Skip to main content
Open Biology logoLink to Open Biology
. 2019 Jun 12;9(6):180265. doi: 10.1098/rsob.180265

Understanding intellectual disability and autism spectrum disorders from common mouse models: synapses to behaviour

Vijaya Verma 1,, Abhik Paul 1,, Anjali Amrapali Vishwanath 1,, Bhupesh Vaidya 1,, James P Clement 1,
PMCID: PMC6597757  PMID: 31185809

Abstract

Normal brain development is highly dependent on the timely coordinated actions of genetic and environmental processes, and an aberration can lead to neurodevelopmental disorders (NDDs). Intellectual disability (ID) and autism spectrum disorders (ASDs) are a group of co-occurring NDDs that affect between 3% and 5% of the world population, thus presenting a great challenge to society. This problem calls for the need to understand the pathobiology of these disorders and to design new therapeutic strategies. One approach towards this has been the development of multiple analogous mouse models. This review discusses studies conducted in the mouse models of five major monogenic causes of ID and ASDs: Fmr1, Syngap1, Mecp2, Shank2/3 and Neuroligins/Neurnexins. These studies reveal that, despite having a diverse molecular origin, the effects of these mutations converge onto similar or related aetiological pathways, consequently giving rise to the typical phenotype of cognitive, social and emotional deficits that are characteristic of ID and ASDs. This convergence, therefore, highlights common pathological nodes that can be targeted for therapy. Other than conventional therapeutic strategies such as non-pharmacological corrective methods and symptomatic alleviation, multiple studies in mouse models have successfully proved the possibility of pharmacological and genetic therapy enabling functional recovery.

Keywords: autism spectrum disorders, intellectual disability, SYNGAP1, fragile X mental retardation protein, MECP2, NEUROLIGIN

1. Introduction

The human brain is a complex organ with a wide array of functions. An adult brain has approximately 86 billion neurons and 85 billion non-neuronal cells [1]. Synchronized activity among neuronal and non-neuronal cells enables us to perform from mundane yet straightforward tasks to an overly complicated range of activities. Development of the human brain is a tightly regulated process. Any change can lead to precarious and detrimental developmental deficits such as neurodevelopmental disorders (NDDs). To name a few, NDDs include, but are not limited to, autism spectrum disorder (ASD), intellectual disability (ID) and attention deficit hyperactivity disorder (ADHD), which affect 3–4% of the world's population [2]. On average, NDD is diagnosed when a child is six months to 1 year old in the absence of well-defined biomarkers as the child is not fulfilling developmental milestones. The Diagnostic and Statistical Manual of Mental Disorders (DSM V), published by the American Psychiatric Association in 2013, has suggested the following for the diagnosis of ID: children often present with difficulty in learning and memory, and exhibit deficits in self-care and social behaviour [3].

Studies over the past few decades have shown that there is a strong genetic correlation between specific genes encoding protein synthesis that regulates synaptic function and ASD/ID [4]. Mutations in such genes, along with gene–gene and gene–environmental interactions, are responsible for ASDs and ID. The resulting pathophysiological mechanisms of many of these overlap with those of ID and ASD, such as SYNGAP1, FMRP, MECP2, NEUROLIGINS/NEURONEXINS and SHANK2/3. Apart from the overlapping pathophysiology, patients with ASD and ID have some comorbid conditions which include schizophrenia, allergic disorders, food sensitivity and autoimmune disorders to name a few [58].

While the distribution of specific proteins such as FMRP is widespread, the SYNGAP1 protein is expressed only in the brain, not in any other organ [9,10]. The variation in the tissue, cellular and subcellular localization and expression of these genes implicated in ID and ASDs may be one of the reasons for the type of phenotypes observed in ID/ASD individuals. Genes such as FMRP present with the syndromic type of ID while others such as SYNGAP1, MECP2 and SHANK3 are mostly linked to the non-syndromic type [11,12]. The environmental factors which can lead to ID and ASDs include the use of certain drugs during pregnancy like valproate and alcohol as well as infections, exposure to heavy metals, such as lead and mercury, and malnutrition [1316]. Although a complex interplay of environmental and genetic factors are known to have a role in the pathophysiology of ID/ASDs, most cases are idiopathic [17].

Precise control of synapse formation and development is essential for correct brain development and function. Abnormalities, if any, can lead to various biochemical and behavioural deficits. In this review, we have discussed the convergence of the pathophysiological hallmarks and phenotypic characteristics with emphasis on the changes in the synaptic morphology (figure 1). For instance, alterations in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor/N-methyl-d-aspartate receptor (AMPA/NMDA) ratio, induction and maintenance of long-term potentiation (LTP) and long-term depression (LTD), and changes in the basal synaptic transmission are some of the common characteristics associated with ID and ASDs of different genetic backgrounds, which are discussed in detail in this review [18]. The current report further highlights the importance of the critical period of development and its alteration due to pathogenic mutations [19].

Figure 1.

Figure 1.

Common pathophysiological features observed in genes implicated in ID/ASDs. Diagram illustrating morphological, synaptic and circuit properties of neurons along with behavioural alterations observed in different key transgenic mouse models. As mentioned in the main discussion, although the mutations were observed in different genes that are implicated in ASDs/ID, there are many common features found in these mutations. Therefore, it is imperative to understand the mechanism of these mutations about neuronal function before prescribing therapeutics to patients with any of these mutations. E/I, excitation–inhibition; GABA, gamma-aminobutyric acid; GDP, giant depolarization potential; D1-MSN, D1 receptors in medium spiny neurons (MSN); AMPAR, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; mEPSC, miniature excitatory postsynaptic current; mIPSC; miniature inhibitory postsynaptic current.

Although the importance of the proper function of neurons during a critical period of development is known, the role of non-neuronal cells such as astrocytes has been barely studied, but this has been changing in recent years. It has been observed that the timing of astrocyte maturation coincides well with the formation of excitatory synapses in the brain. Secretion of molecules such as glypicans by astrocytes helps in the conversion of silent to functional synapses, thereby facilitating the insertion of AMPA receptors [20]. Although the expression of synaptic proteins such as FMRP and NEUROLIGIN has been studied in astrocytes, a complete understanding of their underlying roles in ID and ASDs is still under investigation [21]. Here, we give an overview of the importance of astrocytes during the critical period of development, and how alteration in these impacts neuronal function.

To further our understanding of the physiological relevance of mutations implicated in ID and ASDs, the use of transgenic animal models has provided insights into different pathophysiological aspects of ID and ASDs. These transgenic mice models had been validated at various levels to ensure their efficacy in the replication of not only the pathophysiology but also the behavioural phenotypes [22]. Although there are limitations, they have been useful in understanding the novel mechanisms contributing to the progression and development of these disorders and in finding novel therapeutic targets.

As with most neurological disorders, the exact prevalence and epidemiology of NDDs are not entirely known yet. Meta-analysis studies, which make use of statistical procedures to analyse data from already existing reports, have highlighted the procedural limitations and underreporting cases from several parts of the world [23,24]. However, current studies have shown the prevalence to be higher in low- and middle-income countries, which can be attributed to the lack of essential diagnostic and management resources in these geographical locations [23,24]. Considering that NDDs require early diagnosis, an exhaustive study done in children from birth until 12 years of age found the prevalence to be as high as one in six children in the USA. Moreover, in the same study, the incidence was higher among the males than among females [25].

Nevertheless, according to a recent report by the World Health Organization, about one in every 160 individuals has an ASD/ID [26]. The numbers are remarkably alarming as they are only expected to become worse in the absence of any effective therapeutic strategy. With more laboratories now working on NDDs, insights into possible new therapeutic targets and their mechanisms may aid finding mitigation strategies in the future.

Drug repurposing is one of the impending fields; this involves finding a new indication for an already approved drug [27]. Drug development is an expensive and time-consuming process that could be shortened with the help of this approach. In this review, we have highlighted studies done on drugs with a known indication in some other diseases. Targeting them at the preclinical and clinical stages may prove to be a useful strategy in search of new medications for ID and ASDs.

Although there are several unexplored mechanisms associated with ID/ASDs, the involvement of synaptic function and plasticity in ID and ASDs is well characterized [28]. Technological advancement to measure neuronal activity (electrophysiology and deep-brain imaging) and understanding of the mechanisms which modulate synaptic plasticity may aid in further expanding our knowledge to decipher the pathophysiology of ID and ASDs. In this review, we will discuss the implications of monogenic mutations on the physiological, molecular and biochemical, and morphological aspects of neuronal and non-neuronal development using different mouse models studied over the past few decades.

2. Aetiology/causes of intellectual disability

The causes of ID and ASDs are diverse and involve a range of genetic and environmental factors [29]. Although the nature of the cause for about 60% of all known cases of ID and ASDs remains unknown [30,31], studies where the cause is known have demonstrated that aberrations leading to ID/ASDs mainly occur during the developmental time period, but have a lifelong effect, including in adulthood. The vulnerable range of time includes the pre-, peri- and post-natal stages of development [32]. Environmental stress factors such as poor nutrition, hygiene, infection, familial instability and socio-economic causes may affect brain development, contributing to ID/ASDs [33,34]. For example, it is now believed that oxidative stress as a result of environmental stressors such as heavy metals affects sulfur metabolism, which leads to alteration of the epigenetic mechanisms of gene expression. Hence, the complex interplay of genetic and environmental factors has a crucial role in the pathology of ASDs/ID [17].

Other than environmental factors, about a quarter to half of the identified causes of ID are the result of mutations in genes [32]. These mutations can be either inherited or acquired genetic defects due to metabolic genetic defects as observed in cases such as phenylketonuria and Tay–Sachs disease that can disrupt healthy brain development [33]. These genetic changes can occur on various scales based on size (and thereby ease of detection)—the largest being microscopically visible aberrations at the chromosomal level and 15% of all cases of ID were attributed to these chromosomal defects [30,3538]. This review will provide a brief overview of the current understanding of ID/ASDs with regard to the critical period of development, molecular and biochemical signalling, and electrophysiological aberrations concerning monogenic mutations.

3. The critical period of plasticity

A remarkable property of the brain is its capability to undergo changes based on experiences (stimuli) through a process called synaptic plasticity, particularly during the early stages of development [39]. Synaptic plasticity is a biological process in which a stimulus or experience induces synaptic activity that results in changes in synaptic strength and contributes to learning and memory. It includes the formation, storage and retention of sensory memories, and occurs throughout life starting from birth and continues into adulthood to an extent. This process allows us to do everyday activities such as performing mathematical calculations, decision making and more complex multi-tasking activities, which are coordinated by proper neuronal connections in the brain that act as the central processing unit of the body. However, at the time of birth, there are fewer synaptic connections, but, as the brain matures, there is a massive surge in the number of neuronal connections that enables a child to learn tasks rapidly [40]. During this stage of development, the synaptic connections are highly vulnerable to changes in environmental stimuli. However, at the end of adolescence, only active connections (upon repeated or stronger stimulation) will be strengthened, and less active connections (upon weaker or not repeated stimulation) will be eliminated [41]. For example, a child may learn a new language in a few months to a year, but it might take months to a few years for an adult to master the same language. [42]. These observations postulated the phenomenon of a critical period of neuronal development. Different experiences during early post-natal life determine the process of formation, maturation and elimination of neuronal connections in the brain. Windows of heightened neuronal plasticity during brain development are termed as critical or sensitive periods, which predominantly occur in early life, and, hence, learning any new paradigms is relatively easy and quick in childhood, although it continues at a reduced level in adulthood. A critical period is a time when environmental or sensory input is required for the proper development of particular neuronal connections in different regions of the brain [43,44]. If these connections are unstimulated, the brain function served by that circuit will be permanently compromised [43,44]. For example, the critical period for the visual cortex begins soon after a baby is born in humans or soon after eye opening in animals such as rats or mice. Similarly, the onset of hearing triggers a critical period of development of the auditory cortex in humans. Perturbation in neuronal connections of these regions before the end of the critical period might permanently compromise their function. In the last decade or so, several laboratories have used visual, auditory and thalamocortical regions of the brain as models to understand the importance of the critical period in the plasticity and development of an individual [41,4547].

An important contribution to using the visual cortex as a model came from studies by Hubel and Wiesel in 1962; they emphasized the importance of the stimulus-dependent response by a population of neurons. They observed that, for cortical cells, the most effective stimulus configurations dictated by the spatial arrangement of excitatory and inhibitory regions were long narrow rectangles of light (slits), straight line borders between areas of different brightness (edges) and dark rectangular bars against a light background. To attain the maximum response, the shape, area and orientation of these stimuli were critical [48]. Recordings from different stages of development in normal kittens and those with monocular dominance (MD), where one eyelid was sutured during the early time window of development, concluded that kittens with MD had almost irreversible changes in the functional properties of the visual cortex area V1, suggesting the importance of sensory-dependent activation of neurons during the critical period of development and its relevance to neuronal connections [41]. These studies have demonstrated the importance of the critical period in the visual cortex and the modulation of neuronal plasticity based on experience/sensory information. For a better understanding of the molecular mechanisms underlying the critical period of development and plasticity in the primary visual cortex, studies using various animal models are discussed extensively in [41,4853], and similar information can be obtained for thalamocortical studies from [5457].

Studies have shown that changes at the synaptic receptor number and subunit expression of both excitatory and inhibitory neurons signal the opening or closure of a critical period of neuronal development [58]. For example, based on these studies, NMDARs are considered as one of the molecular determinants of the critical period of plasticity as NMDAR-mediated synaptic transmission is developmentally regulated, and their expression can modify neuronal activity [5861]. In the visual cortex, the percentage of total NMDAR-mediated current can be described by a slow exponential decay between the first and fifth post-natal week. Dark rearing of pups delays the developmental shortening of NMDAR-mediated currents, suggesting that the change in the 2A/2B ratio requires stimulus-based experiences in the visual cortex, and impairs the closure of the critical period. Pups reared in the dark displayed longer duration of NMDAR-mediated currents similar to younger animals bred in normal conditions, suggesting an altered critical period of development [62]. Apart from the visual cortex, various laboratories have studied the critical period of development in the thalamocortical region of the brain. By focusing on the NR2B to NR2A switch, which is developmentally regulated, NMDAR-mediated currents were recorded in layer IV of rat somatosensory cortex and found to be decayed more rapidly in PND7 than in PND3, which further suggests NMDAR-mediated, mainly subunit switch, modulation of the critical period [6365].

Another key factor that regulates the critical period of plasticity during development is the function of gamma-aminobutyric acid (GABA) in the neuron. The function of GABA is carried out by two Cl cotransporters, NKCC1 and KCC2, and its expression varies during the early stages of development [66]. Several studies using the gene disruption method to modulate the function of GABA and chloride concentration suggested the possibility to target NKCC1 and KCC2 for the rewiring of neuronal connections in the brain post-critical period of development, and, hence, a potential target for therapeutics [6769]. In various NDDs, such as ID, ASDs and schizophrenia, one of the common features observed is an imbalance of the excitation–inhibition (E/I) ratio, which can serve as a key factor in understanding the major cause of these disorders, and how manipulating the critical period, particularly targeting GABA, would help to resolve the associated defects [70,71]. It has been known for decades that maturation of the inhibitory cortical circuits in the brain parallels the opening of the critical period [7276]. With the help of glutamate decarboxylase (GAD)–knockout (KO) mice (GAD is a GABA-synthesizing enzyme), it has been demonstrated how inhibitory signalling regulates the critical period. For example, GAD-65 KO mice have impaired ocular dominance shift when one eye is deprived of vision [72]; visions was restored by the application of diazepam (benzodiazepine), which enhances GABA activity, within the critical period window by enhancing GABAR activity. These experiments suggest that ocular dominance plasticity is regulated by inhibitory neuronal signalling in the brain, apart from the excitatory signalling as discussed earlier. GAD-65 KO mice had shown reduced NR2A levels and slower NMDAR-mediated currents in the visual cortex, which plays an important role in the critical period of plasticity [74]. Other studies have focused on the ectopic expression of BDNF in mice, which is also known to regulate GABAergic inhibitory interneurons and related synaptic strength and has been shown to induce early opening and premature closing of the critical period of plasticity [73,77]. These studies suggest that there are different molecular determinants of the critical period of plasticity that are developmentally regulated. Here, we have briefly discussed the role of different proteins, especially GABAR, which are developmentally regulated and, in turn, modulate the critical period of plasticity. Thus, altering GABAR-mediated functions could be a potential therapeutic approach in the rewiring of synaptic connections and rescuing the pathophysiology of NDDs, particularly after a critical period of development, which is one of the major challenges faced by many neuroscientists.

4. Animal models of intellectual disability

The use of animal models to understand disease pathogenesis and to design treatment strategies has been long been a practice in biology and related disciplines. Despite the substantial ethical debate surrounding the use of animals in research, and the argument of interspecific variations, data from animal models remain the most positive attribute of biomedical research [78]. With technological advancements and the advent of transgenic mice, the ability to study more complex biological problems became feasible as a result of genetic modifications [79]. More recently, the use of Cre-Lox technology to generate tissue-specific KOs and other methods of modulating gene transcription in vivo has further contributed to our understanding of the mechanisms of human disease [80,81].

Although it is not possible to mimic all aspects of a disorder or disease in any one animal model, a suitable animal model should be able to replicate the clinical hallmarks of the disease with the paramount degree of robustness. Therefore, for a given condition, a range of animal models are usually defined, characterized and then validated. A good animal model should meet the following criteria: internal validity, external validity, construct validity, face validity and predictive validity. Internal validity refers to the reliability and reproducibility of the model with regard to consistency in the experimental measurements. Face validity describes the degree of similarity between the symptoms shown in human populations and those expressed in the animal model. Predictive validity involves the extrapolation of a particular experimental manipulation done in a species or a specific situation to the other species and situation. Construct validity refers to the degree of similarity in the mechanisms underlying the behavioural similarities between the animal model and those seen in patient populations. External validity involves the generalizability of the results obtained in the animal studies in relation to the general population [22,82].

Different reviews have discussed the commonly available and used animal models for ID [8284]. Some of the widely used mouse models that replicate the pathophysiology of ID are listed in table 1 along with the morphological, biochemical and behavioural alterations associated with them. Nevertheless, citing the failure of pharmacological agents in those clinical trials which otherwise showed efficacy in the preclinical studies, it becomes clear that the existing models have their limitations [79]. Hence, there is a need to look for new animal models of ID/ASDs that display different mutations and not only reproduce the clinical features of a disease but also guarantee a higher degree of translational success.

Table 1.

Tabulation of behavioural, synaptic and biochemical alterations used in transgenic mouse models of different ID/ASD-related genes (Fmr1, Syngap1, Shank, Neuroligin3 and Mecp2). LTD, long-term depression; Pv, parvalbumin; N/D, not defined or determined; EPSC, excitatory postsynaptic current; MSN, medium spiny neurons; IPSC, inhibitory postsynaptic current; E/I, exctiation–inhibitory; GTP, guanosine-5′-triphosphate.

genetic modification behavioural changes changes in synaptic morphology and function biochemical alterations references
Syngap1 mutation
 exon 7/8 in Syngap1+/−mice
B6.129-Syngap1tm1Rlh/J
MGI: 3822367
stereotypic behaviour, anxiety↓, memory deficits and social interaction↓ LTP↓,
AMPA/NMDA↑
N/D [85,86]
 exon 4–9 in Syngap1+/−mice,
Syngap1tm1.1Mabk
MGI: 3511175
mEPSPs↑
early maturation of the spines
altered clustering of PSD-95 protein and their movement into the spine head, dysregulation of Ras, activation of the Rho family of GTP-binding proteins and phosphatidylinositol-3- kinase [87]
 exon 5/6 and 7/8 in Syngap1+/−mice
B6.129S2 Syngap1tm2Geno/RumbJ
MGI: 5796355
STOCK Syngap1tm1.1Geno/RumbJ
MGI: 5796354
seizure threshold↓, altered context discrimination behaviour, locomotor activity↑ early spine maturation, AMPA/NMDA↑,
LTP↓
N/D [88]
Fmr1 mutations
Fmr1-KO (neomycin cassette inserted into exon 5)
B6.129P2-Fmr1tm1Cgr/J
MGI: 2162650
cognition↓ and activity↑ seizure threshold↓, sensitivity to sensory stimuli, anxiety↑ social interaction↓ spine density, immature thin, elongated spines↑ group I mGluR-mediated LTD↑ local protein synthesis↑ [89,90]
Fmr1-KO2 (germline ablation of promoter and first coding exon)
Fmr1tm1.1Cidz
MGI: 3808885
hyperactivity
altered emotional processing
memory deficits
hypersensory response
ultrasonic vocalizations↓
spine heads↓ and wider spine necks AMPA/NMDA↓
NMDAR-mediated LTP↓
[9193]
Fmr1-CKO (promoter and first coding exon are floxed, can be removed with conditional cre-expression)
Fmr1tm1Cidz
MGI: 3603442
hippocampus-dependent learning deficits
cerebellar eyelid conditioning↓
immature spine number↑ LTD↑ [94,95]
Mecp2 mutation
Mecp2 KO
B6.129P2(C)-Mecp2tm1.1Bird/J
MGI: 2165230
movement↓, improper gait, hind limb clasping, respiratory disorder number of dopaminergic neurons↓,
soma size↓,
precocious opening of critical period in visual cortex and accelerated maturation of GABAergic PV(+) neurons
deficit in GABA and glutamate synthesis pathway, spatio-temporal alteration of NMDAR expression, alteration in activity-dependent global chromatin dynamics [96102]
Mecp2-CKO TH-Cre, Mecp2 flox
B6.129P2-Mecp2tm1Bird/J
MGI: 3702570
total distance and vertical activity in open field↓, performance in dowel walking test↓ dopamine, norepinephrine, serotonin release↓ expression of TH and Tph2 [103]
MeCP2 KI
FVB-Tg(MECP2)1Hzo/J
MGI: 3817212
homozygous animals show tremors, gait ataxia↑
heterozygous animals show rescue from RTT-like symptoms
neuronal cell number and brain size is rescued to wild-type littermates N/D [104]
 MeCP2R168X point mutation;
STOCK Mecp2tm1.1Jtc/SchvJ
MGI: 5568127
stereotyped behaviour↑, hypoactivity, breathing problems N/D no change in the Ube3A mRNA level [105]
Shank3 mutation
Shank3 e4–9 homozygous
B6.129S7-Shank3tm1Yhj/J
MGI: 5295948
social interaction↓, repetitive behaviour↑, impaired memory activity-dependent redistribution of GluA1 AMPAR↓, thin long dendritic spines↑, LTP↓ GKAP, PSD95, Homer protein level↓ [106]
Shank3 e4–9 heterozygous
B6[36]-Shank3tm1.2Bux/J
MGI: 5317118
social behaviour, social sniffing, ultrasonic vocalization↓ mEPSC, basal neurotransmission, LTP↓ AMPAR expression↓ [107]
Shank3B−/−
B6.129-Shank3tm2Gfng/J
MGI: 5444207
repetitive grooming↑, social interaction↓ complexity↑ of dendritic length, dendritic arborization↑ and ↓surface area of MSN, caudate volume, cortico-striatal synaptic transmission, mEPSC frequency in MSN SAPAP3, PSD93, Homer, NR2B, GluA2, NR2A expression↓ [108]
Neuroligin
Neuroligin3 R451C knockin mice
B6.129-Nlgn3tm1Sud/J
MGI: 3820515
rotarod-mediated motor behaviour↑ dendritic complexity and dendritic branching in hippocampus↑. mEPSC in CA1, mIPSC in somatosensory cortex, LTP↑. mIPSC in CA3, GABA release, GDP frequency↑. GABAergic synaptic transmission↑ and IPSC amplitude↓ in barrel cortex and hippocampus. IPSC and E/I ratio in D1-MSN  NLGN3 protein misfolding and trafficking defects, NLGN3 expression was 90%↓. Alteration of NMDAR subunit composition and expression of NMDAR subunit 2B↑. IPSC amplitude and success rate at the same synapse failed to respond to AM251 (CB1 receptor antagonist) [109114]
Neuroligin3 R704C knockin mice STOCK Nlgn3tm3.1Sud/J
MGI: 5437466
AMPAR-mediated synaptic response↓ and unaltered NMADR or GABAR-mediated response. Unaltered NMDAR-mediated LTP, EPSC frequency↓ and NMDA/AMPA, in cultured hippocampal neurons↑ levels of AMPAR subunits GluA1 and GluA3↑ [115]

5. Gene mutation in intellectual disability and autistic spectrum disorders

5.1. SYNGAP1

SYNGAP1 is a 135 kDa protein that was shown by Chen et al. [116] and Kim et al. [10] to be one of the targets of phosphorylation by Ca2+/calmodulin-dependent protein kinase II (CaMKII) in the post-synaptic density (PSD) in the rat brain. It has several different isoforms which arise as a result of alternative splicing owing to different start sites. Though the existence of SYNGAP1 isoforms was first identified in 1998, detailed characterization and analysis were done much later [82,116,117]. The functions of different isoforms and physiological functions of Syngap1, particularly in regard to the Syngap1 mutation and its implications in ID and ASDs, are extensively discussed in another comprehensive review by Jeyabalan & Clement [82].

