Skip to main content
Neural Plasticity logoLink to Neural Plasticity
. 2012 Apr 30;2012:486402. doi: 10.1155/2012/486402

Plasticity and mTOR: Towards Restoration of Impaired Synaptic Plasticity in mTOR-Related Neurogenetic Disorders

Tanjala T Gipson 1,2,3,*, Michael V Johnston 1,2,3,4,5,6
PMCID: PMC3350854  PMID: 22619737

Abstract

Objective. To review the recent literature on the clinical features, genetic mutations, neurobiology associated with dysregulation of mTOR (mammalian target of rapamycin), and clinical trials for tuberous sclerosis complex (TSC), neurofibromatosis-1 (NF1) and fragile X syndrome (FXS), and phosphatase and tensin homolog hamartoma syndromes (PTHS), which are neurogenetic disorders associated with abnormalities in synaptic plasticity and mTOR signaling. Methods. Pubmed and Clinicaltrials.gov were searched using specific search strategies. Results/Conclusions. Although traditionally thought of as irreversible disorders, significant scientific progress has been made in both humans and preclinical models to understand how pathologic features of these neurogenetic disorders can be reduced or reversed. This paper revealed significant similarities among the conditions. Not only do they share features of impaired synaptic plasticity and dysregulation of mTOR, but they also share clinical features—autism, intellectual disability, cutaneous lesions, and tumors. Although scientific advances towards discovery of effective treatment in some disorders have outpaced others, progress in understanding the signaling pathways that connect the entire group indicates that the lesser known disorders will become treatable as well.

1. Introduction

Brain plasticity, the developing brain's ability to change in response to either positive experiences or negative experiences, is a critical component of pediatric neurology. The major types of plasticity in the developing brain include adaptive plasticity—occurs in response to learning or recovering from injury or disability; impaired plasticity—results from brain injury due to an acquired or neurogenetic disorder; maladaptive plasticity—a plastic response leading to a new disorder; plasticity as the brain's Achilles' heel—a mechanism, such as selective vulnerability of neurons, which creates risk for injury [1]. Basic cellular mechanisms of plasticity include overproduction of neurons followed by reduction via apoptosis [2]; continued production of new cells from stem cells in the hippocampus and lateral ventricle throughout life [3]; activity-dependent synaptic plasticity through receptor trafficking; activity-dependent production of growth factors; overproduction of synapses and axodendritic connections followed by pruning, activity-dependent stabilization of dendrites and axons; regulation of DNA expression by epigenetic regulation [1]. Although there are many disorders associated with impaired plasticity, this paper will highlight the clinical features, neurobiology associated with dysregulation of mTOR, preclinical studies, and clinical trials in tuberous sclerosis complex (TSC), neurofibromatosis-1 (NF1), and fragile X syndrome (FXS), as well as phosphatase and tensin homolog hamartoma syndromes (PTHS), neurogenetic disorders linked by abnormalities in synaptic plasticity and mTOR (mammalian target of rapamycin) signaling.

2. Methods

Pubmed was searched using the following search strategies: mTOR and/or neurology; mTOR and/or plasticity; mTOR and/or TSC; mTOR and/or NF1; mTOR and/or FXS; mTOR and/or PTHS; plasticity and/or neurology; plasticity and/or TSC; plasticity and/or NF1; plasticity and/or FXS; plasticity and/or PTHS. Clinicaltrials.gov was searched by disorder without language or country of origin restrictions for active studies through 11/30/11.

2.1. Tuberous Sclerosis Complex (TSC)

2.1.1. Clinical Features

Tuberous sclerosis complex (TSC) has an incidence of 1/6000 and may be defined clinically by the presence or absence of major and minor features associated with the disorder and genetically by spontaneous or inherited mutations in TSC1 or TSC2. Major neurologic features include brain lesions-subependymal nodules, subependymal giant cell astrocytomas, and cortical tubers, intractable epilepsy in 60–90% [46], autism in up to 61% [7, 8], intellectual disability in 45% [9], and self-injury in 10% [10]. TSC has also been associated with pulmonary, cardiac, and cutaneous lesions (Table 1).

Table 1.

Diagnostic criteria.

(a) TSC. Definite TSC: two major or one major and two minor features; probable; TSC: one major and one minor feature; possible TSC: one major or two or more minor features

Major features Minor features
Cortical tubers Dental enamel pits
Subependymal nodules Hamartomatous rectal polyps
Subependymal giant cell astrocytoma Bone cysts
Hypomelanotic macules (3 or more) Cerebral white matter radial migration lines
Shagreen patch Gingival fibromas
Facial angiofibromas or forehead plaque Nonrenal hamartoma
Multiple renal nodular hamartomas Retinal achromatic patches
Nontraumatic ungual or periungual fibromas “Confetti” skin lesions
Cardiac rhabdomyoma Multiple renal cysts
Pulmonary lymphangiomyomatosis and/or renal angiomyolipomas

(b) NF1. Presence of two or more clinical features

Family history of NF1 Neurofibromas or plexiform neurofibromas
Six or more cafe-au-lait spots Axillary or groin freckling
Lisch nodules Skeletal abnormalities—tibial dysplasia or shin bone thinning
Optic glioma

(c) FXS

Full mutation >200 CGG repeats
Premutation 50–230 CGG repeats

(d) PTHS (Only Cowden syndrome has diagnostic criteria). Cowden syndrome. Operational diagnosis: mucocutaneous lesion alone if: 6 or more facial papules, 3 or more trichilemmoma; cutaneous facial papules and oral mucosal papillomatosis; oral mucosal papillomatosis and acral keratosis, or 6 or more palmoplantar keratosis; or two or major criteria, including macrocephaly or adult Lhermitte-Duclos disease; or one major or three minor criteria; or four minor criteria

Pathognomic criteria Major criteria Minor criteria
Adult Lhermitte-Duclos Breast cancer Intellectual disability
Mucocutaneous lesions Thyroid cancer Other thyroid lesions
Macrocephaly GI hamartomas
Endometrial cancer Fibrocystic breast disease
Lipomas; fibromas
Genitourinary tumors or malformations

EIF4E (No diagnostic criteria).

2.1.2. Neurobiology of mTOR Dysregulation

Overexpression of the serine/threonine protein kinase mammalian target of rapamycin (mTOR) results from disruption of either TSC1 or TSC2. Typically, TSC1 and TSC2 form a complex, which inhibits Rheb (ras homologue expressed in brain), an activator of mTOR. The consequences of mTOR overexpression include abnormally rapid cell growth and hyperactivation of mRNA translation, which may lead to impaired synaptic plasticity in TSC [11] (Figure 1).

Figure 1.

