including ASD, suggesting a relationship between these disorders and ASD. A subset of ASD individuals with extreme macrocephaly were found to have the mutated PTEN gene, consequently referred to as “macrocephaly/autism syndrome” (MIM #605309) (51,52). These conditions are now considered to result from PTEN gene mutations. They are recognized as a spectrum called PTEN hamartoma tumor syndrome (PHTS) (53). Approximately 7%–17% of individuals with ASD and macrocephaly and 1%–5% of those with ASD carry mutations of the PTEN gene (51,54,55). A recent comparative study found that core ASD symptoms were similar between ASD with PTEN mutations and ASD with nonsyndromic ASD and macrocephaly. Those with the PTEN mutation, however, had less severe symptoms, suggesting distinct pathogenetic mechanisms that underlie PHTS (56).
The PTEN gene encodes phosphatase and tensin homolog, a protein that negatively regulates the cancer-related PI3K/AKT/mTOR signaling pathway (57). PI3K is activated by inputs from receptor tyrosine kinase (e.g., insulin and such growth factors as insulin-like growth factor-1) and converts PIP2 (phosphatidylinositol 4,5-bisphosphate) to PIP3 (phosphatidylinositol (3,4,5)-trisphosphate). An increase in PIP3 facilitates the activation of Akt and inhibition of TSC1/TSC2, leading to the suppression of mTORC1 activity. This explains how deletion of the PTEN gene results in mTORC1 activation and ASD. The presence or absence of involvement of the PI3K/Akt pathway may also suggest why TSC and PHTS are distinct with regard to the predisposition to benign and malignant tumors.
Pten deletion in mice revealed the significance of PTEN in the pathogenesis of macrocephaly and ASD-related abnormal behavior through constitutive mTORC1 hyperactivation. Pten+/− mice are a model of PHTS in human patients. These mice exhibited ASD-related deficits in social interaction and mild macrocephaly (58, 59, 60). Deletion of the Pten gene in neurons in the cerebral cortex and hippocampus recapitulated macrocephaly and ASD-related abnormal behavior, such as less interest in other mice, and these effects were associated with an increase in Akt and S6 phosphorylation in the brain (61).
The recovery of these behavioral deficits in these Pten mutant mice was associated with mTORC1 inhibition. ASD-related social deficits in Pten+/− mice were reversed by the pharmacological inhibition of S6, similar to mTORC1 inhibition (60). In neuron-specific Pten knockout mice, chronic treatment with rapamycin prevented the development of brain hypertrophy and ASD-related abnormal behavior and reduced S6 phosphorylation (62). ASD-like behavior in frontal robe–specific Pten knockout mice was improved by the genetic removal of rictor but not raptor (63), suggesting that mTORC1 inhibition by rapamycin may have only a limited effect in correcting the phenotype that is associated with Pten mutations. Different brain region involvement and different dosages of the Pten mutation may explain these variable effects.
Fragile X Syndrome
Fragile X syndrome (FXS) (MIM #300624) is one of the most common causes of hereditary intellectual disability. Its phenotype includes intellectual disability, ASD, anxiety, and physical features, such as macrocephaly and macroorchidism in males (64). Unlike the diseases that are discussed above, the genetic defect in most FXS cases is the elongation of CGG repeats in the 5′-untranslated region of the FMR1 gene, located on the X chromosome (65,66). Typically, individuals with FXS have a CGG repeat size greater than 200 in the FMR1 gene, called a “full mutation” (67), whereas a shorter FMR1 repeat size (between 55 and 200, premutation) is also relevant to the characteristic movement disorder, called fragile X–associated tremor/ataxia syndrome (68). FXS as diagnosed by detection of the full mutation is found in 1 in 7100 males and 1 in 11,000 females (69). Approximately 30% of male patients with FXS have ASD (2).
In FXS, the elongation of CGG repeats decreases expression of the FMR1 gene. The protein product of the FMR1 gene, FMRP (fragile X mental retardation protein), is an RNA binding protein that represses the translation of postsynaptic components of neurons (70,71). One of the binding partners of FMRP is CYFIP1 (cytoplasmic FMRP-interacting protein 1). The FMRP-CYFIP1 complex binds to eIF4E and inhibits the initiation of translation, which is abolished in FXS (72). As noted above, eIF4E-mediated translation initiation is also suppressed by the sequestration of eIF4E by 4E-BP. mTORC1 hyperactivity reduces 4E-BP binding to eIF4E, resulting in the enhancement of eIF4E-mediated translation initiation. Uninhibited eIF4E-mediated initiation of translation may play a role in the development of ASD in FXS.
Fmr1 knockout animals have been extensively investigated to clarify their behavioral similarity to human patients with FXS (73). ASD-related impairments in social interaction and repetitive behavior were observed in different studies of Fmr1 knockout mice and rats, along with audiogenic seizures and other cognitive deficits (74,75). The molecular pathophysiology of FXS was first revealed with regard to mGlu5, one of two group I metabotropic glutamate receptors (mGluRs), along with mGlu1. Phenotypic alterations in Fmr1 knockout mice, such as impairments in memory and excessive protein synthesis in the hippocampus, were prevented by reducing mGlu5 expression (76). The pharmacological inhibition of mGlu1 and mGlu5 also effectively reduced repetitive behavior in Fmr1 knockout mice. mGlu5 inhibition was superior to mGlu1 inhibition in improving motor learning in the rotarod test and abolishing audiogenic seizures (77).
The dysregulation of eIF4E-mediated translation initiation is involved in FXS and TSC (78), suggesting that FXS and TSC may share a molecular mechanism that leads to ASD. Analyses of the brain in Fmr1 knockout mice revealed higher messenger RNA levels of a set of FMRP target genes, including Mtor and Nf1 (79). The brains of Fmr1 knockout mice showed increases in PI3K (80), ERK (81), and mTORC1 activity (79,82). These alterations were mediated by an increase in mGlu5 activity in Fmr1 knockout mice (83). The associated increase in basal protein synthesis was normalized by ERK inhibition but not by PI3K or mTORC1 inhibition (81,83). The long-term administration of rapamycin also failed to correct behavioral deficits in Fmr1 knockout mice (84). Although there is a molecular pathophysiology interaction between FXS and TSC, these findings suggest that the pharmacological treatment of neuropsychiatric manifestations of FXS should be directed toward inhibiting mGlu5 rather than mTORC1.
Neurofibromatosis 1
Neurofibromatosis 1 (NF1) (MIM #162200) is the most prevalent neurocutaneous syndrome, which is diagnosed in 1 in 3000–4000 individuals. NF1 and numerous other genetic disorders, such as Noonan syndrome, share activation of the RAS/MEK/ERK pathway; thus, they are collectively called RASopathies (85). Its phenotype includes characteristic skin lesions and tumors that occur mainly in peripheral nerves (86). A higher risk of ASD has long been suspected in individuals with NF1. Recent observational studies reported that 15%–30% of patients with NF1 have ASD (87,88).
The NF1 gene product neurofibromin functions as a GTPase-activating protein for RAS. Neurofibromin negatively regulates the signaling pathway from RAS by converting the active GTP-bound form to the inactive GDP-bound form (89). Mutations of the NF1 gene thus result in RAS activation and downstream ERK activation. The activation of ERK, in turn, represses the TSC1/2 complex, finally leading to mTORC1 hyperactivity (86). Nf1+/− mice were found to have cognitive deficits (90), social impairments (91,92), and an increase in ultrasonic vocalizations (93). To date, the efficacy of reversing the NF1 phenotype has been investigated by inhibiting ERK (discussed below), whereas mTORC1 inhibition has not been tested.