5.1.1. Behavioural changes associated with Syngap1 mutation

Syngap1+/− was found to be associated with several behavioural abnormalities, including cognitive and learning deficits, reduced seizure threshold, hyperactivity and increased locomotion [118,119]. Behavioural tests such as the Morris water maze, radial arm maze, spontaneous alternation test and the Y-maze novel arm test have been used to compare learning- and memory-related impairments in Syngap1+/− mice [87,120]. Syngap1+/− mice showed a significant decline in working memory; however, the performance was comparable to wild-type (WT) animals in the reference memory tasks, suggesting only specific memories are impaired [121]. These mice further presented with deficits in the remote memory when tested on the contextual fear learning procedure [119]. These findings have been verified physiologically by Clement et al. [88] by measuring basal synaptic transmission from the dentate gyrus and relating it to learning and memory deficits observed in these mice and patients.

Apart from cognitive decline, Syngap1+/− mice also exhibited stereotypic behaviour, hyperactivity and reduced anxiety-like behaviour, which was estimated experimentally using an open field test and elevated plus maze [85,121]. These mice spent more time in the open arms of the elevated plus maze than the WT animals, thereby displaying enhanced overall locomotion [85,119,121]. These behavioural abnormalities suggest impairment in learning and memory. To further validate these findings related to the Syngap1 heterozygous mutation, a study made use of the cliff avoidance test in which latency to jump off the cliff was higher in the case of the Syngap1+/− mice than in the normal WT animals, suggesting the lack of competency to judge the depth [122]. Similarly, patients with the SYNGAP1 mutation exhibited a non-syndromic form of ID that had been linked to a decline in sociability, and this was reinforced with the help of sociability tests done in transgenic mice [85].

Studies from patients with the SYNGAP1 heterozygous mutation have shown that approximately 80% of patients have epileptic seizures [82,123,124]. Indeed, electroencephalogram monitoring combined with video monitoring assays in patients showed spontaneous abnormal cortical activity in patients with the Syngap1 heterozygous mutation [119]. Similarly, Syngap1+/− mice exhibited a reduction in the seizure threshold and induction of myoclonic seizures as a result of mutations in the Syngap1 gene, which is in agreement with the epileptic seizures observed in patients [88,119]. Additionally, reduced levels of SYNGAP1 in the inhibitory GABAergic neurons exhibited reduced inhibitory synaptic activity and cortical gamma oscillation power and resulted in cognitive and social deficits [125].

A recent report has shown the impairment of sensory processing and touch-related deficits observed in patients as a result of reduced activity within the upper lamina somatosensory cortex (SSC) circuits in Syngap1+/− mice [126]. On the contrary, an earlier finding suggested that Syngap1 heterozygous mutation resulted in increased overall excitability of neurons [126]. These studies demonstrate the fact that Syngap1+/− mutations lead to abnormal neuronal activity, thereby causing excitatory and inhibitory imbalance.

In addition, the clinical findings were similar to the behavioural phenotypes observed in mice. For example, patients with SYNGAP1+/− reported delated psychomotor symptoms and developmental delays [127129]. They also manifested reduced seizure threshold and increased chances of epileptogenesis [124,129131]. Identification of different splice variants of the SYNGAP1 gene in patients would help to understand the implication of this gene mutation in humans and the translational success of studies done in rodents [132,133].

5.1.2. Synaptic function and morphology

SYNGAP1 protein has been shown to regulate postsynaptic cytoskeletal changes and AMPA receptor trafficking onto the surface of the postsynaptic membrane [86]. It was reported that Syngap1−/− mice died within a week of their birth, while the survival of Syngap1+/− mice was similar to WT [86]. The latter further showed a reduction in LTP in comparison with the WT littermates, suggesting impaired learning and memory abilities [86]. Non-viability of the Syngap1−/− mice has been attributed to increased apoptosis as a result of CASPASE-3 activation [134]. This could explain the reasons for not identifying any homozygous mutations in human patients, to date, and corroborates well with the preclinical mouse model, suggesting its impact on translational research. Syngap1+/− mice, on the other hand, exhibited premature spine maturation, leading to an overall increase in neuronal excitability [87]. Research has also pointed out the involvement of SYNGAP1 in the ACTIN-mediated steady-state regulation of spine morphology, which is necessary for spine maturation [135]. Clement et al. [88] confirmed the association of behavioural abnormalities with premature spine maturation in the hippocampus. They observed a higher number of mushroom-shaped spines and a lesser number of stubby spines at the beginning of the second post-natal week in comparison with the WT littermates [88]. This confirmed the role of SYNGAP1 in the control of cytoskeleton rearrangement and spine maturation. A follow-up study went on to suggest the crucial role of SYNGAP1 during the critical period of development [136]. Apart from early spine maturation as previously reported, somatosensory neurons in the Syngap1+/− mice showed adult neuron-like characteristics, including increased arbour complexity, total length and occupational volume [19]. Similar observations were made in another report, which studied the development of layer II/III of the medial prefrontal cortex (mPFC) in Syngap1+/− mice. They found an elevated AMPA/NMDA ratio during the early stages of development, which was correlated to the prematuration of excitatory synapses in the cortex [136].

Maturation of the spines from filopodia to mushroom-shaped requires un-silencing of synapses mediated by an increase in the insertion of AMPA receptors. This change leads to an increase in the level of basal synaptic transmission, which has already been discussed in detail elsewhere [137]. It was initially demonstrated that the basal synaptic transmission remained unaffected in the adult Syngap1+/− mice. However, a follow-up study by Clement et al. [88] showed that AMPAR-mediated currents increased in P14–16, equivalent to that of WT, but otherwise remained unchanged in young (P7–9) and adult mice [88]. This suggested that the alteration in the insertion of AMPA receptors could lead to changes in the excitatory and inhibitory balance and altered the critical period of development which eventually causes various behavioural defects as reported by patients with SYNGAP1+/−.

As a part of the NMDAR-mediated signalling pathway, a heterozygous mutation in Syngap1 may impair the NMDAR-mediated current. However, it remained unaffected when NMDAR-mediated currents were measured from different stages of development, suggesting a role for Syngap1 in synapse formation and function without altering the characteristics of NMDARs [138]. An in vitro study performed later went on to show an increase in the amplitude and frequency of the mEPSCs as a result of an increase in the number of AMPA receptors at the post-synapse and, hence, the surge in the AMPAR-mediated current [87]. In a similar study done in vivo, comparable results were obtained, though only at the P14–16 stages [88]. These studies further suggested that an increase in the AMPA/NMDA ratio at P14–16 in Syngap1+/− mice correlated with an increase in the number of functional synapses in the hippocampus. Experiments involving other areas of the brain such as mPFC have also yielded similar results with an overall increase in glutamatergic activity [119].

As a result of increased basal synaptic transmission and excitability due to increased AMPA receptor insertion at the post-synapse, LTP generation and maintenance were impaired in the adult Syngap1+/− mice [86,119,138]. On the other hand, the effect of Syngap1+/− on LTD induction has been studied less extensively. Acute application of NMDA had been widely used to induce LTD in acute brain slices [139]. The same protocol when used in Syngap1+/− mice yielded poor maintenance of the LTD in comparison with the WT control animals where stable LTD was maintained throughout the experiment [135]. Nevertheless, the paired-pulse protocol used elsewhere did not show significant differences in LTD induction in Syngap1+/− mice, which suggests that the release probability was not affected by SYNGAP1 and it was an altered function of post-synapses [86]. To study the role mGluR-mediated LTD, Barnes et al. [18] stimulated group I mGluRs with dihydroxyphenylglycine (DHPG) in the hippocampus and showed mGluR-LTD was significantly increased independent of protein synthesis in Syngap1+/− mice at PND 25–32. Our unpublished data, at the time of writing this review, further confirmed that increased mGluR-LTD is persistent in adulthood. However, the mechanisms of how SYNGAP1 regulates mGluR-LTD are yet to be elucidated. One of the possible mechanisms proposed by Barnes et al. suggests the presence of convergence in the biochemical signalling pathway downstream of mGluR- and NMDAR-mediated signalling proteins, although this needs to be further investigated in detail.

5.1.3. Biochemical pathways and their alteration in the SYNGAP1 haploinsufficiency

SYNGAP1 is a postsynaptic protein that is downstream of NMDA receptors and the postsynaptic scaffolding protein PSD-95 [10]. It negatively regulates RAS-GTPase activity, thereby regulating the insertion of AMPARs at the post-synapses [10]. Furthermore, its activity is regulated by the phosphorylation of CaMKII [10]. Based on these initial findings, the possible phosphorylation sites of SYNGAP1 were recognized, and the levels of phosphorylation increased activation of NMDARs [140]. Studies performed using Syngap1+/− mice to identify the signalling cascades downstream of SYNGAP1 demonstrated that it is a negative regulator of extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) signalling and a positive modulator of the p38–MAPK signalling pathway, thereby regulating activity-induced synaptic plasticity (figure 2 compares the signalling mechanisms impaired in different mutations in ID that are discussed in this review) [138,141]. Additionally, the activity of proteins such as p21-activated kinase [27], RAC and p-Cofilin, which are regulated by the SYNGAP1, were also elevated in Syngap1+/− mice under the basal conditions [135]. Thus, SYNGAP1 regulates spine morphology and function by modulating cytoskeletal dynamics. It was clear from these studies that the signalling pathways downstream of NMDARs were impaired in Syngap1+/−, causing various sensory, cognitive and social deficits as observed in patients.

Figure 2.

Figure 2.

Illustration depicting primary signalling mechanisms of different neuronal and astrocytic proteins encoded by genes implicated in ID/ASDs. Activation of the pre-synapse leads to neurotransmitter release. The neurotransmitter binds to the corresponding receptors; these allow the influx of divalent ions that trigger several downstream signalling cascades. (1) Activation of group I mGluRs leads to dephosphorylation of FMRP by PP2A. This dephosphorylation displaces FMRP from mRNA promoting their translation. Simultaneously, PP2A inhibits extracellular signal-regulated kinase (ERK)-mediated protein synthesis. On a slower time scale, mGluR activation stimulates the mTOR pathway, which, consequently, re-phosphorylates FMRP and inhibits mRNA translation. (2) NMDAR activation leads to phosphorylation of CaMKII, which in turn phosphorylates SYNGAP1and traffics AMPARs to the postsynaptic membrane via ERK. (3) SHANK3 is expressed downstream of group I mGluRs and regulates signalling via HOMER, which might also regulate NMDAR-mediated signalling. In addition, SHANK3 interacts with NMDAR via GKAP and PSD95, thereby regulating synaptic plasticity. (4) NEUROLIGIN3 interacts directly with NEUREXINS and maintains the stability of the excitatory synapse, whereas NEUROLIGIN2 regulates inhibitory synapse function via GEPHYRIN and COLLYBISTIN. (5) MeCP2 regulates expression of different plasticity-related genes in neurons as well as in astrocytes.

5.1.4. The critical period of plasticity

Syngap1 is shown to have a developmentally regulated expression in mice wherein the expression peaks at PND14 and stabilizes subsequently. In Syngap1+/−, at PND14–16, the SYNGAP1 expression was half of that of WT. Electrophysiological studies in acute hippocampal brain slices have shown an increased AMPA/NMDA ratio which was further corroborated by an increased number of mushroom-shaped spines at PND14, increased basal synaptic transmission and increased mEPSC and mIPSC (increase in amplitude and frequency, which suggests that the number of functional AMPAR/GABAR, respectively, in the post-synapse) at PND14 in Syngap1+/− when compared with WT (matures at PND21) [88]. Overall, this study suggests that early maturation of dendritic spines, which indirectly illustrates the altered critical period of plasticity and development in Syngap1+/−. Clement et al. in 2012 [88], using the conditional Cre-lox system to create haploinsufficiency of SYNGAP1, measured AMPA/NMDA in neonatal and young adult mice. Surprisingly, they observed that the AMPA/NMDA ratio in Syngap1+/− young adults was the same as WT adults, which contradicts increased AMPA/NMDA for neonatal mice. This study further confirms that the critical period of SYNGAP1 protein function is in the first two weeks of hippocampal development. In 2013, a similar study [136] by the same group demonstrated an altered critical period of development in mPFC and thalamocortical connections in Syngap1+/− during development, further validating early excitatory synaptic maturation in mPFC and, thereby, restricting the duration of the critical period of plasticity window [136]. This study further suggests that, owing to a mutation in Syngap1+/−, an altered critical period of plasticity and early maturation of dendritic spine structures might prevent remapping of connections, particularly to any experience, during development. In a study by Aceti et al. [19], tracking of dendritic arborization in the somatosensory cortex in Syngap1+/− at PND21 unveiled several adult-like features of neurons, such as higher order branching, arborization and adult-like dendritic length, suggesting early maturation of neurons [19]. A filopodia to spine structure transition is often associated with functional remapping of sensory circuits in response to experiences [142]. Whisker deprivation indicated a 2.5-fold increase in filopodia density at PND21 in WT which was absent in Syngap1+/−; this further suggests a limited capacity to organize cortical circuits or remapping cortical circuits in Syngap1+/−. To further study the developmental regulation of SYNGAP1 and early maturation of neuronal spines, Aceti and group did genetic rescue by inducible Cre-allele post-critical period. Different behavioural phenotypes such as risk-taking, novelty-induced hyperlocomotion and long-term memory were rescued in neonatal mice (tamoxifen injected in PND1), whereas the young adult (PND21) counterpart showed partial rescue of brain dysfunction in Syngap1+/− [19]. Overall, these studies suggest that there is indeed a heightened period of development and plasticity of SYNGAP1 protein in different brain regions within the critical period window, which is irreversible in Syngap1.

5.2. FMR1

Fragile X syndrome is the most common monogenic form of ID [143,144]. It is an X-linked disorder [145] and, therefore, generally affects males more prevalently (1 in 4000) than females (1 in 8000) [146,147]. It is caused by the loss of function of the fragile X mental retardation protein (FMRP) [148]. The absence of FMRP is most often the result of transcriptional silencing of the locus containing the FMR1 gene [149]. Typically, the FMR1 gene consists of a polymorphic repeat region present in the 5′-UTR (untranslated) non-coding region of the FMR1 gene [148,150]. It consists of a stretch of trinucleotide CGG repeats that varies between six and 54 in normal alleles [150]. Such trinucleotide repeats were found to be susceptible to expansion and contraction events during the process of DNA replication [151,152]. When this region consists of more than 200 trinucleotide repeats, it leads to epigenetic silencing owing to hypermethylation of the repeat region, and the neighbouring CpG islands present in the promoter region leading to heterochromatin formation [149,153156]. The formation of condensed heterochromatin in this region creates a microscopically visible constriction in the corresponding site on the X-chromosome, from which the disease derives its name [148,157]. Alleles containing 54–200 CGG repeats do not lead to silencing but are more likely to expand further and, thus, are termed pre-mutations [158]. Although pre-mutations do not cause fragile X syndrome, they were found to be responsible for causing fragile-X-related disorders such as fragile-X associated tremors/ataxia syndrome (FXTAS) [159,160] and fragile X-related primary ovarian insufficiency (FXPOI) [161,162] (box 1).

Box 1. Mechanism of trinucleotide expansion.

Although the mechanism is unknown, it is thought to occur as a result of the formation of secondary hairpin loop structures by these repeats in the daughter strand. During replication, DNA polymerase synthesizes a new complementary strand of DNA along the original parent strand, which then bind to each other. When a long stretch of DNA has newly synthesized trinucleotide repeat-sequence contacts and attaches to itself due to self-complementarity, it forms a secondary hairpin loop structure, leaving the just-copied parent strand unbound. As a result of this gap, the DNA polymerase slips back and re-replicates the same sequence. In this manner, the daughter strand now contains an extra set of trinucleotide repeats, i.e. expansion takes place [151].

Other than having an ID, patients with fragile X syndrome exhibit a range of morphological abnormalities such as macroorchidism (enlarged testis), macrocephaly, elongated face, prominent jaws and forehead, and a highly arched palate and contain loose connective tissue leading to highly extendable joints, flat feet and soft skin [163165]. To further understand the pathophysiology and aetiology of this disorder, the most commonly used mouse model is the Fmr1-KO mouse, which was first developed in 1994 [166].

5.2.1. Behavioural changes associated with Fmr1 mutation

Patients with fragile X syndrome display a variety of neuropsychiatric symptoms that mainly include cognitive deficits, delayed language development, hyperactivity, social anxiety, impulsivity and a subset of autistic behaviours such as stereotypic and repetitive behaviour, shyness, poor eye contact and hypersensitivity to sound. Other than ID/ASD, fragile X syndrome frequently co-occurs with additional neuropsychiatric disorders such as epilepsy, sleep disorders and ADHD [163,164,167171].

Numerous studies have been performed to study the emergence and nature of behavioural characteristics in the Fmr1-KO mouse model. Various types of fear-conditioning studies that test memory (hippocampus, amygdala and mPFC dependent) have revealed mild impairments in Fmr1-KO mice [172176]. Additionally, in the Morris water maze test, which requires intact long-term hippocampus-dependent memory, Fmr1-KO showed learning deficits during the phases of acquisition and reversal memory [166,177]. Fmr1-KO mice failed to discriminate during the novel object recognition task, a well-established short-term memory task [178,179]. Additionally, Fmr1-KO mice showed exaggerated inhibitory (passive) avoidance extinction, demonstrating impaired emotional memory processing [173,180182]. Thus, the Fmr1-KO mice were found to display various forms of memory impairments and cognitive deficits such as increased locomotor activity, anxiety, poor communication based on the ultrasound test, repetitive behaviour, defective sensory motor gating and seizures and to prefer social isolation [166,173,183192]. Fmr1-KO mice further showed increased stereotypic and repetitive behaviours as observed from self-grooming [186,193] and marble-burying tendencies [194]. Based on these studies, it is evident that the biochemical, molecular, physiological and behavioural findings in mice are similar to phenotypes observed in human patients. Therefore, many of the Fmr1 mouse models are useful tools to study the effect of monogenic mutations in pathophysiology and synaptic deficits as well as behaviour, and can be a potential preclinical model to find novel therapeutic targets [195,196]. However, caution should be shown to determine a suitable mouse model as it should fulfil the criteria discussed in the animal model section earlier.

5.2.2. Synaptic function and morphology

During development, spine morphology gradually changes from thin, motile immature, elongated spines to larger, more stable, mature, stubby, mushroom-shaped spines [197199]. Golgi staining of post-mortem brain tissue of patients with fragile X syndrome revealed dysgenesis in the spine morphology. Neurons in these tissues showed increased spine density, most of which were thin and elongated, indicating an immature spinal phenotype [200]. A similar observation was made in neurons from multiple cortical areas [201]. Fmr1-KO mice also exhibit a similar spine morphology such as the increased density of thin, long, immature spines in many cortical regions and the hippocampus [202,203]. This change in the spines may result from impaired synaptic turnover and maturation of spines during development, leading to retention of an immature spinal phenotype during the adult stages [204]. Studies have indeed found that increased spine turnover fails to decrease after the first two weeks of post-natal development and persists until four weeks of age, and even until adulthood [205,206]. The number of mature dendritic spines (those opposed to pre-synapse) is also reduced in Frmr1-KO mice [207]. This general lack of spine maturation is corroborated using electrophysiological evidence [208,209].

The morphology of spines correlates with the functional properties of synapses such as plasticity. Studies of basal synaptic transmission in Fmr1-KO mice did not show any difference in mEPSC amplitude [210], input–output (IO) curve or paired-pulse ratios (PPRs). However, a difference was observed in evoked spontaneous events, indicating that knockdown of FMRP does not change intrinsic synaptic electrophysiological properties, but reduces or delays the number of functional synaptic connections [210]. Fmr1-KO mice also showed a reduced AMPA/NMDA receptor ratio during the developmental stages [209,211]. Concerning plasticity, Fmr1-KO mice exhibited enhanced group I mGluR-dependent LTD [212]. However, no impairments were observed in NMDAR-mediated LTD [213].

On the other hand, NMDAR-mediated LTP was impaired in many cortical areas of Fmr1-KO mice. In the hippocampus, NMDAR-mediated LTP induced by theta-burst stimulation was also reduced in Fmr1-KO mice [214218]. Both mGluR-mediated LTD and sustained theta-burst-mediated LTP require local synaptic protein synthesis, showing that FMRP may play a role in the regulation of synaptic protein synthesis. Large-scale forms of plasticity such as homeostatic plasticity have also been reported to be impaired in Fmr1-KO mice [219,220]. Other than glutamatergic synaptic impairments, Fmr1-KO mice further show reduced dopamine- [221] and GABAR-mediated signalling, and the number of GABARs were altered in Fmr1-KO mice [222]. These alterations in GABAR cause aberrant GABAR-mediated signalling, contributing to an altered E/I balance [223226]. These studies suggest that the impaired group I mGluR-mediated protein synthesis and altered E/I balance lead to the decreased seizure threshold and learning and memory deficits observed in patients.

Impaired synaptic morphology and function may lead to abnormal neuronal circuit phenotype and subsequent behaviours. The E/I balance is altered in Fmr1-KO mice [226]. In general, reduced inhibition and increased excitation levels were observed in these mice, corroborating the increased susceptibility to seizures and epileptogenesis [90,227229]. Thus, these alterations in the spine morphology and synaptic function could lead to ID and ASDs in human patients.

5.2.3. Biochemical pathways

The FMR1 gene present at locus Xq27.3 in humans codes for the FMRP protein [148]. FMRP was found to be majorly expressed in the brain and testes [230]. It consists of three RNA binding domains—two of which are K homology domains (KH1 and KH2), the third being the RGG (arginine–glycine–glycine) domain [231,232]. Thus, FMRP binds to RNA and regulates many of its dynamics. FMRP binds to almost approximately 4% of the neuronal RNA population and regulates the expression of their proteins, thereby affecting many neuronal and synaptic properties [231,232]. Once bound to mRNA, FMRP negatively regulates its translation [233]. This is hypothesized to take place by one or a combination of three mechanisms: (i) FMRP binds to secondary G-quadruplex structures in mRNA, stalling ribosomes [234237]; (ii) FMRP recruits CYFIP-mediated inhibition of translation [238]; and (iii) FMRP recruits inhibitory miRNA containing AGO2 (Argonaute 2) complex to the mRNA, leading to RNA-induced silencing complex (RISC)-induced silencing [239242]. FMRP additionally has nuclear import and export signals [243]. FMRP has been reported to bind mRNA in the nucleus and regulate RNA's transport to dendritic spines [244,245]. Finally, the two tandem Agenet domains were found to recognize trimethylated serine residues, and, thereby, have been reported to interact with histones and modify chromatin dynamics in the nucleus [245,246]. It is evident from these studies that FMRP binds to different RNA domains, thereby regulating the strength of synapses in response to a stimulus.

An important physiological function of FMRP in neurons is the activity-dependent group I mGluR-mediated repression of local protein synthesis in dendritic spines [247]. FMRP, in its phosphorylated form, represses translation [248,249]. Neurotransmitter-mediated activation of group I mGluRs leads to the activation of a phosphatase PP2A which dephosphorylates FMRP [250]. This change in PP2A leads to removal of FMRP from the mRNA, enabling local protein synthesis of various plasticity-related proteins, including those involved in AMPAR endocytosis [249,251,252]. On a slower time scale, mGluR activates mTOR, which, in turn, inactivates PP2A and activates S6 kinase, leading to re-phosphorylation of FMRP and repression of translation [253,254]. Thus, knockout of Fmr1 leads to elevated levels of protein synthesis, causing dendritic spine dysmorphogenesis and impaired mGluR-mediated LTD [161,255]. In conclusion, such changes at the biochemical and molecular level impacts synaptic function and behaviour.