Figure 1

Pathways associated with neurogenetic conditions linked by mTOR and impaired synaptic plasticity. AKT-v—akt murine thymoma viral oncogene homolog 1; AMPK—adenosine monophosphate kinase; ATP—adenosine triphosphate; EIF4E/4EBP1—eukaryotic translation initiation factor 4E-binding protein 1; ERK—extracellular signal-related kinase; FKB12-FK506—binding protein family; FMRP—fragile X mental retardation protein; GBL/LST8—(mTOR-associated protein, LST8 homolog); GDP—guanosine diphosphate; GTP—guanosine triphosphate; GDP—guanosine diphosphate; HIF1α—hypoxia inducible factor 1, alpha subunit IGF—insulin-like growth factor; IGFR—insulin-like growth factor receptor; IRS—insulin receptor substrate; LKB1—serine threonine kinase 11; mTORC1—mammalian target of rapamycin complex 1; mTORC2—mammalian target of rapamycin complex 2; MEK—dual specificity mitogen-activated protein kinase 1; NF-1—neurofibromatosis 1; P/+ Thr308—phosphorylation of threonine, position 308; P/+ Ser473—phosphorylation of serine, position 473; PDK1—pyruvate dehydrogenase kinase, isozyme 1; PDK 2—pyruvate dehydrogenase kinase, isozyme 2; PI3K—phosphoinositide-3-kinase; PIKE—phosphoinositide 3-kinase enhancer; PIP1—p21-activated protein kinase-interacting protein 1 (PAK1 interacting protein 1); PIP2—phosphatidylinositol 4,5-biphosphate; PIP3—phosphatidylinositol (3,4,5)-triphosphate; PTEN—phosphatase and tensin homolog; RAS—Ras p21 protein activator 1 or RAS GTPase activating protein; REDD1—regulated in development and DNA damage responses; RHEB—Ras homologue expressed in brain; RTK—receptor tyrosine kinase; S6K—ribosomal protein S6 kinase; TSC—tuberous sclerosis complex; VEGF—vascular endothelial growth factor; VHL—von Hippel Lindau.

2.1.3. Preclinical Models

Impaired synaptic plasticity as a consequence of a disruption in either TSC1 or TSC2 has been supported by results from preclinical studies. Abnormalities in long-term potentiation (LTP) and long-term depression (LTD) were found in the Tsc2+/ Eker rat, which carries a spontaneous germline mutation [12]. Abnormal late-phase LTP induction and hippocampal-dependent learning deficits was observed in Tsc2+/− adult mice and improved after rapamycin treatment [13].

Metabotropic glutamate receptor-mediated long-term depression (mGluR-LTD) was impaired in Tsc2+/ mice and related to decreased translation of proteins required for stabilization of LTD. Potentiation of mGluR5 activity led to restoration of normal LTD [14]. mGluR-LTD was also deficient in a neuronal model of Tsc1 [15].

Prolonged neuronal hyperexcitability, typically associated with epilepsy, has also been supported by recent studies as a possible mechanism of impaired synaptic plasticity in TSC [16]. Hyperexcitability in cortical tubers has been linked to abnormalities of glutamate receptor expression [17]. This hyperexcitability was maintained despite the absence of cortical tubers from brain sections of an individual with TSC and Tsc1synapsin conditionalknockout mice, a neuronal model of TSC in an earlier study [18, 19]. An astrocyte-specific model of TSC, Tsc1GFAP conditional knockout mice, was also characterized by abnormally elevated glutamate [20]. Astrocytic dysfunction in the uptake of extracellular potassium may explain the hyperexcitability in this model [21].

2.1.4. Clinical Trials

Guided by preclinical observations, investigators have completed studies to reduce the burden of neurologic disease in individuals with TSC. A clinical trial of everolimus for subependymal giant cell astrocytomas (SEGAs) achieved the primary outcome of reduction in the size of SEGAs, supporting similar results from a case series [2224]. Everolimus is now FDA-approved for reduction in the size of SEGAs that are nonsurgically resectable. Positive outcomes from these studies have led investigators to consider rapamycin for additional neurologic conditions, such as autism [25]. The ability of everolimus to improve cognition is currently under investigation (http://www.clinicaltrials.gov/; NCT01289912).

2.2. Neurofibromatosis 1 (NF1)

2.2.1. Clinical Features

Neurofibromatosis 1 (NF1), a disease caused by an inherited mutation in NF1, has an incidence of 1 in 3500 [26]. NF1 can be diagnosed by identification of the genetic mutation or the presence of two or more clinical features—family history of NF1; six or more cafe-au-lait spots; neurofibromas; plexiform neurofibromas; axillary or groin freckling; Lisch nodules (a hamartomatous nodule of melanocytes on the iris); skeletal abnormalities such as tibial dysplasia or thinning of the shin bone; or optic glioma. Associated conditions include cognitive impairments, pilocytic astrocytomas, and neuropathological abnormalities characterized by MRI hyperintensities, megalencephaly, and thalamic lesions. Cognitive impairment is the most common source of neurological impairment in children with NF1, affecting as many as 81% of children [27]. Neuropathological abnormalities associated with impaired cognition have been identified in some cases. Megalencephaly associated primarily with increased white matter volume was identified in individuals with NF1-associated ADHD [28]. Abnormalities in gray matter volume and enlargement of the corpus callosum have also been associated with cognitive impairment [29]. NF1 has also been characterized by the presence of MRI T2-hyperintensities (nonenhancing bright areas of unknown etiology), sometimes referred to as UBOs (unidentified bright objects). An early study employing sibling comparison found distribution of these lesions to be predictive of lower IQ [30]. Subsequent studies have also supported the role of these lesions in cognition [31, 32]. A longitudinal profile revealed changes in these lesions with childhood regression followed by recurrence in early adolescence [33].

2.2.2. Neurobiology of mTOR Dysregulation

Disinhibited RAS MAPK signaling underlies the molecular basis of disease, and mTOR hyperactivity has also been identified in preclinical models [34]. NF1 encodes neurofibromin, a GTP-ase activating protein, which normally leads to inactivation of Ras. Mutations in neurofibromin lead to overactivation of Ras activity, followed by enhanced activation of the Ras-MAPK signaling pathway as well as PI3K and ERK 1/2 which both inactivate the TSC1/TSC2 complex releasing inhibition of Rheb and allowing activation of mTOR. However, there may be pathways leading to dysregulation of mTOR in NF1 that differ from other conditions [34]. mTOR hyperactivity in Nf1 leads to increased astrocyte proliferation, an effect not shared by preclinical models of Pten, Tsc1, Tsc2, or overexpression of Rheb [35]. Phospho-histone-H3 rather than phosphor-S6 or Ki67 correlated with response to rapamycin in Nf1 mice [36]. Neurofibromin also interacts with caveolin-1 [37] and nucleophosmin [38].

2.2.3. Preclinical Models

Long-term potentiation was impaired by increased hippocampal inhibitory transmission in mice heterozygous for a germline mutation in Nf1 (Nf1+/−). However, restoration of LTP deficits and reversal of cognitive impairments was achieved with pharmacological inhibition of Ras using lovastatin, an HMG CoA reductase inhibitor [39] and BMS 191563, a farnesyltransferase inhibitor [40]. Farnesyltransferase inhibitors demonstrated inhibition of Rheb and subsequent inhibition of mTOR in Tsc1−/− and Tsc2−/− mouse embryonic fibroblasts [41]. Inhibition of ERK also led to restoration of early-phase and long-term LTP [42].