In Utero Exposure to Valproic Acid
Exposure to antiepileptic drugs and psychotropic drugs during pregnancy is also associated with ASD (see the Supplement). Valproic acid (VPA) has long been used as an antiepileptic drug, particularly for generalized epilepsies (94), and several medical conditions, such as migraine (95) and bipolar disorder (96). The use of VPA during pregnancy can be considered an environmental stressor that increases the risk of ASD. Compared with use of other antiepileptic drugs, in utero exposure to VPA was significantly associated with ASD (4), a lower IQ at age 6 years (97), and a higher risk of congenital malformations in offspring (98). Females who are able to be pregnant are now recommended to avoid using VPA as much as possible (99).
The condition in which in utero exposure to VPA results in ASD has been replicated in rodents by giving a single high dose of VPA during pregnancy (100). One of the major mechanisms that causes these ASD-related deficits is through the inhibitory actions of VPA on histone deacetylase (HDAC) (101). The critical role of HDAC inhibition in development of the ASD-related phenotype was demonstrated by comparing mice that were exposed to VPA and valpromide, a VPA analog that is devoid of HDAC activity (102). VPA also activates the PI3K/Akt/mTORC1 pathway (103). Mice and rats that were exposed to VPA in utero exhibited an increase in mTORC1 activity in the brain and ASD-related social deficits. These deficits were reversed by postnatal treatment with rapamycin (104, 105, 106), indicating that constitutively active mTORC1 disrupts social behavior in these rodents.
Trajectories From mTORC1 Dysregulation to ASD
The abovementioned causes of ASD, both genetic and environmental, share the dysregulation of mTORC1-mediated signaling. However, other mechanisms may also be involved in the development of ASD, such as elevations of Akt activation in PHTS. Several proteins that are targeted by mTORC1 may give rise to ASD-related phenotypes.
S6Ks—Protein Synthesis
One of the central functions of mTORC1 is to control, likely by upregulating, protein synthesis through the phosphorylation of S6Ks (14). The hippocampus in Fmr1 knockout mice showed higher rates of protein synthesis (107). Crossing Fmr1 knockout mice with S6K1 knockout mice eliminated the dysregulation of protein synthesis and reversed ASD-related behavioral deficits (74). These findings suggest that the elevation of S6K activity elicits excessive protein synthesis and ASD-related deficits in FXS. In contrast, hippocampal slices from Tsc2+/− mice had lower rates of global protein synthesis (108). A reduction of the protein synthesis rate was observed in different cerebral regions in freely moving Tsc2+/− mice (109), despite the fact that S6K was activated in these mice (40,41). These inconsistent findings raise the possibility of complex pathways that regulate protein synthesis that is associated with mTORC1 and S6K1 activation.
4E-BPs—Initiation of Translation
The translation of a set of synaptic proteins is repressed by 4E-BPs that bind to eIF4E. The mTORC1 phosphorylation of 4E-BPs releases its inhibition of eIF4E and stimulates translation initiation (15). The germline knockout of 4E-BP2, the major form of 4E-BP in the mammalian brain, engendered ASD-related behavioral deficits that were associated with the exacerbation of mGlu5 long-term depression (LTD) (110,111). ASD-related social impairment was also observed in eIF4E transgenic mice, along with an increase in cap-dependent translation and the enhancement of LTD (112). These behavioral, synaptic, and translational defects were normalized by administration of the eIF4E inhibitor 4EGI-1 (110,112) and an mGlu5 inhibitor (111). Interestingly, cognitive impairment in Fmr1 knockout mice was also rescued by 4EGI-1 (113), possibly through suppression of the eIF4E-mediated increase in CYFIP1-induced translation. The above evidence suggests a causal role for eIF4E hyperactivity in cognitive deficits and ASD.
ULK1—Autophagy
Autophagy is a cellular pathway that removes unnecessary proteins and damaged organelles. One of the most notable regulatory pathways is the PI3K/Akt/mTORC1 pathway. Inputs from this pathway suppress autophagy initiation by phosphorylating ULK1 and autophagy factor Atg13, which is necessary for autophagosome formation (19,114,115). Once initiated, autophagy progresses through the involvement of several other ATG proteins (114,116).
The comparison of postmortem brain samples from individuals with ASD and those without ASD revealed higher mTORC1 activity and impairments in autophagy in ASD (117). A similar suppression of autophagy was replicated in the brain in Tsc2+/− mice (117) and neurons that lacked Tsc1 and Tsc2 (117,118). VPA exposure in utero also caused mTORC1 activation and ULK1 phosphorylation, resulting in a decrease in autophagy in the brain. Rapamycin rescued social behaviors and restored ULK1 activity and autophagy (106). These findings suggest that mTORC1 hyperactivation through different mechanisms result in deficits in autophagy and social behaviors.
Interestingly, Tsc2-null neurons exhibited an increase in AMP (adenosine monophosphate) levels AMPK activation, implying energetic stress in the absence of Tsc1/2. This then led to direct ULK1 stimulation and mTORC1-independent autolysosome accumulation. Similar changes were observed in mice that lacked Tsc1 in Purkinje cells and human cortical tuber samples (118). Autophagy regulation in response to energetic stress may be an mTORC1-independent backup mechanism, suggesting the potential of therapeutically targeting this mechanism using adenosine triphosphate–competitive mTOR inhibitors and the biguanide metformin (119).
mGlu5-Mediated Synaptic Function
As discussed above, the abnormal enhancement of mGlu5 activity was first revealed in FXS (76) and then studied in TSC because these two diseases share ASD and mTORC1 hyperactivity. To date, the findings with regard to mGlu5 activity in TSC is inconsistent. First, the Tsc2+/− hippocampus was found to have a decrease in mGlu5 LTD and reduced basal protein synthesis as well as cognitive deficits, in clear contrast to FXS. Deficient mGluR LTD and associated abnormalities were restored by rapamycin and a positive allosteric modulator of mGlu5 (108). Later, however, an conflicting finding was reported. Tsc2+/− hippocampus showed an increase in mGlu5 LTD and impairments in reverse learning. These deficits were reversed by an mGlu5 inhibitor, similar to Fmr1 knockout mice (120). Another strain of Tsc2 mutant mice was investigated, in which the level of TSC2 protein was reduced to approximately 7% relative to wild-type mice. These mutant mice exhibited hyperactivity and epileptic seizures, which were improved by mGlu5 inhibition with an mGlu5 negative allosteric modulator and aggravated by mGlu5 potentiation with another mGlu5 positive allosteric modulator (121). mGluR LTD was maintained in S6K1 knockout mice but enhanced in S6K2 knockout mice with or without S6K1 deletion (122). mGlu5 dysfunction likely underlies cognitive deficits, including in ASD. Further clarification is required to ascertain whether mGlu5 LTD is enhanced or deficient in response to mTORC1 hyperactivity.
Development of Pharmacotherapy for mTOR-Related ASD in Humans
Challenging Experiences in FXS and NF1
The development of pharmacological therapeutics for neuropsychiatric problems that are associated with the aforementioned diseases were first attempted in FXS. The first candidate was an mGluR inhibitor, such as MPEP, and another was a GABAB (gamma-aminobutyric acid B) receptor agonist. Audiogenic seizures in Fmr1 knockout mice were inhibited by the GABAB receptor agonist baclofen and worsened by a GABAB receptor antagonist (123). This line of preclinical evidence prompted researchers to conduct clinical trials with mGlu5 inhibitors and a number of compounds, including GABAB receptor agonists, for neuropsychiatric symptoms in FXS (124). Despite the clinical expectations based on preclinical results, no therapeutic efficacy was found in randomized clinical trials with FXS participants who were treated with the selective mGlu5 inhibitor mavoglurant (125) and GABAB receptor agonist arbaclofen (126). There was one promising report of another clinical trial of mavoglurant. In this trial, visual attention and pupil reactivity while viewing photographs of faces were used as primary outcomes, and mavoglurant treatment resulted in significant improvement (127).