5.2.4. The critical period of plasticity

The critical period of plasticity is often found to be disrupted in many of the NDDs. One of the first studies in the somatosensory cortex by Harlow et al. [256] has shown the impairment of the critical period of plasticity in the Fmr1-KO mouse model. Behaviourally, Fmr1-KO mice showed altered sensory processing, as discussed above. Previous studies have shown an abundance of long thin immature dendritic spines in the somatosensory cortex [257,258]. However, these observations were correlative and unclear regarding the mechanism for various behavioural abnormalities. Using voltage clamp recordings, Harlow et al. [256] measured the ratio of NMDAR- and AMPAR-mediated current (NMDA/AMPA ratio) from the spiny stellate cells of the somatosensory cortex. In WT mice, the NMDA/AMPA ratio decreased progressively from PND4 to PND7, marking the closure of the critical period, whereas in Fmr1−/Y mice the NMDA/AMPA ratio increased between PND4 and P7 and returned to the WT level at PND10–14 [256]. In addition, loss of LTP induction was shown to be a manifestation of the closure of the critical period in the somatosensory cortex [63], which was delayed in Fmr1−/Y mice [256]. These data suggest a delay in the maturation of the thalamocortical synapses in fragile X syndrome, which could be the reason for impaired sensory processing, learning and memory. Along the same lines, altered ocular dominance plasticity in the visual cortex of Fmr1−/Y mice was shown by measuring visual evoked potentials (VEPs) [182,259]. Later, a maladaptive auditory response manifested by patients with fragile X syndrome and in Fmr1-KO animals was shown to be a result of the impaired critical period of plasticity in the primary auditory cortex [229]. In conclusion, alteration in the critical period of synaptic plasticity is a significant contributor to the behavioural and synaptic pathophysiology, which may prevent neuronal remapping in the fragile X syndrome.

5.3. MeCP2

X-linked heterozygous mutations in methyl CpG binding protein 2 (MeCP2) has been shown to cause Rett syndrome (RTT) in humans [260263]. MeCP2 has one of the longest known 3′-UTRs of the human genome and contains four exons [264]. It is believed that the diverse and complex function of MeCP2 partly lies in its 3′-UTR. Studies have shown that different polyadenylation signals also bring changes in the expression pattern of MeCP2 [265].

5.3.1. Behavioural alterations

Studies have shown that approximately 1 in 10 000 females is affected by RTT [266268]. Studies on human patients between 7 and 18 months of age showed phenotypes of deteriorated higher cognitive and social functions, stagnancy of brain development, severe dementia, autism, ataxia and repetitive hand movements [266268]. Constitutive MeCP2 hemizygous KO and conditional KO mouse models showed motor deficits such as improper gait, hindlimb clasping, irregular breathing [98] and microcephaly [98,269], and did not survive for more than 12 weeks after birth. Apart from the motor and social impairment, a mouse model of Mecp2 also manifested impaired hippocampus-dependent spatial memory, and contextual fear memory [270]. Post-mitotic neuron-specific KO and overexpression of Mecp2 displayed a similar phenotype to patients with RTT. These animals showed deficits in motor behaviour, an increase in anxiety-related behaviour, impaired social interaction and alteration in learning and memory [269,271]. In addition, a C-terminal deletion mouse model of MeCP2 also displayed a similar kind of phenotype, with impaired motor learning, social deficits and epileptic seizures [272]. All these results describe successful representation of RTT in mouse models, opening the possibility for effective pre-clinical studies.

5.3.2. Synaptic function and morphology

Post-mortem studies from patients with RTT revealed reduced axonal and dendritic processes and decreased dendritic spine density in CA1 pyramidal neurons [273,274]. A similar phenotype, i.e. reduced neuronal soma size and decreased dendritic arborization, was observed in the cortical pyramidal neurons of layer II/III of the MeCP2 null mouse [275]. Thus, reduced complexity and size of the neurons could be one of the underlying reasons for the impaired behavioural phenotypes in the animal models similar to the patients. Considering the function of MeCP2, it can be speculated that dysregulation in the expression of many genes such as Bdnf, which is important for neuronal growth, may lead to altered neuronal morphology in Mecp2 mutant mouse models. However, a mouse model overexpressing MeCP2 had shown similar spine morphology to the MeCP2 null mouse and patients with RTT. This study suggests that the excess level of the protein affects the synaptic functions, which, in turn, may lead to learning and memory deficits [276].

Patients with RTT show cognitive deficits, which could be due to impairment in the synaptic functions [266268]. Synaptic functions were altered in the MeCP2 mutant mouse models, consistent with the structural and behavioural alterations [277279], thereby causing E/I imbalance. In addition, modification in basal synaptic transmission has resulted in impaired hippocampal LTP, not only in KO but also in knockin studies [270,279,280]. It is clear from these studies that any fluctuation in the level of MeCP2 can alter not only behaviour but also synaptic function.

5.3.3. Biochemical pathways

MeCP2 regulates transcription through DNA methylation and histone acetylation. As the name suggests, it binds to the methylated cytosine residues on DNA through its methyl CpG binding domain (MBD) [281283]. Another domain, the transcriptional repression domain (TRD), interacts with HDACs and mSin3a to regulate transcription of the downstream genes [281]. These proteins act as a corepressor of transcription. Therefore, the interactions are necessary to bring about transcription regulation through MeCP2. MeCP2 is shown to regulate many genes such as Bdnf, Dlx5, Dlx6, Reln and Ube3A, which are crucial for neuronal maturation and development, and the protein product is enriched in synapses [284287].

MeCP2 is a master regulator of the transcription that regulates the neuronal maturation process by controlling the expression of the genes mentioned above. Besides its role in transcription regulation, MeCP2 has been shown to localize in heterochromatin by associating with chromatin remodelling complexes such as SWI/SNF, ATRX and histone methyl-transferase [282,288291]. Association of MeCP2 with these different complexes allows the possibility of a global role in the regulation of gene expression. For a detailed review of the biochemical functions of MEPC2, please refer to Singh et al. [292] and Guy et al. [293].

Therefore, MeCP2, being a transcription regulator, regulates the expression of a wide array of synaptic plasticity-related genes. Hence, it modulates the neuronal/synaptic function which is crucial for healthy brain functions. However, a perturbation in MeCP2-mediated regulation due to a pathogenic mutation alters synaptic function and behaviour, resulting in RTT-related pathophysiology.

5.3.4. The critical period of plasticity

MeCP2 is required at different stages of brain development from increasing dendritic complexity to synaptogenesis and astrocyte maturation. Disrupting MECP2 at any of these stages has been linked to RTT-like phenotypes [294]. The maturation of the cortical GABA inhibitory circuitry, particularly parvalbumin+ (PV+) fast-spiking interneurons, is a key regulator for the initiation and termination of the critical period. MECP2 KOs exhibited accelerated functional maturation of PV interneurons, which correlated with a precocious onset and closure of the critical period and deficient binocular visual function in mature animals [99]. The findings to validate the role of MeCP2 in the critical period came from another study which observed that specific Mecp2 deletion in GABAergic PV cells abolished the visual experience-dependent plasticity during the critical period in post-natal development of the visual cortex, while conditional Mecp2 deletion in somatostatin-expressing GABAergic cells or glutamatergic pyramidal cells had no such effect [295]. Their study demonstrated that, during the critical period, selective deletion of the RTT-related gene Mecp2 in GABAergic PV neurons could result in defective inhibitory PV neuronal circuits in the developing visual cortex, which leads to the absence of experience-dependent critical period plasticity [295].

5.4. SHANK

SH3 and multiple ankyrin repeat domain proteins (SHANKs), also known as ProSAPs, are a family of post-synaptic scaffolding proteins present in the excitatory glutamatergic synapses [296]. Being a primary scaffolding protein, SHANKs organize other proteins in the synapse. Thus, they are essential for normal neuronal development and function [297]. Mutations in different Shank genes (SHANK1, SHANK2 and SHANK3), especially SHANK2 and SHANK3, are associated with ASDs [298], and co-occurrences with ID [299]. A recent meta-analysis study has proposed that around 1% of all patients with ID/ASDs had a mutation in SHANK genes [300]. Thus, it is vital to understand the pathophysiology of such mutations to develop the correct treatment.

5.4.1. Behavioural alterations

A study on the Shank1 null mouse model showed reduced social interaction (social sniffing), reduced ultrasonic vocalization, increased self-grooming and repetitive behaviour, enhanced spatial learning and impaired fear conditioning [301,302]. Although deletions in exons 6–7 and 7 of Shank2 mouse models showed contradictory deficits in the synaptic functions, comparable behavioural phenotypes were seen. Both the mouse models displayed increased locomotor activity, increased anxiety-like behaviour and impaired social behaviour [303,304]. However, exon 7 deletion of Shank2 resulted in increased self-grooming but normal working memory [304].

A mouse model of Shank3 with exon 4–9 deletion showed impaired social behaviour, reduced ultrasonic vocalization, increased self-grooming and impaired novel object recognition [107,305]. Other models of Shank3 mutation displayed similar behavioural deficits, which are summarized in table 1. Thus, it is evident that the mouse models for different Shank mutations can represent many of the behavioural deficits observed in humans.

5.4.2. Synaptic function and morphology

Mutation in the PDZ domain of Shank3 showed reduced dendritic spine density during maturation, whereas mutation in the Ank and Sh3 domains led to a reduced spine head volume [306]. Therefore, SHANK3 plays an essential role in dendritic spine maturation and morphology. An in vitro study using cultured rat neurons showed impairment in mGluR5-dependent plasticity and signalling, demonstrating the importance of SHANK3 downstream to mGluR5-mediated signalling [307]. A mouse model of Shank1 exhibited reduced basal synaptic transmission, whereas LTP and LTD were not altered [302]. On the contrary, opposing effects were observed when exon 6–7 and exon 7 were deleted from Shank2, suggesting that there may be a possibility for an isoform/transcript-specific function of this gene, but it is unclear whether splice variation has any implications for human patients. A study by Schmeisser et al. [304] showed reduced spine density and synaptic transmission in the CA1 region of the hippocampus when exon 7 was deleted. In addition, a decreased I/O ratio and mEPSC frequency were seen in the exon 7 deletions, suggesting an impairment in pre-synaptic neurotransmitter release upon exon 7 deletion. I/O is the measure of basal synaptic transmission, indicating a corresponding output to every input provided to a neuron. In contrast to the increased NMDA/AMPA ratio in exon 7 deletion, decreased NMDA/AMPA was observed in the exon 6–7 deletion model of Shank2 mutation [303,304]. Again, these observations show that the different exons in the gene might have opposite effects on the synaptic functions.

A Shank3 mouse model with exon 4–9 deletion had severe defects in synaptic function. For example, activity-mediated spine remodelling was impaired in the hippocampal CA1 region, leading to learning and memory dysfunctions. AMPAR functions were affected in these mouse models, which was manifested by reduced AMPAR-mediated basal transmission. However, the study showed that the decreased mEPSC amplitude was accompanied by a subsequent increase in mEPSC frequency. The increase in mEPSC frequency suggests an impairment in the level of pre-synaptic function or an increase in the number of functional synapses. Owing to impaired AMPAR-mediated synaptic function, LTP was reduced significantly. These results can be corroborated with impaired learning and memory observed in patients. However, no change in NMDAR- and mGluR-mediated LTD was observed in these mouse models [107,305]. These data reiterate the fact that SHANK3 plays an essential role in synaptic signalling and function. Also, the data indicate the role of SHANK3 not only in mGluR-mediated signalling but also in AMPAR-mediated synaptic transmission. Several other mouse models of Shank3 show deficits in different synaptic functions and are summarized in table 1.

5.4.3. Biochemical pathways and signalling

SHANK proteins are postsynaptic scaffolding proteins associated with the PSD complex in excitatory glutamatergic synapses [308310]. Structurally, the SHANK family of proteins consists of five distinct motifs/domains: ankyrin repeat domain (ANK), Src homology 3 (SH3) domain, PSD-95/disc-large/ZO-1 (PDZ) domain, proline-rich (Pro) and sterile alpha motif (SAM) domain [311]. Studies that have taken place over decades have identified at least 30 proteins interacting with SHANK proteins, including different receptors, ion channels, cytoskeletal proteins and signalling molecules [297,307,308,312314]. A study on Shank3 KO showed an alteration in the mGluR5-HOMER scaffolding that in turn affected the neuronal connections in the brain [315]. However, knockdown of Shank3 led to decreased mGluR5-mediated phosphorylation of ERK1/2 and CREB [307]. These independent observations link the importance of SHANK3 in mGluR5-mediated signalling and, in turn, are associated with regulation of synaptic functions.

Mouse models of the Shank1 mutation showed a decreased synapse-associated protein-90/postsynaptic density-95 associated protein (SAPAP), guanylate kinase-associated protein (GKAP) and HOMER protein level in the PSD complex [302]. Exon 6–7 deletion in the Shank2 model showed reduced phosphorylation of calcium/calmodulin-dependent kinase II (CaMKII), extracellular signal-regulated kinase (ERK) and the AMPAR subunit, GluA1, but increased expression of the NMDAR subunit, GluN1 [303]. Deletion of exon 7 from Shank2 increased NMDAR subunit, GluN2B, expression in the hippocampus, whereas, in the striatum, GluN1, GluN2A, SHANK3 and GluA1 levels were increased [304]. These data show that there could be a different function for SHANKs in different brain regions. Also, by modulating the expression of different NMDAR subunits, synaptic properties involved in learning and memory were modulated by different SHANKs. Studies from a mouse model of Shank3 with exon 4–9 deletion showed a robust reduction in the level of GluA1 subunit [107,305]. Shank3 knockdown in cultured hippocampal neurons showed reduced expression of mGluR5 but not NMDAR or ERK [307], whereas analysis from the striatal PSD fractions of the Shank3B−/− mouse model showed a decreased level of NR2A, NR2B, GluA2 and HOMER [108]. However, other mouse models of Shank3 also show related biochemical alteration (table 1). Based on the studies above, it is evident that the SHANK family of proteins have diverse roles in cellular signalling at different brain regions. In conclusion, the SHANK family of proteins differentially regulate synaptic function by modulating different receptor subunit expression, and, thus, regulate neuronal function, learning and memory, and behaviour.

5.4.4. The critical period of plasticity

An in vivo study has shown that loss of Shank3 led to impairment in the ability of visual cortical circuit recovery following sensory input deprivation [316]. Also, the homeostatic plasticity of neuronal circuits was disrupted in the Shank3 KO model, which hints towards perturbation of the critical period in Shank3 mutation [316]. However, using a conditional knockin mouse model, Mei et al. [317] showed re-expression of SHANK3 in adulthood restored spine density and synaptic functions in the striatum. Repetitive grooming and impaired social interaction were improved, whereas no improvement was seen in anxiety-like behaviour and motor behaviour [317]. From these studies, we can speculate that there was no major effect of Shank3 perturbation on the critical period of development. However, another possibility could be delayed closure of the critical period, which may persist until adulthood in the Shank3 KO, but this is unclear. This may explain the partial rescue of behavioural phenotypes observed after restoring Shank3 expression in adulthood.

5.5. Neuroligins

For almost two decades, it has been known that, to hold a particular synapse together for communications between neurons to occur, there are synaptic adhesion molecules such as NEUROLIGINS present in the post-synapse [318,319], and NEUREXINS, present in the pre-synapses, are known to hold a particular synapse together [320322]. Mutations in NEUREXIN and NEUROLIGIN genes increase the likeliness of affecting synapse formation, as well as function [318,319,323,324]. NRXN1, NRXN2 and NRXN3, in mammals encode for NEUREXINS, α-NEUREXIN and β-NEUREXIN, depending on their promoters [320,321]. NEUROLIGINS generally interact with β-NEUREXIN isoforms [318]. NEUROLIGINS, on the other hand, are encoded by five genes, NLGN1, NLGN2, NLGN3, NLGN4 and NLGN4Y, in humans [318,319,325,326]. Based on immunostaining and biochemical analysis, subcellular localization of NLGN1 was found to be present at the excitatory synapses, and the expression level is low at birth but increases during post-natal days 1–8 and remains relatively high in later stages of development in mouse [323]. NLGN2 and NLGN4 were shown to be expressed in inhibitory synapses [327,328], whereas NLGN3 was expressed in both excitatory as well as inhibitory synapses [329]. With the help of different binding partners, such as PSD-95 [330], MAGUK [331] and GluN1 [332] at excitatory synapses and GEPHYRIN [242,327] and COLLYBISTIN [333,334] at inhibitory synapses, all NEUROLIGINS are required to maintain synapse number/density and to regulate maturation and differentiation of synapses.

NEUROLIGINS were recognized to induce the formation of functional synapses in early 2000. These studies, using non-neuronal cells expressing NEUROLIGIN, revealed that NLGN1 and NLGN2 alone could trigger the formation of pre-synaptic structures such as clustering of synaptic vesicles in the axon terminals of central nervous system (CNS) neurons [335]. The role of NEUROLIGINS was further validated by dissecting functional characteristics, such as NMDAR-mediated excitatory postsynaptic currents and NMDAR-dependent LTP, which are a cellular correlate of learning and memory [336,337]. This study suggests a vital role of NEUROLIGINS in learning and memory. Moreover, Varoqueaux et al. [338] have shown that the Neuroligin mouse model dies after birth due to a reduced neuronal network activity and reduced glutamatergic and GABAergic synapse formation/function resulting in respiratory failure [338], suggesting the importance of NEUROLIGINS not only in synapse formation and maturation but also in neuronal function. In vitro electrophysiological studies have shown altered mIPSC amplitude as well as frequency, suggesting a reduced number of functional GABA receptors at the post-synapse resulting in altered E/I. Furthermore, decreased excitatory and inhibitory synapses were observed in the downregulation of NLGN1 and NLGN2 [339]. The introduction of exogenous NLGN increased both mEPSCs and mIPSCs indicates an increase in excitatory and inhibitory synaptic function (increased number of functional AMPAR and GABAR in the post-synapse, respectively) [340], implying the critical role of NEUROLIGINS in maintaining E/I balance.

Clinically, NLGN1 genetic variants are associated with disorders, such as ASDs [341,342], Alzheimer's disease (AD) [343] and post-traumatic stress disorder (PTSD) [344]. Genome sequence studies in humans have shown dysfunction in NLGN2 to be associated with ASD and schizophrenia. Studies were done by assessing the developmental history of a patient with a rare missense mutation, R215H, which revealed that the patient had psychotic symptoms such as self-laughing and talking, auditory hallucinations and delusions. However, the patient's sibling was a carrier of this mutation, suggesting that the R215H mutation is inheritable and had incomplete penetrance [345,346].

5.5.1. Behavioural alterations

Impaired spatial working memory by using the Morris water maze in either loss of Nlgn1 or overexpression of Nlgn1 was observed, which further implies the constitutive requirement of Nlgn1 for learning and memory [347,348]. Conditional KO of Nlgn1 in the CA1 region of the hippocampus in new-born (P0) or P21 resulted in impaired NMDAR-type and L-type Ca2+ channel-dependent LTP, further validating the loss of spatial working memory [349]. Nlgn1 KO mice created by targeted deletion of exon sequences covering the translational start site 380 bp of the 5′ coding sequence of NLGN1 and by homologous recombination in embryonic stem cells exhibited increased repetitive behaviour such as grooming, impaired social interaction and altered pain sensation [347].

Overexpression of Nlgn2 in transgenic mice had displayed diverse behavioural deficits such as reduced lifespan, limb clasping, offspring viability, repetitive behaviour, anxiety and impaired social interactions [350]. Based on the inhibitory avoidance (IA) behaviour paradigm, widely used for studying fear memories, Ye et al. [351] had found increased expression of NLGN1 and NLGN2 in quantitative immunoblot analyses after training of rats for IA. This altered expression of NLGN1 and NLGN2 suggests a role for both Nlgn1 and Nlgn2 in memory consolidation [351]. Further studies were performed to understand the role of Nlgn2 in memory formation and behaviour. Overexpression of Nlgn2 in the hippocampus had shown an increase in adult neurogenesis but decreased performance in the water maze task, suggesting an impaired working memory [352]. Nlgn2 KO mice showed reduced anxiety, increased impulsivity in the elevated plus maze and reduced fear conditioning with an increased ratio of evoked E/I synaptic currents [353,354]. These reports proved the importance of Nlgn2 in diverse behavioural functions by regulating inhibitory synapse function and plasticity in the mPFC, which is essential for anxiety and fear memory. In addition, Nlgn2 KO mice exhibited an irregular breathing pattern, suggesting its role in regulating lung and heart functions [338].

As discussed in the earlier section regarding the significant role of Nlgn4 KO in synaptic function, other studies have shown perturbations in general behavioural patterns, such as visible platform training in the Morris water maze (for vision), buried food finding (for olfaction), sucrose preference (taste), startle response (hearing), prepulse inhibition (sensorimotor gating), rotarod (locomotor activity and balance), hole board (exploratory behaviour), object preference, open field, hidden platform training in the Morris water maze, cued and contextual fear conditioning, and reversal training in the Morris water maze but lacked seizure propensity. These studies demonstrate a selective deficit in social interaction in Nlgn4 KO as seen in patients with ASDs [109,355,356].

5.5.2. Synaptic function and morphology

A transgenic mouse model of Nlgn1 overexpression led to an increase in excitatory dendritic spine and synapse number, E/I ratio and synaptic transmission in the hippocampus. Additionally, overexpression or downregulation of Nlgn1 has been shown to have impaired long-term potentiation (LTP), suggesting importance for Nlgn1 in learning and memory [348,357]. Moreover, studies from Caenorhabditis elegans lacking nrxn-1 or nlgn-1 have been shown to mediate retrograde synaptic signalling that inhibits neurotransmitter release at neuromuscular junctions, which might affect the activity of neurons in response to stimuli [358]. Nlgn1 KO mice made by targeted deletion (exon sequences covering the translational start site 380 bp of the 5′ coding sequence of Nlgn1) by homologous recombination in embryonic stem cells showed no change in PPRs, and basal synaptic transmission but altered LTP and AMPA/NMDA [347]. These studies from different mouse models show that Nlgn1 is necessary for AMPAR dynamics and, thus, the alteration would impair learning and memory.

E/I imbalance is one of the significant characteristic features of ID and ASDs that causes various physiological and behavioural deficits. In fact, in Nlgn2 transgenic mice, the E/I ratio was found to be decreased in the PFC along with the increased frequency of miniature inhibitory synaptic currents, which suggests an inclination towards potentiation of inhibitory synapses and, thereby, shifting towards altered inhibition [350]. Nlgn2 knockdown was associated with abolished GABAergic function from excitation to inhibition switch in cortical neurons based on Ca2+ imaging studies that show a gradual decrease of GABAR-evoked Ca2+ response in developing neurons along with the decreased frequency of mIPSC and mEPSC, suggesting a reduction in excitatory and inhibitory receptors. However, overexpression of KCC2, the potassium-chloride co-transporter, partially rescued synaptic currents, suggesting a role of NLGN2 in excitatory as well as inhibitory synaptic function [359].

Along with these earlier studies, another study has reported that Nlgn2 KO mice have developmental delays such as delayed eye-opening period, vocalization in pups and reduced body length in these Nlgn2 KO mice [360]. Furthermore, these mice displayed diminished inhibitory synaptic transmission with no change in synapse number in the ventrolateral medulla [333]. These studies suggest that Nlgn2 is an essential constituent of inhibitory synapses and necessary for the formation, maintenance and function of inhibitory neurons.

To further study the role of Nlgn3 mutations in synaptic function and neuronal development in vivo, lentiviral-mediated knockdown of Nlgn3 in the CA1 region of the hippocampus at P0 and P21 was performed, and excitatory basal synaptic transmission was unaffected [361]. On the contrary, a primary neuronal culture study has shown that overexpression of Nlgn3 increased inhibitory postsynaptic currents (IPSCs), suggesting an increase in expression of GABAergic currents [362]. Patch clamp recordings from the hippocampus, somatosensory cortex and cerebellum of Nlgn3 KO mice have revealed increased mIPSC (increased number of functional GABA receptors) and decreased mEPSC (decreased number of functional AMPA receptors) in the hippocampus, suggesting an increase in inhibitory activity as the number of functional GABARs was higher when compared with excitatory activity. Additionally, decreased mEPSC and impaired mGluR-mediated LTD in the cerebellum was also observed, which suggests a differential function of Nlgn3 in different regions of the brain [110,363].

An Nlgn3, wherein R451C was overexpressed, increase in mIPSC frequency (increase in the number of functional GABARs) was associated with altered GABA release probability, concomitant with an increase in giant depolarizing potential (GDP), a neuronal network-related activity mediated by GABAR [111]. These studies further validate the importance of Nlgn3 in network-based activities in immature neurons. Since GABAR-mediated network activity, as well as GABAergic synaptic transmission, was impaired in R451C, it is essential to understand whether the effects observed were a global GABAR-mediated phenomenon or any specific GABAergic cell types involved in it. Using a paired whole-cell patch clamp between one PV expressing basket cells and either spiny neurons/pyramidal neurons, IPSCs measured from PV neurons displayed impaired amplitude and frequency in the hippocampus as well as in the barrel cortex in Nlgn3 R451C knockin mice [112,113]. In addition, these studies found that the IPSC was reduced, leading to an altered E/I ratio in D1-medium spiny neurons (D1-MSN) in R451C mutant mice [114]. These studies show that the NLGN3 mutation has a stronger effect on PV cells comparatively, and is vital for D1-MSN-mediated synaptic transmission.