2.2.4. Clinical Trials

Simvastatin in children with NF1 improved object assembly, a secondary outcome in a randomized trial, but there was no difference in primary outcome [43]. Preliminary results of a subsequent of lovastatin in children with NF1 revealed improvement in verbal and nonverbal memory [44].

2.3. Fragile X Syndrome (FXS)

2.3.1. Clinical Features

Fragile X syndrome (FXS) is the leading cause of inherited intellectual disability and has a full mutation gene frequency of 1 in 2500 [45, 46]. Associated neurologic conditions include autism, anxiety, and ADHD [47, 48]. Definitive diagnosis relies on genetic confirmation and individuals may be classified as full mutation if there are greater than 200 CGG repeats within the promoter of the fragile X mental retardation-1 gene (FMR1) and premutation if there are 50 to 230 repeats [49].

2.3.2. Neurobiology of mTOR Dysregulation

These abnormal CGG repeats result in suppression of FMR1 gene transcription and subsequently reduced to absent fragile X mental retardation protein (FMRP) [50, 51]. Loss of FMRP releases inhibition of PIKE, which activates PI3K and leads to increased mTOR activity. The “mGluR theory” proposes that elevation of group I mGluRs (mGluR1 and mGluR5) glutamate receptors leading to reduced insertion of AMPA receptors into the postsynaptic membrane is one of the central mechanisms of impaired synaptic plasticity in FXS, and this has been supported in experimental models [52]. Increased mGluR5 activity and reduced insertion of AMPA receptors leads to long-term depression (LTD) due to reduced AMPA-mediated synaptic activity.

2.3.3. Preclinical Models

Using preclinical models, specific interactions among synaptic proteins and FMRP have been identified. Initially, abnormal synaptic translation of CaMKIIa, PSD-95, and GluR1/2 mRNAs was observed in the Fmr1 knockout mouse [53]. Subsequent studies revealed regulation of expression of PSD-95 by FMRP, miR125a, and mGluR.

Phosphorylation of FMRP induces the creation of an AGO2-miR125a complex, which inhibits PSD-95 mRNA. mGluR stimulation, however, causes dephosphorylation of FMRP, which leads to activation of translation of PSD-95. In Fmr1 KO mice, miR125a is reduced in addition to the reduction in FMRP [54]. In addition to hyperactivity of group 1 mGluR and mGluR-LTD, abnormally increased signaling of mTOR in hippocampus was discovered in Fmr1 KO mice, providing a link between mGluR elevation and abnormalities in synaptic plasticity leading to cognitive impairment. Loss of FMRP releases inhibition of PIKE, which activates PI3K and leads to increased mTOR activity as measured by four methods. Abnormally increased mTOR leads to an abnormal increase in cap-dependent translation of synaptic proteins and subsequent abnormalities in synaptic plasticity. Inhibition of PI3K activity resulted in normal levels of phosphorylated mTOR. Increased PTEN activity, mediated by dephosphorylation, was discovered in Fmr1 KO mice and may serve as a feedback inhibition to compensate for abnormally increased PI3K since PTEN dephosphorylates PI3K, which reduces phosphorylation and activation of Akt [55].

In a Drosophila model of FXS, treatment with mGluR antagonists during development resulted in reversal of neuropathology, abnormal courtship behavior, and impaired memory. Partial reversal of impaired memory and abnormal courtship behavior without change in neuropathology was seen in treated adults. Conversely, treatment led to impairment in wild-type control flies [56].

Reduction in genetic function of mGluR5, achieved by crossing Fmr1 mutant mice with heterozygous mGluR5 mutant mice, rescued many of the core phenotypic features in the Fmr1 KO mouse [57]. Treatment of Fmr1 KO mice with either an mGluR1 antagonist (JNJ) or an mGluR5 antagonist (MPEP) led to similar, but slightly different neurologic and behavioral improvements. Marble burying, a measure of repetitive behavior, was reduced without reduction in activity in Fmr1 KO and WT mice. MPEP eliminated audiogenic-induced seizures. Motor learning also improved with MPEP in Fmr1 KO mice. Prepulse inhibition, a measure of sensorimotor gating, known to be abnormally increased in Fmr1 KO mice was not affected by JNJ or MPEP [58]. Abnormalities in prepulse inhibition were linked to abnormalities in presynaptic short-term plasticity in mice models of schizophrenia [59].

Prolonged UP states, a marker of cortical hyperexcitability in Fmr1 KO mice was found to be due to a non-translation-related function of mGluR5, and treatment with MPEP reversed this phenomenon [60]. In addition to long-term postsynaptic plasticity, abnormalities in short-term presynaptic plasticity were also identified in Fmr1 KO mice and may also contribute to cognitive impairment [61]. Another approach utilized GABAA receptor agonist in Fmr1 KO mice, resulting in restoration of amygdala-based deficits in neuronal excitability, reduced prepulse inhibition, and alleviation of hyperactivity [62]. The behavioral effects of genetic reduction of mGluR1 and mGluR5 by 50% were observed in Fmr1 KO mice. Reduction in mGluR1 led to decreased activity, whereas reduction in mGluR5 led to decreased active social behavior and decreased thermal sensitivity. Neither genetic reduction resulted in changes in memory, motor responses, sensorimotor gating, audiogenic seizures, and responses related to anxiety and perseveration [58].