As noted above, the central pathophysiology of NF1 is disinhibition of the RAS/MEK/ERK pathway. The HMG-CoA (β-hydroxy-β-methylglutaryl–coenzyme A) inhibitors lovastatin and simvastatin inactivated ERK (128) and ameliorated cognitive dysfunction in Nf1+/− mice (129). Based on these findings, independent randomized clinical trials were conducted with lovastatin and simvastatin. Some of these trials reported improvements in cognitive function (130,131), but others failed to find therapeutic efficacy in ASD and neuropsychiatric symptoms (132, 133, 134, 135).
mTORC1 Inhibitors in Clinical Studies of ASD
Despite the abundance of evidence in rodent models, clinical studies are limited with regard to the effects of mTORC1 inhibitors on neuropsychiatric phenotypes. Anecdotal case reports have described that ASD and behavioral problems were improved by everolimus that was used for subependymal giant cell astrocytoma (136) and drug-resistant epilepsy (137). Changes in ASD symptoms and cognitive function were evaluated as secondary outcomes in clinical studies of mTORC1 inhibitors for TSC-associated lesions. Some studies observed improvements in cognitive function and ASD-related behavior. Patients with TSC-associated renal and pulmonary lesions exhibited improvements in neurocognitive performance after sirolimus therapy (138). Studies of everolimus for drug-resistant epilepsy in individuals with TSC revealed improvements in ASD-related behavior in an open-label study (139) and a randomized study (140). Another open-label study of subependymal giant cell astrocytoma reported a negative effect of everolimus on neuropsychological function (141). Later, a randomized controlled trial was conducted, in which children with TSC who had ASD or other neurodevelopmental disorders but no drug-resistant epilepsy were enrolled to investigate the effect of everolimus for these conditions. Everolimus did not improve ASD or neuropsychological deficits (142).
At the time of writing this article, we found no reports of changes in ASD-related behaviors by mTORC1 inhibitors in patients with PTEN-related disorders. One randomized controlled trial will be conducted for PHTS and comorbid ASD using everolimus (143).
Concerns about using mTORC1 inhibitors for ASD include their adverse effects. Some are mild (e.g., stomatitis and hyperlipidemia), but others can be serious (e.g., immunosuppression and interstitial lung disease) (137, 138, 139, 140, 141, 142, 143). The benefits of treating ASD and risk of adverse effects should be cautiously weighed.
Conclusions
In this review, the central role of constitutively active mTORC1-mediated signal transduction was discussed in the context of ASD and its molecular pathophysiology. We presented rodent models of human disorders and the current state of developing pharmacological therapeutics. Currently, mTORC1 inhibitors are the most promising drugs that can improve ASD, which have been widely used in patients with TSC and other diseases. However, it remains unclear whether their beneficial effects on ASD and related neurocognitive conditions surpass their adverse effects, which can be severe. Preclinical evidence is abundant, but further clinical experience is needed with regard to the efficacy of mTORC1 inhibitors and other promising agents, such as mGlu5 inhibitors, in ameliorating ASD.
Acknowledgments and Disclosures
This work was supported by a grant from the Japan Society for the Promotion of Science KAKENHI (Grant No. 21H03028 [to KI]).
We thank Michael Arends for proofreading the manuscript.
The authors report no biomedical financial interests or potential conflicts of interest.
Footnotes
Supplementary material cited in this article is available online at https://doi.org/10.1016/j.bpsgos.2021.08.005.
Supplementary Material
References
- 1.American Psychiatric Association . 5th ed. American Psychiatric Publishing; Washington, DC: 2013. Diagnostic and Statistical Manual of Mental Disorders. [Google Scholar]
- 2.Richards C., Jones C., Groves L., Moss J., Oliver C. Prevalence of autism spectrum disorder phenomenology in genetic disorders: A systematic review and meta-analysis. Lancet Psychiatry. 2015;2:909–916. doi: 10.1016/S2215-0366(15)00376-4. [DOI] [PubMed] [Google Scholar]
- 3.Rylaarsdam L., Guemez-Gamboa A. Genetic causes and modifiers of autism spectrum disorder. Front Cell Neurosci. 2019;13:385. doi: 10.3389/fncel.2019.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Christensen J., Grønborg T.K., Sørensen M.J., Schendel D., Parner E.T., Pedersen L.H., Vestergaard M. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309:1696–1703. doi: 10.1001/jama.2013.2270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Agrawal S., Rao S.C., Bulsara M.K., Patole S.K. Prevalence of autism spectrum disorder in preterm infants: A meta-analysis. Pediatrics. 2018;142 doi: 10.1542/peds.2018-0134. [DOI] [PubMed] [Google Scholar]
- 6.Bockaert J., Marin P. mTOR in brain physiology and pathologies. Physiol Rev. 2015;95:1157–1187. doi: 10.1152/physrev.00038.2014. [DOI] [PubMed] [Google Scholar]
- 7.Sarbassov D.D., Ali S.M., Kim D.H., Guertin D.A., Latek R.R., Erdjument-Bromage H., et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol. 2004;14:1296–1302. doi: 10.1016/j.cub.2004.06.054. [DOI] [PubMed] [Google Scholar]
- 8.Gaubitz C., Oliveira T.M., Prouteau M., Leitner A., Karuppasamy M., Konstantinidou G., et al. Molecular basis of the rapamycin insensitivity of target of rapamycin complex 2. Mol Cell. 2015;58:977–988. doi: 10.1016/j.molcel.2015.04.031. [DOI] [PubMed] [Google Scholar]
- 9.Jozwiak J., Jozwiak S., Wlodarski P. Possible mechanisms of disease development in tuberous sclerosis. Lancet Oncol. 2008;9:73–79. doi: 10.1016/S1470-2045(07)70411-4. [DOI] [PubMed] [Google Scholar]
- 10.Dibble C.C., Cantley L.C. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015;25:545–555. doi: 10.1016/j.tcb.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mendoza M.C., Er E.E., Blenis J. The Ras-ERK and PI3K-mTOR pathways: Cross-talk and compensation. Trends Biochem Sci. 2011;36:320–328. doi: 10.1016/j.tibs.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sukumaran A., Choi K., Dasgupta B. Insight on transcriptional regulation of the energy sensing AMPK and biosynthetic mTOR pathway genes. Front Cell Dev Biol. 2020;8:671. doi: 10.3389/fcell.2020.00671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang Y., Gao X., Saucedo L.J., Ru B., Edgar B.A., Pan D. Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat Cell Biol. 2003;5:578–581. doi: 10.1038/ncb999. [DOI] [PubMed] [Google Scholar]
- 14.Magnuson B., Ekim B., Fingar D.C. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012;441:1–21. doi: 10.1042/BJ20110892. [DOI] [PubMed] [Google Scholar]
- 15.Qin X., Jiang B., Zhang Y. 4E-BP 1, a multifactor regulated multifunctional protein. Cell Cycle. 2016;15:781–786. doi: 10.1080/15384101.2016.1151581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dossou A.S., Basu A. The emerging roles of mTORC1 in macromanaging autophagy. Cancers. 2019;11:1422. doi: 10.3390/cancers11101422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Annett S., Moore G., Robson T. FK506 binding proteins and inflammation related signalling pathways; Basic biology, current status and future prospects for pharmacological intervention. Pharmacol Ther. 2020;215:107623. doi: 10.1016/j.pharmthera.2020.107623. [DOI] [PubMed] [Google Scholar]
