Abstract
Fragile X spectrum disorder (FXSD) includes: fragile X syndrome (FXS), fragile X-associated tremor ataxia syndrome (FXTAS) and fragile X-associated primary ovarian insufficiency (FXPOI), as well as other medical, psychiatric and neurobehavioral problems associated with the premutation and gray zone alleles. FXS is the most common monogenetic cause of autism (ASD) and intellectual disability (ID). The understanding of the neurobiology of FXS has led to many targeted-treatment trials in FXS. The first wave of phase II clinical trials in FXS were designed to target the mGluR5 pathway; however the results did not show significant efficacy and the trials were terminated. The advances in the understanding of the GABA system in FXS have shifted the focus of treatment trials to GABA agonists, and a new wave of promising clinical trials is under way. Ganaxolone and allopregnanolone (GABA agonists) have been studied in individuals with FXSD and are currently in phase II trials. Both allopregnanolone and ganaxolone may be efficacious in treatment of FXS and FXTAS, respectively. Allopregnanolone, ganaxolone, riluzole, gaboxadol, tiagabine, and vigabatrin are potential GABAergic treatments. The lessons learned from the initial trials have not only shifted the targeted system, but also have refined the design of clinical trials. The results of these new trials will likely impact further clinical trials for FXS and other genetic disorders associated with ASD.
Keywords: GABA, GABAA, GABA system, Fragile X Spectrum Disorder, FXSD, Fragile X Syndrome, FXS, Fragile X-associated Tremor Ataxia Syndrome, FXTAS, Targeted treatments, Clinical trials, ASD treatments, Autism
INTRODUCTION
The recently proposed term “fragile X spectrum disorder” (FXSD) highlights the continuity of clinical phenotypes from the gray zone (45-54 CGG repeats in the promoter of the FMR1 gene, located in the X chromosome) throughout the premutation range (55-200) and into the full mutation range (>200). The FXSD term emerged due to an overlap of symptoms across the CGG repeat range, CGG repeat range. Developmental problems similar to those with fragile X syndrome (FXS) including intellectual disability (ID), autism spectrum disorders (ASD) and seizures can occur in some children with the premutation [1-3]; and FXTAS, typically associated with the premutation, has now been observed in individuals with the gray zone mutation [4,5] and full mutation with lack of methylation or mosaicism [6-8].
Since the initial description of the fragile X syndrome (FXS) by Lubs and colleagues [9] almost five decades ago, considerable advances in the understanding of the phenotype-genotype and the neurobiology of FXS have been made. FXS is the leading mono- genic form of ASD and ID in males and presents with typical facial dymorphism in the majority of older individuals but in only 30% of children. Intellectual disability occurs in 85% of males (mean IQ is 40) and 25% of females (IQ below 70). In addition, about 60% of males with FXS have a diagnosis of ASD [10, 11]. The physical features are long and narrow face, large and prominent ears, high arched palate, hyperextensible finger joints, pectus excavatum, flat feet, soft skin and mitral valve prolapse. Other signs include low muscle tone, seizures and pubertal macroorchidism [12-14]. Studies show that the clinical features of individuals with FXS [14, 15] are the result of the FMRP (FMR1 encoded protein) deficit [16] seen in the full mutation [17] and abnormal methylation of the promoter and the gene [18, 19]. Males with the full mutation have little or absent production of FMR1 mRNA and FMRP [20]. Females have variable levels of FMR1 mRNA and FMRP, related to the X- chromosome activation ratio (the percentage of cells with the normal X as the active X chromosome) [21].
FMRP, an RNA binding protein, is in part a key translational suppressor and a transport regulator of several mRNAs that are important for synaptic plasticity [22]. FMRP acutely regulates metabotropic glutamate receptor (mGluR)-stimulated protein synthesis and long-term synaptic depression (mGluR-LTD) [23]. In the absence of FMRP, there is an increased number of long and immature dendritic spines of neurons in the Fmr1 knockout (KO) mice [24, 25]. The mGluR5 pathway plays a role on the development of long-term depression (LTD) in FXS, which in turn weaken long-term memory consolidation [26-28]. These advances in understanding the neurobiology of FXS have led to studies of targeted treatments that can rescue many features of FXS in the Fmr1 KO mice and in other animal models [22, 29-33]. In the past decade, human clinical trials for FXS based on the use of mGluR5 antagonists were conducted; however, due to their lack of efficacy these trials were abandoned [32, 34, 35]. Since then, the focus has shifted to the GABA system [36]. Studies in the Fmr1 KO mouse demonstrated down-regulation of the GABA system with decreased levels of many of the GABA receptors and proteins that are related to the synthesis and metabolism of GABA [30, 37, 38]. These findings have led to clinical trials of GABAergic drugs in FXS. In this review we will discuss the GABA deficits observed in FXSD as well as and potential GABAergic compounds and ongoing related clinical trials.
In 1991 the second FXSD fragile X-associated primary ovarian insufficiency (FXPOI) was described and was linked to the premutation allele [39, 40]. About 20% of female carriers have FXPOI, which is defined as menopause before the age of 40. The mechanism that causes FXPOI is not known, but it has been proposed that the premutation leads to the accumulation of ovarian toxic products [41, 42]. The third FXSD, also linked to the premutation allele, was described in 2001 [43] and subsequently named fragile X- associated tremor ataxia syndrome (FXTAS) [44, 45].
Clinical features of FXTAS include progressive kinetic tremor, gait ataxia, executive function and memory deficits, peripheral neuropathy, and parkinsonism; with associated variable features, such as cognitive decline into dementia, dysautonomia, and psychiatric problems, including depression, apathy, and anxiety [43, 46- 48]. Many individuals with the premutation develop neurological and cognitive problems—including executive function deficits, memory problems, neuropathy, immune mediated problems, migraines, hypertension, and psychiatric symptoms, particularly anxiety—before the onset of tremor and ataxia [49-55]. Neuroimaging in FXTAS reveals white matter disease and diffusion tensor imaging (DTI) abnormalities of both the cerebellum and cerebrum [56- 61]; atrophy of the basal ganglia and brain stem [61, 62], cerebellum spongiosis [59,63,64], demyelination in the white matter, and iron accumulation in choroid plexus and basal ganglia [65]. The pathological hallmark of FXTAS is the presence of intranuclear inclusions in neurons and astrocytes throughout the central nervous system and peripheral nervous system [62, 66, 67]. Peripheral organ inclusions are found in the gastro-intestinal system, heart, testicles, adrenals and pancreas; and their presence correlate to many of the symptoms that are commonly seen in carriers, including cardiac arrhythmias, hypertension, irritable bowel syndrome, erectile dysfunction, and low testosterone levels [66, 68]. Although FXTAS is a disorder associated with aging in carriers of the FMR1 premutation, FXTAS can occur earlier in adult life, particularly if another disease process is occurring, such as substance abuse that may exacerbate the pathological process of FXTAS [69-71].
FXTAS is thought to be the result of mRNA toxicity from the elevated FMR1 mRNA levels, a chronic DNA damage repair mechanism, and in part to lower FMRP levels [72]. The elevated levels of mRNA can form hairpin structures which are sticky and lead to the sequestration of many proteins that are important for neuronal function and the formation of pathognomonic inclusion bodies in neurons and astrocytes throughout the CNS, the peripheral nervous system, and other organs [8, 73]. Animal model studies show that mouse hippocampal premutation neurons have a cluster burst firing activity related to the GABA deficits; and allopreg- nanolone, a GABAA agonist, eliminates the abnormal burst firing [74]. A clinical trial of allopregnanolone in premutation carriers with FXTAS is under development and portends the potential use of GABAergic treatments in premutation carriers.