2.3.4. Clinical Trials

Human studies have led to the identification of the behavioral/cognitive profile of Fragile X as well an endophenotype of autism in Fragile X distinguished by social withdrawal [6365]. Comparison of patients with FXS with and without autism supported the previously identified endophenotype of social withdrawal in FXS-associated autism by the finding of decrease in the left temporal gyrification index, an indicator of cortico-cortical connectivity and organization [66]. Recent significant scientific discoveries have culminated in human clinical trials targeting different aspects of the neurobiological impairments in FXS. AFQ056, an mGluR5 antagonist, resulted in different responses dependent upon the methylation status of FMR1. Patients with full methylation of FMR1 and no detectable FMR1 mRNA in the blood responded positively to treatment with improvement in inappropriate speech, stereotypic behavior, and hyperactivity [67]. Additional trials focused on antagonizing mGluR5 include a trial of fenobam, which reduced anxiety, hyperarousal, improved accuracy in continuous performance tasks, and prepulse inhibition of startle [68]; acamprosate, which in three young adult patients, resulted in improvement in communication and global clinical improvement (CGI-I) [69]. Results are pending from an open label phase I study of STX107 (Seaside Therapeutics) and a phase II trial of RO4917523 (Hoffman-LaRoche) (clinicaltrials.gov). Other mechanisms that may lead to repair of the impaired plasticity associated with FXS have also been examined. Phospholipase C and glycogen synthase kinase-3, linked to Gp1 mGluR signaling, have been targeted using lithium, which resulted in improvement in cognition and adaptive skills [70]. Ampalex (CX516) is an ampakine (binds AMPA receptors) that increases hippocampal LTP by slowing receptor deactivation [71, 72]. Evaluation of ampalex in a placebo-controlled phase II trial for Fragile X-associated autism did not reveal differences in the primary outcome of memory or any of the secondary outcomes: overall functioning, attention/executive functioning, language, or behavior [73]. Minocycline, a broad spectrum antibiotic and analogue of tetracycline, has been found to have neuronal effects. In C57BL/6J mice, minocycline increased phosphorylation of GluR1 and subsequent insertion of AMPA receptors in vivo and vitro [74]. Study of minocycline in Fmr1 KO mice revealed behavioral improvement: reduction in anxiety and improved exploration as well as neuropathological improvement—dendritic spine maturation associated with inhibition of abnormally elevated matrix metalloproteinase-9 (MMP-9) in hippocampal neurons [75]. The observations in Fmr1 KO mice were supported in a Drosophila model of FXS where treatment with minocycline or genetic elimination of mmp1 reverses synaptic structural abnormalities [76]. Open-label treatment with minocycline in individuals with FXS led to significant improvement in irritability [77]. Cholinergic deficits in FXS, confirmed in individuals by 1H magnetic resonance spectroscopy, were targeted using donepezil in an open label study with noted improvement in continuous naming, attention difficulties, and total ABC score as well as irritability and hyperactivity [78]. Reduction in glutamate using riluzole in an open-label study corrected abnormal activation of ERK; however, improvement in the primary outcome-repetitive, compulsive behavior was not achieved [79]. A single-dose, placebo-controlled trial of oxytocin for social anxiety in FXS resulted in improvement in eye gaze towards the examiner in a social challenge [80]. Aripiprazole, an atypical antipsychotic that is a partial D2 and 5-HT1A agonist as well as a 5-HT2A antagonist, improved scores on CGI-I and ABC-irritability [81]. Baclofen, a GABAB receptor agonist, inhibited seizures in Fmr1 mice [82]. A phase II, randomized double-blind study of arbaclofen has been completed with results pending, and a phase III study of arbaclofen is now recruiting (http://www.clinicaltrials.gov/).

2.4. PTEN-Associated Conditions

2.4.1. Clinical Features

Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a phosphatase which limits cell growth by apoptosis and cell cycle arrest. Conditions linked by a genetic mutation in PTEN have been collectively termed phosphatase and tensin homologue hamartoma syndromes (PTHS) and include Juvenile Polyposis, Lhermitte-Duclos disease, Bannayan-Riley-Ruvalcaba, Cowden Syndrome, Proteus-syndrome, and Proteus-like conditions. Cowden syndrome and Bannayan-Riley-Ruvalcaba have been associated with autism and intellectual disability.

Cowden syndrome has an estimated prevalence of 1/200,000 and may be diagnosed by the presence of either pathognomonic criteria or a specific combination of major and minor criteria. Severe and progressive macrocephaly (>2 S.D.) associated with autism should prompt consideration of the diagnosis and led to publication of the first reported case of Cowden syndrome-associated autism and epilepsy [83]. A similar pattern with the addition of a lipoma and thyroid adenoma led to the identification of Bannayan-Riley-Ruvalcaba Syndrome (BRRS) in a nine-year-old girl [84]. A retrospective review of 114 patients analyzed for PTEN mutations revealed mutations in 18% of those with macrocephaly in addition to either ASD or ID [85]. This contrasts with a PTEN mutation in one of eighty-eight children (1%) with macrocephaly and ASD [86]. BRRS does not have established diagnostic criteria; however, macrocephaly is usually the most striking feature. Identification of Cowden and Bannayan-Riley-Ruvalcaba syndrome in the same family raises the possibility of the two syndromes being the same syndrome with variation in phenotypic expression [87].

Functional analysis of the consequence of PTEN germline mutations from individuals with autism spectrum disorders was compared to PTEN germline mutations in individuals with PTHS in a humanized yeast-based bioassay and revealed greater preservation of PTEN PIP3 phosphatase activity in those with ASD [88].

2.4.2. Neurobiology of mTOR Dysregulation

PTEN is important in mTOR signaling since it removes a phosphate from phosphatidylinositol 3,4,5-triphosphate (PIP3). This conversion from PIP3 to PIP2 negates the activity of PI3K and results in elevation of mTOR since the processes downstream—Akt activation, Akt-mediated phosphorylation and inhibition of TSC2, release inhibition of Rheb which activates mTOR.

2.4.3. Preclinical Models

Evidence of impaired synaptic plasticity in PTEN mutations has been identified in Pten conditional knockout mice. Neuropathological features include enlarged neuronal nuclei and cell bodies, increased density of dendritic spines, abnormalities in axonal myelination, and weakening of excitatory synaptic transmission in hippocampal neurons between CA3 and CA1 as evidenced by impaired EPSPs, normal presynaptic function, and reduced long-term potentiation [89]. Cre-driven deletion of Pten in cortical and hippocampal neurons of mice was associated with hyperactivity of the mTOR pathway as well as hypersensitivity to stimuli, social interaction abnormalities, ectopic dendrites, increased axonal synapses, and macrocephaly associated with neuronal hypertrophy [90].

2.4.4. Clinical Trials

A pilot study is now recruiting for an open label trial of sirolimus, an mTOR inhibitor, in adult patients with Cowden syndrome, tumors, and germline PTEN mutations (http://www.clinicaltrials.gov/).

2.5. EIF4E-Associated Autism

Synaptic translation mediated by EIF4E is a common and final process of the pathways associated with PTEN, mTOR, and FMRP and serves a critical role in learning and memory [91, 92]. Linkage to chromosome 4q, the region containing EIF4E has been shown in genome-wide linkage studies [93, 94]. After identification of a translocation involving the region containing EIF4E in a young boy with autistic regression, investigators screened for mutations among families with two autistic siblings and found EIF4E mutations in two related families [95].

3. Conclusions

Review of recent literature reveals significant advances in our ability to understand the pathogenesis of several neurogenetic conditions associated with intellectual disability and autism that have been considered to be idiopathic and untreatable. In this paper, we have highlighted recent discoveries in neurogenetic conditions united primarily by dysregulation of mTOR and evidence of impaired synaptic plasticity (Table 2). In addition to autism and intellectual disability, some of these conditions also share an association with cutaneous lesions and tumor development. Based on this knowledge, it is reasonable to hope that these disorders could become treatable in the near future. Investigators have already begun the process of connected research, as exemplified by the work of Auerbach et al. who simultaneously examined models of TSC and FXS and created a model by crossing the two models to discover that the same intervention, modulating metabotropic glutamate receptor 5, demonstrates efficacy for both models in opposing directions [14]. Continuing to examine the link between these disorders is likely to lead to a greater chance of discovery for all of them. Tools needed to translate basic science research into clinical trials which yield definitive results include refined genotypic and phenotypic characterization, detailed knowledge of the natural history of the conditions, knowledge of optimal therapeutic windows, valid biomarkers, and expertise in clinical trials.

Table 2.

Mechanisms of impaired synaptic plasticity, mTOR dysregulation, and therapeutic targets.