Moreover, the same knockin mouse model of Nlgn3, R451C, showed increased dendritic complexity and branching in stratum radiatum. These mice also showed increased excitatory basal synaptic transmission, LTP, NMDA/AMPA and mEPSC in the CA1 region of the hippocampus, suggesting, unlike KO models of Nlgn3, R451C function majorly affects glutamatergic synapse [110]. In contrast to previous studies, another mouse model of Nlgn3, R704C, displayed a decreased AMPAR-mediated synaptic response, rendering the NMADR- or GABAR-mediated response unaltered. Additionally, NMDAR-mediated LTP was associated with reduced EPSC frequency and increased NMDA/AMPA ratio in cultured hippocampal neurons, suggesting that an R704 mutation affects differently the inhibitory as well as excitatory synapses [115]. Overall, different mutant mouse models of Nlgn3 depict region- and synapse-specific function. Therefore, specificity in neuronal types and synapses may help Nlgn3 to execute different functions efficiently in various parts of the brain.

Nlgn4 is another essential gene that plays a significant role in synapse formation, development and function. To further understand the role of Nlgn4 in neuronal function, Jamain and group [355] have demonstrated reduced ultrasonic vocalizations in Nlgn4 KO males. The Nlgn4 KO is the result of chimeric non-functional protein. It contains a small fraction of the esterase domain that cannot bind to Nrxn upon contact with a female in oestrous cycle, suggesting a lack of ability to attract the opposite gender despite being fertile. A magnetic resonance imaging volumetric study demonstrated a reduction in total brain volume, particularly in the cerebellum and brainstem [355]. Nlgn4 KO showed a reduced decay in glycinergic mIPSC, impaired inhibition, altered firing and decreased β-wave amplitude in retinal cells, demonstrating that Nlgn4 localizes to glycinergic post-synapses and plays an essential role in encoding stimuli in the retinal network [328]. The Nlgn4 R87 W mutation abolished NLGN4-induced synapse formation and function, particularly in modulating synaptic strength [364]. An important hallmark of several NDDs, such as ID/ASDs, is impaired synapse formation and function. As NLGN4 is implicated in ASDs (reviewed extensively in [356]), a point mutation in Nlgn4 causes ID/ASDs through the loss of function. A study identified a frameshift de novo mutation, 1186insT in NLGN4, in two siblings with ASD and Asperger syndrome in a Swedish family, and that linked NLGN3 and NLGN4 to ASDs. Another Swedish family of two siblings with ASD and Asperger syndrome were identified with a C to T transition in NLGN3. This mutation had led to changes in highly conserved arginine to cysteine (R451C), an integral part of the esterase domain that is necessary for interaction with NRXNs [365]. Therefore, these studies have demonstrated that mutations in different Nlgn genes affect the basic synaptic transmission, which can have lasting implications for the pathophysiology in ID/ASDs.

5.5.3. Biochemical pathways

NEUREXIN and NEUROLIGIN contain an intracellular PDZ-binding domain that mediates interactions with synaptic scaffolding proteins, such as calcium/calmodulin-dependent serine protein kinase (CASK) [366], and Munc18 interacting protein; lin10/X11 [367] (for NEUREXINS) and PSD95 (for NEUROLIGINS) [330]. The synaptic function of NEUROLIGIN depends on cis clustering of NEUROLIGIN molecules, which requires a crucial integral esterase like ectodomain that can interact with NEUREXIN and execute synaptic functions [368]. PSD-95, a critical scaffolding protein interacting with NEUROLIGIN, recruits different synaptic protein/receptors like NMDAR, which activate the downstream signalling necessary for learning and memory. It also recruits specific adaptor proteins such as GKAP [369] that, in turn, interacts with SHANK3 [356,369]. This interaction is necessary for normal synaptic functions and is indirectly driven by different synaptic partners. Sequence homology studies have revealed that NLGN1, NLGN3 and NLGN4 were similar, unlike NLGN2, which is predominantly expressed in inhibitory synapses [318,319]. NLGN2 was shown to stabilize inhibitory synapses with the help of scaffolding protein GEPHYRIN, which interacts with an α2 subunit of GABARs [370,371] and helps in the maintenance and function of GABAergic synapses. Overall, NLGN4 is considered as one of the primary receptors involved in learning and memory formation, along with evolutionary conserved other NLGNs.

5.5.4. The critical period of plasticity

During brain development, NLGN1, NLGN2, NLGN3 and NLGN4 expression increases from embryonic to post-natal days before reaching a plateau around three weeks in the mouse hippocampus [355]. This study suggests that NLGNs have a precise developmental window of expression and, thus, could be considered as one of the determinants of a critical period of development in the brain, which could have implications in ID/ASDs [356]. To further corroborate NLGNs in the critical period of development, monocular and binocular deprivations (MD, BD) in mice were performed. Increased spatial acuity by measuring visually evoked potentials was observed in young adult R451C Nlgn3 mutant mice. Immunohistochemical analysis revealed an increased number of puncta of GAD65. This increase suggests an alteration in the number of GABAergic interneurons, thereby resulting in elevated inhibition that led to decreased E/I, and, thus, increased acuity in mutant mice. R451C also showed permanent loss of acuity on prolonged MD as WT, but the relative loss was more for the mutant mice, suggesting a longer window for plasticity. This alteration observed in MD might lead to the abnormal opening of a critical period of plasticity and impaired local circuit connections [355]. Studies have further tried to dissect the mechanism of NEUROLIGIN functions in synapse development and neuronal functions using different animal models such as KO, knockin and transgenic mice.

6. Metaplasticity

Metaplasticity is a term which refers to the higher order of synaptic plasticity, i.e. plasticity of the synaptic plasticity [372]. It includes processes that lead to physiological and biochemical changes, altering the neuron's ability to induce and maintain synaptic plasticity [373]. Different mechanisms have been proposed to explain metaplasticity depending on the location and the type of synapse or receptors of interest [373]. To date, there are no reports stating the direct involvement of metaplasticity in any gene mutations implicated in ID or ASDs. However, it has been proposed that synaptic defects and memory deficits associated with ID and ASDs may be due to an inability to undergo metaplasticity during various developmental stages [374]. It has also been demonstrated that hippocampal metaplasticity is required for the formation of temporal associative memories [375]. Although studies done to investigate the effect of metaplasticity in animal models are limited, owing to its role in the maintenance of LTP and LTD it may be crucial for learning and memory impairments in ID.

7. Glial cells in intellectual disability

Other than neuronal cells, the brain consists of non-neuronal cells called glia. The three main types of glia are—oligodendrocytes (responsible for axonal myelination), microglia (responsible for immunity-related functions) and astrocytes (responsible for maintaining homeostasis in the brain) [376].

7.1. Astrocytes

Astrocytes are star-shaped cells initially only known to provide neuronal support. However, research over the past few decades has increasingly revealed the importance of astrocytes, and their multifaceted role in the brain [20,377,378]. Astrocytes regulate post-natal neurogenesis [379,380] and maintain homeostasis of many factors such as ions, neurotransmitters, water and extracellular matrix [381384]. They regulate many aspects of the blood–brain barrier [385,386] and glucose metabolism (supply and storage) in the brain [387,388]. They protect against damaging factors such as pathogens, reactive oxygen species and excitotoxicity [389391]. Most importantly, astrocytes wrap around synapses and finely regulate all aspects of synaptic dynamics such as formation, maturation, plasticity and even elimination [378,392395].

Proteins of many genes implicated in ASDs and ID are expressed in astrocytes. Fmr1 is one such gene that encodes FMRP. In astrocytes, FMRP is expressed during early and mid-post-natal developmental stages, indicating its probable role in brain development and fragile X pathology [396,397]. Indeed, studies have shown that Fmr1-KO astrocytes were able to induce pathogenic delayed maturation and synaptic protein expression in WT neurons. Vice versa, WT astrocytes can rescue abnormal spinal and dendritic phenotype in Fmr1-KO neurons [398,399]. Astrocyte-specific knockdown of Fmr1 led to increased spine density in cortical neurons and can be rescued by restoring astrocytic FMRP levels [21]. A study on MECP2 has also been shown to be expressed in astrocytes, and similar co-culture studies demonstrate astroglia contribution to disease pathology [400,401]. Other than these proteins, only NEUROLIGINS are expressed in astrocytes, where they contribute to the formation of astrocyte morphology and neuronal synapses [402]. Disease-related studies for Neuroligins, Shank and Syngap1 remain to be carried out.

7.2. Microglia and oligodendrocytes

Microglia are the resident macrophages of the brain. They are of mesodermal origin and migrate into the brain during development [376,403]. Other than immune surveillance, microglia help in synaptic pruning (through complement-mediated phagocytosis or TREM2 signalling), clearance of apoptotic neural debris during development and synaptic plasticity (through secreted factors) [404,405]. On the other hand, oligodendrocytes originate from neural tissue and differentiate from oligodendrocyte precursor cells. They differentiate soon after neuronal migration and start wrapping neuronal axons with lipid-rich myelin, providing insulation and dividing the axon into multiple domains. The altered activity helps in regulation of the propagating action potentials and, in turn, neuronal circuit properties [406]. Oligodendrocytes also provide metabolic support and maintain ion homeostasis of the axon [407,408]. Not many studies have been performed on the relationship between genes implicated in ID/ASDs and microglia/oligodendrocytes. However, FMRP has been demonstrated to express in both cell types [397]. Further studies regarding how these mutations (implicated in ID and ASDs) affect microglia and oligodendrocytes, and how this, in turn, contributes to the pathophysiology of these disorders remains an open and exciting topic for investigation.

8. Current and upcoming therapeutic options targeting synaptic abnormalities

A therapeutic intervention targeting the underlying causes of ID and ASDs is not yet available in the clinic, but preclinical trials are going on in many laboratories. Drugs are, nevertheless, used to provide symptomatic relief from anxiety, epilepsy, depression, and cognitive and social dysfunctions in ID/ASDs. Response to already existing treatment options is known to be variable and is reported to have some side effects owing to their off-target interactions [29,30]. Coadministration of different medications with irrational prescription and use was another concern that affects people with ID/ASDs. More often than not, polypharmacy is a common practice and may lead to adverse drug reactions [31,32].

Although some of the currently followed strategies have prolonged the life of patients, their quality of life has not improved substantially [33]. Hence, there is a need to search for a new therapeutic intervention which can provide a cure, or at least alleviate the symptoms better than the existing therapeutic strategies for patients with ASDs and ID.

Different mutations, as discussed earlier, and drugs targeting these had undergone rigorous testing in preclinical trials on different mouse models. These have provided insights into the underlying mechanisms associated with the efficacy of these drugs for the treatment of ID/ASDs. Some of these approaches are summarized in table 2. However, their translational success is yet to be validated in clinical trials.

Table 2.

Upcoming therapeutic approaches for the treatment of ID. This includes drugs which have shown some promise in the preclinical studies for the treatment of ID. HMG-Co-A, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase.

s.no. drug known mechanism of action structure approval status target in ID references
1 lovastatin statins (HMG-Co-A inhibitor) graphic file with name rsob-9-180265-i1.jpg approved for hypercholesterolaemia FMRP and SYNGAP1-mediated RAS-ERK1/2 activation [32,34]
2 rapamycin mTOR inhibitor graphic file with name rsob-9-180265-i2.jpg approved as an immunomodulator the modulator of SYNGAP1-mediated mTOR signalling [33]
3 ganaxolone the positive allosteric modulator of GABAA receptors graphic file with name rsob-9-180265-i3.jpg phase 3 clinical trial for CDKL5 deficiency disorder the positive modulator of FMRP-mediated GABAA receptor expression [30]
4 valproate inhibitor of GABA transaminase and that of voltage-gated Na+ channels graphic file with name rsob-9-180265-i4.jpg approved for mania and epilepsy rescues from the SHANK-3 overexpression-mediated manic-like behaviour [35]
5 oxytocin peptide hormone which plays a role in milk ejection Cys – Tyr – Ile – Gln – Asn – Cys – Pro – Leu – Gly – NH2 approved for improvement or facilitation of uterine contractions during birth reverses the behavioural deficits in Shank-3-deficient mice [30]
6 D-cycloserine an antibiotic which is an inhibitor of the bacterial cell wall synthesis graphic file with name rsob-9-180265-i5.jpg approved antibiotic for the treatment of tuberculosis reverses the repetitive and stereotyped grooming behavioural deficits in Neuroligin 1 knockout mice [36]

9. Conclusion

ID and ASDs are prevalent NDDs that have proven to be complicated and challenging in several aspects. Since they often present with a highly variable and overlapping spectrum of symptoms and syndromes, defining a distinct set of diagnostic criteria has been difficult for clinicians and scientists. However, studies in mouse models of monogenic causes of ID and ASDs have proved to be immensely helpful in the construction of the pathophysiology of these disorders, through a bottom-up approach. These studies have demonstrated that ID/ASDs with diverse causal origins have intersecting aetiologies that might be responsible for the observed shared phenotypes. Opposing cellular phenotypes observed in these disorders highlight the importance and need for balanced and timely developmental processes at all systemic levels.

Further studies have employed these aspects for the development of genetic and pharmacological therapeutic strategies (creation of mouse models with counteracting mutations to re-establish balance and the testing of drugs targeting common neurological pathways). However, only a fraction have been uncovered in the understanding of these disorders, and require further studies for improved diagnosis, treatment and prognosis. As highlighted in this review, there are several areas which remain unexplored and could play an essential role in the pathophysiology of ID/ASDs. Additionally, the role of non-neuronal cells such as astrocytes, oligodendrocytes and microglia have also not been studied in detail concerning mutations in ID/ASDs. The augmented critical period is another characteristic modality altered in many forms of ID/ASDs. The study of precise mechanisms for a better understanding of this phase of development could be useful for rescue during the later period of life. Because the diagnosis is delayed during a critical period of development, reversing neuronal connections becomes difficult, which is one of the significant questions lingering in the minds of neuroscientists. Preclinical studies in this regard can warrant some useful clues for the translational success of small molecules being tested for efficacy in ID/ASDs. However, many drugs still fail in clinical trials even after ameliorating disease pathology in the preclinical mouse models. Poor experimental design with inadequate sample size could be one of the reasons for failure at the later stages.

Another critical point is the variability in the intrinsic metabolic and biochemical pathways among different animal strains and species that lead to changes in drug pharmacokinetics and pharmacodynamics across systems. These factors influence how a potential therapeutic candidate molecule can be metabolized by the animal model, and how this is different in human beings. One viable alternative to overcome the above issues is to use patient-derived IPSCs, which have been considered as a model in the last decade or so. However, to acquire an all-round understanding of ID/ASDs, it is crucial to study in vivo, i.e. animal models in combination with patient-derived IPSCs. Such a combinatorial study can fulfil the existing gap in our knowledge about ID/ASDs and show the way for future therapeutic strategies.

Supplementary Material

Reviewer comments

Data accessibility

This article does not contain any additional data.

Authors' contribution

V.V., A.P., A.A.V. and B.V. contributed equally to this review. V.V. wrote the sections related to NEUROLIGINS and the critical period of development; A.P. wrote the sections related to MECP2 and SHANK mutation, A.A.V. wrote the sections related to the FMRP, aetiology, contribution of non-neuronal cells and conclusion part of the review. B.V. wrote the sections pertaining to the introduction, SYNGAP1, animal models of ID, metaplasticity and therapeutic approaches. All authors contributed equally to the figures and tables. J.P.C. edited the manuscript. All the authors provided the conceptual idea for the review.

Competing interests

All authors declare no conflict of interest.

Funding

The work was supported by grants to J.P.C. by DST-SERB (SERB/JC/4518), DBT-JNCASR (LSRET-JNC/JC/4531) and intramural funding from the Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.