GABA IN THE CORTEX
While the glutamatergic hypothesis of FXS proposes an exaggerated excitatory mGluR signaling, the GABAergic hypothesis is anchored in decreased GABA signaling. These two hypotheses combined suggest that an excitatory/inhibitory imbalance underlies the pathophysiology of FXS. GABA is the major inhibitory neurotransmitter in the adult mammalian brain and is synthesized and released by interneurons in the cerebral cortex. These interneurons are the only source of GABA and the main provider of inhibitory signaling to the cerebral cortex. All interneurons in the cortex are inhibitory with the exception of spiny stellate cells. Inhibitory interneurons include a wide variety of subpopulations that can be classified based on their morphology, marker expression, and electrophysiological properties. GABA is synthesized by two isoforms of glutamic acid decarboxylase (GAD), GAD65 and GAD67. Both isoforms are present in most interneurons, but GAD67 is located the cytoplasm, whereas GAD65 is located in membranes and nerve endings. It has been suggested that GAD67 preferentially synthesizes cytoplasmic GABA, while GAD65 synthesized GABA for vesicular release [75]. Therefore, the level of expression of GAD65 vs. that of GAD67 could be an indicator of interneuronal capacity to produce and release GABA. GABA is secreted into the synaptic space and binds to transmembrane receptors in pre- and postsynaptic neuronal processes, normally inducing hyperpolarizing events. There are two GABA receptors, the ionotropic GABAA receptor and the metabotropic GABAB receptor. In the adult brain, GABA acts primarily through activation of the fast-hyperpolarizing GABAA receptors that are ligand-gated chloride ion channels comprised of a, B, y and 8 subunits in a heteropentameric structure [76, 77]. GABAB receptors are responsible for the later and slower component of inhibitory transmission [78] and are composed of two subunits, 1 and 2. GABA is released into the extracellular space, where there is reuptake by the GABA transporter GAT1 to the intracellular space. The balance between the different components of the GABA system modulates the inhibitory state of the CNS circuitry and an alteration of this balance induces a change of the excitatory/inhibitory balance in the brain and therefore circuitry malfunction.
GABA SYSTEM RELATED TREATMENT FOR FXS
Animal Models
The standard animal model for the study of FXS is the Fmr1 KO mouse. Studies investigating the amount of the GABA neurotransmitter in the Fmr1 KO mice have not come into an agreement. While, an increase in the amount of GABA was described in brainstem [79], a decrease in GABA has been noted in the cortex and cerebellum in these mice [37, 80]. The number of GABA-synthesizing cells, the inhibitory interneurons, was reported unchanged in the cortex; however the number of a specific type of interneuronal subpopulation, the PV+ cells, were decreased in the cortex of Fmr1 KO mice [37, 81]. Increasing evidence also sup- ports a role for GABAA receptors in the pathophysiology of FXTAS [82]. Fmr1 KO mice present with underexpression of a series of messengers and proteins of GABA receptor subunits (a to p subunits in cortex, hippocampus, cerebellum, and other brain region [83-89]. It has been suggested that the absence of FMRP in mice leads to dysregulation of the mRNAs of some GABAA receptors that would normally be bound to it [36]. GABA transporter proteins (GAT1-4), GABA degradation enzymes (succinate semialdehyde dehydrogenase and GABA transaminase), and GABA receptor-clustering proteins (gephyrin) have also been studied in the Fmr1 KO mice, being mostly down-regulated [82, 84]. However, the level of GAD65/67 protein was increased in the cortex, hippocampus, brainstem and cerebellum [87, 88], while it was reported to be decreased in the amygdala of the Fmr1 KO mice [37]. A GABAB receptor subunit is also down-regulated in the Fmr1 KO mice [90]. The expression of genes encoding enzymes and proteins important in the synthesis, transport, and degradation of GABA or related to the clustering of GABAA receptors has also been examined in FXTAS. Cerebellar RNA over-expression of several GABAA receptor subunits (a1, a3, a4, B1, B2 and y2) and proteins involved in the GABA metabolism (GAD1, ssadh and gephyrin) in a CGG repeat premutation knock-in mouse, a model for FXTAS, was reported [82]. Because no differential expression of the GABA-related genes was observed in the cerebellum of the knock- in premutation mouse, the authors speculated that it is unlikely that a reduction of FMRP levels is responsible for these findings and hypothesized that it may be due to an RNA effect [82]. More studies are needed in order to understand the state of the GABAergic system in both FXS and FXTAS. Unfortunately, similar studies in postmortem tissue of patients with FXS are lacking at this point. Studies in humans are necessary in order to better understand these diseases and generate novel and efficient treatments for FXS and premutation involvement.
One of the main targets for treatments for FXS is the GABAA receptor. Several modulators of GABAA activity have been tested in preclinical studies in the Fmr1 KO mice, including gaboxadol, and neurosteroids such as allopregnanolone or similar synthetic steroids. Gaboxadol is a superagonist of 8 containing GABAA receptor [91]. Gaboxadol-treated Fmr1 KO mice experienced an in- crease in the axon-potential threshold and a decrease of hyperactivity [92]. Additionally, alphaxalone and ganaxolone, synthetic neurosteroids, prevented seizures and corrected repetitive behavior in the Fmr1 KO mice [93]. The GABAB receptor has also been targeted for treatment in the Fmr1 KO mouse and in patients. Arbaclofen, a selective GABAB agonist, reversed social behavior deficits and elevated protein synthesis in Fmr1 KO mice [94, 95].
No GABA-related preclinical treatments have been performed for FXTAS. However, Zhengyu Cao and collaborators evaluated the influence of allopregnanolone, a positive allosteric modulator of the postsynaptic GABAA receptors [96], on the electric firing pattern of premutation hippocampal neurons [74]. They found that allopregnanolone suppressed the clustered burst firing pattern of premutation hippocampal neurons, reducing the spike rate as well as the mean burst duration, in a concentration dependent and reversible manner. Allopregnanolone and ganaxolone are agonists (allosteric modulators) of GABAA receptors [97, 98] that are currently being studied as a FXS treatment in humans. Acomprosate, riluzole, gaboxadol, tiagabine, and vigabatrin are other potential GABAergic treatments.
HUMAN STUDIES
(Table 1. summarizes the studies in individuals with FXSD).
Table 1.
Drug | Efficacy in FXSD |
---|---|
Acamprosate | Improvements in language and behavior [101] Improvements in social behavior and inattention/hyperactivity [102] |
Arbaclofen | Improvements in social function and behavior in children with FXS [103] |
Allopregnanolone | Initiating for adults with FXTAS |
Ganaxolone | Ongoing study in children with FXS (ClinicalTrials.gov Identifier: NCT01725152) |
Gaboxadol | No studies but recently licensed for initiation of phase 1 and 2 studies in FXS and Angelman syndrome |
Riluzole | Clinical response in 1/6, Improvements in attention and hyperactivity, correction in ERK activation (open label) [129]. One patient with FXTAS improved in the ICARS [128] |
Tiagabine | No studies |
Vigabatrin | No studies |
Acamprosate
Acamprosate, an FDA-approved drug for the maintenance of abstinence from alcohol use in adults, is an agent with potential pleiotropic effects impacting glutamate and GABA neurotransmission [99]. In animal studies, acamprosate acts as an antagonist at the NMDA glutamate receptor and exhibits GABAA agonism [99, 100]. The open-label treatment with acamprosate in three patients with FXS and a comorbid diagnosis of ASD showed that all three patients improved in linguistic communication [101].
A prospective open-label 10-week trial of acamprosate was also conducted in 12 youth aged 6-17 years (mean age: 11.9 years) with FXS. Acamprosate use (mean dose: 1,054 ± 422 mg/day) was generally safe and well tolerated and was associated with a significant improvement in social behavior and a reduction in inattention/hyperactivity. An increase in brain-derived neurotrophic factor (BDNF) occurred with treatment and BDNF may be a useful bio- marker in future acamprosate studies [102].