Condition Gene (chromosome) Mechanism of impaired synaptic plasticity impairment mTOR physiology Therapeutic targets
TSC TSC 1 (9) or
TSC2 (16)
↓mGluR-LTD ↑mTOR mTOR antagonists
mGluR 5 agonist

NF1 NF1 (17) ↓LTP↑GABA ↑mTOR Ras antagonists
ERK antagonists

FXS FMR1 (X) ↑mGluR-LTD ↑mTOR mGluR5 antagonists
mGluR5/mGluR1 genetic reduction
GABAA receptor agonist
PLC/GSK3 antagonist (lithium)
AMPA receptor agonist
MMP 9 antagonist

PTHS PTEN (10) ↓LTP ↑mTOR mTOR antagonists

EIF4E-associated autism EIF4E (4) unknown Downstream of mTOR None developed

References

  • 1.Johnston MV, Ishida A, Ishida WN, Matsushita HB, Nishimura A, Tsuji M. Plasticity and injury in the developing brain. Brain and Development. 2009;31(1):1–10. doi: 10.1016/j.braindev.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Haydar TF, Bambrick LL, Krueger BK, Rakic P. Organotypic slice cultures for analysis of proliferation, cell death, and migration in the embryonic neocortex. Brain Research Protocols. 1999;4(3):425–437. doi: 10.1016/s1385-299x(99)00033-1. [DOI] [PubMed] [Google Scholar]
  • 3.Kernie SG, Parent JM. Forebrain neurogenesis after focal ischemic and traumatic brain injury. Neurobiology of Disease. 2010;37(2):267–274. doi: 10.1016/j.nbd.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Gomez M, Sampson JR, Whittemore VH. Tuberous Sclerosis Complex. 3rd edition. Oxford, UK: Oxford University Press; 1999. [Google Scholar]
  • 5.Jansen FE, Vincken KL, Algra A, et al. Cognitive impairment in tuberous sclerosis complex is a multifactorial condition. Neurology. 2008;70(12):916–923. doi: 10.1212/01.wnl.0000280579.04974.c0. [DOI] [PubMed] [Google Scholar]
  • 6.Rosner M, Hanneder M, Siegel N, Valli A, Hengstschläger M. The tuberous sclerosis gene products hamartin and tuberin are multifunctional proteins with a wide spectrum of interacting partners. Mutation Research. 2008;658(3):234–246. doi: 10.1016/j.mrrev.2008.01.001. [DOI] [PubMed] [Google Scholar]
  • 7.Gillberg IC, Gillberg C, Ahlsen G. Autistic behaviour and attention deficits in tuberous sclerosis: a population-based study. Developmental Medicine and Child Neurology. 1994;36(1):50–56. doi: 10.1111/j.1469-8749.1994.tb11765.x. [DOI] [PubMed] [Google Scholar]
  • 8.Webb DW, Fryer AE, Osborne JP. Morbidity associated with tuberous sclerosis: a population study. Developmental Medicine and Child Neurology. 1996;38(2):146–155. doi: 10.1111/j.1469-8749.1996.tb12086.x. [DOI] [PubMed] [Google Scholar]
  • 9.Joinson C, O’Callaghan FJ, Osborne JP, Martyn C, Harris T, Bolton PF. Learning disability and epilepsy in an epidemiological sample of individuals with tuberous sclerosis complex. Psychological Medicine. 2003;33(2):335–344. doi: 10.1017/s0033291702007092. [DOI] [PubMed] [Google Scholar]
  • 10.Staley BA, Montenegro MA, Major P, et al. Self-injurious behavior and tuberous sclerosis complex: frequency and possible associations in a population of 257 patients. Epilepsy and Behavior. 2008;13(4):650–653. doi: 10.1016/j.yebeh.2008.07.010. [DOI] [PubMed] [Google Scholar]
  • 11.Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends in Neurosciences. 2010;33(2):67–75. doi: 10.1016/j.tins.2009.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Von Der Brelie C, Waltereit R, Zhang L, Beck H, Kirschstein T. Impaired synaptic plasticity in a rat model of tuberous sclerosis. European Journal of Neuroscience. 2006;23(3):686–692. doi: 10.1111/j.1460-9568.2006.04594.x. [DOI] [PubMed] [Google Scholar]
  • 13.Ehninger D, Han S, Shilyansky C, et al. Reversal of learning deficits in a TSC 2+/- mouse model of tuberous sclerosis. Nature Medicine. 2008;14(8):843–848. doi: 10.1038/nm1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Auerbach BD, Osterweil EK, Bear MF. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature. 2011;480(7375):63–68. doi: 10.1038/nature10658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bateup HS, Takasaki KT, Saulnier JL, Denefrio CL, Sabatini BL. Loss of Tsc1 in vivo impairs hippocampal mGluR-LTD and increases excitatory synaptic function. Journal of Neuroscience. 2011;31(24):8862–8869. doi: 10.1523/JNEUROSCI.1617-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Napolioni V, Moavero R, Curatolo P. Recent advances in neurobiology of tuberous sclerosis complex. Brain and Development. 2009;31(2):104–113. doi: 10.1016/j.braindev.2008.09.013. [DOI] [PubMed] [Google Scholar]
  • 17.Talos DM, Kwiatkowski DJ, Cordero K, Black PM, Jensen FE. Cell-specific alterations of glutamate receptor expression in tuberous sclerosis complex cortical tubers. Annals of Neurology. 2008;63(4):454–465. doi: 10.1002/ana.21342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Meikle L, Talos DM, Onda H, et al. A mouse model of tuberous sclerosis: neuronal loss of Tsc1 causes dysplastic and ectopic neurons, reduced myelination, seizure activity, and limited survival. Journal of Neuroscience. 2007;27(21):5546–5558. doi: 10.1523/JNEUROSCI.5540-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang Y, Greenwood JSF, Calcagnotto ME, Kirsch HE, Barbaro NM, Baraban SC. Neocortical hyperexcitability in a human case of tuberous sclerosis complex and mice lacking neuronal expression of Tsc1. Annals of Neurology. 2007;61(2):139–152. doi: 10.1002/ana.21058. [DOI] [PubMed] [Google Scholar]
  • 20.Zeng LH, Ouyang Y, Gazit V, et al. Abnormal glutamate homeostasis and impaired synaptic plasticity and learning in a mouse model of tuberous sclerosis complex. Neurobiology of Disease. 2007;28(2):184–196. doi: 10.1016/j.nbd.2007.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jansen LA, Uhlmann EJ, Crino PB, Gutmann DH, Wong M. Epileptogenesis and reduced inward rectifier potassium current in tuberous sclerosis complex-1-deficient astrocytes. Epilepsia. 2005;46(12):1871–1880. doi: 10.1111/j.1528-1167.2005.00289.x. [DOI] [PubMed] [Google Scholar]
  • 22.Franz DN, Leonard J, Tudor C, et al. Rapamycin causes regression of astrocytomas in tuberous sclerosis complex. Annals of Neurology. 2006;59(3):490–498. doi: 10.1002/ana.20784. [DOI] [PubMed] [Google Scholar]
  • 23.Franz DN. Everolimus: an mTOR inhibitor for the treatment of tuberous sclerosis. Expert Review of Anticancer Therapy. 2011;11(8):1181–1192. doi: 10.1586/era.11.93. [DOI] [PubMed] [Google Scholar]