References

  • 1.Azevedo FA, Carvalho LR, Grinberg LT, Farfel JM, Ferretti RE, Leite RE, Filho WJ, Lent R, Herculano-Houzel S. 2009. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J. Comp. Neurol. 513, 532–541. ( 10.1002/cne.21974) [DOI] [PubMed] [Google Scholar]
  • 2.Carninci P, et al. 2005. The transcriptional landscape of the mammalian genome. Science 309, 1559–1563. ( 10.1126/science.1112014) [DOI] [PubMed] [Google Scholar]
  • 3.American Psychiatric Association. 2013. Diagnostic and statistical manual of mental disorders; DSM-V. Washington, DC: American Psychiatric Association. [Google Scholar]
  • 4.Yoo H. 2015. Genetics of autism spectrum disorder: current status and possible clinical applications. Exp. Neurobiol. 24, 257–272. ( 10.5607/en.2015.24.4.257) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kohane IS, et al. 2012. The co-morbidity burden of children and young adults with autism spectrum disorders. PLoS ONE 7, e33224 ( 10.1371/journal.pone.0033224) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rapoport J, Chavez A, Greenstein D, Addington A, Gogtay N. 2009. Autism spectrum disorders and childhood-onset schizophrenia: clinical and biological contributions to a relation revisited. J. Am. Acad. Child Adolesc. Psychiatry 48, 10–18. ( 10.1097/CHI.0b013e31818b1c63) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hsiao EY, McBride SW, Chow J, Mazmanian SK, Patterson PH. 2012. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl Acad. Sci. USA 109, 12 776–12 781. ( 10.1073/pnas.1202556109) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schieve LA, Gonzalez V, Boulet SL, Visser SN, Rice CE, Braun KVN, Boyle CA. 2012. Concurrent medical conditions and health care use and needs among children with learning and behavioral developmental disabilities, National Health Interview Survey, 2006–2010. Res. Dev. Disabil. 33, 467–476. ( 10.1016/j.ridd.2011.10.008) [DOI] [PubMed] [Google Scholar]
  • 9.Weiler IJ, et al. 1997. Fragile X mental retardation protein is translated near synapses in response to neurotransmitter activation. Proc. Natl Acad. Sci. USA 94, 5395–5400. ( 10.1073/pnas.94.10.5395) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kim JH, Liao D, Lau L-F, Huganir RL. 1998. SynGAP: a synaptic RasGAP that associates with the PSD-95/SAP90 protein family. Neuron 20, 683–691. ( 10.1016/S0896-6273(00)81008-9) [DOI] [PubMed] [Google Scholar]
  • 11.Hamdan FF, et al. 2011. Excess of de novo deleterious mutations in genes associated with glutamatergic systems in nonsyndromic intellectual disability. Am. J. Hum. Genet. 88, 306–316. ( 10.1016/j.ajhg.2011.02.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gomot M, et al. 2003. MECP2 gene mutations in non-syndromic X-linked mental retardation: phenotype–genotype correlation. Am. J. Med. Genet. A 123, 129–139. ( 10.1002/ajmg.a.20247) [DOI] [PubMed] [Google Scholar]
  • 13.Christensen J, Grønborg TK, Sørensen MJ, Schendel D, Parner ET, Pedersen LH, Vestergaard M. 2013. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA 309, 1696–1703. ( 10.1001/jama.2013.2270) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mukherjee R, Layton M, Yacoub E, Turk J. 2011. Autism and autistic traits in people exposed to heavy prenatal alcohol: data from a clinical series of 21 individuals and nested case control study. Adv. Ment. Health Intellect. Disabil 5, 42–49. ( 10.5042/amhid.2011.0015) [DOI] [Google Scholar]
  • 15.Gorini F, Muratori F, Morales MA. 2014. The role of heavy metal pollution in neurobehavioral disorders: a focus on autism. Rev. J. Autism Dev. Disord. 1, 354–372. ( 10.1007/s40489-014-0028-3) [DOI] [Google Scholar]
  • 16.Kawicka A, Regulska-Ilow B. 2013. How nutritional status, diet and dietary supplements can affect autism. A review. Roczniki Państwowego Zakładu Higieny 64, 1–12. [PubMed] [Google Scholar]
  • 17.Deth R, Muratore C, Benzecry J, Power-Charnitsky V-A, Waly M. 2008. How environmental and genetic factors combine to cause autism: a redox/methylation hypothesis. Neurotoxicology 29, 190–201. ( 10.1016/j.neuro.2007.09.010) [DOI] [PubMed] [Google Scholar]
  • 18.Barnes SA, et al. 2015. Convergence of hippocampal pathophysiology in Syngap+/− and Fmr1−/y mice. J. Neurosci. 35, 15 073–15 081. ( 10.1523/JNEUROSCI.1087-15.2015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Aceti M, et al. 2015. Syngap1 haploinsufficiency damages a postnatal critical period of pyramidal cell structural maturation linked to cortical circuit assembly. Biol. Psychiatry 77, 805–815. ( 10.1016/j.biopsych.2014.08.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Clarke LE, Barres BA. 2013. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321. ( 10.1038/nrn3484) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Higashimori H, Schin CS, Chiang MS, Morel L, Shoneye TA, Nelson DL, Yang Y. 2016. Selective deletion of astroglial FMRP dysregulates glutamate transporter GLT1 and contributes to fragile X syndrome phenotypes in vivo. J. Neurosci. 36, 7079–7094. ( 10.1523/JNEUROSCI.1069-16.2016) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.van der Staay FJ, Arndt SS, Nordquist RE. 2009. Evaluation of animal models of neurobehavioral disorders. Behav. Brain Funct. 5, 11 ( 10.1186/1744-9081-5-11) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bitta M, Kariuki SM, Abubakar A, Newton CR. 2017. Burden of neurodevelopmental disorders in low and middle-income countries: a systematic review and meta-analysis. Wellcome Open Res. 2, 121 ( 10.12688/wellcomeopenres.13540.3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Maulik PK, Mascarenhas MN, Mathers CD, Dua T, Saxena S. 2011. Prevalence of intellectual disability: a meta-analysis of population-based studies. Res. Dev. Disabil. 32, 419–436. ( 10.1016/j.ridd.2010.12.018) [DOI] [PubMed] [Google Scholar]
  • 25.Boyle CA, Boulet S, Schieve LA, Cohen RA, Blumberg SJ, Yeargin-Allsopp M, Visser S, Kogan MD. 2011. Trends in the prevalence of developmental disabilities in US children, 1997–2008. Pediatrics. 127, 1034–1042. ( 10.1542/peds.2010-2989) [DOI] [PubMed] [Google Scholar]
  • 26.World Health Organization. Autism spectrum disorders. 2017. [cited 2018 8th December]. See https://www.who.int/news-room/fact-sheets/detail/autism-spectrum-disorders
  • 27.Pushpakom S, et al. 2018. Drug repurposing: progress, challenges and recommendations. Nat. Rev. Drug Discov. 18, 41–58. ( 10.1038/nrd.2018.168) [DOI] [PubMed] [Google Scholar]
  • 28.Zoghbi HY, Bear MF. 2012. Synaptic dysfunction in neurodevelopmental disorders associated with autism and intellectual disabilities. Cold Spring Harb. Perspect. Biol. 4, a009886 ( 10.1101/cshperspect.a009886) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Moog U. 2005. The outcome of diagnostic studies on the etiology of mental retardation: considerations on the classification of the causes. Am. J. Med. Genet. A 137, 228–231. ( 10.1002/ajmg.a.30841) [DOI] [PubMed] [Google Scholar]
  • 30.Ellison JW, Rosenfeld JA, Shaffer LG. 2013. Genetic basis of intellectual disability. Annu. Rev. Med. 64, 441–450. ( 10.1146/annurev-med-042711-140053) [DOI] [PubMed] [Google Scholar]
  • 31.Srivastava AK, Schwartz CE. 2014. Intellectual disability and autism spectrum disorders: causal genes and molecular mechanisms. Neurosci. Biobehav. Rev. 46(Pt 2), 161–174. ( 10.1016/j.neubiorev.2014.02.015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Srour M, Shevell M. 2014. Genetics and the investigation of developmental delay/intellectual disability. Arch. Dis. Child. 99, 386–389. ( 10.1136/archdischild-2013-304063) [DOI] [PubMed] [Google Scholar]
  • 33.Katz G, Lazcano-Ponce E. 2008. Intellectual disability: definition, etiological factors, classification, diagnosis, treatment and prognosis. Salud Publica Mex. 50(Suppl. 2), s132–s141. [DOI] [PubMed] [Google Scholar]
  • 34.Reichenberg A, et al. 2016. Discontinuity in the genetic and environmental causes of the intellectual disability spectrum. Proc. Natl Acad. Sci. USA 113, 1098–1103. ( 10.1073/pnas.1508093112) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vissers LE, Gilissen C, Veltman JA. 2016. Genetic studies in intellectual disability and related disorders. Nat. Rev. Genet. 17, 9–18. ( 10.1038/nrg3999) [DOI] [PubMed] [Google Scholar]
  • 36.Pinto D, et al. 2010. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 466, 368–372. ( 10.1038/nature09146) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kaufman L, Ayub M, Vincent JB. 2010. The genetic basis of non-syndromic intellectual disability: a review. J. Neurodev. Disord. 2, 182–209. ( 10.1007/s11689-010-9055-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chelly J, Mandel JL. 2001. Monogenic causes of X-linked mental retardation. Nat. Rev. Genet. 2, 669–680. ( 10.1038/35088558) [DOI] [PubMed] [Google Scholar]
  • 39.Bliss TV, Collingridge GL. 1993. A synaptic model of memory: long-term potentiation in the hippocampus. Nature 361, 31–39. ( 10.1038/361031a0) [DOI] [PubMed] [Google Scholar]
  • 40.Levitt P. 2003. Structural and functional maturation of the developing primate brain. J. Pediatr. 143, S35–S45. ( 10.1067/S0022-3476(03)00400-1) [DOI] [PubMed] [Google Scholar]
  • 41.Wiesel TN, Hubel DH. 1963. Single-cell responses in striate cortex of kittens deprived of vision in one eye. J. Neurophysiol. 26, 1003–1017. ( 10.1152/jn.1963.26.6.1003) [DOI] [PubMed] [Google Scholar]
  • 42.Stiles J, Jernigan TL. 2010. The basics of brain development. Neuropsychol. Rev. 20, 327–348. ( 10.1007/s11065-010-9148-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hensch TK. 2005. Critical period plasticity in local cortical circuits. Nat. Rev. Neurosci. 6, 877–888. ( 10.1038/nrn1787) [DOI] [PubMed] [Google Scholar]
  • 44.Hensch TK, Bilimoria PM. 2012. Re-opening windows: manipulating critical periods for brain development. Cerebrum 2012, 11. [PMC free article] [PubMed] [Google Scholar]
  • 45.Hubel DH. 1957. Tungsten microelectrode for recording from single units. Science 125, 549–550. ( 10.1126/science.125.3247.549) [DOI] [PubMed] [Google Scholar]
  • 46.Hubel DH, Wiesel TN. 1968. Receptive fields and functional architecture of monkey striate cortex. J. Physiol. 195, 215–243. ( 10.1113/jphysiol.1968.sp008455) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hubel DH, Wiesel TN. 1970. The period of susceptibility to the physiological effects of unilateral eye closure in kittens. J. Physiol. 206, 419–436. ( 10.1113/jphysiol.1970.sp009022) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hubel DH, Wiesel TN. 1962. Receptive fields, binocular interaction and functional architecture in the cat's visual cortex. J. Physiol. 160, 106–154. ( 10.1113/jphysiol.1962.sp006837) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Horton JC, Hocking DR. 1997. Timing of the critical period for plasticity of ocular dominance columns in macaque striate cortex. J. Neurosci. 17, 3684–3709. ( 10.1523/JNEUROSCI.17-10-03684.1997) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Smith SL, Trachtenberg JT. 2007. Experience-dependent binocular competition in the visual cortex begins at eye opening. Nat. Neurosci. 10, 370–375. ( 10.1038/nn1844) [DOI] [PubMed] [Google Scholar]
  • 51.Crowley JC, Katz LC. 2000. Early development of ocular dominance columns. Science 290, 1321–1324. ( 10.1126/science.290.5495.1321) [DOI] [PubMed] [Google Scholar]
  • 52.Faguet J, Maranhao B, Smith SL, Trachtenberg JT. 2009. Ipsilateral eye cortical maps are uniquely sensitive to binocular plasticity. J. Neurophysiol. 101, 855–861. ( 10.1152/jn.90893.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Espinosa JS, Stryker MP. 2012. Development and plasticity of the primary visual cortex. Neuron 75, 230–249. ( 10.1016/j.neuron.2012.06.009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Erzurumlu RS, Gaspar P. 2012. Development and critical period plasticity of the barrel cortex. Eur. J. Neurosci. 35, 1540–1553. ( 10.1111/j.1460-9568.2012.08075.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lendvai B, Stern EA, Chen B, Svoboda K. 2000. Experience-dependent plasticity of dendritic spines in the developing rat barrel cortex in vivo. Nature 404, 876–881. ( 10.1038/35009107) [DOI] [PubMed] [Google Scholar]
  • 56.Maravall M, Petersen RS, Fairhall AL, Arabzadeh E, Diamond ME. 2007. Shifts in coding properties and maintenance of information transmission during adaptation in barrel cortex. PLoS Biol. 5, e19 ( 10.1371/journal.pbio.0050019) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Shoykhet M, Land PW, Simons DJ. 2005. Whisker trimming begun at birth or on postnatal day 12 affects excitatory and inhibitory receptive fields of layer IV barrel neurons. J. Neurophysiol. 94, 3987–3995. ( 10.1152/jn.00569.2005) [DOI] [PubMed] [Google Scholar]
  • 58.Cull-Candy S, Brickley S, Farrant M. 2001. NMDA receptor subunits: diversity, development and disease. Curr. Opin Neurobiol. 11, 327–335. ( 10.1016/S0959-4388(00)00215-4) [DOI] [PubMed] [Google Scholar]
  • 59.Livingston FS, Mooney R. 1997. Development of intrinsic and synaptic properties in a forebrain nucleus essential to avian song learning. J. Neurosci. 17, 8997–9009. ( 10.1523/JNEUROSCI.17-23-08997.1997) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Flint AC, Maisch US, Weishaupt JH, Kriegstein AR, Monyer H. 1997. NR2A subunit expression shortens NMDA receptor synaptic currents in developing neocortex. J. Neurosci. 17, 2469–2476. ( 10.1523/JNEUROSCI.17-07-02469.1997) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Roberts EB, Ramoa AS. 1999. Enhanced NR2A subunit expression and decreased NMDA receptor decay time at the onset of ocular dominance plasticity in the ferret. J. Neurophysiol. 81, 2587–2591. ( 10.1152/jn.1999.81.5.2587) [DOI] [PubMed] [Google Scholar]
  • 62.Carmignoto G, Vicini S. 1992. Activity-dependent decrease in NMDA receptor responses during development of the visual cortex. Science 258, 1007–1011. ( 10.1126/science.1279803) [DOI] [PubMed] [Google Scholar]
  • 63.Crair MC, Malenka RC. 1995. A critical period for long-term potentiation at thalamocortical synapses. Nature 375, 325–328. ( 10.1038/375325a0) [DOI] [PubMed] [Google Scholar]
  • 64.Lu W, Man H, Ju W, Trimble WS, MacDonald JF, Wang YT. 2001. Activation of synaptic NMDA receptors induces membrane insertion of new AMPA receptors and LTP in cultured hippocampal neurons. Neuron 29, 243–254. ( 10.1016/S0896-6273(01)00194-5) [DOI] [PubMed] [Google Scholar]
  • 65.Agmon A, O'Dowd DK. 1992. NMDA receptor-mediated currents are prominent in the thalamocortical synaptic response before maturation of inhibition. J. Neurophysiol. 68, 345–349. ( 10.1152/jn.1992.68.1.345) [DOI] [PubMed] [Google Scholar]
  • 66.Ben-Ari Y. 2014. The GABA excitatory/inhibitory developmental sequence: a personal journey. Neuroscience 279, 187–219. ( 10.1016/j.neuroscience.2014.08.001) [DOI] [PubMed] [Google Scholar]
  • 67.Fagiolini M, Hensch TK. 2000. Inhibitory threshold for critical-period activation in primary visual cortex. Nature 404, 183–186. ( 10.1038/35004582) [DOI] [PubMed] [Google Scholar]
  • 68.Sipila ST, Huttu K, Yamada J, Afzalov R, Voipio J, Blaesse P, Kaila K. 2009. Compensatory enhancement of intrinsic spiking upon NKCC1 disruption in neonatal hippocampus. J. Neurosci. 29, 6982–6988. ( 10.1523/JNEUROSCI.0443-09.2009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hubner CA, Stein V, Hermans-Borgmeyer I, Meyer T, Ballanyi K, Jentsch TJ. 2001. Disruption of KCC2 reveals an essential role of K-Cl cotransport already in early synaptic inhibition. Neuron 30, 515–524. ( 10.1016/S0896-6273(01)00297-5) [DOI] [PubMed] [Google Scholar]
  • 70.Baroncelli L, Braschi C, Spolidoro M, Begenisic T, Maffei L, Sale A. 2011. Brain plasticity and disease: a matter of inhibition. Neural Plast. 2011, 286073 ( 10.1155/2011/286073) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bavelier D, Levi DM, Li RW, Dan Y, Hensch TK. 2010. Removing brakes on adult brain plasticity: from molecular to behavioral interventions. J. Neurosci. 30, 14 964–14 971. ( 10.1523/JNEUROSCI.4812-10.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hensch TK, Fagiolini M, Mataga N, Stryker MP, Baekkeskov S, Kash SF. 1998. Local GABA circuit control of experience-dependent plasticity in developing visual cortex. Science 282, 1504–1508. ( 10.1126/science.282.5393.1504) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hanover JL, Huang ZJ, Tonegawa S, Stryker MP. 1999. Brain-derived neurotrophic factor overexpression induces precocious critical period in mouse visual cortex. J. Neurosci. 19, RC40 ( 10.1523/JNEUROSCI.19-22-j0003.1999) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kanold PO. 2009. Subplate neurons: crucial regulators of cortical development and plasticity. Front. Neuroanat. 3, 16 ( 10.3389/neuro.05.016.2009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Southwell DG, Froemke RC, Alvarez-Buylla A, Stryker MP, Gandhi SP. 2010. Cortical plasticity induced by inhibitory neuron transplantation. Science 327, 1145–1148. ( 10.1126/science.1183962) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Toyoizumi T, Miyamoto H, Yazaki-Sugiyama Y, Atapour N, Hensch TK, Miller KD. 2013. A theory of the transition to critical period plasticity: inhibition selectively suppresses spontaneous activity. Neuron 80, 51–63. ( 10.1016/j.neuron.2013.07.022) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Huang ZJ, Kirkwood A, Pizzorusso T, Porciatti V, Morales B, Bear MF, Maffei L, Tonegawa S. 1999. BDNF regulates the maturation of inhibition and the critical period of plasticity in mouse visual cortex. Cell 98, 739–755. ( 10.1016/S0092-8674(00)81509-3) [DOI] [PubMed] [Google Scholar]
  • 78.Andersen ML, Winter LM. 2017. Animal models in biological and biomedical research-experimental and ethical concerns. An. Acad. Bras. Cienc. 91(Suppl. 1), e20170238 ( 10.1590/0001-3765201720170238) [DOI] [PubMed] [Google Scholar]
  • 79.Erickson CA, et al. 2017. Fragile X targeted pharmacotherapy: lessons learned and future directions. J. Neurodev. Disord. 9, 7 ( 10.1186/s11689-017-9186-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Orban PC, Chui D, Marth JD. 1992. Tissue-and site-specific DNA recombination in transgenic mice. Proc. Natl Acad. Sci. USA 89, 6861–6865. ( 10.1073/pnas.89.15.6861) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Doetschman T, Azhar M. 2012. Cardiac-specific inducible and conditional gene targeting in mice. Circ. Res. 110, 1498–1512. ( 10.1161/CIRCRESAHA.112.265066) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jeyabalan N, Clement JP. 2016. SYNGAP1: mind the gap. Front. Cell. Neurosci. 10, 32 ( 10.3389/fncel.2016.00032) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Scorza CA, Cavalheiro EA. 2011. Animal models of intellectual disability: towards a translational approach. Clinics 66, 55–63. ( 10.1590/S1807-59322011001300007) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ogden KK, Ozkan ED, Rumbaugh G. 2016. Prioritizing the development of mouse models for childhood brain disorders. Neuropharmacology 100, 2–16. ( 10.1016/j.neuropharm.2015.07.029) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Guo X, Hamilton PJ, Reish NJ, Sweatt JD, Miller CA, Rumbaugh G. 2009. Reduced expression of the NMDA receptor-interacting protein SynGAP causes behavioral abnormalities that model symptoms of schizophrenia. Neuropsychopharmacology 34, 1659–1672. ( 10.1038/npp.2008.223) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kim JH, Lee H-K, Takamiya K, Huganir RL. 2003. The role of synaptic GTPase-activating protein in neuronal development and synaptic plasticity. J. Neurosci. 23, 1119–1124. ( 10.1523/JNEUROSCI.23-04-01119.2003) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Vazquez LE, Chen H-J, Sokolova I, Knuesel I, Kennedy MB. 2004. SynGAP regulates spine formation. J. Neurosci. 24, 8862–8872. ( 10.1523/JNEUROSCI.3213-04.2004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Clement JP, et al. 2012. Pathogenic SYNGAP1 mutations impair cognitive development by disrupting maturation of dendritic spine synapses. Cell 151, 709–723. ( 10.1016/j.cell.2012.08.045) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Oostra B, Bakker C, Reyniers E. 1994. FMR1 knockout mice: a model to study fragile X mental retardation. Am. J. Hum. Genet. 55, 23–33. [Google Scholar]
  • 90.Dahlhaus R. 2018. Of men and mice: modeling the fragile X syndrome. Front. Mol. Neurosci. 11, 41 ( 10.3389/fnmol.2018.00041) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mientjes E, et al. 2006. The generation of a conditional Fmr1 knock out mouse model to study Fmrp function in vivo. Neurobiol. Dis. 21, 549–555. ( 10.1016/j.nbd.2005.08.019) [DOI] [PubMed] [Google Scholar]
  • 92.Gaudissard J, Ginger M, Premoli M, Memo M, Frick A, Pietropaolo S. 2017. Behavioral abnormalities in the Fmr1-KO2 mouse model of fragile X syndrome: the relevance of early life phases. Autism Res. 10, 1584–1596. ( 10.1002/aur.1814) [DOI] [PubMed] [Google Scholar]
  • 93.Wijetunge LS, Angibaud J, Frick A, Kind PC, Nägerl UV. 2014. Stimulated emission depletion (STED) microscopy reveals nanoscale defects in the developmental trajectory of dendritic spine morphogenesis in a mouse model of fragile X syndrome. J. Neurosci. 34, 6405–6412. ( 10.1523/JNEUROSCI.5302-13.2014) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Koekkoek S, et al. 2005. Deletion of FMR1 in Purkinje cells enhances parallel fiber LTD, enlarges spines, and attenuates cerebellar eyelid conditioning in fragile X syndrome. Neuron. 47, 339–352. ( 10.1016/j.neuron.2005.07.005) [DOI] [PubMed] [Google Scholar]
  • 95.Guo W, et al. 2011. Ablation of Fmrp in adult neural stem cells disrupts hippocampus-dependent learning. Nat. Med. 17, 559–565. ( 10.1038/nm.2336) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.El-Khoury R, Panayotis N, Matagne V, Ghata A, Villard L, Roux J-C. 2014. GABA and glutamate pathways are spatially and developmentally affected in the brain of Mecp2-deficient mice. PLoS ONE 9, e92169 ( 10.1371/journal.pone.0092169) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Blue ME, Kaufmann WE, Bressler J, Eyring C, O'Driscoll C, Naidu S, Johnston MV. 2011. Temporal and regional alterations in NMDA receptor expression in Mecp2-null mice. Anat. Rec. 294, 1624–1634. ( 10.1002/ar.21380) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Guy J, Hendrich B, Holmes M, Martin JE, Bird A. 2001. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 27, 322–326. ( 10.1038/85899) [DOI] [PubMed] [Google Scholar]
  • 99.Krishnan K, Wang B-S, Lu J, Wang L, Maffei A, Cang J, Huang ZJ. 2015. MeCP2 regulates the timing of critical period plasticity that shapes functional connectivity in primary visual cortex. Proc. Natl Acad. Sci. USA 112, E4782–E4791. ( 10.1073/pnas.1506499112) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Abdala AP, Dutschmann M, Bissonnette JM, Paton JF. 2010. Correction of respiratory disorders in a mouse model of Rett syndrome. Proc. Natl Acad. Sci. USA 107, 18 208–18 213. ( 10.1073/pnas.1012104107) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Carouge D, Host L, Aunis D, Zwiller J, Anglard P. 2010. CDKL5 is a brain MeCP2 target gene regulated by DNA methylation. Neurobiol. Dis. 38, 414–424. ( 10.1016/j.nbd.2010.02.014) [DOI] [PubMed] [Google Scholar]