Arbaclofen
Arbaclofen, a selective GABAB agonist, was evaluated in a randomized, double-blind, placebo-controlled crossover study in 63 subjects (55 male), ages 6 to 39 years, with FXS. A post-hoc subgroup of 27 subjects with more severe social impairment showed improvements on the Vineland II-Socialization raw score, on the ABC-Social Avoidance scale, and on all global measures. In this exploratory study, arbaclofen did not show a benefit on irritability in FXS [103]. This study led to an 8-week open-label trial enrolling 32 children and adolescents with either Autistic Disorder or Pervasive Developmental Disorder-Not Otherwise Specified without FXS, and a score 2:17 on the Aberrant Behavior Checklist (ABC)- Irritability subscale [104]. Arbaclofen was generally well tolerated. The most common adverse events were agitation and irritability, which typically resolved without dose changes, and were often felt to represent spontaneous variation in underlying symptoms. Improvements were observed on several outcome measures in this exploratory trial, including the ABC-Irritability (the primary end- point) and the Lethargy/Social Withdrawal subscales, the Social Responsiveness Scale, the CY-BOCS-PDD, and clinical global impression scales. However, a subsequent 12-week double-blind controlled trial of arbaclofen in individuals with ASD aged 5 to 21 did not demonstrate efficacy [105], leading to the dissolution of the company. The phase III double-blind controlled trial of arbaclofen in adults with FXS also did not demonstrate efficacy; however, the controlled trial in children with FXS aged 5 to 11 did demonstrate limited efficacy in two secondary measures (ABC FX-Irritability scale and the parent stress scale), although the primary outcome measure, ABC FX-Social Avoidance Scale, did not demonstrate efficacy [106]. Although arbaclofen may have limited efficacy in young children with FXS, it is no longer available for research study.
Allopregnanlone
Allopregnanolone is a neurosteroid and positive modulator of GABAA receptors. Functions associated with this compound in- clude stimulation of neurogenesis in the hippocampus [107], reversal of hippocampal-dependent learning and memory problems [108], neuroprotection by reducing the expression of the proapoptotic protein caspase 3 [109, 110] and inhibiting the mitochondrial permeability transition pores, a key process in the intrinsic pathway of apoptosis [111]. As previously mentioned, preliminary data demonstrate abnormal network bursting activity in hippocampal neurons cultured from premutation mice in addition to up-regulation of several proteins that are related to the RNA toxicity caused by the elevated FMR1 mRNA seen in premutation carriers [74]. Both the RNA toxicity and the persistent bursting are a plausible mechanism for neurodegeneration that may lead to the neurological symptoms in FXTAS. Allopregnanolone, by potentiating GABAA-receptor signaling, has been shown to eliminate the up-regulation of proteins and the abnormal neuronal bursting in premutation neurons that leads to neuroprotection [74, 112].
Since Alzheimer’s disease (AD) is sometimes associated with FXTAS [55, 113], and postmortem neuropathological studies of brains of individuals with FXTAS have demonstrated changes consistent with AD, it is important to review some of the studies of allopregnanolone in AD. Preliminary data demonstrated in an AD mouse model and human progenitor cells that allopregnanolone improved neuronal proliferation [114], reversed neurogenic and cognitive deficits [115], promoted regeneration and reduced beta-amyloid burden [116], restored hippocampal dependent learning, and improved memory and survival [108]. In addition, allopregnanolone is reduced in the prefrontal and temporal cortex in AD. Currently this drug is being studied in AD (ClinicalTrials.gov Identifier: NCT02221622), traumatic brain injury (ClinicalTrials.gov Identifier: NCT01673828) and mild traumatic brain injury (ClinicalTrials.gov Identifier: NCT01336413).
In summary, allopregnanolone can prevent neuronal loss and also stimulate the generation of new neurons from neural stem and progenitor cells. This compound is considered a targeted treatment for FXTAS and may counteract the development of neurological disability by improving executive function deficits, memory deficits, and psychiatric symptoms, particularly anxiety (a hallmark of premutation carriers) [50].
Currently, there is an IND for an open trial to assess the safety and efficacy of allopregnanolone as a new treatment for adults with FXTAS and neurological problems. Premutation carriers ages 50- 85 who present with tremor and/or ataxia in addition to executive function deficits will be enrolled. The participants will receive escalating doses of allopregnanolone IV infusions once a week for three weeks to reach the highest tolerated dose and continue for 12 weeks. Primary outcome measures include the California Verbal Learning Test. Secondary end points are other cognitive measures, such as the Behavioral Dyscontrol Scale (BDS-2), CANTAB battery, COWAT, ERP measures, and a tremor/balance measure, the CATSYS, used in previous treatment studies of FXTAS [117]. It is hoped that this treatment will stabilize the progression of FXTAS and improve memory and executive function deficits in addition to anxiety that these carriers have.
Ganaxolone
Ganaxolone, also known as CCD-1042, is a 3B-methylated synthetic analog of the progesterone metabolite allopregnanolone, which is a neuroactive steroid, and is a potent and selective positive allosteric modulator of the GABAA receptor [98]. Ganaxolone is beneficial for the treatment of seizures in children and adults [87, 96, 98, 118, 119] and also has sedative and anxiolytic effects. Unlike benzodiazepines, there does not appear to be tolerance to the anticonvulsant effects of ganaxolone [118, 120, 121]. In humans, ganaxolone is well tolerated [118, 121-125], with side- effects including sedation, dizziness, fatigue, and headache. Six hundred thirty one adults (males and females, age 18 to 69 years old) and 79 pediatric subjects (males and female, age 6 months to 15 years) have been exposed to ganaxolone [126]. There were 214 subjects in Phase I studies and 498 in Phase II epilepsy and migraine studies. The epilepsy studies involved more than 100 patients and generated data supportive of ganaxolone’s efficacy and safety in the treatment of both children and adults suffering from refractory epilepsy. In a placebo-controlled adult epilepsy trial, adverse events were similar to placebo. Overall, ganaxolone was considered to be well tolerated in clinical trials, and the evidence of efficacy in epilepsy was sufficient for the drug to be advanced to additional clinical testing. However it has never been tested as an anxiolytic. As previously mentioned, ganaxolone is considered a targeted specific treatment of FXS because the GABAA receptors are dramatically down-regulated in FXS together with other proteins involved in the metabolism of GABA. Recent evidence indicates that deficits in GABA-mediated inhibition may underlie many of the key symptoms in FXS, including seizures, anxiety, and autistic behaviors [82,84,93,127]. As GABAA receptors are the major inhibitory receptors in the brain and are specifically involved in processes that are disturbed in fragile X, including neuronal excitability (leading to enhanced seizure susceptibility), anxiety, sleep, and learning, enhancement of the function of GABAA receptors may have major therapeutic benefits for FXS. Currently, there is a phase II proof-of-concept clinical trial of ganaxolone being conducted. This study is a double-blind, randomized, placebo-controlled, crossover trial to investigate ganaxolone treatment in children and adolescents with FXS (ClinicalTrials.gov Identifier: NCT01725152). Sixty subjects (aged 6-17 years) are being randomized to receive either ganaxolone or placebo for six weeks and then crossed over to the opposite treatment for another six weeks. The aims of the study are to assess the safety, tolerability, and efficacy of ganaxolone for treatment of anxiety and attention in individuals with FXS. The hypothesis is that ganaxolone treatment compared to placebo will improve anxiety and attention as measured by the several neuropsychological and psychometric tests. The ganaxolone oral suspension dose is titrated from 3 mg/kg up to 12 mg/kg, with maximum of 1500 mg/day; the placebo and ganaxolone are given in three divided doses daily. The primary outcome measure used is the Clinical Global Impression- Improvement scale (CGI-I). Secondary outcome measures include Pediatric Anxiety Rating Scale (PARS), Visual Analog Scale, Aberrant Behavior Checklist and the Swanson, Nolan, and Pelham-IV questionnaire (SNAP-IV). Other outcome measures are the KiTAP-Test of Attentional Performance for Children, social gaze (eye tracking) and event-related brain potentials (ERP). The study was started in 2012 and it will be completed in 2015.
Riluzole
Riluzole is hypothesized to have an inhibitory effect on glutamate release, block excitotoxic effects of glutamate, and potentiate postsynaptic GABAA receptor function. The pleiotropic effects of riluzole may antagonize common mechanisms underlying chronic cerebellar ataxia, a debilitating and untreatable consequence of various diseases including FXTAS. In a randomized, double-blind, placebo-controlled pilot trial, 40 patients presenting with cerebellar ataxias of different etiologies (one of them with FXTAS) were randomly assigned to riluzole (100 mg/day) or placebo for 8 weeks. The number of patients with a 5-point drop in the International Cooperative Ataxia Rating Scale (ICARS) was significantly higher in the riluzole group than in the placebo group after 4 weeks. The participant with FXTAS showed the aforementioned improvement [128]. In a second trial, riluzole was associated with clinical response in 1 of 6 adult subjects with FXS (17%), in a six-week open- label prospective pilot study (100 mg/day). Significant improvement was only noted on the ADHD Rating Scale-IV (which became non-significant when corrected for multiple comparisons). Overall, riluzole use was not associated with significant clinical improvement in FXS [129].