  • 24.Krueger DA, Care MM, Holland K, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. The New England Journal of Medicine. 2010;363(19):1801–1811. doi: 10.1056/NEJMoa1001671. [DOI] [PubMed] [Google Scholar]
  • 25.Ehninger D, Silva AJ. Rapamycin for treating tuberous sclerosis and autism spectrum disorders. Trends in Molecular Medicine. 2011;17(2):78–87. doi: 10.1016/j.molmed.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Huson S. The neurofibromatoses: classification, clinical features and genetic counselling. In: Kaufmann D, editor. Neurofibromatoses (Monographs in Human Genetics) 1st edition. Basel, Switzerland: S. Karger A.G.; 2008. pp. 1–20. [Google Scholar]
  • 27.Hyman SL, Shores A, North KN. The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005;65(7):1037–1044. doi: 10.1212/01.wnl.0000179303.72345.ce. [DOI] [PubMed] [Google Scholar]
  • 28.Cutting LE, Cooper KL, Koth CW, et al. Megalencephaly in NF1: predominantly white matter contribution and mitigation by adhd. Neurology. 2002;59(9):1388–1394. doi: 10.1212/01.wnl.0000032370.68306.8a. [DOI] [PubMed] [Google Scholar]
  • 29.Pride N, Payne JM, Webster R, Shores EA, Rae C, North KN. Corpus callosum morphology and its relationship to cognitive function in neurofibromatosis type 1. Journal of Child Neurology. 2010;25(7):834–841. doi: 10.1177/0883073809350723. [DOI] [PubMed] [Google Scholar]
  • 30.Denckla MB, Hofman K, Mazzocco MMM, et al. Relationship between T2-weighted hyperintensities (unidentified bright objects) and lower iqs in children with neurofibromatosis-1. American Journal of Medical Genetics A. 1996;67(1):98–102. doi: 10.1002/(SICI)1096-8628(19960216)67:1<98::AID-AJMG17>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 31.Chabernaud C, Sirinelli D, Barbier C, et al. Thalamo-striatal T2-weighted hyperintensities (unidentified bright objects) correlate with cognitive impairments in neurofibromatosis type 1 during childhood. Developmental Neuropsychology. 2009;34(6):736–748. doi: 10.1080/87565640903265137. [DOI] [PubMed] [Google Scholar]
  • 32.Hyman SL, Gill DS, Shores EA, Steinberg A, North KN. T2 hyperintensities in children with neurofibromatosis type 1 and their relationship to cognitive functioning. Journal of Neurology, Neurosurgery and Psychiatry. 2007;78(10):1088–1091. doi: 10.1136/jnnp.2006.108134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kraut MA, Gerring JP, Cooper KL, Thompson RE, Denckla MB, Kaufman WE. Longitudinal evolution of unidentified bright objects in children with neurofibromatosis-1. American Journal of Medical Genetics A. 2004;129(2):113–119. doi: 10.1002/ajmg.a.20656. [DOI] [PubMed] [Google Scholar]
  • 34.Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Research. 2005;65(7):2755–2760. doi: 10.1158/0008-5472.CAN-04-4058. [DOI] [PubMed] [Google Scholar]
  • 35.Banerjee S, Crouse NR, Emnett RJ, Gianino SM, Gutmann DH. Neurofibromatosis-1 regulates mTOR-mediated astrocyte growth and glioma formation in a TSC/Rheb-independent manner. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(38):15996–16001. doi: 10.1073/pnas.1019012108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Banerjee S, Gianino SM, Gao F, Christians U, Gutmann DH. Interpreting mammalian target of rapamycin and cell growth inhibition in a genetically engineered mouse model of NF1-deficient astrocytes. Molecular Cancer Therapeutics. 2011;10(2):279–291. doi: 10.1158/1535-7163.MCT-10-0654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Boyanapalli M, Lahoud OB, Messiaen L, et al. Neurofibromin binds to caveolin-1 and regulates ras, fak, and akt. Biochemical and Biophysical Research Communications. 2006;340(4):1200–1208. doi: 10.1016/j.bbrc.2005.12.129. [DOI] [PubMed] [Google Scholar]
  • 38.Sandsmark DK, Zhang H, Hegedus B, Pelletier CL, Weber JD, Gutmann DH. Nucleophosmin mediates mammalian target of rapamycin-dependent actin cytoskeleton dynamics and proliferation in neurofibromin-deficient astrocytes. Cancer Research. 2007;67(10):4790–4799. doi: 10.1158/0008-5472.CAN-06-4470. [DOI] [PubMed] [Google Scholar]
  • 39.Li W, Cui Y, Kushner SA, et al. The hmg-coa reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Current Biology. 2005;15(21):1961–1967. doi: 10.1016/j.cub.2005.09.043. [DOI] [PubMed] [Google Scholar]
  • 40.Costa RM, Federov NB, Kogan JH, et al. Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature. 2002;415(6871):526–530. doi: 10.1038/nature711. [DOI] [PubMed] [Google Scholar]
  • 41.Gau CL, Kato-Stankiewicz J, Jiang C, Miyamoto S, Guo L, Tamanoi F. Farnesyltransferase inhibitors reverse altered growth and distribution of actin filaments in tsc-deficient cells via inhibition of both rapamycin-sensitive and -insensitive pathways. Molecular Cancer Therapeutics. 2005;4(6):918–926. doi: 10.1158/1535-7163.MCT-04-0347. [DOI] [PubMed] [Google Scholar]
  • 42.Guilding C, McNair K, Stone TW, Morris BJ. Restored plasticity in a mouse model of neurofibromatosis type 1 via inhibition of hyperactive erk and creb. European Journal of Neuroscience. 2007;25(1):99–105. doi: 10.1111/j.1460-9568.2006.05238.x. [DOI] [PubMed] [Google Scholar]
  • 43.Krab LC, de Goede-Bolder A, Aarsen FK, et al. Effect of simvastatin on cognitive functioning in children with neurofibromatosis type 1: a randomized controlled trial. Journal of the American Medical Association. 2008;300(3):287–294. doi: 10.1001/jama.300.3.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Acosta MT, Kardel PG, Walsh KS, Rosenbaum KN, Gioia GA, Packer RJ. Lovastatin as treatment for neurocognitive deficits in neurofibromatosis type 1: phase I study. Pediatric Neurology. 2011;45(4):241–245. doi: 10.1016/j.pediatrneurol.2011.06.016. [DOI] [PubMed] [Google Scholar]
  • 45.Hagerman RJ, Amiri K, Cronister A. Fragile X checklist. American Journal of Medical Genetics A. 1991;38(2-3):283–287. doi: 10.1002/ajmg.1320380223. [DOI] [PubMed] [Google Scholar]
  • 46.Hagerman PJ. The fragile X prevalence paradox. Journal of Medical Genetics. 2008;45(8):498–499. doi: 10.1136/jmg.2008.059055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hagerman RJ, Hagerman PJ. Fragile X syndrome: a model of gene-brain-behavior relationships. Molecular Genetics and Metabolism. 2001;74(1-2):89–97. doi: 10.1006/mgme.2001.3225. [DOI] [PubMed] [Google Scholar]