  • 102.Singleton MK, Gonzales ML, Leung KN, Yasui DH, Schroeder DI, Dunaway K, LaSalle JM. 2011. MeCP2 is required for global heterochromatic and nucleolar changes during activity-dependent neuronal maturation. Neurobiol. Dis. 43, 190–200. ( 10.1016/j.nbd.2011.03.011) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Samaco RC, et al. 2009. Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities. Proc. Natl Acad. Sci. USA 106, 21 966–21 971. ( 10.1073/pnas.0912257106) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Luikenhuis S, Giacometti E, Beard CF, Jaenisch R. 2004. Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. Proc. Natl Acad. Sci. USA 101, 6033–6038. ( 10.1073/pnas.0401626101) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lawson-Yuen A, Liu D, Han L, Jiang ZI, Tsai GE, Basu AC, Picker J, Feng J, Coyle JT. 2007. Ube3a mRNA and protein expression are not decreased in Mecp2R168X mutant mice. Brain Res. 1180, 1–6. ( 10.1016/j.brainres.2007.08.039) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang X, et al. 2011. Synaptic dysfunction and abnormal behaviors in mice lacking major isoforms of Shank3. Hum. Mol. Genet. 20, 3093–3108. ( 10.1093/hmg/ddr212) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bozdagi O, et al. 2010. Haploinsufficiency of the autism-associated Shank3 gene leads to deficits in synaptic function, social interaction, and social communication. Mol. Autism 1, 15 ( 10.1186/2040-2392-1-15) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Peca J, Feliciano C, Ting JT, Wang W, Wells MF, Venkatraman TN, Lascola CD, Fu Z, Feng G. 2011. Shank3 mutant mice display autistic-like behaviours and striatal dysfunction. Nature 472, 437–442. ( 10.1038/nature09965) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tabuchi K, Blundell J, Etherton MR, Hammer RE, Liu X, Powell CM, Sudhof TC. 2007. A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 318, 71–76. ( 10.1126/science.1146221) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Etherton M, Foldy C, Sharma M, Tabuchi K, Liu X, Shamloo M, Malenka RC, Sudhof TC. 2011. Autism-linked neuroligin-3 R451C mutation differentially alters hippocampal and cortical synaptic function. Proc. Natl Acad. Sci. USA 108, 13 764–13 769. ( 10.1073/pnas.1111093108) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pizzarelli R, Cherubini E. 2013. Developmental regulation of GABAergic signalling in the hippocampus of neuroligin 3 R451C knock-in mice: an animal model of autism. Front. Cell. Neurosci. 7, 85 (doi:0.3389/fncel.2013.00085) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cellot G, Cherubini E. 2014. Reduced inhibitory gate in the barrel cortex of Neuroligin3R451C knock-in mice, an animal model of autism spectrum disorders. Physiol Rep. 2, e12077 ( 10.14814/phy2.12077) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Foldy C, Malenka RC, Sudhof TC. 2013. Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron 78, 498–509. ( 10.1016/j.neuron.2013.02.036) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Rothwell PE, Fuccillo MV, Maxeiner S, Hayton SJ, Gokce O, Lim BK, Fowler SC, Malenka RC, Sudhof TC. 2014. Autism-associated neuroligin-3 mutations commonly impair striatal circuits to boost repetitive behaviors. Cell 158, 198–212. ( 10.1016/j.cell.2014.04.045) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Etherton MR, Tabuchi K, Sharma M, Ko J, Sudhof TC. 2011. An autism-associated point mutation in the neuroligin cytoplasmic tail selectively impairs AMPA receptor-mediated synaptic transmission in hippocampus. EMBO J. 30, 2908–2919. ( 10.1038/emboj.2011.182) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Chen H-J, Rojas-Soto M, Oguni A, Kennedy MB. 1998. A synaptic Ras-GTPase activating protein (p135 SynGAP) inhibited by CaM kinase II. Neuron 20, 895–904. ( 10.1016/S0896-6273(00)80471-7) [DOI] [PubMed] [Google Scholar]
  • 117.McMahon A, et al. 2012. SynGAP isoforms exert opposing effects on synaptic strength. Nat. Commun. 3, 900 ( 10.1038/ncomms1900) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Parker MJ, et al. 2015. De novo, heterozygous, loss-of-function mutations in SYNGAP1 cause a syndromic form of intellectual disability. Am. J. Med. Genet. A 167, 2231–2237. ( 10.1002/ajmg.a.37189) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Ozkan ED, et al. 2014. Reduced cognition in Syngap1 mutants is caused by isolated damage within developing forebrain excitatory neurons. Neuron 82, 1317–1333. ( 10.1016/j.neuron.2014.05.015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Muhia M, Willadt S, Yee BK, Feldon J, Paterna J-C, Schwendener S, Vogt K, Kennedy MB, Knuesel I. 2012. Molecular and behavioral changes associated with adult hippocampus-specific SynGAP1 knockout. Learn. Mem. 19, 268–281. ( 10.1101/lm.026351.112) [DOI] [PubMed] [Google Scholar]
  • 121.Muhia M, Yee BK, Feldon J, Markopoulos F, Knuesel I. 2010. Disruption of hippocampus-regulated behavioural and cognitive processes by heterozygous constitutive deletion of SynGAP. Eur. J. Neurosci. 31, 529–543. ( 10.1111/j.1460-9568.2010.07079.x) [DOI] [PubMed] [Google Scholar]
  • 122.Kilinc M, Creson T, Rojas C, Aceti M, Ellegood J, Vaissiere T, Lerch JP, Rumbaugh G. 2018. Species-conserved SYNGAP1 phenotypes associated with neurodevelopmental disorders. Mol. Cell. Neurosci. 91, 140–150. ( 10.1016/j.mcn.2018.03.008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hamdan FF, et al. 2009. Mutations in SYNGAP1 in autosomal nonsyndromic mental retardation. N. Engl. J. Med. 360, 599–605. ( 10.1056/NEJMoa0805392) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mignot C, et al. 2016. Genetic and neurodevelopmental spectrum of SYNGAP1-associated intellectual disability and epilepsy. J. Med. Genet. 53, 511–522. ( 10.1136/jmedgenet-2015-103451) [DOI] [PubMed] [Google Scholar]
  • 125.Berryer MH, et al. 2016. Decrease of SYNGAP1 in GABAergic cells impairs inhibitory synapse connectivity, synaptic inhibition and cognitive function. Nat. Commun. 7, 13340 ( 10.1038/ncomms13340) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Michaelson SD, et al. 2018. SYNGAP1 heterozygosity disrupts sensory processing by reducing touch-related activity within somatosensory cortex circuits. Nat. Neurosci. 21, 1–13. ( 10.1038/s41593-018-0268-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Kimura Y, Akahira-Azuma M, Harada N, Enomoto Y, Tsurusaki Y, Kurosawa K. 2018. Novel SYNGAP1 variant in a patient with intellectual disability and distinctive dysmorphisms. Congenit. Anom. 58, 188–190. ( 10.1111/cga.12273) [DOI] [PubMed] [Google Scholar]
  • 128.Berryer MH, et al. 2013. Mutations in SYNGAP1 cause intellectual disability, autism, and a specific form of epilepsy by inducing haploinsufficiency. Hum. Mutat. 34, 385–394. ( 10.1002/humu.22248) [DOI] [PubMed] [Google Scholar]
  • 129.Writzl K, Knegt AC. 2013. 6p21.3 microdeletion involving the SYNGAP1 gene in a patient with intellectual disability, seizures, and severe speech impairment. Am. J. Med. Genet. A 161, 1682–1685. ( 10.1002/ajmg.a.35930) [DOI] [PubMed] [Google Scholar]
  • 130.von Stülpnagel C, Funke C, Haberl C, Hörtnagel K, Jüngling J, Weber YG, Staudt M, Kluger G. 2015. SYNGAP1 mutation in focal and generalized epilepsy: a literature overview and a case report with special aspects of the EEG. Neuropediatrics 46, 287–291. ( 10.1055/s-0035-1554098) [DOI] [PubMed] [Google Scholar]
  • 131.Lo-Castro A, Curatolo P. 2014. Epilepsy associated with autism and attention deficit hyperactivity disorder: is there a genetic link? Brain Dev. 36, 185–193. ( 10.1016/j.braindev.2013.04.013) [DOI] [PubMed] [Google Scholar]
  • 132.Prchalova D, Havlovicova M, Sterbova K, Stranecky V, Hancarova M, Sedlacek Z. 2017. Analysis of 31-year-old patient with SYNGAP1 gene defect points to importance of variants in broader splice regions and reveals developmental trajectory of SYNGAP1-associated phenotype: case report. BMC Med. Genet. 18, 62 ( 10.1186/s12881-017-0425-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dyment D, et al. 2015. Whole-exome sequencing broadens the phenotypic spectrum of rare pediatric epilepsy: a retrospective study. Clin. Genet. 88, 34–40. ( 10.1111/cge.12464) [DOI] [PubMed] [Google Scholar]
  • 134.Knuesel I, Elliott A, Chen HJ, Mansuy IM, Kennedy MB. 2005. A role for synGAP in regulating neuronal apoptosis. Eur. J. Neurosci. 21, 611–621. ( 10.1111/j.1460-9568.2005.03908.x) [DOI] [PubMed] [Google Scholar]
  • 135.Carlisle HJ, Manzerra P, Marcora E, Kennedy MB. 2008. SynGAP regulates steady-state and activity-dependent phosphorylation of cofilin. J. Neurosci. 28, 13 673–13 683. ( 10.1523/JNEUROSCI.4695-08.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Clement JP, Ozkan ED, Aceti M, Miller CA, Rumbaugh G. 2013. SYNGAP1 links the maturation rate of excitatory synapses to the duration of critical-period synaptic plasticity. J. Neurosci. 33, 10 447–10 452. ( 10.1523/JNEUROSCI.0765-13.2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Sheng M, Lee SH. 2001. AMPA receptor trafficking and the control of synaptic transmission. Cell 105, 825–828. ( 10.1016/S0092-8674(01)00406-8) [DOI] [PubMed] [Google Scholar]
  • 138.Komiyama NH, et al. 2002. SynGAP regulates ERK/MAPK signaling, synaptic plasticity, and learning in the complex with postsynaptic density 95 and NMDA receptor. J. Neurosci. 22, 9721–9732. ( 10.1523/JNEUROSCI.22-22-09721.2002) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Norris CM, Korol DL, Foster TC. 1996. Increased susceptibility to induction of long-term depression and long-term potentiation reversal during aging. J. Neurosci. 16, 5382–5392. ( 10.1523/JNEUROSCI.16-17-05382.1996) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Oh JS, Manzerra P, Kennedy MB. 2004. Regulation of the neuron-specific Ras GTPase activating protein, synGAP, by Ca2+/calmodulin-dependent protein kinase II. J. Biol. Chem. 279, 17 980–17 988. [DOI] [PubMed] [Google Scholar]
  • 141.Rumbaugh G, Adams JP, Kim JH, Huganir RL. 2006. SynGAP regulates synaptic strength and mitogen-activated protein kinases in cultured neurons. Proc. Natl Acad. Sci. USA 103, 4344–4351. ( 10.1073/pnas.0600084103) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Trachtenberg JT, Chen BE, Knott GW, Feng G, Sanes JR, Welker E, Svoboda K. 2002. Long-term in vivo imaging of experience-dependent synaptic plasticity in adult cortex. Nature 420, 788–794. ( 10.1038/nature01273) [DOI] [PubMed] [Google Scholar]
  • 143.Rousseau F, Rouillard P, Morel ML, Khandjian EW, Morgan K. 1995. Prevalence of carriers of premutation-size alleles of the FMRI gene—and implications for the population genetics of the fragile X syndrome. Am. J. Hum. Genet. 57, 1006–1018. [PMC free article] [PubMed] [Google Scholar]
  • 144.Hersh JH, Saul RA, Committee on Genetics . 2011. Health supervision for children with fragile X syndrome. Pediatrics 127, 994–1006. ( 10.1542/peds.2010-3500) [DOI] [PubMed] [Google Scholar]
  • 145.Martin JP, Bell J. 1943. A pedigree of mental defect showing sex-linkage. J. Neurol. Psychiatry 6, 154–157. ( 10.1136/jnnp.6.3-4.154) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wijetunge LS, Chattarji S, Wyllie DJ, Kind PC. 2013. Fragile X syndrome: from targets to treatments. Neuropharmacology 68, 83–96. ( 10.1016/j.neuropharm.2012.11.028) [DOI] [PubMed] [Google Scholar]
  • 147.Sherman SL, et al. 1985. Further segregation analysis of the fragile X syndrome with special reference to transmitting males. Hum. Genet. 69, 289–299. ( 10.1007/BF00291644) [DOI] [PubMed] [Google Scholar]
  • 148.Verkerk AJ, et al. 1991. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 65, 905–914. ( 10.1016/0092-8674(91)90397-h) [DOI] [PubMed] [Google Scholar]
  • 149.Sutcliffe JS, Nelson DL, Zhang F, Pieretti M, Caskey CT, Saxe D, Warren ST. 1992. DNA methylation represses FMR-1 transcription in fragile X syndrome. Hum. Mol. Genet. 1, 397–400. ( 10.1093/hmg/1.6.397) [DOI] [PubMed] [Google Scholar]
  • 150.Fu YH, et al. 1991. Variation of the CGG repeat at the fragile X site results in genetic instability: resolution of the Sherman paradox. Cell 67, 1047–1058. ( 10.1016/0092-8674(91)90283-5) [DOI] [PubMed] [Google Scholar]
  • 151.McMurray CT. 2010. Mechanisms of trinucleotide repeat instability during human development. Nat. Rev. Genet. 11, 786–799. ( 10.1038/nrg2828) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Hartenstine MJ, Goodman MF, Petruska J. 2000. Base stacking and even/odd behavior of hairpin loops in DNA triplet repeat slippage and expansion with DNA polymerase. J. Biol. Chem. 275, 18 382–18 390. ( 10.1074/jbc.275.24.18382) [DOI] [PubMed] [Google Scholar]
  • 153.Oberle I, Rousseau F, Heitz D, Kretz C, Devys D, Hanauer A, Boue J, Bertheas MF, Mandel JL. 1991. Instability of a 550-base pair DNA segment and abnormal methylation in fragile X syndrome. Science 252, 1097–1102. ( 10.2307/2876286) [DOI] [PubMed] [Google Scholar]
  • 154.Coffee B, Zhang F, Warren ST, Reines D. 1999. Acetylated histones are associated with FMR1 in normal but not fragile X-syndrome cells. Nat. Genet. 22, 98–101. ( 10.1038/8807) [DOI] [PubMed] [Google Scholar]
  • 155.Coffee B, Zhang F, Ceman S, Warren ST, Reines D. 2002. Histone modifications depict an aberrantly heterochromatinized FMR1 gene in fragile X syndrome. Am. J. Hum. Genet. 71, 923–932. ( 10.1086/342931) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pieretti M, Zhang FP, Fu YH, Warren ST, Oostra BA, Caskey CT, Nelson DL. 1991. Absence of expression of the FMR-1 gene in fragile X syndrome. Cell 66, 817–822. ( 10.1016/0092-8674(91)90125-i) [DOI] [PubMed] [Google Scholar]
  • 157.Sutherland GR. 1977. Fragile sites on human chromosomes: demonstration of their dependence on the type of tissue culture medium. Science 197, 265–266. ( 10.2307/1744517) [DOI] [PubMed] [Google Scholar]
  • 158.Heitz D, Devys D, Imbert G, Kretz C, Mandel JL. 1992. Inheritance of the fragile X syndrome: size of the fragile X premutation is a major determinant of the transition to full mutation. J. Med. Genet. 29, 794–801. ( 10.1136/jmg.29.11.794) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Jacquemont S, et al. 2004. Penetrance of the fragile X-associated tremor/ataxia syndrome in a premutation carrier population. JAMA 291, 460–469. ( 10.1001/jama.291.4.460) [DOI] [PubMed] [Google Scholar]
  • 160.Tassone F, Hagerman RJ, Garcia-Arocena D, Khandjian EW, Greco CM, Hagerman PJ. 2004. Intranuclear inclusions in neural cells with premutation alleles in fragile X associated tremor/ataxia syndrome. J. Med. Genet. 41, e43 ( 10.1136/jmg.2003.012518) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Santoro MR, Bray SM, Warren ST. 2012. Molecular mechanisms of fragile X syndrome: a twenty-year perspective. Annu. Rev. Pathol. 7, 219–245. ( 10.1146/annurev-pathol-011811-132457) [DOI] [PubMed] [Google Scholar]
  • 162.Sherman SL. 2000. Premature ovarian failure in the fragile X syndrome. Am. J. Med. Genet. 97, 189–194. () [DOI] [PubMed] [Google Scholar]
  • 163.Hagerman RJ, Hagerman PJ. 2002. The fragile X premutation: into the phenotypic fold. Curr. Opin Genet. Dev. 12, 278–283. ( 10.1016/s0959-437x(02)00299-x) [DOI] [PubMed] [Google Scholar]
  • 164.Hagerman RJ, et al. 2009. Advances in the treatment of fragile X syndrome. Pediatrics 123, 378–390. ( 10.1542/peds.2008-0317) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Hagerman RJ, Amiri K, Cronister A. 1991. Fragile X checklist. Am. J. Med. Genet. 38, 283–287. ( 10.1002/ajmg.1320380223) [DOI] [PubMed] [Google Scholar]
  • 166.Bakker CE, et al. 1994. Fmr1 knockout mice: a model to study fragile X mental retardation. Cell 78, 23–33. ( 10.1016/0092-8674(94)90569-x) [DOI] [PubMed] [Google Scholar]
  • 167.Lubs HA. 1969. A marker X chromosome. Am. J. Hum. Genet. 21, 231–244. [PMC free article] [PubMed] [Google Scholar]
  • 168.Lubs HA, Watson M, Breg R, Lujan E. 1984. Restudy of the original marker X family. Am. J. Med. Genet. 17, 133–144. ( 10.1002/ajmg.1320170108) [DOI] [PubMed] [Google Scholar]
  • 169.Cantu JM, Scaglia HE, Medina M, Gonzalez-Diddi M, Morato T, Moreno ME, Perez-Palacios G. 1976. Inherited congenital normofunctional testicular hyperplasia and mental deficiency. Hum. Genet. 33, 23–33. ( 10.1007/bf00447283) [DOI] [PubMed] [Google Scholar]
  • 170.Mattei JF, Mattei MG, Aumeras C, Auger M, Giraud F. 1981. X-linked mental retardation with the fragile X. A study of 15 families. Hum. Genet. 59, 281–289. ( 10.1007/bf00295459) [DOI] [PubMed] [Google Scholar]
  • 171.Davids JR, Hagerman RJ, Eilert RE. 1990. Orthopaedic aspects of fragile-X syndrome. J. Bone Joint Surg. Am. 72, 889–896. ( 10.2106/00004623-199072060-00015) [DOI] [PubMed] [Google Scholar]
  • 172.Paradee W, Melikian HE, Rasmussen DL, Kenneson A, Conn PJ, Warren ST. 1999. Fragile X mouse: strain effects of knockout phenotype and evidence suggesting deficient amygdala function. Neuroscience 94, 185–192. ( 10.1016/s0306-4522(99)00285-7) [DOI] [PubMed] [Google Scholar]
  • 173.Ding Q, Sethna F, Wang H. 2014. Behavioral analysis of male and female Fmr1 knockout mice on C57BL/6 background. Behav. Brain Res. 271, 72–78. ( 10.1016/j.bbr.2014.05.046) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Zhao MG, Toyoda H, Ko SW, Ding HK, Wu LJ, Zhuo M. 2005. Deficits in trace fear memory and long-term potentiation in a mouse model for fragile X syndrome. J. Neurosci. 25, 7385–7392. ( 10.1523/JNEUROSCI.1520-05.2005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Dobkin C, Rabe A, Dumas R, El Idrissi A, Haubenstock H, Brown WT. 2000. Fmr1 knockout mouse has a distinctive strain-specific learning impairment. Neuroscience 100, 423–429. ( 10.1016/s0306-4522(00)00292-x) [DOI] [PubMed] [Google Scholar]
  • 176.Van Dam D, D'Hooge R, Hauben E, Reyniers E, Gantois I, Bakker CE, Oostra BA, Kooy RF, De Deyn PP. 2000. Spatial learning, contextual fear conditioning and conditioned emotional response in Fmr1 knockout mice. Behav. Brain Res. 117, 127–136. ( 10.1016/s0166-4328(00)00296-5) [DOI] [PubMed] [Google Scholar]
  • 177.Kooy RF, et al. 1996. Transgenic mouse model for the fragile X syndrome. Am. J. Med. Genet. 64, 241–245. () [DOI] [PubMed] [Google Scholar]
  • 178.Ventura R, Pascucci T, Catania MV, Musumeci SA, Puglisi-Allegra S. 2004. Object recognition impairment in Fmr1 knockout mice is reversed by amphetamine: involvement of dopamine in the medial prefrontal cortex. Behav. Pharmacol. 15, A28 ( 10.1097/00008877-200409000-00107) [DOI] [PubMed] [Google Scholar]
  • 179.King MK, Jope RS. 2013. Lithium treatment alleviates impaired cognition in a mouse model of fragile X syndrome. Genes Brain Behav. 12, 723–731. ( 10.1111/gbb.12071) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Veeraragavan S, Bui N, Perkins JR, Yuva-Paylor LA, Carpenter RL, Paylor R. 2011. Modulation of behavioral phenotypes by a muscarinic M1 antagonist in a mouse model of fragile X syndrome. Psychopharmacology (Berl.) 217, 143–151. ( 10.1007/s00213-011-2276-6) [DOI] [PubMed] [Google Scholar]
  • 181.Michalon A, Sidorov M, Ballard TM, Ozmen L, Spooren W, Wettstein JG, Jaeschke G, Bear MF, Lindemann L. 2012. Chronic pharmacological mGlu5 inhibition corrects fragile X in adult mice. Neuron 74, 49–56. ( 10.1016/j.neuron.2012.03.009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Dolen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, Bear MF. 2007. Correction of fragile X syndrome in mice. Neuron 56, 955–962. ( 10.1016/j.neuron.2007.12.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Peier AM, McIlwain KL, Kenneson A, Warren ST, Paylor R, Nelson DL. 2000. (Over)correction of FMR1 deficiency with YAC transgenics: behavioral and physical features. Hum. Mol. Genet. 9, 1145–1159. ( 10.1093/hmg/9.8.1145) [DOI] [PubMed] [Google Scholar]
  • 184.Spencer CM, Alekseyenko O, Serysheva E, Yuva-Paylor LA, Paylor R. 2005. Altered anxiety-related and social behaviors in the Fmr1 knockout mouse model of fragile X syndrome. Genes Brain Behav. 4, 420–430. ( 10.1111/j.1601-183X.2005.00123.x) [DOI] [PubMed] [Google Scholar]
  • 185.Mineur YS, Sluyter F, de Wit S, Oostra BA, Crusio WE. 2002. Behavioral and neuroanatomical characterization of the Fmr1 knockout mouse. Hippocampus 12, 39–46. ( 10.1002/hipo.10005) [DOI] [PubMed] [Google Scholar]
  • 186.McNaughton CH, Moon J, Strawderman MS, Maclean KN, Evans J, Strupp BJ. 2008. Evidence for social anxiety and impaired social cognition in a mouse model of fragile X syndrome. Behav. Neurosci. 122, 293–300. ( 10.1037/0735-7044.122.2.293) [DOI] [PubMed] [Google Scholar]
  • 187.Dahlhaus R, El-Husseini A. 2010. Altered neuroligin expression is involved in social deficits in a mouse model of the fragile X syndrome. Behav. Brain Res. 208, 96–105. ( 10.1016/j.bbr.2009.11.019) [DOI] [PubMed] [Google Scholar]
  • 188.Rotschafer SE, Trujillo MS, Dansie LE, Ethell IM, Razak KA. 2012. Minocycline treatment reverses ultrasonic vocalization production deficit in a mouse model of fragile X syndrome. Brain Res. 1439, 7–14. ( 10.1016/j.brainres.2011.12.041) [DOI] [PubMed] [Google Scholar]
  • 189.Roy S, Watkins N, Heck D. 2012. Comprehensive analysis of ultrasonic vocalizations in a mouse model of fragile X syndrome reveals limited, call type specific deficits. PLoS ONE 7, e44816 ( 10.1371/journal.pone.0044816) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Musumeci SA, Ferri R, Elia M, Colognola RM, Bergonzi P, Tassinari CA. 1991. Epilepsy and fragile X syndrome: a follow-up study. Am. J. Med. Genet. 38, 511–513. ( 10.1002/ajmg.1320380276) [DOI] [PubMed] [Google Scholar]
  • 191.Musumeci SA, Bosco P, Calabrese G, Bakker C, De Sarro GB, Elia M, Ferri R, Oostra BA. 2000. Audiogenic seizures susceptibility in transgenic mice with fragile X syndrome. Epilepsia 41, 19–23. ( 10.1111/j.1528-1157.2000.tb01499.x) [DOI] [PubMed] [Google Scholar]
  • 192.Paylor R, Yuva-Paylor LA, Nelson DL, Spencer CM. 2008. Reversal of sensorimotor gating abnormalities in Fmr1 knockout mice carrying a human Fmr1 transgene. Behav. Neurosci. 122, 1371–1377. ( 10.1037/a0013047) [DOI] [PubMed] [Google Scholar]
  • 193.Pietropaolo S, Guilleminot A, Martin B, D'Amato FR, Crusio WE. 2011. Genetic-background modulation of core and variable autistic-like symptoms in Fmr1 knock-out mice. PLoS ONE 6, e17073 ( 10.1371/journal.pone.0017073) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Spencer CM, Alekseyenko O, Hamilton SM, Thomas AM, Serysheva E, Yuva-Paylor LA, Paylor R. 2011. Modifying behavioral phenotypes in Fmr1KO mice: genetic background differences reveal autistic-like responses. Autism Res. 4, 40–56. ( 10.1002/aur.168) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kazdoba TM, Leach PT, Silverman JL, Crawley JN. 2014. Modeling fragile X syndrome in the Fmr1 knockout mouse. Intractable Rare Dis. Res. 3, 118–133. ( 10.5582/irdr.2014.01024) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Melancia F, Trezza V. 2018. Modelling fragile X syndrome in the laboratory setting: a behavioral perspective. Behav. Brain Res. 350, 149–163. ( 10.1016/j.bbr.2018.04.042) [DOI] [PubMed] [Google Scholar]
  • 197.Alvarez VA, Sabatini BL. 2007. Anatomical and physiological plasticity of dendritic spines. Annu. Rev. Neurosci. 30, 79–97. ( 10.1146/annurev.neuro.30.051606.094222) [DOI] [PubMed] [Google Scholar]
  • 198.Zuo Y, Lin A, Chang P, Gan WB. 2005. Development of long-term dendritic spine stability in diverse regions of cerebral cortex. Neuron 46, 181–189. ( 10.1016/j.neuron.2005.04.001) [DOI] [PubMed] [Google Scholar]
  • 199.Roberts TF, Tschida KA, Klein ME, Mooney R. 2010. Rapid spine stabilization and synaptic enhancement at the onset of behavioural learning. Nature 463, 948–952. ( 10.1038/nature08759) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Rudelli RD, Brown WT, Wisniewski K, Jenkins EC, Laure-Kamionowska M, Connell F, Wisniewski HM. 1985. Adult fragile X syndrome. Clinico-neuropathologic findings. Acta Neuropathol. 67, 289–295. ( 10.1007/BF00687814) [DOI] [PubMed] [Google Scholar]
  • 201.Irwin SA, et al. 2001. Abnormal dendritic spine characteristics in the temporal and visual cortices of patients with fragile-X syndrome: a quantitative examination. Am. J. Med. Genet. 98, 161–167. () [DOI] [PubMed] [Google Scholar]
  • 202.Grossman AW, Aldridge GM, Weiler IJ, Greenough WT. 2006. Local protein synthesis and spine morphogenesis: fragile X syndrome and beyond. J. Neurosci. 26, 7151–7155. ( 10.1523/JNEUROSCI.1790-06.2006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Comery TA, Harris JB, Willems PJ, Oostra BA, Irwin SA, Weiler IJ, Greenough WT. 1997. Abnormal dendritic spines in fragile X knockout mice: maturation and pruning deficits. Proc. Natl Acad. Sci. USA 94, 5401–5404. ( 10.1073/pnas.94.10.5401) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Galvez R, Gopal AR, Greenough WT. 2003. Somatosensory cortical barrel dendritic abnormalities in a mouse model of the fragile X mental retardation syndrome. Brain Res. 971, 83–89. ( 10.1016/s0006-8993(03)02363-1) [DOI] [PubMed] [Google Scholar]