CONCLUDING REMARKS
Even though many targeted clinical trials have been completed, the treatment of children with FXS continues to require special education, speech and language therapy, behavioral intervention, sensory integration, occupational therapy, and physical therapy. In addition, a variety of medications other than targeted treatments such as stimulants, guanfacine, aripiprazole, rispiradone and selective serotonin reuptake inhibitors (SSRIs) are also required [130]. In regards to FXTAS there is currently no disease-modifying intervention although, several medications can help with the symptomatic treatment [55, 131, 132]. There are many challenges at many levels in the discovery of efficacious medications that target specific protein deficits related to genetic syndromes such as FMRP in FXS.
FMRP has many functions, although it mainly acts as a translational repressor; FMRP can also enhance translation, transport and stabilize mRNAs among other functions. FMRP is thought to enhance GABA-receptors expression, so that its absence will lower GABA receptors. Neurobiological studies have demonstrated GABA deficits in both those with FXS and premutation disorders including FXTAS. The advances in the understanding of the GABA system and neurobiology in FXS is now leading to a new wave of clinical trials, and the experience of the previous trials has help researchers and physician scientists to make modifications in the design of the trials that may potentially identify specific benefits in the target population. The criteria for inclusion and outcome measures in clinical trials, in particular, are areas of active research.
The GABAergic compounds described here, particularly the neurosteroids, will hopefully demonstrate safety and efficacy in the studies of FXSD including both those with FXS and premutation disorders. Both allopregnanolone and ganaxolone hold promise to treat FXSDs, and studies with gaboxidol are planned. It is important to remark that other agents that indirectly influence the GABA deficits in FXS include the IGF-1 analogue, which is currently undergoing multicenter trials in FXS [133, 134], and cannabinoids [135], for which trials will begin soon in FXS. It is likely that GABAergic agents will continue to be part of a multimodality treatment for FXSDs for many years to come. The results of these new trials will likely impact further trials for FXSD and other genetic or idiopathic disorders. Further studies are needed to determine the safety and efficacy of GABA-agonist treatments for FXSD. The future looks promising for GABA-agonist compounds for FXSD including FXS and FXTAS.
ACKNOWLEDGEMENTS
We appreciate the support of Patrick Adams for making important edits and formatting this manuscript. This work was supported by grants from the National Fragile X Foundation, NICHD grant HD036071, Department of Defense grant PR101054, the Health and Human Services Administration for Developmental Disabilities grant 90DD0596, NIMH R01 MH094681 to VMC and by the Seaver Foundation; Dr. Lozano is an NIH (GM082773) and Seaver Faculty Scholar.
Footnotes
CONFLICT OF INTEREST
Dr. Hagerman has received funding from Novartis, Roche, Neuren, and Alcobra for treatment trials in fragile X syndrome. She has also consulted with Roche/Genentech, Alcobra, and Novartis regarding treatment trials in fragile X syndrome.
REFERENCES
- [1].Farzin F, Perry H, Hessl D, et al. Autism spectrum disorders and attention-deficit/hyperactivity disorder in boys with the fragile X premutation. J Develop Behav Pediatr. 2006;27(2 Suppl):S137–44. doi: 10.1097/00004703-200604002-00012. [DOI] [PubMed] [Google Scholar]
- [2].Chonchaiya W, Au J, Schneider A, et al. Increased prevalence of seizures in boys who were probands with the FMR1 premutation and co-morbid autism spectrum disorder. Hum Genet. 2012;131(4):581–9. doi: 10.1007/s00439-011-1106-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Lozano R, Rosero CA, Hagerman RJ. Fragile X spectrum disorders. Intractable Rare Dis Res. 2014;3(4):134–46. doi: 10.5582/irdr.2014.01022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Liu Y, Winarni TI, Zhang L, Tassone F, Hagerman RJ. Fragile X-associated tremor/ataxia syndrome (FXTAS) in grey zone carriers. Clin Genet. 2013;84(1):74–7. doi: 10.1111/cge.12026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Hall D, Tassone F, Klepitskaya O, Leehey M. Fragile X-associated tremor ataxia syndrome in FMR1 gray zone allele carriers. Mov Disord. 2012;27(2):296–300. doi: 10.1002/mds.24021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Loesch DZ, Sherwell S, Kinsella G, et al. Fragile X-associated tremor/ataxia phenotype in a male carrier of unmethylated full mutation in the FMR1 gene. Clin Genet. 2012;82(1):88–92. doi: 10.1111/j.1399-0004.2011.01675.x. [DOI] [PubMed] [Google Scholar]
- [7].Santa Maria L, Pugin A, Alliende MA, et al. FXTAS in an unmethylated mosaic male with fragile X syndrome from Chile. Clin Genet. 2014;86(4):378–82. doi: 10.1111/cge.12278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Pretto DI, Hunsaker MR, Cunningham CL, Greco CM, Hagerman RJ, Noctor SC, et al. Intranuclear inclusions in a fragile X mosaic male. Transl Neurodegener. 2013;2(1):10. doi: 10.1186/2047-9158-2-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Lubs HA. A marker X chromosome. Am J Hum Genet. 1969;21(3):231–44. [PMC free article] [PubMed] [Google Scholar]
- [10].Hatton DD, Sideris J, Skinner M, et al. Autistic behavior in children with fragile X syndrome: Prevalence, stability, and the impact of FMRP. Am J Med Genet A. 2006;140(17):1804–13. doi: 10.1002/ajmg.a.31286. [DOI] [PubMed] [Google Scholar]
- [11].Harris SW, Hessl D, Goodlin-Jones B, et al. Autism profiles of males with fragile X syndrome. Am J Ment Retard. 2008;113(6):427–38. doi: 10.1352/2008.113:427-438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Davids JR, Hagerman RJ, Eilert RE. Orthopaedic aspects of fragile-X syndrome. J Bone Joint Surg Am. 1990;72(6):889–96. [PubMed] [Google Scholar]
- [13].Pyeritz RE, Stamberg J, Thomas GH, Bell BB, Zahka KG, Bernhardt BA. The marker Xq28 syndrome ("Fragile-X Syndrome") in a retarded man with mitral valve prolapse. Johns Hopkins Med J. 1982;151(5):231–7. [PubMed] [Google Scholar]
- [14].Hagerman RJ. The physical and behavioral phenotype. In: Hager- man RJ, Hagerman PJ, editors. Fragile X syndrome: Diagnosis, treatment, and research. 3rd ed The Johns Hopkins Univer- sity Press; Baltimore: 2002. pp. 3–109. [Google Scholar]
- [15].Berry-Kravis E, Grossman AW, Crnic LS, Greenough WT. Under- standing fragile X syndrome. Curr Paediatr. 2002;12(4):316–24. [Google Scholar]
- [16].Loesch DZ, Huggins RM, Hagerman RJ. Phenotypic variation and FMRP levels in fragile X. Ment Retard Dev Disabil Res Rev. 2004;10(1):31–41. doi: 10.1002/mrdd.20006. [DOI] [PubMed] [Google Scholar]
- [17].Strom CM, Crossley B, Redman JB, et al. Molecular testing for Fragile X Syndrome: lessons learned from 119,232 tests performed in a clinical laboratory. Genet Med. 2007;9(1):46–51. doi: 10.1097/gim.0b013e31802d833c. [DOI] [PubMed] [Google Scholar]