  • 48.Rogers SJ, Wehner EA, Hagerman R. The behavioral phenotype in fragile X: symptoms of autism in very young children with fragile X syndrome, idiopathic autism, and other developmental disorders. Journal of Developmental and Behavioral Pediatrics. 2001;22(6):409–417. doi: 10.1097/00004703-200112000-00008. [DOI] [PubMed] [Google Scholar]
  • 49.Verkerk AJMH, Pieretti M, Sutcliffe JS, 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(5):905–914. doi: 10.1016/0092-8674(91)90397-h. [DOI] [PubMed] [Google Scholar]
  • 50.Devys D, Lutz Y, Rouyer N, Bellocq JP, Mandel JL. The FMR-1 protein is cytoplasmic, most abundant in neurons and appears normal in carriers of a fragile X premutation. Nature Genetics. 1993;4(4):335–340. doi: 10.1038/ng0893-335. [DOI] [PubMed] [Google Scholar]
  • 51.Penagarikano O, Mulle JG, Warren ST. The pathophysiology of fragile X syndrome. Annual Review of Genomics and Human Genetics. 2007;8:109–129. doi: 10.1146/annurev.genom.8.080706.092249. [DOI] [PubMed] [Google Scholar]
  • 52.Bear MF, Huber KM, Warren ST. The mGluR theory of fragile X mental retardation. Trends in Neurosciences. 2004;27(7):370–377. doi: 10.1016/j.tins.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 53.Muddashetty RS, Kelić S, Gross C, Xu M, Bassell GJ. Dysregulated metabotropic glutamate receptor-dependent translation of AMPA receptor and postsynaptic density-95 mRNAs at synapses in a mouse model of fragile X syndrome. Journal of Neuroscience. 2007;27(20):5338–5348. doi: 10.1523/JNEUROSCI.0937-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Muddashetty RS, Nalavadi VC, Gross C, et al. Reversible inhibition of PSD-95 mRNA translation by miR-125a, FMRP phosphorylation, and mGluR signaling. Molecular Cell. 2011;42(5):673–688. doi: 10.1016/j.molcel.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sharma A, Hoeffer CA, Takayasu Y, et al. Dysregulation of mTOR signaling in fragile X syndrome. Journal of Neuroscience. 2010;30(2):694–702. doi: 10.1523/JNEUROSCI.3696-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.McBride SMJ, Choi CH, Wang Y, et al. Pharmacological rescue of synaptic plasticity, courtship behavior, and mushroom body defects in a drosophila model of fragile X syndrome. Neuron. 2005;45(5):753–764. doi: 10.1016/j.neuron.2005.01.038. [DOI] [PubMed] [Google Scholar]
  • 57.Dölen G, Osterweil E, Rao BSS, et al. Correction of fragile X syndrome in mice. Neuron. 2007;56(6):955–962. doi: 10.1016/j.neuron.2007.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Thomas AM, Bui N, Graham D, Perkins JR, Yuva-Paylor LA, Paylor R. Genetic reduction of group 1 metabotropic glutamate receptors alters select behaviors in a mouse model for fragile X syndrome. Behavioural Brain Research. 2011;223(2):310–321. doi: 10.1016/j.bbr.2011.04.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Blundell J, Kaeser PS, Südhof TC, Powell CM. RIM1α and interacting proteins involved in presynaptic plasticity mediate prepulse inhibition and additional behaviors linked to schizophrenia. Journal of Neuroscience. 2010;30(15):5326–5333. doi: 10.1523/JNEUROSCI.0328-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.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. Journal of Neuroscience. 2011;31(40):14223–14234. doi: 10.1523/JNEUROSCI.3157-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Deng PY, Sojka D, Klyachko VA. Abnormal presynaptic short-term plasticity and information processing in a mouse model of fragile X syndrome. Journal of Neuroscience. 2011;31(30):10971–10982. doi: 10.1523/JNEUROSCI.2021-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Olmos-Serrano JL, Corbin JG, Burns MP. The GABAA receptor agonist THIP ameliorates specific behavioral deficits in the mouse model of fragile X syndrome. Developmental Neuroscience. 2011;33(5):395–403. doi: 10.1159/000332884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Budimirovic DB, Bukelis I, Cox C, Gray RM, Tierney E, Kaufmann WE. Autism spectrum disorder in fragile X syndrome: differential contribution of adaptive socialization and social withdrawal. American Journal of Medical Genetics A. 2006;140(17):1814–1826. doi: 10.1002/ajmg.a.31405. [DOI] [PubMed] [Google Scholar]
  • 64.Budimirovic DB, Kaufmann WE. What can we learn about autism from studying fragile X syndrome? Developmental Neuroscience. 2011;33(5):379–394. doi: 10.1159/000330213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kaufmann WE, Cortell R, Kau ASM, et al. Autism spectrum disorder in fragile X syndrome: communication, social interaction, and specific behaviors. American Journal of Medical Genetics A. 2004;129(3):225–234. doi: 10.1002/ajmg.a.30229. [DOI] [PubMed] [Google Scholar]
  • 66.Meguid NA, Fahim C, Sami R, et al. Cognition and lobar morphology in full mutation boys with fragile X syndrome. Brain and Cognition. 2012;78(1):74–84. doi: 10.1016/j.bandc.2011.09.005. [DOI] [PubMed] [Google Scholar]
  • 67.Jacquemont S, Curie A, des Portes V, et al. Epigenetic modification of the fmr1 gene in fragile X syndrome is associated with differential response to the mGluR5 antagonist AFQ056. Science Translational Medicine. 2011;3(64) doi: 10.1126/scitranslmed.3001708. Article ID 64ra1. [DOI] [PubMed] [Google Scholar]
  • 68.Berry-Kravis E, Hessl D, Coffey S, et al. A pilot open label, single dose trial of fenobam in adults with fragile X syndrome. Journal of Medical Genetics. 2009;46(4):266–271. doi: 10.1136/jmg.2008.063701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Erickson CA, Mullett JE, McDougle CJ. Brief report: acamprosate in fragile X syndrome. Journal of Autism and Developmental Disorders. 2010;40(11):1412–1416. doi: 10.1007/s10803-010-0988-9. [DOI] [PubMed] [Google Scholar]
  • 70.Berry-Kravis E, Sumis A, Hervey C, et al. Open-label treatment trial of lithium to target the underlying defect in fragile X syndrome. Journal of Developmental and Behavioral Pediatrics. 2008;29(4):293–302. doi: 10.1097/DBP.0b013e31817dc447. [DOI] [PubMed] [Google Scholar]
  • 71.Arai A, Kessler M, Xiao P, Ambros-Ingerson J, Rogers G, Lynch G. A centrally active drug that modulates AMPA receptor gated currents. Brain Research. 1994;638(1-2):343–346. doi: 10.1016/0006-8993(94)90669-6. [DOI] [PubMed] [Google Scholar]