- 18.MacKeigan J.P., Krueger D.A. Differentiating the mTOR inhibitors everolimus and sirolimus in the treatment of tuberous sclerosis complex. Neuro Oncol. 2015;17:1550–1559. doi: 10.1093/neuonc/nov152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hua H., Kong Q., Zhang H., Wang J., Luo T., Jiang Y. Targeting mTOR for cancer therapy. J Hematol Oncol. 2019;12:71. doi: 10.1186/s13045-019-0754-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bourneville D. Sclérose tubéreuse des circonvolutions cérébrales: idiotie et épilepsie hémiplégique. Arch Neurol. 1880;1:81–91. [Google Scholar]
- 21.Pringle J.J. A case of congenital adenoma sebaceum. Br J Dermatol. 1890;2:1–14. [Google Scholar]
- 22.van Slegtenhorst M., de Hoogt R., Hermans C., Nellist M., Janssen B., Verhoef S., 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]
- 23.European Chromosome 16 Tuberous Sclerosis Consortium Identification and characterization of the tuberous sclerosis gene on chromosome 16. Cell. 1993;75:1305–1315. doi: 10.1016/0092-8674(93)90618-z. [DOI] [PubMed] [Google Scholar]
- 24.Northrup H., Krueger D.A., International Tuberous Sclerosis Complex Consensus Group Tuberous sclerosis complex diagnostic criteria update: Recommendations of the 2012 International Tuberous Sclerosis Complex Consensus Conference. Pediatr Neurol. 2013;49:243–254. doi: 10.1016/j.pediatrneurol.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.O’Callaghan F.J., Shiell A.W., Osborne J.P., Martyn C.N. Prevalence of tuberous sclerosis estimated by capture-recapture analysis. Lancet. 1998;351:1490. doi: 10.1016/S0140-6736(05)78872-3. [DOI] [PubMed] [Google Scholar]
- 26.Hallett L., Foster T., Liu Z., Blieden M., Valentim J. Burden of disease and unmet needs in tuberous sclerosis complex with neurological manifestations: Systematic review. Curr Med Res Opin. 2011;27:1571–1583. doi: 10.1185/03007995.2011.586687. [DOI] [PubMed] [Google Scholar]
- 27.Krueger D.A., Northrup H., International Tuberous Sclerosis Complex Consensus Group Tuberous sclerosis complex surveillance and management: Recommendations of the 2012 International tuberous sclerosis Complex Consensus Conference. Pediatr Neurol. 2013;49:255–265. doi: 10.1016/j.pediatrneurol.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Curatolo P., Moavero R., de Vries P.J. Neurological and neuropsychiatric aspects of tuberous sclerosis complex. Lancet Neurol. 2015;14:733–745. doi: 10.1016/S1474-4422(15)00069-1. [DOI] [PubMed] [Google Scholar]
- 29.de Vries P.J., Hunt A., Bolton P.F. The psychopathologies of children and adolescents with tuberous sclerosis complex (TSC): A postal survey of UK families. Eur Child Adolesc Psychiatry. 2007;16:16–24. doi: 10.1007/s00787-006-0570-3. [DOI] [PubMed] [Google Scholar]
- 30.Loomes R., Hull L., Mandy W.P.L. What is the male-to-female ratio in autism spectrum disorder? A systematic review and meta-analysis. J Am Acad Child Adolesc Psychiatry. 2017;56:466–474. doi: 10.1016/j.jaac.2017.03.013. [DOI] [PubMed] [Google Scholar]
- 31.Jones A.C., Daniells C.E., Snell R.G., Tachataki M., Idziaszczyk S.A., Krawczak M., et al. Molecular genetic and phenotypic analysis reveals differences between TSC1 and TSC2 associated familial and sporadic tuberous sclerosis. Hum Mol Genet. 1997;6:2155–2161. doi: 10.1093/hmg/6.12.2155. [DOI] [PubMed] [Google Scholar]
- 32.Dabora S.L., Jozwiak S., Franz D.N., Roberts P.S., Nieto A., Chung J., 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]
- 33.Lewis J.C., Thomas H.V., Murphy K.C., Sampson J.R. Genotype and psychological phenotype in tuberous sclerosis. J Med Genet. 2004;41:203–207. doi: 10.1136/jmg.2003.012757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Numis A.L., Major P., Montenegro M.A., Muzykewicz D.A., Pulsifer M.B., Thiele E.A. 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]
- 35.Kobayashi T., Hirayama Y., Kobayashi E., Kubo Y., Hino O. A germline insertion in the tuberous sclerosis (Tsc2) gene gives rise to the Eker rat model of dominantly inherited cancer. Nat Genet. 1995;9:70–74. doi: 10.1038/ng0195-70. [DOI] [PubMed] [Google Scholar]
- 36.Kobayashi T., Minowa O., Kuno J., Mitani H., Hino O., Noda T. Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germ-line Tsc2 mutation in mice. Cancer Res. 1999;59:1206–1211. [PubMed] [Google Scholar]
- 37.Kobayashi T., Minowa O., Sugitani Y., Takai S., Mitani H., Kobayashi E., et al. A germ-line Tsc1 mutation causes tumor development and embryonic lethality that are similar, but not identical to, those caused by Tsc2 mutation in mice. Proc Natl Acad Sci U S A. 2001;98:8762–8767. doi: 10.1073/pnas.151033798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Goorden S.M., van Woerden G.M., van der Weerd L., Cheadle J.P., Elgersma Y. Cognitive deficits in Tsc1+/- mice in the absence of cerebral lesions and seizures. Ann Neurol. 2007;62:648–655. doi: 10.1002/ana.21317. [DOI] [PubMed] [Google Scholar]
- 39.Onda H., Lueck A., Marks P.W., Warren H.B., Kwiatkowski D.J. Tsc2+/- mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background. J Clin Invest. 1999;104:687–695. doi: 10.1172/JCI7319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ehninger D., Han S., Shilyansky C., Zhou Y., Li W., Kwiatkowski D.J., et al. Reversal of learning deficits in a Tsc2+/- mouse model of tuberous sclerosis. Nat Med. 2008;14:843–848. doi: 10.1038/nm1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sato A., Kasai S., Kobayashi T., Takamatsu Y., Hino O., Ikeda K., Mizuguchi M. Rapamycin reverses impaired social interaction in mouse models of tuberous sclerosis complex. Nat Commun. 2012;3:1292. doi: 10.1038/ncomms2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Waltereit R., Japs B., Schneider M., de Vries P.J., Bartsch D. Epilepsy and Tsc2 haploinsufficiency lead to autistic-like social deficit behaviors in rats. Behav Genet. 2011;41:364–372. doi: 10.1007/s10519-010-9399-0. [DOI] [PubMed] [Google Scholar]
- 43.Schneider M., de Vries P.J., Schönig K., Rößner V., Waltereit R. mTOR inhibitor reverses autistic-like social deficit behaviours in adult rats with both Tsc2 haploinsufficiency and developmental status epilepticus. Eur Arch Psychiatry Clin Neurosci. 2017;267:455–463. doi: 10.1007/s00406-016-0703-8. [DOI] [PubMed] [Google Scholar]
- 44.Tsai P.T., Hull C., Chu Y., Greene-Colozzi E., Sadowski A.R., Leech J.M., 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]
- 45.Reith R.M., McKenna J., Wu H., Hashmi S.S., Cho S.H., Dash P.K., Gambello M.J. 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]
- 46.Zeng L.H., Xu L., Gutmann D.H., Wong M. Rapamycin prevents epilepsy in a mouse model of tuberous sclerosis complex. Ann Neurol. 2008;63:444–453. doi: 10.1002/ana.21331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zeng L.H., Rensing N.R., Zhang B., Gutmann D.H., Gambello M.J., Wong M. Tsc2 gene inactivation causes a more severe epilepsy phenotype than Tsc1 inactivation in a mouse model of tuberous sclerosis complex. Hum Mol Genet. 2011;20:445–454. doi: 10.1093/hmg/ddq491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Marsh D.J., Kum J.B., Lunetta K.L., Bennett M.J., Gorlin R.J., Ahmed S.F., et al. PTEN mutation spectrum and genotype-phenotype correlations in Bannayan-Riley-Ruvalcaba syndrome suggest a single entity with Cowden syndrome. Hum Mol Genet. 1999;8:1461–1472. doi: 10.1093/hmg/8.8.1461. [DOI] [PubMed] [Google Scholar]