- [18].Tassone F, Hagerman RJ, Chamberlain WD, Hagerman PJ. Transcription of the FMR1 gene in individuals with fragile X syndrome. Am J Med Genet. 2000;97(3):195–203. doi: 10.1002/1096-8628(200023)97:3<195::AID-AJMG1037>3.0.CO;2-R. [DOI] [PubMed] [Google Scholar]
- [19].Tassone F, Longshore J, Zunich J, Steinbach P, Salat U, Taylor AK. Tissue-specific methylation differences in a fragile X premutation carrier. Clin Genet. 1999;55(5):346–51. doi: 10.1034/j.1399-0004.1999.550508.x. [DOI] [PubMed] [Google Scholar]
- [20].Godler DE, Tassone F, Loesch DZ, et al. Methylation of novel markers of fragile X alleles is inversely correlated with FMRP expression and FMR1 activation ratio. Hum Mol Genet. 2010;19(8):1618–32. doi: 10.1093/hmg/ddq037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Tassone F, Hagerman RJ, Ikle DN, et al. FMRP expression as a potential prognostic indicator in fragile X syndrome. Am J Med Genet. 1999;84(3):250–61. [PubMed] [Google Scholar]
- [22].Darnell JC, Klann E. The translation of translational control by FMRP: therapeutic targets for FXS. Nat Neurosci. 2013;16(11):1530–6. doi: 10.1038/nn.3379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Fernandez E, Rajan N, Bagni C. The FMRP regulon: from targets to disease convergence. Front Neurosci. 2013;7:191. doi: 10.3389/fnins.2013.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Bagni C, Greenough WT. From mRNP trafficking to spine dysmorphogenesis: the roots of fragile X syndrome. Nat Rev. 2005;6(5):376–87. doi: 10.1038/nrn1667. [DOI] [PubMed] [Google Scholar]
- [25].Banerjee P, Schoenfeld BP, Bell AJ, et al. Short- and long-term memory are modulated by multiple isoforms of the fragile X mental retardation protein. J Neurosci. 2010;30(19):6782–92. doi: 10.1523/JNEUROSCI.6369-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Niere F, Wilkerson JR, Huber KM. Evidence for a fragile X mental retardation protein-mediated translational switch in metabotropic glutamate receptor-triggered Arc translation and long-term depression. J Neurosci. 2012;32(17):5924–36. doi: 10.1523/JNEUROSCI.4650-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Sharma A, Hoeffer CA, Takayasu Y, et al. Dysregulation of mTOR signaling in fragile X syndrome. J Neurosci. 2010;30(2):694–702. doi: 10.1523/JNEUROSCI.3696-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Bear MF, Huber KM, Warren ST. The mGluR theory of fragile X mental retardation. Trends Neurosci. 2004;27(7):370–77. doi: 10.1016/j.tins.2004.04.009. [DOI] [PubMed] [Google Scholar]
- [29].Wang L, Berry-Kravis E, Wang P, Rathmell B, Carpenter R, Bear M. Arbaclofen treatment is associated with global behavioral improvement in fragile X syndrome (FXS): results of a randomized, controlled phase II trial. J Dev Behav Pediatr. in press. [Google Scholar]
- [30].Lozano R, Hare EB, Hagerman RJ. Modulation of the GABAergic pathway for the treatment of fragile X syndrome. Neuropsychiatr Dis Treat. 2014;10:1769–79. doi: 10.2147/NDT.S42919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Greiss-Hess L, Hagerman RJ. Sertraline Treatment for Young Children with FXS - Preliminary Findings. 14th International Fragile X Conference; Anaheim, CA. [Google Scholar]
- [32].Hare EB, Hagerman RJ, Lozano R. Targeted treatments in fragile X syndrome. Expert Opin Orphan Drugs. 2014;2(6):531–43. [Google Scholar]
- [33].Leigh MJ, Nguyen DV, Mu Y, et al. A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile X syndrome. J Develop Behav Pediatr. 2013;34(3):147–55. doi: 10.1097/DBP.0b013e318287cd17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Scharf SH, Jaeschke G, Wettstein JG, Lindemann L. Metabotropic glutamate receptor 5 as drug target for Fragile X syndrome. Curr Opin Pharmacol. 2015;20:124–34. doi: 10.1016/j.coph.2014.11.004. [DOI] [PubMed] [Google Scholar]
- [35].Jacquemont S, Berry-Kravis E, Hagerman R, et al. The challenges of clinical trials in fragile X syndrome. Psychopharmacology (Berl) 2014;231(6):1237–50. doi: 10.1007/s00213-013-3289-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].D'Hulst C, Kooy RF. The GABAA receptor: a novel target for treatment of fragile X? Trends Neurosci. 2007;30(8):425–31. doi: 10.1016/j.tins.2007.06.003. [DOI] [PubMed] [Google Scholar]
- [37].Olmos-Serrano JL, Paluszkiewicz SM, Martin BS, Kaufmann WE, Corbin JG, Huntsman MM. Defective GABAergic neurotransmission and pharmacological rescue of neuronal hyperexcitability in the amygdala in a mouse model of fragile X syndrome. J Neurosci. 2010;30(29):9929–38. doi: 10.1523/JNEUROSCI.1714-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Paluszkiewicz SM, Martin BS, Huntsman MM. Fragile X syndrome: the GABAergic system and circuit dysfunction. Dev Neu- rosci. 2011;33(5):349–64. doi: 10.1159/000329420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Gersak K, Meden-Vrtovec H, Peterlin B. Fragile X premutation in women with sporadic premature ovarian failure in Slovenia. Hum Reprod. 2003;18(8):1637–40. doi: 10.1093/humrep/deg327. [DOI] [PubMed] [Google Scholar]
- [40].Cronister A, Schreiner R, Wittenberger M, Amiri K, Harris K, Hagerman RJ. Heterozygous fragile X female: historical, physical, cognitive, and cytogenetic features. Am J Med Genet. 1991;38(2- 3):269–74. doi: 10.1002/ajmg.1320380221. [DOI] [PubMed] [Google Scholar]
- [41].Hunter JE, Epstein MP, Tinker SW, Charen KH, Sherman SL. Fragile X-associated primary ovarian insufficiency: evidence for additional genetic contributions to severity. Genet Epidemiol. 2008;32(6):553–9. doi: 10.1002/gepi.20329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Wheeler AC, Bailey DB, Berry-Kravis E, et al. Associated features in females with an FMR1 premutation. J Neurodev Disord. 2014;6 doi: 10.1186/1866-1955-6-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Hagerman RJ, Leehey M, Heinrichs W, et al. Intention tremor, parkinsonism, and generalized brain atrophy in male carriers of fragile X. Neurology. 2001;57(1):127–30. doi: 10.1212/wnl.57.1.127. [DOI] [PubMed] [Google Scholar]
- [44].Jacquemont S, Farzin F, Hall D, et al. Aging in individuals with the FMR1 mutation. Am J Ment Retard. 2004;109(2):154–64. doi: 10.1352/0895-8017(2004)109<154:AIIWTF>2.0.CO;2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Jacquemont S, Hagerman RJ, Hagerman PJ, Leehey MA. Fragile-X syndrome and fragile X-associated tremor/ataxia syndrome: two faces of FMR1. Lancet Neurol. 2007;6(1):45–55. doi: 10.1016/S1474-4422(06)70676-7. [DOI] [PubMed] [Google Scholar]
- [46].Berry-Kravis E, Abrams L, Coffey SM, et al. Fragile X-associated tremor/ataxia syndrome: clinical features, genetics, and testing guidelines. Mov Disord. 2007;22(14):2018–30. doi: 10.1002/mds.21493. [DOI] [PubMed] [Google Scholar]
- [47].Jacquemont S, Hagerman RJ, Leehey M, et al. Fragile X premutation tremor/ataxia syndrome: molecular, clinical, and neuroimaging correlates. Am J Hum Genet. 2003;72(4):869–78. doi: 10.1086/374321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Tassone F, Berry-Kravis EM, editors. The Fragile X-Associated Tremor Ataxia Syndrome (FXTAS) Springer Sci- ence+Business Media; New York, NY: 2010. [Google Scholar]