  • 205.Cruz-Martin A, Crespo M, Portera-Cailliau C. 2010. Delayed stabilization of dendritic spines in fragile X mice. J. Neurosci. 30, 7793–7803. ( 10.1523/JNEUROSCI.0577-10.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Pan F, Aldridge GM, Greenough WT, Gan WB. 2010. Dendritic spine instability and insensitivity to modulation by sensory experience in a mouse model of fragile X syndrome. Proc. Natl Acad. Sci. USA 107, 17 768–17 773. ( 10.1073/pnas.1012496107) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Antar LN, Li C, Zhang H, Carroll RC, Bassell GJ. 2006. Local functions for FMRP in axon growth cone motility and activity-dependent regulation of filopodia and spine synapses. Mol. Cell. Neurosci. 32, 37–48. ( 10.1016/j.mcn.2006.02.001) [DOI] [PubMed] [Google Scholar]
  • 208.Huber K. 2007. Fragile X syndrome: molecular mechanisms of cognitive dysfunction. Am. J. Psychiatry. 164, 556 ( 10.1176/ajp.2007.164.4.556) [DOI] [PubMed] [Google Scholar]
  • 209.Pfeiffer BE, Huber KM. 2007. Fragile X mental retardation protein induces synapse loss through acute postsynaptic translational regulation. J. Neurosci. 27, 3120–3130. ( 10.1523/JNEUROSCI.0054-07.2007) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Braun K, Segal M. 2000. FMRP involvement in formation of synapses among cultured hippocampal neurons. Cereb. Cortex. 10, 1045–1052. ( 10.1093/cercor/10.10.1045) [DOI] [PubMed] [Google Scholar]
  • 211.Pilpel Y, Kolleker A, Berberich S, Ginger M, Frick A, Mientjes E, Oostra BA, Seeburg PH. 2009. Synaptic ionotropic glutamate receptors and plasticity are developmentally altered in the CA1 field of Fmr1 knockout mice. J. Physiol. 587, 787–804. ( 10.1113/jphysiol.2008.160929) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Huber KM, Kayser MS, Bear MF. 2000. Role for rapid dendritic protein synthesis in hippocampal mGluR-dependent long-term depression. Science 288, 1254–1257. ( 10.1126/science.288.5469.1254) [DOI] [PubMed] [Google Scholar]
  • 213.Huber KM, Gallagher SM, Warren ST, Bear MF. 2002. Altered synaptic plasticity in a mouse model of fragile X mental retardation. Proc. Natl Acad. Sci. USA 99, 7746–7750. ( 10.1073/pnas.122205699) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Huang YY, Kandel ER. 2005. Theta frequency stimulation induces a local form of late phase LTP in the CA1 region of the hippocampus. Learn. Mem. 12, 587–593. ( 10.1101/lm.98905) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Huang YY, Kandel ER. 2005. Theta frequency stimulation up-regulates the synaptic strength of the pathway from CA1 to subiculum region of hippocampus. Proc. Natl Acad. Sci. USA 102, 232–237. ( 10.1073/pnas.0408368102) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Shang Y, Wang H, Mercaldo V, Li X, Chen T, Zhuo M. 2009. Fragile X mental retardation protein is required for chemically-induced long-term potentiation of the hippocampus in adult mice. J. Neurochem. 111, 635–646. ( 10.1111/j.1471-4159.2009.06314.x) [DOI] [PubMed] [Google Scholar]
  • 217.Hu H, Qin Y, Bochorishvili G, Zhu Y, van Aelst L, Zhu JJ. 2008. Ras signaling mechanisms underlying impaired GluR1-dependent plasticity associated with fragile X syndrome. J. Neurosci. 28, 7847–7862. ( 10.1523/JNEUROSCI.1496-08.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Lauterborn JC, Rex CS, Kramar E, Chen LY, Pandyarajan V, Lynch G, Gall CM. 2007. Brain-derived neurotrophic factor rescues synaptic plasticity in a mouse model of fragile X syndrome. J. Neurosci. 27, 10 685–10 694. ( 10.1523/JNEUROSCI.2624-07.2007) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Soden ME, Chen L. 2010. Fragile X protein FMRP is required for homeostatic plasticity and regulation of synaptic strength by retinoic acid. J. Neurosci. 30, 16 910–16 921. ( 10.1523/JNEUROSCI.3660-10.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Zhang Z, et al. 2018. The fragile X mutation impairs homeostatic plasticity in human neurons by blocking synaptic retinoic acid signaling. Sci. Transl. Med. 10, eaar4338 ( 10.1126/scitranslmed.aar4338) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Wang H, et al. 2008. FMRP acts as a key messenger for dopamine modulation in the forebrain. Neuron 59, 634–647. ( 10.1016/j.neuron.2008.06.027) [DOI] [PubMed] [Google Scholar]
  • 222.Gao F, Qi L, Yang Z, Yang T, Zhang Y, Xu H, Zhao H. 2018. Impaired GABA neural circuits are critical for fragile X syndrome. Neural Plast. 2018, 8423420 ( 10.1155/2018/8423420) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Centonze D, et al. 2008. Abnormal striatal GABA transmission in the mouse model for the fragile X syndrome. Biol. Psychiatry. 63, 963–973. ( 10.1016/j.biopsych.2007.09.008) [DOI] [PubMed] [Google Scholar]
  • 224.Gibson JR, Bartley AF, Hays SA, Huber KM. 2008. Imbalance of neocortical excitation and inhibition and altered UP states reflect network hyperexcitability in the mouse model of fragile X syndrome. J. Neurophysiol. 100, 2615–2626. ( 10.1152/jn.90752.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Curia G, Papouin T, Seguela P, Avoli M. 2009. Downregulation of tonic GABAergic inhibition in a mouse model of fragile X syndrome. Cereb. Cortex. 19, 1515–1520. ( 10.1093/cercor/bhn159) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Tyzio R, et al. 2014. Oxytocin-mediated GABA inhibition during delivery attenuates autism pathogenesis in rodent offspring. Science 343, 675–679. ( 10.1126/science.1247190) [DOI] [PubMed] [Google Scholar]
  • 227.Contractor A, Klyachko VA, Portera-Cailliau C. 2015. Altered neuronal and circuit excitability in fragile X syndrome. Neuron 87, 699–715. ( 10.1016/j.neuron.2015.06.017) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Doll CA, Vita DJ, Broadie K. 2017. Fragile X mental retardation protein requirements in activity-dependent critical period neural circuit refinement. Curr. Biol. 27, 2318–2330 e2313. ( 10.1016/j.cub.2017.06.046) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Kim H, Gibboni R, Kirkhart C, Bao S. 2013. Impaired critical period plasticity in primary auditory cortex of fragile X model mice. J. Neurosci. 33, 15 686–15 692. ( 10.1523/JNEUROSCI.3246-12.2013) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Devys D, Lutz Y, Rouyer N, Bellocq JP, Mandel JL. 1993. The FMR-1 protein is cytoplasmic, most abundant in neurons and appears normal in carriers of a fragile X premutation. Nat. Genet. 4, 335–340. ( 10.1038/ng0893-335) [DOI] [PubMed] [Google Scholar]
  • 231.Ashley CT Jr, Wilkinson KD, Reines D, Warren ST. 1993. FMR1 protein: conserved RNP family domains and selective RNA binding. Science 262, 563–566. ( 10.1126/science.7692601) [DOI] [PubMed] [Google Scholar]
  • 232.Siomi H, Siomi MC, Nussbaum RL, Dreyfuss G. 1993. The protein product of the fragile X gene, FMR1, has characteristics of an RNA-binding protein. Cell 74, 291–298. ( 10.1016/0092-8674(93)90420-u) [DOI] [PubMed] [Google Scholar]
  • 233.Khandjian EW, Corbin F, Woerly S, Rousseau F. 1996. The fragile X mental retardation protein is associated with ribosomes. Nat. Genet. 12, 91–93. ( 10.1038/ng0196-91) [DOI] [PubMed] [Google Scholar]
  • 234.Darnell JC, Jensen KB, Jin P, Brown V, Warren ST, Darnell RB. 2001. Fragile X mental retardation protein targets G quartet mRNAs important for neuronal function. Cell 107, 489–499. ( 10.1016/S0092-8674(01)00566-9) [DOI] [PubMed] [Google Scholar]
  • 235.Darnell JC, Fraser CE, Mostovetsky O, Stefani G, Jones TA, Eddy SR, Darnell RB. 2005. Kissing complex RNAs mediate interaction between the fragile-X mental retardation protein KH2 domain and brain polyribosomes. Genes Dev. 19, 903–918. ( 10.1101/gad.1276805) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Schaeffer C, Bardoni B, Mandel JL, Ehresmann B, Ehresmann C, Moine H. 2001. The fragile X mental retardation protein binds specifically to its mRNA via a purine quartet motif. EMBO J. 20, 4803–4813. ( 10.1093/emboj/20.17.4803) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Darnell JC, et al. 2011. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 146, 247–261. ( 10.1016/j.cell.2011.06.013) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Napoli I, et al. 2008. The fragile X syndrome protein represses activity-dependent translation through CYFIP1, a new 4E-BP. Cell 134, 1042–1054. ( 10.1016/j.cell.2008.07.031) [DOI] [PubMed] [Google Scholar]
  • 239.Caudy AA, Myers M, Hannon GJ, Hammond SM. 2002. Fragile X-related protein and VIG associate with the RNA interference machinery. Genes Dev. 16, 2491–2496. ( 10.1101/gad.1025202) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Ishizuka A, Siomi MC, Siomi H. 2002. A drosophila fragile X protein interacts with components of RNAi and ribosomal proteins. Genes Dev. 16, 2497–2508. ( 10.1101/gad.1022002) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Plante I, Davidovic L, Ouellet DL, Gobeil LA, Tremblay S, Khandjian EW, Provost P. 2006. Dicer-derived microRNAs are utilized by the fragile X mental retardation protein for assembly on target RNAs. J. Biomed. Biotechnol. 2006, 64347 ( 10.1155/JBB/2006/64347) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Graf ER, Zhang X, Jin SX, Linhoff MW, Craig AM. 2004. Neurexins induce differentiation of GABA and glutamate postsynaptic specializations via neuroligins. Cell 119, 1013–1026. ( 10.1016/j.cell.2004.11.035) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Eberhart DE, Malter HE, Feng Y, Warren ST. 1996. The fragile X mental retardation protein is a ribonucleoprotein containing both nuclear localization and nuclear export signals. Hum. Mol. Genet. 5, 1083–1091. ( 10.1093/hmg/5.8.1083) [DOI] [PubMed] [Google Scholar]
  • 244.Bramham CR, Wells DG. 2007. Dendritic mRNA: transport, translation and function. Nat. Rev. Neurosci. 8, 776–789. ( 10.1038/nrn2150) [DOI] [PubMed] [Google Scholar]
  • 245.Maurer-Stroh S, Dickens NJ, Hughes-Davies L, Kouzarides T, Eisenhaber F, Ponting CP. 2003. The Tudor domain 'Royal Family': Tudor, plant Agenet, Chromo, PWWP and MBT domains. Trends Biochem. Sci. 28, 69–74. ( 10.1016/S0968-0004(03)00004-5) [DOI] [PubMed] [Google Scholar]
  • 246.Adams-Cioaba MA, Guo Y, Bian C, Amaya MF, Lam R, Wasney GA, Vedadi M, Xu C, Min J. 2010. Structural studies of the tandem Tudor domains of fragile X mental retardation related proteins FXR1 and FXR2. PLoS ONE 5, e13559 ( 10.1371/journal.pone.0013559) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Bassell GJ, Warren ST. 2008. Fragile X syndrome: loss of local mRNA regulation alters synaptic development and function. Neuron 60, 201–214. ( 10.1016/j.neuron.2008.10.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Ceman S, O'Donnell WT, Reed M, Patton S, Pohl J, Warren ST. 2003. Phosphorylation influences the translation state of FMRP-associated polyribosomes. Hum. Mol. Genet. 12, 3295–3305. ( 10.1093/hmg/ddg350) [DOI] [PubMed] [Google Scholar]
  • 249.Muddashetty RS, Nalavadi VC, Gross C, Yao X, Xing L, Laur O, Warren ST, Bassell GJ. 2011. Reversible inhibition of PSD-95 mRNA translation by miR-125a, FMRP phosphorylation, and mGluR signaling. Mol. Cell. 42, 673–688. ( 10.1016/j.molcel.2011.05.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Narayanan U, Nalavadi V, Nakamoto M, Pallas DC, Ceman S, Bassell GJ, Warren ST. 2007. FMRP phosphorylation reveals an immediate-early signaling pathway triggered by group I mGluR and mediated by PP2A. J. Neurosci. 27, 14 349–14 357. ( 10.1523/JNEUROSCI.2969-07.2007) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Cheever A, Ceman S. 2009. Phosphorylation of FMRP inhibits association with dicer. RNA 15, 362–366. ( 10.1261/rna.1500809) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Nakamoto M, Nalavadi V, Epstein MP, Narayanan U, Bassell GJ, Warren ST. 2007. Fragile X mental retardation protein deficiency leads to excessive mGluR5-dependent internalization of AMPA receptors. Proc. Natl Acad. Sci. USA 104, 15 537–15 542. ( 10.1073/pnas.0707484104) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Narayanan U, Nalavadi V, Nakamoto M, Thomas G, Ceman S, Bassell GJ, Warren ST. 2008. S6K1 phosphorylates and regulates fragile X mental retardation protein (FMRP) with the neuronal protein synthesis-dependent mammalian target of rapamycin (mTOR) signaling cascade. J. Biol. Chem. 283, 18 478–18 482. ( 10.1074/jbc.C800055200) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Sharma A, Hoeffer CA, Takayasu Y, Miyawaki T, McBride SM, Klann E, Zukin RS. 2010. Dysregulation of mTOR signaling in fragile X syndrome. J. Neurosci. 30, 694–702. ( 10.1523/JNEUROSCI.3696-09.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Bear MF, Huber KM, Warren ST. 2004. The mGluR theory of fragile X mental retardation. Trends Neurosci. 27, 370–377. ( 10.1016/j.tins.2004.04.009) [DOI] [PubMed] [Google Scholar]
  • 256.Harlow EG, Till SM, Russell TA, Wijetunge LS, Kind P, Contractor A. 2010. Critical period plasticity is disrupted in the barrel cortex of FMR1 knockout mice. Neuron 65, 385–398. ( 10.1016/j.neuron.2010.01.024) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Nimchinsky EA, Oberlander AM, Svoboda K. 2001. Abnormal development of dendritic spines in FMR1 knock-out mice. J. Neurosci. 21, 5139–5146. ( 10.1523/JNEUROSCI.21-14-05139.2001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Galvez R, Greenough WT. 2005. Sequence of abnormal dendritic spine development in primary somatosensory cortex of a mouse model of the fragile X mental retardation syndrome. Am. J. Med. Genet. A 135, 155–160. ( 10.1002/ajmg.a.30709) [DOI] [PubMed] [Google Scholar]
  • 259.Frenkel MY, Bear MF. 2004. How monocular deprivation shifts ocular dominance in visual cortex of young mice. Neuron 44, 917–923. ( 10.1016/j.neuron.2004.12.003) [DOI] [PubMed] [Google Scholar]
  • 260.Pacala SW, et al. 2001. Consistent land- and atmosphere-based U.S. carbon sink estimates. Science 292, 2316–2320. ( 10.1126/science.1057320) [DOI] [PubMed] [Google Scholar]
  • 261.Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. 1999. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 23, 185–188. ( 10.1038/13810) [DOI] [PubMed] [Google Scholar]
  • 262.Shibayama A, et al. 2004. MECP2 structural and 3'-UTR variants in schizophrenia, autism and other psychiatric diseases: a possible association with autism. Am. J. Med. Genet. B Neuropsychiatr. Genet. 128B, 50–53. ( 10.1002/ajmg.b.30016) [DOI] [PubMed] [Google Scholar]
  • 263.Campos M Jr, Abdalla CB, dos Santos AV, Pestana CP, dos Santos JM, Santos-Reboucas CB, Pimentel MM. 2009. A MECP2 mutation in a highly conserved aminoacid causing mental retardation in a male. Brain Dev. 31, 176–178. ( 10.1016/j.braindev.2008.07.001) [DOI] [PubMed] [Google Scholar]
  • 264.Reichwald K, Thiesen J, Wiehe T, Weitzel J, Poustka WA, Rosenthal A, Platzer M, Stratling WH, Kioschis P. 2000. Comparative sequence analysis of the MECP2-locus in human and mouse reveals new transcribed regions. Mamm. Genome. 11, 182–190. ( 10.1007/s003350010035) [DOI] [PubMed] [Google Scholar]
  • 265.Coy JF, Sedlacek Z, Bachner D, Delius H, Poustka A. 1999. A complex pattern of evolutionary conservation and alternative polyadenylation within the long 3′-untranslated region of the methyl-CpG-binding protein 2 gene (MeCP2) suggests a regulatory role in gene expression. Hum. Mol. Genet. 8, 1253–1262. ( 10.1093/hmg/8.7.1253) [DOI] [PubMed] [Google Scholar]
  • 266.Hagberg B, Aicardi J, Dias K, Ramos O. 1983. A progressive syndrome of autism, dementia, ataxia, and loss of purposeful hand use in girls: Rett's syndrome: report of 35 cases. Ann. Neurol. 14, 471–479. ( 10.1002/ana.410140412) [DOI] [PubMed] [Google Scholar]
  • 267.Schonewolf-Greulich B, Stahlhut M, Larsen JL, Syhler B, Bisgaard AM. 2017. Functional abilities in aging women with Rett syndrome—the Danish cohort. Disabil. Rehabil. 39, 911–918. ( 10.3109/09638288.2016.1170896) [DOI] [PubMed] [Google Scholar]
  • 268.Parisi L, Di Filippo T, Roccella M. 2016. The quality of life in girls with Rett syndrome. Ment Illn. 8, 6302 ( 10.4081/mi.2016.6302) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Chen RZ, Akbarian S, Tudor M, Jaenisch R. 2001. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat. Genet. 27, 327–331. ( 10.1038/85906) [DOI] [PubMed] [Google Scholar]
  • 270.Moretti P, et al. 2006. Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J. Neurosci. 26, 319–327. ( 10.1523/JNEUROSCI.2623-05.2006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Gemelli T, Berton O, Nelson ED, Perrotti LI, Jaenisch R, Monteggia LM. 2006. Postnatal loss of methyl-CpG binding protein 2 in the forebrain is sufficient to mediate behavioral aspects of Rett syndrome in mice. Biol. Psychiatry. 59, 468–476. ( 10.1016/j.biopsych.2005.07.025) [DOI] [PubMed] [Google Scholar]
  • 272.Shahbazian M, Young J, Yuva-Paylor L, Spencer C, Antalffy B, Noebels J, Armstrong D, Paylor R, Zoghbi H. 2002. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3. Neuron 35, 243–254. ( 10.1016/S0896-6273(02)00768-7) [DOI] [PubMed] [Google Scholar]
  • 273.Armstrong DD. 2002. Neuropathology of Rett syndrome. Ment. Retard. Dev. Disabil. Res. Rev. 8, 72–76. ( 10.1002/mrdd.10027) [DOI] [PubMed] [Google Scholar]
  • 274.Chapleau CA, Calfa GD, Lane MC, Albertson AJ, Larimore JL, Kudo S, Armstrong DL, Percy AK, Pozzo-Miller L. 2009. Dendritic spine pathologies in hippocampal pyramidal neurons from Rett syndrome brain and after expression of Rett-associated MECP2 mutations. Neurobiol. Dis. 35, 219–233. ( 10.1016/j.nbd.2009.05.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Kishi N, Macklis JD. 2004. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol. Cell. Neurosci. 27, 306–321. ( 10.1016/j.mcn.2004.07.006) [DOI] [PubMed] [Google Scholar]
  • 276.Zhou Z, et al. 2006. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 52, 255–269. ( 10.1016/j.neuron.2006.09.037) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Nelson ED, Kavalali ET, Monteggia LM. 2006. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr. Biol. 16, 710–716. ( 10.1016/j.cub.2006.02.062) [DOI] [PubMed] [Google Scholar]
  • 278.Chao HT, Zoghbi HY, Rosenmund C. 2007. MeCP2 controls excitatory synaptic strength by regulating glutamatergic synapse number. Neuron 56, 58–65. ( 10.1016/j.neuron.2007.08.018) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Asaka Y, Jugloff DG, Zhang L, Eubanks JH, Fitzsimonds RM. 2006. Hippocampal synaptic plasticity is impaired in the Mecp2-null mouse model of Rett syndrome. Neurobiol. Dis. 21, 217–227. ( 10.1016/j.nbd.2005.07.005) [DOI] [PubMed] [Google Scholar]
  • 280.Collins AL, Levenson JM, Vilaythong AP, Richman R, Armstrong DL, Noebels JL, Sweatt JD, Zoghbi HY. 2004. Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Hum. Mol. Genet. 13, 2679–2689. ( 10.1093/hmg/ddh282) [DOI] [PubMed] [Google Scholar]
  • 281.Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, Bird A. 1998. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389. ( 10.1038/30764) [DOI] [PubMed] [Google Scholar]
  • 282.Fuks F, Hurd PJ, Wolf D, Nan X, Bird AP, Kouzarides T. 2003. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J. Biol. Chem. 278, 4035–4040. ( 10.1074/jbc.M210256200) [DOI] [PubMed] [Google Scholar]
  • 283.Free A, Wakefield RI, Smith BO, Dryden DT, Barlow PN, Bird AP. 2001. DNA recognition by the methyl-CpG binding domain of MeCP2. J. Biol. Chem. 276, 3353–3360. ( 10.1074/jbc.M007224200) [DOI] [PubMed] [Google Scholar]
  • 284.Chen WG, Chang Q, Lin Y, Meissner A, West AE, Griffith EC, Jaenisch R, Greenberg ME. 2003. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 302, 885–889. ( 10.1126/science.1086446) [DOI] [PubMed] [Google Scholar]
  • 285.Martinowich K, Hattori D, Wu H, Fouse S, He F, Hu Y, Fan G, Sun YE. 2003. DNA methylation-related chromatin remodeling in activity-dependent BDNF gene regulation. Science 302, 890–893. ( 10.1126/science.1090842) [DOI] [PubMed] [Google Scholar]
  • 286.Horike S, Cai S, Miyano M, Cheng JF, Kohwi-Shigematsu T. 2005. Loss of silent-chromatin looping and impaired imprinting of DLX5 in Rett syndrome. Nat. Genet. 37, 31–40. ( 10.1038/ng1491) [DOI] [PubMed] [Google Scholar]
  • 287.Jordan C, Li HH, Kwan HC, Francke U. 2007. Cerebellar gene expression profiles of mouse models for Rett syndrome reveal novel MeCP2 targets. BMC Med. Genet. 8, 36 ( 10.1186/1471-2350-8-36) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Harikrishnan KN, et al. 2005. Brahma links the SWI/SNF chromatin-remodeling complex with MeCP2-dependent transcriptional silencing. Nat. Genet. 37, 254–264. ( 10.1038/ng1516) [DOI] [PubMed] [Google Scholar]
  • 289.Nan X, Hou J, Maclean A, Nasir J, Lafuente MJ, Shu X, Kriaucionis S, Bird A. 2007. Interaction between chromatin proteins MECP2 and ATRX is disrupted by mutations that cause inherited mental retardation. Proc. Natl Acad. Sci. USA 104, 2709–2714. ( 10.1073/pnas.0608056104) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Lewis JD, Meehan RR, Henzel WJ, Maurer-Fogy I, Jeppesen P, Klein F, Bird A. 1992. Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell 69, 905–914. ( 10.1016/0092-8674(92)90610-O) [DOI] [PubMed] [Google Scholar]
  • 291.Nan X, Tate P, Li E, Bird A. 1996. DNA methylation specifies chromosomal localization of MeCP2. Mol. Cell. Biol. 16, 414–421. ( 10.1128/MCB.16.1.414) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Singh J, Saxena A, Christodoulou J, Ravine D. 2008. MECP2 genomic structure and function: insights from ENCODE. Nucleic Acids Res. 36, 6035–6047. ( 10.1093/nar/gkn591) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Guy J, Cheval H, Selfridge J, Bird A. 2011. The role of MeCP2 in the brain. Annu. Rev. Cell Dev. Biol. 27, 631–652. ( 10.1146/annurev-cellbio-092910-154121) [DOI] [PubMed] [Google Scholar]
  • 294.Du F, Nguyen MVC, Karten A, Felice CA, Mandel G, Ballas N. 2016. Acute and crucial requirement for MeCP2 function upon transition from early to late adult stages of brain maturation. Hum. Mol. Genet. 25, 1690–1702. ( 10.1093/hmg/ddw038) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.He LJ, Liu N, Cheng TL, Chen XJ, Li YD, Shu YS, Qiu ZL, Zhang XH. 2014. Conditional deletion of Mecp2 in parvalbumin-expressing GABAergic cells results in the absence of critical period plasticity. Nat. Commun. 5, 5036 ( 10.1038/ncomms6036) [DOI] [PubMed] [Google Scholar]
  • 296.Monteiro P, Feng G. 2017. SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat. Rev. Neurosci. 18, 147–157. ( 10.1038/nrn.2016.183) [DOI] [PubMed] [Google Scholar]
  • 297.Naisbitt S, Kim E, Tu JC, Xiao B, Sala C, Valtschanoff J, Weinberg RJ, Worley PF, Sheng M. 1999. Shank, a novel family of postsynaptic density proteins that binds to the NMDA receptor/PSD-95/GKAP complex and cortactin. Neuron 23, 569–582. ( 10.1016/S0896-6273(00)80809-0) [DOI] [PubMed] [Google Scholar]
  • 298.Durand CM, et al. 2007. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat. Genet. 39, 25–27. ( 10.1038/ng1933) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Soorya L, et al. 2013. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol. Autism 4, 18 ( 10.1186/2040-2392-4-18) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Leblond CS, et al. 2014. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet. 10, e1004580 ( 10.1371/journal.pgen.1004580) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Silverman JL, Turner SM, Barkan CL, Tolu SS, Saxena R, Hung AY, Sheng M, Crawley JN. 2011. Sociability and motor functions in Shank1 mutant mice. Brain Res. 1380, 120–137. ( 10.1016/j.brainres.2010.09.026) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Hung AY, et al. 2008. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J. Neurosci. 28, 1697–1708. ( 10.1523/JNEUROSCI.3032-07.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Won H, et al. 2012. Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature 486, 261–265. ( 10.1038/nature11208) [DOI] [PubMed] [Google Scholar]
  • 304.Schmeisser MJ, et al. 2012. Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/Shank2. Nature 486, 256–260. ( 10.1038/nature11015) [DOI] [PubMed] [Google Scholar]
  • 305.Yang M, et al. 2012. Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adolescent Shank3 null mutant mice. J. Neurosci. 32, 6525–6541. ( 10.1523/JNEUROSCI.6107-11.2012) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Roussignol G, Ango F, Romorini S, Tu JC, Sala C, Worley PF, Bockaert J, Fagni L. 2005. Shank expression is sufficient to induce functional dendritic spine synapses in aspiny neurons. J. Neurosci. 25, 3560–3570. ( 10.1523/JNEUROSCI.4354-04.2005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Verpelli C, et al. 2011. Importance of Shank3 protein in regulating metabotropic glutamate receptor 5 (mGluR5) expression and signaling at synapses. J. Biol. Chem. 286, 34 839–34 850. ( 10.1074/jbc.M111.258384) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Grabrucker AM. 2014. A role for synaptic zinc in ProSAP/Shank PSD scaffold malformation in autism spectrum disorders. Dev. Neurobiol. 74, 136–146. ( 10.1002/dneu.22089) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Sheng M, Kim E. 2000. The Shank family of scaffold proteins. J. Cell Sci. 113(Pt 11), 1851–1856. [DOI] [PubMed] [Google Scholar]