- 49.Zhou X.P., Waite K.A., Pilarski R., Hampel H., Fernandez M.J., Bos C., et al. Germline PTEN promoter mutations and deletions in Cowden/Bannayan-Riley-Ruvalcaba syndrome result in aberrant PTEN protein and dysregulation of the phosphoinositol-3-kinase/Akt pathway. Am J Hum Genet. 2003;73:404–411. doi: 10.1086/377109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhou X.P., Marsh D.J., Morrison C.D., Chaudhury A.R., Maxwell M., Reifenberger G., Eng C. Germline inactivation of PTEN and dysregulation of the phosphoinositol-3-kinase/Akt pathway cause human Lhermitte-Duclos disease in adults. Am J Hum Genet. 2003;73:1191–1198. doi: 10.1086/379382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Butler M.G., Dasouki M.J., Zhou X.P., Talebizadeh Z., Brown M., Takahashi T.N., et al. Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet. 2005;42:318–321. doi: 10.1136/jmg.2004.024646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.McBride K.L., Varga E.A., Pastore M.T., Prior T.W., Manickam K., Atkin J.F., Herman G.E. Confirmation study of PTEN mutations among individuals with autism or developmental delays/mental retardation and macrocephaly. Autism Res. 2010;3:137–141. doi: 10.1002/aur.132. [DOI] [PubMed] [Google Scholar]
- 53.Blumenthal G.M., Dennis P.A. PTEN hamartoma tumor syndromes. Eur J Hum Genet. 2008;16:1289–1300. doi: 10.1038/ejhg.2008.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Varga E.A., Pastore M., Prior T., Herman G.E., McBride K.L. The prevalence of PTEN mutations in a clinical pediatric cohort with autism spectrum disorders, developmental delay, and macrocephaly. Genet Med. 2009;11:111–117. doi: 10.1097/GIM.0b013e31818fd762. [DOI] [PubMed] [Google Scholar]
- 55.Zhou J., Parada L.F. PTEN signaling in autism spectrum disorders. Curr Opin Neurobiol. 2012;22:873–879. doi: 10.1016/j.conb.2012.05.004. [DOI] [PubMed] [Google Scholar]
- 56.Busch R.M., Srivastava S., Hogue O., Frazier T.W., Klaas P., Hardan A., et al. Neurobehavioral phenotype of autism spectrum disorder associated with germline heterozygous mutations in PTEN. Transl Psychiatry. 2019;9:253. doi: 10.1038/s41398-019-0588-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Georgescu M.M. PTEN tumor suppressor network in PI3K-Akt pathway control. Genes Cancer. 2010;1:1170–1177. doi: 10.1177/1947601911407325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Clipperton-Allen A.E., Page D.T. Pten haploinsufficient mice show broad brain overgrowth but selective impairments in autism-relevant behavioral tests. Hum Mol Genet. 2014;23:3490–3505. doi: 10.1093/hmg/ddu057. [DOI] [PubMed] [Google Scholar]
- 59.Séjourné J., Llaneza D., Kuti O.J., Page D.T. Social behavioral deficits coincide with the onset of seizure susceptibility in mice lacking serotonin receptor 2c. PLoS One. 2015;10 doi: 10.1371/journal.pone.0136494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Huang W.C., Chen Y., Page D.T. Hyperconnectivity of prefrontal cortex to amygdala projections in a mouse model of macrocephaly/autism syndrome. Nat Commun. 2016;7:13421. doi: 10.1038/ncomms13421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kwon C.H., Luikart B.W., Powell C.M., Zhou J., Matheny S.A., Zhang W., et al. Pten regulates neuronal arborization and social interaction in mice. Neuron. 2006;50:377–388. doi: 10.1016/j.neuron.2006.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhou J., Blundell J., Ogawa S., Kwon C.H., Zhang W., Sinton C., 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]
- 63.Chen C.J., Sgritta M., Mays J., Zhou H., Lucero R., Park J., et al. Therapeutic inhibition of mTORC2 rescues the behavioral and neurophysiological abnormalities associated with Pten-deficiency. Nat Med. 2019;25:1684–1690. doi: 10.1038/s41591-019-0608-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kidd S.A., Lachiewicz A., Barbouth D., Blitz R.K., Delahunty C., McBrien D., et al. Fragile X syndrome: A review of associated medical problems. Pediatrics. 2014;134:995–1005. doi: 10.1542/peds.2013-4301. [DOI] [PubMed] [Google Scholar]
- 65.Verkerk A.J., Pieretti M., Sutcliffe J.S., Fu Y.H., Kuhl D.P., Pizzuti A., et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell. 1991;65:905–914. doi: 10.1016/0092-8674(91)90397-h. [DOI] [PubMed] [Google Scholar]
- 66.Fu Y.H., Kuhl D.P., Pizzuti A., Pieretti M., Sutcliffe J.S., Richards S., et al. Variation of the CGG repeat at the fragile X site results in genetic instability: Resolution of the Sherman paradox. Cell. 1991;67:1047–1058. doi: 10.1016/0092-8674(91)90283-5. [DOI] [PubMed] [Google Scholar]
- 67.Naumann A., Hochstein N., Weber S., Fanning E., Doerfler W. A distinct DNA-methylation boundary in the 5′-upstream sequence of the FMR1 promoter binds nuclear proteins and is lost in fragile X syndrome. Am J Hum Genet. 2009;85:606–616. doi: 10.1016/j.ajhg.2009.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Brouwer J.R., Willemsen R., Oostra B.A. The FMR1 gene and fragile X-associated tremor/ataxia syndrome. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:782–798. doi: 10.1002/ajmg.b.30910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hunter J., Rivero-Arias O., Angelov A., Kim E., Fotheringham I., Leal J. Epidemiology of fragile X syndrome: A systematic review and meta-analysis. Am J Med Genet A. 2014;164A:1648–1658. doi: 10.1002/ajmg.a.36511. [DOI] [PubMed] [Google Scholar]
- 70.Richter J.D., Bassell G.J., Klann E. Dysregulation and restoration of translational homeostasis in fragile X syndrome. Nat Rev Neurosci. 2015;16:595–605. doi: 10.1038/nrn4001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hagerman R.J., Berry-Kravis E., Hazlett H.C., Bailey D.B., Jr., Moine H., Kooy R.F., et al. Fragile X syndrome. Nat Rev Dis Primers. 2017;3:17065. doi: 10.1038/nrdp.2017.65. [DOI] [PubMed] [Google Scholar]
- 72.Napoli I., Mercaldo V., Boyl P.P., Eleuteri B., Zalfa F., De Rubeis S., et al. The fragile X syndrome protein represses activity-dependent translation through CYFIP1, a new 4E-BP. Cell. 2008;134:1042–1054. doi: 10.1016/j.cell.2008.07.031. [DOI] [PubMed] [Google Scholar]
- 73.Gantois I., Popic J., Khoutorsky A., Sonenberg N. Metformin for treatment of fragile X syndrome and other neurological disorders. Annu Rev Med. 2019;70:167–181. doi: 10.1146/annurev-med-081117-041238. [DOI] [PubMed] [Google Scholar]
- 74.Bhattacharya A., Kaphzan H., Alvarez-Dieppa A.C., Murphy J.P., Pierre P., Klann E. Genetic removal of P70 S6 kinase 1 corrects molecular, synaptic, and behavioral phenotypes in fragile X syndrome mice. Neuron. 2012;76:325–337. doi: 10.1016/j.neuron.2012.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hamilton S.M., Green J.R., Veeraragavan S., Yuva L., McCoy A., Wu Y., et al. Fmr1 and Nlgn3 knockout rats: Novel tools for investigating autism spectrum disorders. Behav Neurosci. 2014;128:103–109. doi: 10.1037/a0035988. [DOI] [PubMed] [Google Scholar]