- [49].Hagerman RJ, Coffey SM, Maselli R, et al. Neuropathy as a presenting feature in fragile X-associated tremor/ataxia syndrome. Am J Med Genet A. 2007;143(19):2256–60. doi: 10.1002/ajmg.a.31920. [DOI] [PubMed] [Google Scholar]
- [50].Bourgeois JA, Coffey SM, Rivera SM, et al. A review of fragile X premutation disorders: expanding the psychiatric perspective. J Clin Psychiatry. 2009;70(6):852–62. doi: 10.4088/JCP.08m04476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Bourgeois JA, Seritan AL, Casillas EM, et al. Lifetime prevalence of mood and anxiety disorders in fragile X premutation carriers. J Clin Psychiatry. 2011;72(2):175–82. doi: 10.4088/JCP.09m05407blu. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Coffey SM, Cook K, Tartaglia N, et al. Expanded clinical phenotype of women with the FMR1 premutation. Am J Med Genet A. 2008;146A(8):1009–16. doi: 10.1002/ajmg.a.32060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Soontarapornchai K, Maselli R, Fenton-Farrell G, et al. Abnormal nerve conduction features in fragile X premutation carriers. Arch Neurol. 2008;65(4):495–8. doi: 10.1001/archneur.65.4.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Grigsby J, Brega AG, Engle K, et al. Cognitive profile of fragile X premutation carriers with and without fragile X-associated tremor/ataxia syndrome. Neuropsychology. 2008;22(1):48–60. doi: 10.1037/0894-4105.22.1.48. [DOI] [PubMed] [Google Scholar]
- [55].Hagerman R, Hagerman P. Advances in clinical and molecular understanding of the FMR1 premutation and fragile X-associated tremor/ataxia syndrome. Lancet Neurol. 2013;12(8):786–98. doi: 10.1016/S1474-4422(13)70125-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Loesch DZ, Cook M, Litewka L, et al. A low symptomatic form of neurodegeneration in younger carriers of the FMR1 premutation, manifesting typical radiological changes. J Med Genet. 2008;45(3):179–81. doi: 10.1136/jmg.2007.054171. [DOI] [PubMed] [Google Scholar]
- [57].Cohen S, Masyn K, Adams J, et al. Molecular and imaging correlates of the fragile X-associated tremor/ataxia syndrome. Neurology. 2006;67(8):1426–31. doi: 10.1212/01.wnl.0000239837.57475.3a. [DOI] [PubMed] [Google Scholar]
- [58].Brunberg JA, Jacquemont S, Hagerman RJ, et al. Fragile X premutation carriers: characteristic MR imaging findings of adult male patients with progressive cerebellar and cognitive dysfunction. AJNR Am J Neuroradiol. 2002;23(10):1757–66. [PMC free article] [PubMed] [Google Scholar]
- [59].Hashimoto R, Javan AK, Tassone F, Hagerman RJ, Rivera SM. A voxel-based morphometry study of grey matter loss in fragile X- associated tremor/ataxia syndrome. Brain. 2011;134(Pt 3):863–78. doi: 10.1093/brain/awq368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Wang JY, Hessl D, Schneider A, Tassone F, Hagerman RJ, Rivera SM. Fragile X-associated tremor/ataxia syndrome: influence of the FMR1 gene on motor fiber tracts in males with normal and premutation alleles. JAMA Neurol. 2013;70(8):1022–9. doi: 10.1001/jamaneurol.2013.2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Wang JY, Hagerman RJ, Rivera SM. A multimodal imaging analysis of subcortical gray matter in fragile X premutation carriers. Mov Disord. 2013;28(9):1278–84. doi: 10.1002/mds.25473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Hashimoto R, Srivastava S, Tassone F, Hagerman RJ, Rivera SM. Diffusion tensor imaging in male premutation carriers of the fragile X mental retardation gene. Mov Disord. 2011;26(7):1329–36. doi: 10.1002/mds.23646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Adams JS, Adams PE, Nguyen D, et al. Volumetric brain changes in females with fragile X-associated tremor/ataxia syndrome (FXTAS) Neurology. 2007;69(9):851–9. doi: 10.1212/01.wnl.0000269781.10417.7b. [DOI] [PubMed] [Google Scholar]
- [64].Adams PE, Adams JS, Nguyen DV, et al. Psychological symptoms correlate with reduced hippocampal volume in fragile X premutation carriers. Am J Med Genet B Neuropsychiatr Genet. 2010;153B(3):775–85. doi: 10.1002/ajmg.b.31046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Ariza J, Steward C, Rueckert F, et al. Dysregulated iron metabolism in the choroid plexus in fragile X-associated tremor/ataxia syndrome. Brain Res. 2015;1598:88–96. doi: 10.1016/j.brainres.2014.11.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Hunsaker MR, Greco CM, Spath MA, et al. Widespread non-central nervous system organ pathology in fragile X premutation carriers with fragile X-associated tremor/ataxia syndrome and CGG knock-in mice. Acta Neuropathol. 2011;122(4):467–79. doi: 10.1007/s00401-011-0860-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Greco CM, Berman RF, Martin RM, et al. Neuropathology of fragile X-associated tremor/ataxia syndrome (FXTAS) Brain. 2006;129(Pt 1):243–55. doi: 10.1093/brain/awh683. [DOI] [PubMed] [Google Scholar]
- [68].Greco CM, Soontrapornchai K, Wirojanan J, Gould JE, Hagerman PJ, Hagerman RJ. Testicular and pituitary inclusion formation in fragile X associated tremor/ataxia syndrome. J Urol. 2007;177(4):1434–7. doi: 10.1016/j.juro.2006.11.097. [DOI] [PubMed] [Google Scholar]
- [69].Muzar Z, Lozano R, Schneider A, et al. Methadone use in a male with the FMRI premutation and FXTAS. Am J Med Genet A. 2015;167(6):1354–9. doi: 10.1002/ajmg.a.37030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Muzar Z, Adams PE, Schneider A, Hagerman RJ, Lozano R. Addictive substances may induce a rapid neurological deterioration in fragile X-associated tremor ataxia syndrome: A report of two cases. Intractable Rare Dis Res. 2014;3(4):162–5. doi: 10.5582/irdr.2014.01023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Martinez-Cerdeno V, Lechpammer M, Lott A, Schneider A, Hagerman R. Fragile X-associated tremor/ataxia syndrome in a 36 year old man. JAMA Neurol. doi: 10.1001/jamaneurol.2015.1138. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Hagerman PJ, Hagerman RJ. Fragile X-associated tremor/ataxia syndrome. Ann N Y Acad Sci. 2015;1338:58–70. doi: 10.1111/nyas.12693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Filley CM, Brown MS, Onderko K, et al. White matter disease and cognitive impairment in FMR1 premutation carriers. Neurology. 2015 doi: 10.1212/WNL.0000000000001612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Cao Z, Hulsizer S, Tassone F, et al. Clustered burst firing in FMR1 premutation hippocampal neurons: amelioration with allopregnanolone. Hum Mol Genet. 2012;21(13):2923–35. doi: 10.1093/hmg/dds118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Soghomonian JJ, Martin DL. Two isoforms of glutamate decarboxylase: why? Trends Pharmacol Sci. 1998;19(12):500–5. doi: 10.1016/s0165-6147(98)01270-x. [DOI] [PubMed] [Google Scholar]
- [76].Macdonald RL, Olsen RW. GABAA receptor channels. Ann Rev Neurosci. 1994;17:569–602. doi: 10.1146/annurev.ne.17.030194.003033. [DOI] [PubMed] [Google Scholar]
- [77].Blednov YA, Benavidez JM, Black M, et al. GABAA receptors containing rho1 subunits contribute to in vivo effects of ethanol in mice. PLoS One. 2014;9(1):e85525. doi: 10.1371/journal.pone.0085525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Couve A, Moss SJ, Pangalos MN. GABAB receptors: a new paradigm in G protein signaling. Mol Cell Neurosci. 2000;16(4):296–312. doi: 10.1006/mcne.2000.0908. [DOI] [PubMed] [Google Scholar]