  • 72.Arai A, Kessler M, Rogers G, Lynch G. Effects of a memory-enhancing drug on dl-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor currents and synaptic transmission in hippocampus. Journal of Pharmacology and Experimental Therapeutics. 1996;278(2):627–638. [PubMed] [Google Scholar]
  • 73.Berry-Kravis E, Krause SE, Block SS, et al. Effect of CX516, an ampa-modulating compound, on cognition and behavior in fragile X syndrome: a controlled trial. Journal of Child and Adolescent Psychopharmacology. 2006;16(5):525–540. doi: 10.1089/cap.2006.16.525. [DOI] [PubMed] [Google Scholar]
  • 74.Imbesi M, Uz T, Manev R, Sharma RP, Manev H. Minocycline increases phosphorylation and membrane insertion of neuronal glur1 receptors. Neuroscience Letters. 2008;447(2-3):134–137. doi: 10.1016/j.neulet.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bilousova TV, Dansie L, Ngo M, et al. Minocycline promotes dendritic spine maturation and improves behavioural performance in the fragile X mouse model. Journal of Medical Genetics. 2009;46(2):94–102. doi: 10.1136/jmg.2008.061796. [DOI] [PubMed] [Google Scholar]
  • 76.Siller SS, Broadie K. Neural circuit architecture defects in a Drosophila model of fragile X syndrome are alleviated by minocycline treatment and genetic removal of matrix metalloproteinase. DMM Disease Models and Mechanisms. 2011;4(5):673–685. doi: 10.1242/dmm.008045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Paribello C, Tao L, Folino A, et al. Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC Neurology. 2010;10, article 91 doi: 10.1186/1471-2377-10-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kesler SR, Lightbody AA, Reiss AL. Cholinergic dysfunction in fragile X syndrome and potential intervention: a preliminary 1h mrs study. American Journal of Medical Genetics A. 2009;149(3):403–407. doi: 10.1002/ajmg.a.32697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Erickson CA, Weng N, Weiler IJ, et al. Open-label riluzole in fragile X syndrome. Brain Research. 2011;1380:264–270. doi: 10.1016/j.brainres.2010.10.108. [DOI] [PubMed] [Google Scholar]
  • 80.Hall SS, Lightbody AA, McCarthy BE, Parker KJ, Reiss AL. Effects of intranasal oxytocin on social anxiety in males with fragile X syndrome. Psychoneuroendocrinology. 2012;37(4):509–518. doi: 10.1016/j.psyneuen.2011.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Erickson CA, Stigler KA, Posey DJ, McDougle CJ. Aripiprazole in autism spectrum disorders and fragile X syndrome. Neurotherapeutics. 2010;7(3):258–263. doi: 10.1016/j.nurt.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Pacey LKK, Heximer SP, Hampson DR. Increased gabab receptor-mediated signaling reduces the susceptibility of fragile X knockout mice to audiogenic seizures. Molecular Pharmacology. 2009;76(1):18–24. doi: 10.1124/mol.109.056127. [DOI] [PubMed] [Google Scholar]
  • 83.Conti S, Condo M, Posar A, et al. Phosphatase and tensin homolog (PTEN) gene mutations and autism: literature review and a case report of a patient with Cowden syndrome, autistic disorder, and epilepsy. Journal of Child Neurology. 2012;27(3):392–397. doi: 10.1177/0883073811420296. [DOI] [PubMed] [Google Scholar]
  • 84.Stein MT, Elias ER, Saenz M, Pickler L, Reynolds A. Autistic spectrum disorder in a 9-year-old girl with macrocephaly. Journal of Developmental and Behavioral Pediatrics. 2010;31(7):632–634. doi: 10.1097/DBP.0b013e3181ef422a. [DOI] [PubMed] [Google Scholar]
  • 85.Varga EA, Pastore M, Prior T, Herman GE, McBride KL. The prevalence of PTEN mutations in a clinical pediatric cohort with autism spectrum disorders, developmental delay, and macrocephaly. Genetics in Medicine. 2009;11(2):111–117. doi: 10.1097/GIM.0b013e31818fd762. [DOI] [PubMed] [Google Scholar]
  • 86.Buxbaum JD, Cai G, Chaste P, et al. Mutation screening of the PTEN gene in patients with autism spectrum disorders and macrocephaly. American Journal of Medical Genetics B. 2007;144(4):484–491. doi: 10.1002/ajmg.b.30493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Perriard J, Saurat JH, Harms M. An overlap of Cowden’s disease and bannayan-riley-ruvalcaba syndrome in the same family. Journal of the American Academy of Dermatology. 2000;42(2):348–350. doi: 10.1016/s0190-9622(00)90109-9. [DOI] [PubMed] [Google Scholar]
  • 88.Rodríguez-Escudero I, Oliver MD, Andrés-Pons A, Molina M, Cid VJ, Pulido R. A comprehensive functional analysis of PTEN mutations: Implications in tumor- and autism-related syndromes. Human Molecular Genetics. 2011;20(21):4132–4142. doi: 10.1093/hmg/ddr337. [DOI] [PubMed] [Google Scholar]
  • 89.Fraser MM, Bayazitov IT, Zakharenko SS, Baker SJ. Phosphatase and tensin homolog, deleted on chromosome 10 deficiency in brain causes defects in synaptic structure, transmission and plasticity, and myelination abnormalities. Neuroscience. 2008;151(2):476–488. doi: 10.1016/j.neuroscience.2007.10.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kwon CH, Luikart BW, Powell CM, et al. Pten regulates neuronal arborization and social interaction in mice. Neuron. 2006;50(3):377–388. doi: 10.1016/j.neuron.2006.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Klann E, Dever TE. Biochemical mechanisms for translational regulation in synaptic plasticity. Nature Reviews Neuroscience. 2004;5(12):931–942. doi: 10.1038/nrn1557. [DOI] [PubMed] [Google Scholar]
  • 92.Richter JD, Sonenberg N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature. 2005;433(7025):477–480. doi: 10.1038/nature03205. [DOI] [PubMed] [Google Scholar]
  • 93.Schellenberg GD, Dawson G, Sung YJ, et al. Evidence for multiple loci from a genome scan of autism kindreds. Molecular Psychiatry. 2006;11(11):1049–1060. doi: 10.1038/sj.mp.4001874. [DOI] [PubMed] [Google Scholar]
  • 94.Yonan AL, Alarcón M, Cheng R, et al. A genomewide screen of 345 families for autism-susceptibility loci. American Journal of Human Genetics. 2003;73(4):886–897. doi: 10.1086/378778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Neves-Pereira M, Müller B, Massie D, et al. Deregulation of eIF4E: a novel mechanism for autism. Journal of Medical Genetics. 2009;46(11):759–765. doi: 10.1136/jmg.2009.066852. [DOI] [PubMed] [Google Scholar]

Articles from Neural Plasticity are provided here courtesy of Wiley

RESOURCES