- 76.Dölen G., Osterweil E., Rao B.S., Smith G.B., Auerbach B.D., Chattarji S., Bear M.F. Correction of fragile X syndrome in mice. Neuron. 2007;56:955–962. doi: 10.1016/j.neuron.2007.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Thomas A.M., Bui N., Perkins J.R., Yuva-Paylor L.A., Paylor R. Group I metabotropic glutamate receptor antagonists alter select behaviors in a mouse model for fragile X syndrome. Psychopharmacology. 2012;219:47–58. doi: 10.1007/s00213-011-2375-4. [DOI] [PubMed] [Google Scholar]
- 78.Hou L., Klann E. Activation of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway is required for metabotropic glutamate receptor-dependent long-term depression. J Neurosci. 2004;24:6352–6361. doi: 10.1523/JNEUROSCI.0995-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Casingal C.R., Kikkawa T., Inada H., Sasaki Y., Osumi N. Identification of FMRP target mRNAs in the developmental brain: FMRP might coordinate Ras/MAPK, Wnt/β-catenin, and mTOR signaling during corticogenesis. Mol Brain. 2020;13:167. doi: 10.1186/s13041-020-00706-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Gross C., Nakamoto M., Yao X., Chan C.B., Yim S.Y., Ye K., et al. Excess phosphoinositide 3-kinase subunit synthesis and activity as a novel therapeutic target in fragile X syndrome. J Neurosci. 2010;30:10624–10638. doi: 10.1523/JNEUROSCI.0402-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Osterweil E.K., Krueger D.D., Reinhold K., Bear M.F. Hypersensitivity to mGluR5 and ERK1/2 leads to excessive protein synthesis in the hippocampus of a mouse model of fragile X syndrome. J Neurosci. 2010;30:15616–15627. doi: 10.1523/JNEUROSCI.3888-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sharma A., Hoeffer C.A., Takayasu Y., Miyawaki T., McBride S.M., Klann E., Zukin R.S. Dysregulation of mTOR signaling in fragile X syndrome. J Neurosci. 2010;30:694–702. doi: 10.1523/JNEUROSCI.3696-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ronesi J.A., Collins K.A., Hays S.A., Tsai N.P., Guo W., Birnbaum S.G., et al. Disrupted Homer scaffolds mediate abnormal mGluR5 function in a mouse model of fragile X syndrome. Nat Neurosci. 2012;15:431–440. doi: 10.1038/nn.3033. S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Saré R.M., Song A., Loutaev I., Cook A., Maita I., Lemons A., et al. Negative effects of chronic rapamycin treatment on behavior in a mouse model of fragile X syndrome. Front Mol Neurosci. 2017;10:452. doi: 10.3389/fnmol.2017.00452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Jafry M., Sidbury R. RASopathies. Clin Dermatol. 2020;38:455–461. doi: 10.1016/j.clindermatol.2020.03.010. [DOI] [PubMed] [Google Scholar]
- 86.Walker J.A., Upadhyaya M. Emerging therapeutic targets for neurofibromatosis type 1. Expert Opin Ther Targets. 2018;22:419–437. doi: 10.1080/14728222.2018.1465931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Garg S., Lehtonen A., Huson S.M., Emsley R., Trump D., Evans D.G., Green J. Autism and other psychiatric comorbidity in neurofibromatosis type 1: Evidence from a population-based study. Dev Med Child Neurol. 2013;55:139–145. doi: 10.1111/dmcn.12043. [DOI] [PubMed] [Google Scholar]
- 88.Walsh K.S., Vélez J.I., Kardel P.G., Imas D.M., Muenke M., Packer R.J., et al. Symptomatology of autism spectrum disorder in a population with neurofibromatosis type 1. Dev Med Child Neurol. 2013;55:131–138. doi: 10.1111/dmcn.12038. [DOI] [PubMed] [Google Scholar]
- 89.Martin G.A., Viskochil D., Bollag G., McCabe P.C., Crosier W.J., Haubruck H., et al. The Gap-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990;63:843–849. doi: 10.1016/0092-8674(90)90150-d. [DOI] [PubMed] [Google Scholar]
- 90.Silva A.J., Frankland P.W., Marowitz Z., Friedman E., Laszlo G.S., Cioffi D., et al. A mouse model for the learning and memory deficits associated with neurofibromatosis type I. Nat Genet. 1997;15:281–284. doi: 10.1038/ng0397-281. [DOI] [PubMed] [Google Scholar]
- 91.Molosh A.I., Johnson P.L., Spence J.P., Arendt D., Federici L.M., Bernabe C., et al. Social learning and amygdala disruptions in Nf1 mice are rescued by blocking p21-activated kinase. Nat Neurosci. 2014;17:1583–1590. doi: 10.1038/nn.3822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Petrella L.I., Cai Y., Sereno J.V., Gonçalves S.I., Silva A.J., Castelo-Branco M. Brain and behaviour phenotyping of a mouse model of neurofibromatosis type-1: An MRI/DTI study on social cognition. Genes Brain Behav. 2016;15:637–646. doi: 10.1111/gbb.12305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Maloney S.E., Chandler K.C., Anastasaki C., Rieger M.A., Gutmann D.H., Dougherty J.D. Characterization of early communicative behavior in mouse models of neurofibromatosis type 1. Autism Res. 2018;11:44–58. doi: 10.1002/aur.1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tomson T., Battino D., Perucca E. Valproic acid after five decades of use in epilepsy: Time to reconsider the indications of a time-honoured drug. Lancet Neurol. 2016;15:210–218. doi: 10.1016/S1474-4422(15)00314-2. [DOI] [PubMed] [Google Scholar]
- 95.Linde M., Mulleners W.M., Chronicle E.P., McCrory D.C. Valproate (valproic acid or sodium valproate or a combination of the two) for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev. 2013;6:CD010611. doi: 10.1002/14651858.CD010611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.McIntyre R.S., Berk M., Brietzke E., Goldstein B.I., López-Jaramillo C., Kessing L.V., et al. Bipolar disorders. Lancet. 2020;396:1841–1856. doi: 10.1016/S0140-6736(20)31544-0. [DOI] [PubMed] [Google Scholar]
- 97.Baker G.A., Bromley R.L., Briggs M., Cheyne C.P., Cohen M.J., García-Fiñana M., et al. IQ at 6 years after in utero exposure to antiepileptic drugs: A controlled cohort study. Neurology. 2015;84:382–390. doi: 10.1212/WNL.0000000000001182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Jentink J., Loane M.A., Dolk H., Barisic I., Garne E., Morris J.K., et al. Valproic acid monotherapy in pregnancy and major congenital malformations. N Engl J Med. 2010;362:2185–2193. doi: 10.1056/NEJMoa0907328. [DOI] [PubMed] [Google Scholar]
- 99.Tomson T., Marson A., Boon P., Canevini M.P., Covanis A., Gaily E., et al. Valproate in the treatment of epilepsy in girls and women of childbearing potential. Epilepsia. 2015;56:1006–1019. doi: 10.1111/epi.13021. [DOI] [PubMed] [Google Scholar]
- 100.Nicolini C., Fahnestock M. The valproic acid-induced rodent model of autism. Exp Neurol. 2018;299:217–227. doi: 10.1016/j.expneurol.2017.04.017. [DOI] [PubMed] [Google Scholar]
- 101.Phiel C.J., Zhang F., Huang E.Y., Guenther M.G., Lazar M.A., Klein P.S. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734–36741. doi: 10.1074/jbc.M101287200. [DOI] [PubMed] [Google Scholar]
- 102.Kataoka S., Takuma K., Hara Y., Maeda Y., Ago Y., Matsuda T. Autism-like behaviours with transient histone hyperacetylation in mice treated prenatally with valproic acid. Int J Neuropsychopharmacol. 2013;16:91–103. doi: 10.1017/S1461145711001714. [DOI] [PubMed] [Google Scholar]