- [79].Gruss M, Braun K. Alterations of amino acids and monoamine metabolism in male Fmr1 knockout mice: a putative animal model of the human fragile X mental retardation syndrome. Neural Plast. 2001;8(4):285–98. doi: 10.1155/NP.2001.285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Davidovic L, Navratil V, Bonaccorso CM, Catania MV, Bardoni B, Dumas ME. A metabolomic and systems biology perspective on the brain of the fragile X syndrome mouse model. Genome Res. 2011;21(12):2190–202. doi: 10.1101/gr.116764.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Selby L, Zhang C, Sun QQ. Major defects in neocortical GABAergic inhibitory circuits in mice lacking the fragile X mental retardation protein. Neurosci Lett. 2007;412(3):227–32. doi: 10.1016/j.neulet.2006.11.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].D'Hulst C, Heulens I, Brouwer JR, et al. Expression of the GABAergic system in animal models for fragile X syndrome and fragile X associated tremor/ataxia syndrome (FXTAS) Brain Res. 2009;1253:176–83. doi: 10.1016/j.brainres.2008.11.075. [DOI] [PubMed] [Google Scholar]
- [83].Gantois I, Vandesompele J, Speleman F, et al. Expression profiling suggests underexpression of the GABA(A) receptor subunit delta in the fragile X knockout mouse model. Neurobiology of disease. 2006;21(2):346–57. doi: 10.1016/j.nbd.2005.07.017. [DOI] [PubMed] [Google Scholar]
- [84].D'Hulst C, De Geest N, Reeve SP, et al. Decreased expression of the GABAA receptor in fragile X syndrome. Brain Res. 2006;1121(1):238–45. doi: 10.1016/j.brainres.2006.08.115. [DOI] [PubMed] [Google Scholar]
- [85].Curia G, Papouin T, Seguela P, Avoli M. Downregulation of tonic GABAergic inhibition in a mouse model of fragile X syndrome. Cereb Cortex. 2009;19(7):1515–20. doi: 10.1093/cercor/bhn159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Hong A, Zhang A, Ke Y, El Idrissi A, Shen CH. Downregulation of GABA(A) beta subunits is transcriptionally controlled by Fmr1p. J Mol Neurosci. 2012;46(2):272–5. doi: 10.1007/s12031-011-9531-5. [DOI] [PubMed] [Google Scholar]
- [87].El Idrissi A, Ding XH, Scalia J, Trenkner E, Brown WT, Dobkin C. Decreased GABA(A) receptor expression in the seizure-prone fragile X mouse. Neurosci Lett. 2005;377(3):141–6. doi: 10.1016/j.neulet.2004.11.087. [DOI] [PubMed] [Google Scholar]
- [88].Adusei DC, Pacey LK, Chen D, Hampson DR. Early developmental alterations in GABAergic protein expression in fragile X knockout mice. Neuropharmacology. 2010;59(3):167–71. doi: 10.1016/j.neuropharm.2010.05.002. [DOI] [PubMed] [Google Scholar]
- [89].Kratovac S, Corbin JG. Developmental changes in expression of inhibitory neuronal proteins in the Fragile X Syndrome mouse basolateral amygdala. Brain Res. 2013;1537:69–78. doi: 10.1016/j.brainres.2013.08.052. [DOI] [PubMed] [Google Scholar]
- [90].Pacey LK, Tharmalingam S, Hampson DR. Subchronic administration and combination metabotropic glutamate and GABAB receptor drug therapy in fragile X syndrome. J Pharmacol Exp Ther. 2011;338(3):897–905. doi: 10.1124/jpet.111.183327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Wafford KA, Ebert B. Gaboxadol--a new awakening in sleep. Curr Opin Pharmacol. 2006;6(1):30–6. doi: 10.1016/j.coph.2005.10.004. [DOI] [PubMed] [Google Scholar]
- [92].Olmos-Serrano JL, Corbin JG, Burns MP. The GABA(A) receptor agonist THIP ameliorates specific behavioral deficits in the mouse model of fragile X syndrome. Dev Neurosci. 2011;33(5):395–403. doi: 10.1159/000332884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Heulens I, D'Hulst C, Van Dam D, De Deyn PP, Kooy RF. Pharmacological treatment of fragile X syndrome with GABAergic drugs in a knockout mouse model. Behav Brain Res. 2012;229(1):244–9. doi: 10.1016/j.bbr.2012.01.031. [DOI] [PubMed] [Google Scholar]
- [94].Qin M, Huang T, Kader M, et al. R-Baclofen Reverses a Social Behavior Deficit and Elevated Protein Synthesis in a Mouse Model of Fragile X Syndrome. Int J Neuropsychopharmacol. 2015 doi: 10.1093/ijnp/pyv034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Henderson C, Wijetunge L, Kinoshita MN, et al. Reversal of disease-related pathologies in the fragile X mouse model by selective activation of GABAB receptors with arbaclofen. Sci Transl Med. 2012;4(152):152ra28. doi: 10.1126/scitranslmed.3004218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Kokate TG, Svensson BE, Rogawski MA. Anticonvulsant activity of neurosteroids: correlation with gamma-aminobutyric acid-evoked chloride current potentiation. J Pharmacol Exp Ther. 1994;270(3):1223–9. [PubMed] [Google Scholar]
- [97].Beekman M, Ungard JT, Gasior M, et al. Reversal of behavioral effects of pentylenetetrazol by the neuroactive steroid ganaxolone. J Pharmacol Exp Ther. 1998;284(3):868–77. [PubMed] [Google Scholar]
- [98].Carter RB, Wood PL, Wieland S, et al. Characterization of the anticonvulsant properties of ganaxolone (CCD 1042; 3alpha- hydroxy-3beta-methyl-5alpha-pregnan-20-one), a selective, high- affinity, steroid modulator of the gamma-aminobutyric acid(A) re- ceptor. J Pharmacol Exp Ther. 1997;280(3):1284–95. [PubMed] [Google Scholar]
- [99].Mann K, Kiefer F, Spanagel R, Littleton J. Acamprosate: recent findings and future research directions. Alcohol Clin Exp Res. 2008;32(7):1105–10. doi: 10.1111/j.1530-0277.2008.00690.x. [DOI] [PubMed] [Google Scholar]
- [100].Pierrefiche O, Daoust M, Naassila M. Biphasic effect of acamprosate on NMDA but not on GABAA receptors in spontaneous rhythmic activity from the isolated neonatal rat respiratory net- work. Neuropharmacology. 2004;47(1):35–45. doi: 10.1016/j.neuropharm.2004.03.004. [DOI] [PubMed] [Google Scholar]
- [101].Erickson CA, Mullett JE, McDougle CJ. Brief report: acamprosate in fragile X syndrome. J Autism Dev Disord. 2010;40(11):1412–6. doi: 10.1007/s10803-010-0988-9. [DOI] [PubMed] [Google Scholar]
- [102].Erickson CA, Wink LK, Ray B, et al. Impact of acamprosate on behavior and brain-derived neurotrophic factor: an open-label study in youth with fragile X syndrome. Psychopharmacology (Berl) 2013;228(1):75–84. doi: 10.1007/s00213-013-3022-z. [DOI] [PubMed] [Google Scholar]
- [103].Berry-Kravis EM, Hessl D, Rathmell B, et al. Effects of STX209 (arbaclofen) on neurobehavioral function in children and adults with fragile X syndrome: a randomized, controlled, phase 2 trial. Sci Transl Med. 2012;4(152):152ra27. doi: 10.1126/scitranslmed.3004214. [DOI] [PubMed] [Google Scholar]
- [104].Erickson CA, Veenstra-Vanderweele JM, Melmed RD, et al. STX209 (arbaclofen) for autism spectrum disorders: an 8-week open-label study. J Autism Dev Disord. 2014;44(4):958–64. doi: 10.1007/s10803-013-1963-z. [DOI] [PubMed] [Google Scholar]
- [105].Veenstra-Vanderweele JM, Sikich L, Melmed RD, et al. Randomized, controlled, phase 2 trial of STX209 for social function in ASD. International Meeting for Autism Research (IMFAR); San Sebastian, Spain. May 1-4, 2013. [Google Scholar]
- [106].Berry-Kravis E. Progress and hurdles in development of targeted treatments for fragile X syndrome. International Meeting for Autism Research (IMFAR); Salt Lake City, UT. May 13-16, 2015. [Google Scholar]