  • 310.Sheng M, Kim E. 2011. The postsynaptic organization of synapses. Cold Spring Harb. Perspect. Biol. 3, a005678 ( 10.1101/cshperspect.a005678) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Jiang YH, Ehlers MD. 2013. Modeling autism by SHANK gene mutations in mice. Neuron 78, 8–27. ( 10.1016/j.neuron.2013.03.016) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Kreienkamp HJ. 2008. Scaffolding proteins at the postsynaptic density: shank as the architectural framework. In Protein–protein interactions as new drug targets (eds Klussmann E, Scott J), pp. 365–380. Handbook of Experimental Pharmacology, vol. 186. Berlin: Springer. [DOI] [PubMed] [Google Scholar]
  • 313.Sakai Y, Shaw CA, Dawson BC, Dugas DV, Al-Mohtaseb Z, Hill DE, Zoghbi HY. 2011. Protein interactome reveals converging molecular pathways among autism disorders. Sci. Transl. Med. 3, 86ra49 ( 10.1126/scitranslmed.3002166) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Tu JC, et al. 1999. Coupling of mGluR/Homer and PSD-95 complexes by the Shank family of postsynaptic density proteins. Neuron 23, 583–592. ( 10.1016/s0896-6273(00)80810-7) [DOI] [PubMed] [Google Scholar]
  • 315.Wang X, et al. 2016. Altered mGluR5-Homer scaffolds and corticostriatal connectivity in a Shank3 complete knockout model of autism. Nat. Commun. 7, 11459 ( 10.1038/ncomms11459) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Tatavarty V, Torrado Pacheco A, Lin H, Miska NJ, Hengen KB, Wagner FF, Turrigiano GG. 2018. Autism-associated Shank3 is essential for homeostatic plasticity and neuronal circuit stability. bioRxiv 365445 ( 10.1101/365445) [DOI]
  • 317.Mei Y, Monteiro P, Zhou Y, Kim JA, Gao X, Fu Z, Feng G. 2016. Adult restoration of Shank3 expression rescues selective autistic-like phenotypes. Nature 530, 481–484. ( 10.1038/nature16971) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Ichtchenko K, Hata Y, Nguyen T, Ullrich B, Missler M, Moomaw C, Sudhof TC. 1995. Neuroligin 1: a splice site-specific ligand for beta-neurexins. Cell 81, 435–443. ( 10.1016/0092-8674(95)90396-8) [DOI] [PubMed] [Google Scholar]
  • 319.Ichtchenko K, Nguyen T, Sudhof TC. 1996. Structures, alternative splicing, and neurexin binding of multiple neuroligins. J. Biol. Chem. 271, 2676–2682. ( 10.1074/jbc.271.5.2676) [DOI] [PubMed] [Google Scholar]
  • 320.Ushkaryov YA, Hata Y, Ichtchenko K, Moomaw C, Afendis S, Slaughter CA, Sudhof TC. 1994. Conserved domain structure of beta-neurexins. Unusual cleaved signal sequences in receptor-like neuronal cell-surface proteins. J. Biol. Chem. 269, 11 987–11 992. [PubMed] [Google Scholar]
  • 321.Ushkaryov YA, Sudhof TC. 1993. Neurexin III alpha: extensive alternative splicing generates membrane-bound and soluble forms. Proc. Natl Acad. Sci. USA 90, 6410–6414. ( 10.1073/pnas.90.14.6410) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Ushkaryov YA, Petrenko AG, Geppert M, Sudhof TC. 1992. Neurexins: synaptic cell surface proteins related to the alpha-latrotoxin receptor and laminin. Science 257, 50–56. ( 10.1126/science.1621094) [DOI] [PubMed] [Google Scholar]
  • 323.Song JY, Ichtchenko K, Sudhof TC, Brose N. 1999. Neuroligin 1 is a postsynaptic cell-adhesion molecule of excitatory synapses. Proc. Natl Acad. Sci. USA 96, 1100–1105. ( 10.1073/pnas.96.3.1100) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Siddiqui TJ, Pancaroglu R, Kang Y, Rooyakkers A, Craig AM. 2010. LRRTMs and neuroligins bind neurexins with a differential code to cooperate in glutamate synapse development. J. Neurosci. 30, 7495–7506. ( 10.1523/JNEUROSCI.0470-10.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Philibert RA, Winfield SL, Sandhu HK, Martin BM, Ginns EI. 2000. The structure and expression of the human neuroligin-3 gene. Gene 246, 303–310. ( 10.1016/S0378-1119(00)00049-4) [DOI] [PubMed] [Google Scholar]
  • 326.Bolliger MF, Frei K, Winterhalter KH, Gloor SM. 2001. Identification of a novel neuroligin in humans which binds to PSD-95 and has a widespread expression. Biochem. J. 356, 581–588. ( 10.1042/bj3560581) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Varoqueaux F, Jamain S, Brose N. 2004. Neuroligin 2 is exclusively localized to inhibitory synapses. Eur. J. Cell Biol. 83, 449–456. ( 10.1078/0171-9335-00410) [DOI] [PubMed] [Google Scholar]
  • 328.Hoon M, et al. 2011. Neuroligin-4 is localized to glycinergic postsynapses and regulates inhibition in the retina. Proc. Natl Acad. Sci. USA 108, 3053–3058. ( 10.1073/pnas.1006946108) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Budreck EC, Scheiffele P. 2007. Neuroligin-3 is a neuronal adhesion protein at GABAergic and glutamatergic synapses. Eur. J. Neurosci. 26, 1738–1748. ( 10.1111/j.1460-9568.2007.05842.x) [DOI] [PubMed] [Google Scholar]
  • 330.Irie M, Hata Y, Takeuchi M, Ichtchenko K, Toyoda A, Hirao K, Takai Y, Rosahl TW, Sudhof TC. 1997. Binding of neuroligins to PSD-95. Science 277, 1511–1515. ( 10.1126/science.277.5331.1511) [DOI] [PubMed] [Google Scholar]
  • 331.Meyer G, Varoqueaux F, Neeb A, Oschlies M, Brose N. 2004. The complexity of PDZ domain-mediated interactions at glutamatergic synapses: a case study on neuroligin. Neuropharmacology 47, 724–733. ( 10.1016/j.neuropharm.2004.06.023) [DOI] [PubMed] [Google Scholar]
  • 332.Budreck EC, et al. 2013. Neuroligin-1 controls synaptic abundance of NMDA-type glutamate receptors through extracellular coupling. Proc. Natl Acad. Sci. USA 110, 725–730. ( 10.1073/pnas.1214718110) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Poulopoulos A, et al. 2009. Neuroligin 2 drives postsynaptic assembly at perisomatic inhibitory synapses through gephyrin and collybistin. Neuron 63, 628–642. ( 10.1016/j.neuron.2009.08.023) [DOI] [PubMed] [Google Scholar]
  • 334.Soykan T, et al. 2014. A conformational switch in collybistin determines the differentiation of inhibitory postsynapses. EMBO J. 33, 2113–2133. ( 10.15252/embj.201488143) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Scheiffele P, Fan J, Choih J, Fetter R, Serafini T. 2000. Neuroligin expressed in nonneuronal cells triggers presynaptic development in contacting axons. Cell 101, 657–669. ( 10.1016/S0092-8674(00)80877-6) [DOI] [PubMed] [Google Scholar]
  • 336.Sara Y, Biederer T, Atasoy D, Chubykin A, Mozhayeva MG, Sudhof TC, Kavalali ET. 2005. Selective capability of SynCAM and neuroligin for functional synapse assembly. J. Neurosci. 25, 260–270. ( 10.1523/JNEUROSCI.3165-04.2005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Fu Z, Washbourne P, Ortinski P, Vicini S. 2003. Functional excitatory synapses in HEK293 cells expressing neuroligin and glutamate receptors. J. Neurophysiol. 90, 3950–3957. ( 10.1152/jn.00647.2003) [DOI] [PubMed] [Google Scholar]
  • 338.Varoqueaux F, Aramuni G, Rawson RL, Mohrmann R, Missler M, Gottmann K, Zhang W, Sudhof TC, Brose N. 2006. Neuroligins determine synapse maturation and function. Neuron 51, 741–754. ( 10.1016/j.neuron.2006.09.003) [DOI] [PubMed] [Google Scholar]
  • 339.Chih B, Engelman H, Scheiffele P. 2005. Control of excitatory and inhibitory synapse formation by neuroligins. Science 307, 1324–1328. ( 10.1126/science.1107470) [DOI] [PubMed] [Google Scholar]
  • 340.Prange O, Wong TP, Gerrow K, Wang YT, El-Husseini A. 2004. A balance between excitatory and inhibitory synapses is controlled by PSD-95 and neuroligin. Proc. Natl Acad. Sci. USA 101, 13 915–13 920. ( 10.1073/pnas.0405939101) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Nakanishi M, Nomura J, Ji X, Tamada K, Arai T, Takahashi E, Bucan M, Takumi T. 2017. Functional significance of rare neuroligin 1 variants found in autism. PLoS Genet. 13, e1006940 ( 10.1371/journal.pgen.1006940) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Glessner JT, et al. 2009. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature 459, 569–573. ( 10.1038/nature07953) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Tristan-Clavijo E, Camacho-Garcia RJ, Robles-Lanuza E, Ruiz A, van der Zee J, Van Broeckhoven C, Hernandez I, Martinez-Mir A, Scholl FG. 2015. A truncating mutation in Alzheimer's disease inactivates neuroligin-1 synaptic function. Neurobiol. Aging. 36, 3171–3175. ( 10.1016/j.neurobiolaging.2015.09.004) [DOI] [PubMed] [Google Scholar]
  • 344.Kilaru V, et al. 2016. Genome-wide gene-based analysis suggests an association between Neuroligin 1 (NLGN1) and post-traumatic stress disorder. Transl Psychiatry. 6, e820 ( 10.1038/tp.2016.69) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Parente DJ, Garriga C, Baskin B, Douglas G, Cho MT, Araujo GC, Shinawi M. 2017. Neuroligin 2 nonsense variant associated with anxiety, autism, intellectual disability, hyperphagia, and obesity. Am. J. Med. Genet. A 173, 213–216. ( 10.1002/ajmg.a.37977) [DOI] [PubMed] [Google Scholar]
  • 346.Sun C, Cheng MC, Qin R, Liao DL, Chen TT, Koong FJ, Chen G, Chen CH. 2011. Identification and functional characterization of rare mutations of the neuroligin-2 gene (NLGN2) associated with schizophrenia. Hum. Mol. Genet. 20, 3042–3051. ( 10.1093/hmg/ddr208) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Blundell J, Blaiss CA, Etherton MR, Espinosa F, Tabuchi K, Walz C, Bolliger MF, Sudhof TC, Powell CM. 2010. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J. Neurosci. 30, 2115–2129. ( 10.1523/JNEUROSCI.4517-09.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Dahlhaus R, Hines RM, Eadie BD, Kannangara TS, Hines DJ, Brown CE, Christie BR, El-Husseini A. 2010. Overexpression of the cell adhesion protein neuroligin-1 induces learning deficits and impairs synaptic plasticity by altering the ratio of excitation to inhibition in the hippocampus. Hippocampus 20, 305–322. ( 10.1002/hipo.20630) [DOI] [PubMed] [Google Scholar]
  • 349.Jiang M, Polepalli J, Chen LY, Zhang B, Sudhof TC, Malenka RC. 2017. Conditional ablation of neuroligin-1 in CA1 pyramidal neurons blocks LTP by a cell-autonomous NMDA receptor-independent mechanism. Mol. Psychiatry. 22, 375–383. ( 10.1038/mp.2016.80) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Hines RM, et al. 2008. Synaptic imbalance, stereotypies, and impaired social interactions in mice with altered neuroligin 2 expression. J. Neurosci. 28, 6055–6067. ( 10.1523/JNEUROSCI.0032-08.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Ye X, Kapeller-Libermann D, Travaglia A, Inda MC, Alberini CM. 2017. Direct dorsal hippocampal-prelimbic cortex connections strengthen fear memories. Nat. Neurosci. 20, 52–61. ( 10.1038/nn.4443) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.van der Kooij MA, et al. 2014. Impaired hippocampal neuroligin-2 function by chronic stress or synthetic peptide treatment is linked to social deficits and increased aggression. Neuropsychopharmacology 39, 1148–1158. ( 10.1038/npp.2013.315) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Liang J, Xu W, Hsu YT, Yee AX, Chen L, Sudhof TC. 2015. Conditional neuroligin-2 knockout in adult medial prefrontal cortex links chronic changes in synaptic inhibition to cognitive impairments. Mol. Psychiatry. 20, 850–859. ( 10.1038/mp.2015.31) [DOI] [PubMed] [Google Scholar]
  • 354.Durieux AM, Horder J, Petrinovic MM. 2016. Neuroligin-2 and the tightrope of excitation/inhibition balance in the prefrontal cortex. J. Neurophysiol. 115, 5–7. ( 10.1152/jn.00703.2015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Jamain S, et al. 2008. Reduced social interaction and ultrasonic communication in a mouse model of monogenic heritable autism. Proc. Natl Acad. Sci. USA 105, 1710–1715. ( 10.1073/pnas.0711555105) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Sudhof TC. 2008. Neuroligins and neurexins link synaptic function to cognitive disease. Nature 455, 903–911. ( 10.1038/nature07456) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Kim J, et al. 2008. Neuroligin-1 is required for normal expression of LTP and associative fear memory in the amygdala of adult animals. Proc. Natl Acad. Sci. USA 105, 9087–9092. ( 10.1073/pnas.0803448105) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Hu Z, Hom S, Kudze T, Tong XJ, Choi S, Aramuni G, Zhang W, Kaplan JM. 2012. Neurexin and neuroligin mediate retrograde synaptic inhibition in C. elegans. Science 337, 980–984. ( 10.1126/science.1224896) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Sun C, Zhang L, Chen G. 2013. An unexpected role of neuroligin-2 in regulating KCC2 and GABA functional switch. Mol. Brain. 6, 23 ( 10.1186/1756-6606-6-23) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Wohr M, Silverman JL, Scattoni ML, Turner SM, Harris MJ, Saxena R, Crawley JN. 2013. Developmental delays and reduced pup ultrasonic vocalizations but normal sociability in mice lacking the postsynaptic cell adhesion protein neuroligin2. Behav. Brain Res. 251, 50–64. ( 10.1016/j.bbr.2012.07.024) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Soler-Llavina GJ, Fuccillo MV, Ko J, Sudhof TC, Malenka RC. 2011. The neurexin ligands, neuroligins and leucine-rich repeat transmembrane proteins, perform convergent and divergent synaptic functions in vivo. Proc. Natl Acad. Sci. USA 108, 16 502–16 509. ( 10.1073/pnas.1114028108) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Gutierrez RC, Hung J, Zhang Y, Kertesz AC, Espina FJ, Colicos MA. 2009. Altered synchrony and connectivity in neuronal networks expressing an autism-related mutation of neuroligin 3. Neuroscience 162, 208–221. ( 10.1016/j.neuroscience.2009.04.062) [DOI] [PubMed] [Google Scholar]
  • 363.Baudouin SJ, et al. 2012. Shared synaptic pathophysiology in syndromic and nonsyndromic rodent models of autism. Science 338, 128–132. ( 10.1126/science.1224159) [DOI] [PubMed] [Google Scholar]
  • 364.Zhang C, et al. 2009. A neuroligin-4 missense mutation associated with autism impairs neuroligin-4 folding and endoplasmic reticulum export. J. Neurosci. 29, 10 843–10 854. ( 10.1523/JNEUROSCI.1248-09.2009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Jamain S, et al. 2003. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat. Genet. 34, 27–29. ( 10.1038/ng1136) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Hata Y, Butz S, Sudhof TC. 1996. CASK: a novel dlg/PSD95 homolog with an N-terminal calmodulin-dependent protein kinase domain identified by interaction with neurexins. J. Neurosci. 16, 2488–2494. ( 10.1523/JNEUROSCI.16-08-02488.1996) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Biederer T, Sudhof TC. 2000. Mints as adaptors. Direct binding to neurexins and recruitment of munc18. J. Biol. Chem. 275, 39 803–39 806. ( 10.1074/jbc.C000656200) [DOI] [PubMed] [Google Scholar]
  • 368.Dean C, Scholl FG, Choih J, DeMaria S, Berger J, Isacoff E, Scheiffele P. 2003. Neurexin mediates the assembly of presynaptic terminals. Nat. Neurosci. 6, 708–716. ( 10.1038/nn1074) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Kim E, Naisbitt S, Hsueh YP, Rao A, Rothschild A, Craig AM, Sheng M. 1997. GKAP, a novel synaptic protein that interacts with the guanylate kinase-like domain of the PSD-95/SAP90 family of channel clustering molecules. J. Cell Biol. 136, 669–678. ( 10.1083/jcb.136.3.669) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Tretter V, Jacob TC, Mukherjee J, Fritschy JM, Pangalos MN, Moss SJ. 2008. The clustering of GABA(A) receptor subtypes at inhibitory synapses is facilitated via the direct binding of receptor alpha 2 subunits to gephyrin. J. Neurosci. 28, 1356–1365. ( 10.1523/JNEUROSCI.5050-07.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Hanus C, Ehrensperger MV, Triller A. 2006. Activity-dependent movements of postsynaptic scaffolds at inhibitory synapses. J. Neurosci. 26, 4586–4595. ( 10.1523/JNEUROSCI.5123-05.2006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Abraham WC, Bear MF. 1996. Metaplasticity: the plasticity of synaptic plasticity. Trends Neurosci. 19, 126–130. ( 10.1016/S0166-2236(96)80018-X) [DOI] [PubMed] [Google Scholar]
  • 373.Abraham WC. 2008. Metaplasticity: tuning synapses and networks for plasticity. Nat. Rev. Neurosci. 9, 387 ( 10.1038/nrn2356) [DOI] [PubMed] [Google Scholar]
  • 374.Megill A, Tran T, Eldred K, Lee NJ, Wong PC, Hoe H-S, Kirkwood A, Lee H-K. 2015. Defective age-dependent metaplasticity in a mouse model of Alzheimer's disease. J. Neurosci. 35, 11 346–11 357. ( 10.1523/JNEUROSCI.5289-14.2015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Xu J, Antion MD, Nomura T, Kraniotis S, Zhu Y, Contractor A. 2014. Hippocampal metaplasticity is required for the formation of temporal associative memories. J. Neurosci. 34, 16 762–16 773. ( 10.1523/JNEUROSCI.2869-13.2014) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Allen NJ, Lyons DA. 2018. Glia as architects of central nervous system formation and function. Science 362, 181–185. ( 10.1126/science.aat0473) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Sofroniew MV, Vinters HV. 2010. Astrocytes: biology and pathology. Acta Neuropathol. 119, 7–35. ( 10.1007/s00401-009-0619-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Allen NJ. 2014. Astrocyte regulation of synaptic behavior. Annu. Rev. Cell Dev. Biol. 30, 439–463. ( 10.1146/annurev-cellbio-100913-013053) [DOI] [PubMed] [Google Scholar]
  • 379.Laywell ED, Steindler DA, Silver DJ. 2007. Astrocytic stem cells in the adult brain. Neurosurg. Clin. N. Am. 18, 21–30, viii ( 10.1016/j.nec.2006.10.003) [DOI] [PubMed] [Google Scholar]
  • 380.Gonzalez-Perez O, Quinones-Hinojosa A. 2012. Astrocytes as neural stem cells in the adult brain. J. Stem. Cells. 7, 181–188. (jsc.2012.7.3.181). [PMC free article] [PubMed] [Google Scholar]
  • 381.Higashi K, Fujita A, Inanobe A, Tanemoto M, Doi K, Kubo T, Kurachi Y. 2001. An inwardly rectifying K(+) channel, Kir4.1, expressed in astrocytes surrounds synapses and blood vessels in brain. Am. J. Physiol. Cell Physiol. 281, C922–C931. ( 10.1152/ajpcell.2001.281.3.C922) [DOI] [PubMed] [Google Scholar]
  • 382.Verkman AS. 2005. More than just water channels: unexpected cellular roles of aquaporins. J. Cell Sci. 118, 3225–3232. ( 10.1242/jcs.02519) [DOI] [PubMed] [Google Scholar]
  • 383.Theis M, Sohl G, Eiberger J, Willecke K. 2005. Emerging complexities in identity and function of glial connexins. Trends Neurosci. 28, 188–195. ( 10.1016/j.tins.2005.02.006) [DOI] [PubMed] [Google Scholar]
  • 384.Bergles DE, Diamond JS, Jahr CE. 1999. Clearance of glutamate inside the synapse and beyond. Curr. Opin. Neurobiol. 9, 293–298. ( 10.1016/S0959-4388(99)80043-9) [DOI] [PubMed] [Google Scholar]
  • 385.Beck DW, Vinters HV, Hart MN, Cancilla PA. 1984. Glial cells influence polarity of the blood-brain barrier. J. Neuropathol. Exp. Neurol. 43, 219–224. ( 10.1097/00005072-198405000-00001) [DOI] [PubMed] [Google Scholar]
  • 386.Abbott NJ, Rönnbäck L, Hansson E. 2006. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 7, 41 ( 10.1038/nrn1824) [DOI] [PubMed] [Google Scholar]
  • 387.Brown AM, Tekkök SB, Ransom BR. 2004. Energy transfer from astrocytes to axons: the role of CNS glycogen. Neurochem. Int. 45, 529–536. ( 10.1016/j.neuint.2003.11.005) [DOI] [PubMed] [Google Scholar]
  • 388.Brown AM, Ransom BR. 2007. Astrocyte glycogen and brain energy metabolism. Glia 55, 1263–1271. ( 10.1002/glia.20557) [DOI] [PubMed] [Google Scholar]
  • 389.Rothstein JD, et al. 1996. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron 16, 675–686. ( 10.1016/S0896-6273(00)80086-0) [DOI] [PubMed] [Google Scholar]
  • 390.Sofroniew MV. 2009. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 32, 638–647. ( 10.1016/j.tins.2009.08.002) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Peters A, Palay SL, Webster HdeF. 1991. The fine structure of the nervous system. Neurons and their supporting cells, 3rd edn. New York, NY: Oxford University Press. [Google Scholar]
  • 392.Stevens B, et al. 2007. The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178. ( 10.1016/j.cell.2007.10.036) [DOI] [PubMed] [Google Scholar]
  • 393.Pfrieger FW, Barres BA. 1997. Synaptic efficacy enhanced by glial cells in vitro. Science 277, 1684–1687. ( 10.1126/science.277.5332.1684) [DOI] [PubMed] [Google Scholar]
  • 394.Pyka M, Wetzel C, Aguado A, Geissler M, Hatt H, Faissner A. 2011. Chondroitin sulfate proteoglycans regulate astrocyte-dependent synaptogenesis and modulate synaptic activity in primary embryonic hippocampal neurons. Eur. J Neurosci. 33, 2187–2202. ( 10.1111/j.1460-9568.2011.07690.x) [DOI] [PubMed] [Google Scholar]
  • 395.Allen NJ. 2012. Glial control of synaptogenesis. In Neuroglia, 3rd edn (eds Kettenmann H, Ransom BR), pp. 388–401. New York, NY: Oxford University Press. [Google Scholar]
  • 396.Wang H, Ku L, Osterhout DJ, Li W, Ahmadian A, Liang Z, Feng Y. 2004. Developmentally-programmed FMRP expression in oligodendrocytes: a potential role of FMRP in regulating translation in oligodendroglia progenitors. Hum. Mol. Genet. 13, 79–89. ( 10.1093/hmg/ddh009) [DOI] [PubMed] [Google Scholar]
  • 397.Gholizadeh S, Halder SK, Hampson DR. 2015. Expression of fragile X mental retardation protein in neurons and glia of the developing and adult mouse brain. Brain Res. 1596, 22–30. ( 10.1016/j.brainres.2014.11.023) [DOI] [PubMed] [Google Scholar]
  • 398.Jacobs S, Doering LC. 2010. Astrocytes prevent abnormal neuronal development in the fragile X mouse. J. Neurosci. 30, 4508–4514. ( 10.1523/JNEUROSCI.5027-09.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Jacobs S, Cheng C, Doering LC. 2016. Hippocampal neuronal subtypes develop abnormal dendritic arbors in the presence of fragile X astrocytes. Neuroscience 324, 202–217. ( 10.1016/j.neuroscience.2016.03.011) [DOI] [PubMed] [Google Scholar]
  • 400.Maezawa I, Swanberg S, Harvey D, LaSalle JM, Jin LW. 2009. Rett syndrome astrocytes are abnormal and spread MeCP2 deficiency through gap junctions. J. Neurosci. 29, 5051–5061. ( 10.1523/JNEUROSCI.0324-09.2009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Jin XR, Chen XS, Xiao L. 2017. MeCP2 deficiency in neuroglia: new progress in the pathogenesis of Rett syndrome. Front. Mol. Neurosci. 10, 316 ( 10.3389/fnmol.2017.00316) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Stogsdill JA, Ramirez J, Liu D, Kim YH, Baldwin KT, Enustun E, Ejikeme T, Ji RR, Eroglu C. 2017. Astrocytic Neuroligins control astrocyte morphogenesis and synaptogenesis. Nature 551, 192–197. ( 10.1038/nature24638) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Ginhoux F, et al. 2010. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845. ( 10.1126/science.1194637) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404.Paolicelli RC, Gross CT. 2011. Microglia in development: linking brain wiring to brain environment. Neuron Glia Biol. 7, 77–83. ( 10.1017/S1740925X12000105) [DOI] [PubMed] [Google Scholar]
  • 405.Vinuesa A, et al. 2018. Early exposure to a high-fat diet impacts on hippocampal plasticity: implication of microglia-derived exosome-like extracellular vesicles. Mol. Neurobiol. ( 10.1007/s12035-018-1435-8) [DOI] [PubMed] [Google Scholar]
  • 406.Almeida RG, Lyons DA. 2017. On myelinated axon plasticity and neuronal circuit formation and function. J. Neurosci. 37, 10 023–10 034. ( 10.1523/JNEUROSCI.3185-16.2017) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407.Larson VA, Mironova Y, Vanderpool KG, Waisman A, Rash JE, Agarwal A, Bergles DE. 2018. Oligodendrocytes control potassium accumulation in white matter and seizure susceptibility. Elife 7, e34829 ( 10.7554/eLife.34829) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.Nave KA, Ehrenreich H. 2014. Myelination and oligodendrocyte functions in psychiatric diseases. JAMA Psychiatry 71, 582–584. ( 10.1001/jamapsychiatry.2014.189) [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Reviewer comments

Data Availability Statement

This article does not contain any additional data.


Articles from Open Biology are provided here courtesy of The Royal Society

RESOURCES