- 103.Gurpur P.B., Liu J., Burkin D.J., Kaufman S.J. Valproic acid activates the PI3K/Akt/mTOR pathway in muscle and ameliorates pathology in a mouse model of Duchenne muscular dystrophy. Am J Pathol. 2009;174:999–1008. doi: 10.2353/ajpath.2009.080537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Qin L., Dai X., Yin Y. Valproic acid exposure sequentially activates Wnt and mTOR pathways in rats. Mol Cell Neurosci. 2016;75:27–35. doi: 10.1016/j.mcn.2016.06.004. [DOI] [PubMed] [Google Scholar]
- 105.Kotajima-Murakami H., Kobayashi T., Kashii H., Sato A., Hagino Y., Tanaka M., et al. Effects of rapamycin on social interaction deficits and gene expression in mice exposed to valproic acid in utero. Mol Brain. 2019;12:3. doi: 10.1186/s13041-018-0423-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Lieberman O.J., Cartocci V., Pigulevskiy I., Molinari M., Carbonell J., Broseta M.B., et al. mTOR suppresses macroautophagy during striatal postnatal development and is hyperactive in mouse models of autism spectrum disorders. Front Cell Neurosci. 2020;14:70. doi: 10.3389/fncel.2020.00070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Qin M., Kang J., Burlin T.V., Jiang C., Smith C.B. Postadolescent changes in regional cerebral protein synthesis: An in vivo study in the FMR1 null mouse. J Neurosci. 2005;25:5087–5095. doi: 10.1523/JNEUROSCI.0093-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Auerbach B.D., Osterweil E.K., Bear M.F. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature. 2011;480:63–68. doi: 10.1038/nature10658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Saré R.M., Huang T., Burlin T., Loutaev I., Smith C.B. Decreased rates of cerebral protein synthesis measured in vivo in a mouse model of tuberous sclerosis complex: Unexpected consequences of reduced tuberin. J Neurochem. 2018;145:417–425. doi: 10.1111/jnc.14311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Gkogkas C.G., Khoutorsky A., Ran I., Rampakakis E., Nevarko T., Weatherill D.B., et al. Autism-related deficits via dysregulated eIF4E-dependent translational control. Nature. 2013;493:371–377. doi: 10.1038/nature11628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Aguilar-Valles A., Matta-Camacho E., Khoutorsky A., Gkogkas C., Nader K., Lacaille J.C., Sonenberg N. Inhibition of group I metabotropic glutamate receptors reverses autistic-like phenotypes caused by deficiency of the translation repressor eIF4E binding protein 2. J Neurosci. 2015;35:11125–11132. doi: 10.1523/JNEUROSCI.4615-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Santini E., Huynh T.N., MacAskill A.F., Carter A.G., Pierre P., Ruggero D., et al. Exaggerated translation causes synaptic and behavioural aberrations associated with autism. Nature. 2013;493:411–415. doi: 10.1038/nature11782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Santini E., Huynh T.N., Longo F., Koo S.Y., Mojica E., D’Andrea L., et al. Reducing eIF4E-eIF4G interactions restores the balance between protein synthesis and actin dynamics in fragile X syndrome model mice. Sci Signal. 2017;10 doi: 10.1126/scisignal.aan0665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Mizushima N., Levine B., Cuervo A.M., Klionsky D.J. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075. doi: 10.1038/nature06639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hosokawa N., Hara T., Kaizuka T., Kishi C., Takamura A., Miura Y., et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20:1981–1991. doi: 10.1091/mbc.E08-12-1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yu L., Chen Y., Tooze S.A. Autophagy pathway: Cellular and molecular mechanisms. Autophagy. 2018;14:207–215. doi: 10.1080/15548627.2017.1378838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Tang G., Gudsnuk K., Kuo S.H., Cotrina M.L., Rosoklija G., Sosunov A., et al. Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits. Neuron. 2014;83:1131–1143. doi: 10.1016/j.neuron.2014.07.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Di Nardo A., Wertz M.H., Kwiatkowski E., Tsai P.T., Leech J.D., Greene-Colozzi 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]
- 119.Kim Y.C., Guan K.L. mTOR: A pharmacologic target for autophagy regulation. J Clin Invest. 2015;125:25–32. doi: 10.1172/JCI73939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Potter W.B., Basu T., O’Riordan K.J., Kirchner A., Rutecki P., Burger C., Roopra A. Reduced juvenile long-term depression in tuberous sclerosis complex is mitigated in adults by compensatory recruitment of mGluR5 and Erk signaling. PLoS Biol. 2013;11 doi: 10.1371/journal.pbio.1001627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kelly E., Schaeffer S.M., Dhamne S.C., Lipton J.O., Lindemann L., Honer M., et al. mGluR5 modulation of behavioral and epileptic phenotypes in a mouse model of tuberous sclerosis complex. Neuropsychopharmacology. 2018;43:1457–1465. doi: 10.1038/npp.2017.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Antion M.D., Hou L., Wong H., Hoeffer C.A., Klann E. mGluR-dependent long-term depression is associated with increased phosphorylation of S6 and synthesis of elongation factor 1A but remains expressed in S6K-deficient mice. Mol Cell Biol. 2008;28:2996–3007. doi: 10.1128/MCB.00201-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Pacey L.K., Heximer S.P., Hampson D.R. Increased GABA(B) receptor-mediated signaling reduces the susceptibility of fragile X knockout mice to audiogenic seizures. Mol Pharmacol. 2009;76:18–24. doi: 10.1124/mol.109.056127. [DOI] [PubMed] [Google Scholar]
- 124.Berry-Kravis E.M., Lindemann L., Jønch A.E., Apostol G., Bear M.F., Carpenter R.L., et al. Drug development for neurodevelopmental disorders: Lessons learned from fragile X syndrome. Nat Rev Drug Discov. 2018;17:280–299. doi: 10.1038/nrd.2017.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Berry-Kravis E., Des Portes V., Hagerman R., Jacquemont S., Charles P., Visootsak J., et al. Mavoglurant in fragile X syndrome: Results of two randomized, double-blind, placebo-controlled trials. Sci Transl Med. 2016;8:321ra5. doi: 10.1126/scitranslmed.aab4109. [DOI] [PubMed] [Google Scholar]
- 126.Berry-Kravis E., Hagerman R., Visootsak J., Budimirovic D., Kaufmann W.E., Cherubini M., et al. Arbaclofen in fragile X syndrome: Results of phase 3 trials. J Neurodev Disord. 2017;9:3. doi: 10.1186/s11689-016-9181-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hessl D., Harvey D., Sansone S., Crestodina C., Chin J., Joshi R., et al. Effects of mavoglurant on visual attention and pupil reactivity while viewing photographs of faces in fragile X syndrome. PLoS One. 2019;14 doi: 10.1371/journal.pone.0209984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Miura S., Matsuo Y., Saku K. Simvastatin suppresses coronary artery endothelial tube formation by disrupting Ras/Raf/ERK signaling. Atherosclerosis. 2004;175:235–243. doi: 10.1016/j.atherosclerosis.2004.04.017. [DOI] [PubMed] [Google Scholar]
- 129.Li W., Cui Y., Kushner S.A., Brown R.A., Jentsch J.D., Frankland P.W., et al. The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Curr Biol. 2005;15:1961–1967. doi: 10.1016/j.cub.2005.09.043. [DOI] [PubMed] [Google Scholar]