- [107].Brinton RD. Neurosteroids as regenerative agents in the brain: therapeutic implications. Nat Rev Endocrinol. 2013;9(4):241–50. doi: 10.1038/nrendo.2013.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Singh C, Liu L, Wang JM, et al. Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice. Neurobiol Aging. 2012;33(8):1493–506. doi: 10.1016/j.neurobiolaging.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Djebaili M, Hoffman SW, Stein DG. Allopregnanolone and progesterone decrease cell death and cognitive deficits after a contusion of the rat prefrontal cortex. Neuroscience. 2004;123(2):349–59. doi: 10.1016/j.neuroscience.2003.09.023. [DOI] [PubMed] [Google Scholar]
- [110].Djebaili M, Guo Q, Pettus EH, Hoffman SW, Stein DG. The neurosteroids progesterone and allopregnanolone reduce cell death, gliosis, and functional deficits after traumatic brain injury in rats. J Neurotrauma. 2005;22(1):106–18. doi: 10.1089/neu.2005.22.106. [DOI] [PubMed] [Google Scholar]
- [111].Sayeed I, Parvez S, Wali B, Siemen D, Stein DG. Direct inhibition of the mitochondrial permeability transition pore: a possible mechanism for better neuroprotective effects of allopregnanolone over progesterone. Brain Res. 2009;1263:165–73. doi: 10.1016/j.brainres.2009.01.045. [DOI] [PubMed] [Google Scholar]
- [112].Palop JJ, Chin J, Roberson ED, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer's disease. Neuron. 2007;55(5):697–711. doi: 10.1016/j.neuron.2007.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Tassone F, Greco CM, Hunsaker MR, et al. Neuropathological, clinical and molecular pathology in female fragile X premutation carriers with and without FXTAS. Genes Brain Behav. 2012;11(5):577–85. doi: 10.1111/j.1601-183X.2012.00779.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Wang JM, Johnston PB, Ball BG, Brinton RD. The neurosteroid allopregnanolone promotes proliferation of rodent and human neural progenitor cells and regulates cell-cycle gene and protein expression. J Neurosci. 2005;25(19):4706–18. doi: 10.1523/JNEUROSCI.4520-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Wang JM, Singh C, Liu L, et al. Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer's disease. Proc Natl Acad Sci USA. 2010;107(14):6498–503. doi: 10.1073/pnas.1001422107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Chen S, Wang JM, Irwin RW, Yao J, Liu L, Brinton RD. Allopregnanolone Promotes Regeneration and Reduces beta-Amyloid Bur- den in a Preclinical Model of Alzheimer's Disease. PLoS One. 2011;6(8):e24293. doi: 10.1371/journal.pone.0024293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Seritan AL, Nguyen DV, Mu Y, et al. Memantine for fragile X-Associated Tremor/Ataxia Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. J Clin Psychiatry. 2014;75(3):264–71. doi: 10.4088/JCP.13m08546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Reddy DS. Neurosteroids: endogenous role in the human brain and therapeutic potentials. Prog Brain Res. 2010;186:113–37. doi: 10.1016/B978-0-444-53630-3.00008-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Monaghan EP, McAuley JW, Data JL. Ganaxolone: a novel positive allosteric modulator of the GABA(A) receptor complex for the treatment of epilepsy. Expert Opin Investig Drugs. 1999;8(10):1663–71. doi: 10.1517/13543784.8.10.1663. [DOI] [PubMed] [Google Scholar]
- [120].Reddy DS, O'Malley BW, Rogawski MA. Anxiolytic activity of progesterone in progesterone receptor knockout mice. Neuropharmacology. 2005;48(1):14–24. doi: 10.1016/j.neuropharm.2004.09.002. [DOI] [PubMed] [Google Scholar]
- [121].Reddy DS. Pharmacology of endogenous neuroactive steroids. Crit Rev Neurobiol. 2003;15(3-4):197–234. doi: 10.1615/critrevneurobiol.v15.i34.20. [DOI] [PubMed] [Google Scholar]
- [122].Kerrigan JF, Shields WD, Nelson TY, et al. Ganaxolone for treating intractable infantile spasms: a multicenter, open-label, add- on trial. Epilepsy Res. 2000;42(2-3):133–9. doi: 10.1016/s0920-1211(00)00170-4. [DOI] [PubMed] [Google Scholar]
- [123].Laxer K, Blum D, Abou-Khalil BW, et al. Assessment of ganaxolone's anticonvulsant activity using a randomized, double-blind, presurgical trial design. Ganaxolone Presurgical Study Group. Epilepsia. 2000;41(9):1187–94. doi: 10.1111/j.1528-1157.2000.tb00324.x. [DOI] [PubMed] [Google Scholar]
- [124].Monaghan EP, Navalta LA, Shum L, Ashbrook DW, Lee DA. Initial human experience with ganaxolone, a neuroactive steroid with antiepileptic activity. Epilepsia. 1997;38(9):1026–31. doi: 10.1111/j.1528-1157.1997.tb01486.x. [DOI] [PubMed] [Google Scholar]
- [125].Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS. Progress report on new antiepileptic drugs: a summary of the Tenth Eilat Conference (EILAT X) Epilepsy Res. 2010;92(2-3):89–124. doi: 10.1016/j.eplepsyres.2010.09.001. [DOI] [PubMed] [Google Scholar]
- [126].Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Perucca E, Tomson T. Progress report on new antiepileptic drugs: a summary of the Eighth Eilat Conference (EILAT VIII) Epilepsy Res. 2007;73(1):1–52. doi: 10.1016/j.eplepsyres.2006.10.008. [DOI] [PubMed] [Google Scholar]
- [127].Kooy F, Heulens I, D'Hulst C, et al. The GABAA receptor as a potential target for therapy of the fragile X syndrome [abstract]. NFXF 12th International FX Conference; Detroit, MI. July 21 - 25. [Google Scholar]
- [128].Ristori G, Romano S, Visconti A, et al. Riluzole in cerebellar ataxia: a randomized, double-blind, placebo-controlled pilot trial. Neurology. 2010;74(10):839–45. doi: 10.1212/WNL.0b013e3181d31e23. [DOI] [PubMed] [Google Scholar]
- [129].Erickson CA, Weng N, Weiler IJ, et al. Open-label riluzole in fragile X syndrome. Brain Res. 2011;1380:264–70. doi: 10.1016/j.brainres.2010.10.108. [DOI] [PubMed] [Google Scholar]
- [130].Hagerman RJ, Berry-Kravis E, Kaufmann WE, et al. Advances in the treatment of fragile X syndrome. Pediatrics. 2009;123(1):378–90. doi: 10.1542/peds.2008-0317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Leehey MA, Berry-Kravis E, Goetz CG, Hagerman R. Clinical Neurological Phenotype of FXTAS. In: Tassone F, Berry-Kravis E, editors. The Fragile X-Associated Tremor Ataxia Syndrome (FXTAS) Springer; New York: 2011. pp. 1–16. [Google Scholar]
- [132].Polussa J, Schneider A, Hagerman R. Molecular Advances Leading to Treatment Implications for Fragile X Premutation Carriers. Brain Disord Ther. 2014;3 doi: 10.4172/2168-975X.1000119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Berry-Kravis E. Mechanism-based treatments in neurodevelopmental disorders: fragile X syndrome. Pediatr Neurol. 2014;50(4):297–302. doi: 10.1016/j.pediatrneurol.2013.12.001. [DOI] [PubMed] [Google Scholar]
- [134].Gross C, Hoffmann A, Bassell GJ, Berry-Kravis EM. Therapeutic Strategies in Fragile X Syndrome: From Bench to Bedside and Back. Neurotherapeutics. 2015 doi: 10.1007/s13311-015-0355-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Fernandez-Ruiz J, Sagredo O, Pazos MR, et al. Cannabidiol for neurodegenerative disorders: important new clinical applications for this phytocannabinoid? Br J Clin Pharmacol. 2013;75(2):323–33. doi: 10.1111/j.1365-2125.2012.04341.x. [DOI] [PMC free article] [PubMed] [Google Scholar]