- 130.Mainberger F., Jung N.H., Zenker M., Wahlländer U., Freudenberg L., Langer S., et al. Lovastatin improves impaired synaptic plasticity and phasic alertness in patients with neurofibromatosis type 1. BMC Neurol. 2013;13:131. doi: 10.1186/1471-2377-13-131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bearden C.E., Hellemann G.S., Rosser T., Montojo C., Jonas R., Enrique N., et al. A randomized placebo-controlled lovastatin trial for neurobehavioral function in neurofibromatosis I. Ann Clin Transl Neurol. 2016;3:266–279. doi: 10.1002/acn3.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ullrich N.J., Payne J.M., Walsh K.S., Cutter G., Packer R., North K., et al. Visual spatial learning outcomes for clinical trials in neurofibromatosis type 1. Ann Clin Transl Neurol. 2020;7:245–249. doi: 10.1002/acn3.50976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Krab L.C., de Goede-Bolder A., Aarsen F.K., Pluijm S.M., Bouman M.J., van der Geest J.N., et al. Effect of simvastatin on cognitive functioning in children with neurofibromatosis type 1: A randomized controlled trial. JAMA. 2008;300:287–294. doi: 10.1001/jama.300.3.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.van der Vaart T., Plasschaert E., Rietman A.B., Renard M., Oostenbrink R., Vogels A., et al. Simvastatin for cognitive deficits and behavioural problems in patients with neurofibromatosis type 1 (NF1-SIMCODA): A randomised, placebo-controlled trial. Lancet Neurol. 2013;12:1076–1083. doi: 10.1016/S1474-4422(13)70227-8. [DOI] [PubMed] [Google Scholar]
- 135.Stivaros S., Garg S., Tziraki M., Cai Y., Thomas O., Mellor J., et al. Randomised controlled trial of simvastatin treatment for autism in young children with neurofibromatosis type 1 (SANTA) Mol Autism. 2018;9:12. doi: 10.1186/s13229-018-0190-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Hwang S.K., Lee J.H., Yang J.E., Lim C.S., Lee J.A., Lee Y.S., et al. Everolimus improves neuropsychiatric symptoms in a patient with tuberous sclerosis carrying a novel TSC2 mutation. Mol Brain. 2016;9:56. doi: 10.1186/s13041-016-0222-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kilincaslan A., Kok B.E., Tekturk P., Yalcinkaya C., Ozkara C., Yapici Z. Beneficial effects of everolimus on autism and attention-deficit/hyperactivity disorder symptoms in a group of patients with tuberous sclerosis complex. J Child Adolesc Psychopharmacol. 2017;27:383–388. doi: 10.1089/cap.2016.0100. [DOI] [PubMed] [Google Scholar]
- 138.Davies D.M., de Vries P.J., Johnson S.R., McCartney D.L., Cox J.A., Serra A.L., et al. Sirolimus therapy for angiomyolipoma in tuberous sclerosis and sporadic lymphangioleiomyomatosis: A phase 2 trial. Clin Cancer Res. 2011;17:4071–4081. doi: 10.1158/1078-0432.CCR-11-0445. [DOI] [PubMed] [Google Scholar]
- 139.Krueger D.A., Wilfong A.A., Holland-Bouley K., Anderson A.E., Agricola K., Tudor C., et al. Everolimus treatment of refractory epilepsy in tuberous sclerosis complex. Ann Neurol. 2013;74:679–687. doi: 10.1002/ana.23960. [DOI] [PubMed] [Google Scholar]
- 140.Mizuguchi M., Ikeda H., Kagitani-Shimono K., Yoshinaga H., Suzuki Y., Aoki M., et al. Everolimus for epilepsy and autism spectrum disorder in tuberous sclerosis complex: EXIST-3 substudy in Japan. Brain Dev. 2019;41:1–10. doi: 10.1016/j.braindev.2018.07.003. [DOI] [PubMed] [Google Scholar]
- 141.Krueger D.A., Care M.M., Holland K., Agricola K., Tudor C., Mangeshkar P., et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. 2010;363:1801–1811. doi: 10.1056/NEJMoa1001671. [DOI] [PubMed] [Google Scholar]
- 142.Overwater I.E., Rietman A.B., Mous S.E., Bindels-de Heus K., Rizopoulos D., Ten Hoopen L.W., et al. A randomized controlled trial with everolimus for IQ and autism in tuberous sclerosis complex. Neurology. 2019;93:e200–e209. doi: 10.1212/WNL.0000000000007749. [DOI] [PubMed] [Google Scholar]
- 143.Hardan A.Y., Jo B., Frazier T.W., Klaas P., Busch R.M., Dies K.A., et al. A randomized double-blind controlled trial of everolimus in individuals with PTEN mutations: Study design and statistical considerations. Contemp Clin Trials Commun. 2021;21:100733. doi: 10.1016/j.conctc.2021.100733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Page D.T., Kuti O.J., Prestia C., Sur M. Haploinsufficiency for Pten and serotonin transporter cooperatively influences brain size and social behavior. Proc Natl Acad Sci U S A. 2009;106:1989–1994. doi: 10.1073/pnas.0804428106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cupolillo D., Hoxha E., Faralli A., De Luca A., Rossi F., Tempia F., Carulli D. Autistic-like traits and cerebellar dysfunction in Purkinje cell PTEN knock-out mice. Neuropsychopharmacology. 2016;41:1457–1466. doi: 10.1038/npp.2015.339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Napoli E., Ross-Inta C., Wong S., Hung C., Fujisawa Y., Sakaguchi D., et al. Mitochondrial dysfunction in Pten haplo-insufficient mice with social deficits and repetitive behavior: Interplay between Pten and p53. PLoS One. 2021;7 doi: 10.1371/journal.pone.0042504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Lugo J.N., Smith G.D., Arbuckle E.P., White J., Holley A.J., Floruta C.M., et al. Deletion of PTEN produces autism-like behavioral deficits and alterations in synaptic proteins. Front Mol Neurosci. 2014;7:27. doi: 10.3389/fnmol.2014.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Amiri A., Cho W., Zhou J., Birnbaum S.G., Sinton C.M., McKay R.M., et al. Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnormalities and alters neurogenesis. J Neurosci. 2012;32:5880–5890. doi: 10.1523/JNEUROSCI.5462-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Clipperton-Allen A.E., Chen Y., Page D.T. Autism-relevant behaviors are minimally impacted by conditional deletion of Pten in oxytocinergic neurons. Autism Res. 2016;9:1248–1262. doi: 10.1002/aur.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Vogt D., Cho K.K.A., Lee A.T., Sohal V.S., Rubenstein J.L.R. The parvalbumin/somatostatin ratio is increased in Pten mutant mice and by human PTEN ASD alleles. Cell Rep. 2015;11:944–956. doi: 10.1016/j.celrep.2015.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Sánchez-Puelles C., Calleja-Felipe M., Ouro A., Bougamra G., Arroyo A., Diez I., et al. PTEN activity defines an axis for plasticity at cortico-amygdala synapses and influences social behavior. Cereb Cortex. 2020;30:505–524. doi: 10.1093/cercor/bhz103. [DOI] [PubMed] [Google Scholar]
- 152.Kramvis I., Mansvelder H.D., Loos M., Meredith R. Hyperactivity, perseveration and increased responding during attentional rule acquisition in the Fragile X mouse model. Front Behav Neurosci. 2013;7:172. doi: 10.3389/fnbeh.2013.00172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gandhi R.M., Kogan C.S., Messier C. 2-Methyl-6-(phenylethynyl) pyridine (MPEP) reverses maze learning and PSD-95 deficits in Fmr1 knock-out mice. Front Cell Neurosci. 2014;8:70. doi: 10.3389/fncel.2014.00070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Wang K., Li N., Xu M., Huang M., Huang F. Glyoxalase 1 inhibitor alleviates autism-like phenotype in a prenatal valproic acid-induced mouse model. ACS Chem Neurosci. 2020;11:3786–3792. doi: 10.1021/acschemneuro.0c00482. [DOI] [PubMed] [Google Scholar]
- 155.Yang J.Q., Yang C.H., Yin B.Q. Combined the GABA-A and GABA-B receptor agonists attenuates autistic behaviors in a prenatal valproic acid-induced mouse model of autism. Behav Brain Res. 2021;403:113094. doi: 10.1016/j.bbr.2020.113094. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

