Abstract
Neurodevelopmental disorders result from impaired development or maturation of the central nervous system. Both genetic and environmental factors can contribute to the pathogenesis of these disorders; however, the exact causes are frequently complex and unclear. Individuals with neurodevelopmental disorders may have deficits with diverse manifestations, including challenges with sensory function, motor function, learning, memory, executive function, emotion, anxiety, and social ability. Although these functions are mediated by multiple brain regions, many of them are dependent on the hippocampus. Extensive research supports important roles of the mammalian hippocampus in learning and cognition. In addition, with its high levels of activity-dependent synaptic plasticity and lifelong neurogenesis, the hippocampus is sensitive to experience and exposure and susceptible to disease and injury. In this review, we first summarize hippocampal deficits seen in several human neurodevelopmental disorders, and then discuss hippocampal impairment including hippocampus-dependent behavioral deficits found in animal models of these neurodevelopmental disorders.
Keywords: Neurodevelopment, neurodevelopmental disorder, hippocampus, animal model, behavior, hippocampal-dependent
1. Introduction
Neurodevelopmental disorders (NDs) are a diverse group of disorders resulting from impaired development or maturation of the central nervous system. The causes of NDs are also varied and complex. NDs can result from mutations of single genes, such as in fragile X syndrome (FXS) and Rett syndrome (RTT), or from environmental exposures or injuries, such as fetal alcohol spectrum disorders (FASD) and perinatal ischemia. Most NDs are likely the result of both genetic alterations and environmental impacts1,2. NDs affect many aspects of biological functions controlled by the brain, including but not limited to challenges with sensory function, motor function, learning, memory, executive function, emotion, anxiety, and social ability. Although these functions are mediated by multiple brain regions, many of them are dependent on proper functioning of the hippocampus, as discussed below.
Extensive research has established critical roles of the mammalian hippocampus in the formation of spatial, temporal, and contextual memories. The hippocampus seems to be important for distinct encoding, retrieval, and consolidation steps during memory formation3-8. For instance, hippocampal mechanisms can directly influence sensorimotor functions, such as locomotor activity and startle reactivity. Aberrant hippocampal function can lead to neuropsychiatric diseases, in particular psychosis 9. In addition, diverse behavioral effects may be associated with different hippocampal subregions. For example, the dorsal hippocampus has a preferential role in certain forms of learning and memory, notably spatial learning, while the ventral hippocampus may have a preferential role in brain processes associated with anxiety-related behaviors 10. Hippocampal neurons send distinct behavior-contingent information selectively to different target areas. Projections from the ventral hippocampal CA1 to the shell of the nucleus accumbens play a necessary and sufficient role in the formation of social memory11. On the other hand, projections from the ventral hippocampus to the basal amygdala mediate contextual fear behavior, and projections from the ventral hippocampus to the central amygdala mediate retrieval of context-dependent memories of fear12. Also, the hippocampus integrates with subcortical sites and the prefrontal cortex to play key roles in executive functions. This functional connectivity enables learning-behavior translation, and is in part why hippocampal dysfunctions results in memory and executive deficits13.
Long-term potentiation (LTP) is a form of protein synthesis-dependent synaptic plasticity and is considered to be the cellular basis of learning and memory14. LTP in the hippocampal CA3-CA1 pyramidal neurons is important for learning, and the strength of hippocampal LTP has been used extensively to evaluate the impact of genetic mutations, exposures, diseases, and injuries on brain functions15
In addition, a unique feature of the mammalian hippocampus is lifelong adult neurogenesis16. In mammalian experimental models, the subgranular zone of the dentate gyrus (DG) of the hippocampus and the subventricular zone (SVZ) of the lateral ventricles have continuous new neuron production, even though major developmental neurogenesis ceases upon birth in most other brain regions. This postnatal hippocampal neurogenesis, termed adult neurogenesis, has been found in nearly all mammalian species analyzed to date16, including in humans17-19. Extensive studies using rodent models have shown that a small number of radial glia-like neural stem cells (NSCs) in the adult hippocampus actively integrate both extrinsic and intrinsic signals to either maintain their quiescent state or divide and give rise to intermediate progenitor cells, which subsequently differentiate into glutamatergic DG granule neurons or astrocytes16. Although the scale of adult neurogenesis is small, with about 700 new cells generated per day in humans, the lifelong addition of these new cells may have a significant impact on behaviors, neural plasticity, and hippocampal function18,19. Animal studies using specific genetic ablation or enhancement of adult hippocampal neurogenesis have provided convincing evidence for the importance of adult neurogenesis in hippocampal-dependent learning and working memory20-22. In addition, a number of recent studies suggest that neurogenesis in dorsal and ventral hippocampus has differential roles in hippocampal-dependent learning and memory23. These findings are consistent with the notion that sub regions of the adult hippocampus mediate different behaviors, with the dorsal hippocampus important for spatial learning and memory and the ventral hippocampus essential for emotional behavior24.
The hippocampus is known to be vulnerable to hypoxia, stress, toxins, and malnutrition, and it plays a clinically relevant role in children who are born preterm or who have been exposed to alcohol, toxins, or drugs during fetal development25-27. Also, as discussed in this review, hippocampal dysfunctions are found in NDs resulting from genetic mutations, including FXS, RTT, and Down syndrome (DS)28-35. The development and progress of noninvasive magnetic resonance imaging (MRI), including volumetric MRI analysis and functional MRI (fMRI), has facilitated the study of hippocampal changes in children with NDs.
Adult neurogenesis is also highly sensitive to genetic mutations and life experiences, providing the cellular basis for gene and environmental interactions that impact behaviors36,37. Numerous animal studies have shown the important roles of adult hippocampal neurogenesis38-43. Enhancement of adult neurogenesis via environmental enrichment, physical exercise, nutrition, inhibition of cells death, etc., leads to improved learning and memory, cognitive ability, and reduced anxiety in animal models44-49. On the other hand, inhibiting adult neurogenesis through genetic mutations, pharmacological inhibition, chronic stress, and disease modeling leads to impaired cognitive functions21,40,50-52. Altered adult neurogenesis has been found in a number of animal models of NDs and neuropsychiatric disorders53. However, due to a lack of effective non-invasive methods adult neurogenesis in humans is mostly assessed indirectly through radio carbon dating 18 and histological analysis of postmortem tissues17,19. Notably, human adult neurogenesis has yet to be assessed in individuals with NDs.
In this review, we first summarize hippocampal deficits seen in several human NDs, and then discuss the hippocampal changes, particularly hippocampus-dependent behavioral deficits, found in animal models of NDs (Table 1).
Table 1.
Rodent Behavioral Tests for Assessing Hippocampal Related Deficits
Functional Domains |
Behavioral Tests |
Description | Hippocam pus- dependent |
Adult neurogenes is- dependent |
Species | Disease models tested |
Example References |
---|---|---|---|---|---|---|---|
Associative Learning |
Contextual Fear Conditioning |
Associate environment with aversive foot shock | Yes | Yes | Mouse Mouse |
FXS RTT |
(Kazdoba et al., 2014) (Stearns et al., 2007) |
Trace Fear Conditioning | Associate foot shock with context in the presence of time interval between stimuli | Yes | Yes | Mouse | FXS | (Guo et al., 2011; Guo et al., 2012; Zhao et al., 2005) | |
Spatial Learning and Working Memory | Barnes Maze | Learn and find hiding hole based on spatial cues | Yes | Yes | Mouse | FASD | (Houle et al., 2017) |
Radial Arm Maze | Distinction between closely spaced arms | Yes | Yes | Mouse | FASD | (Berman and Hannigan, 2000) | |
Morris Water Maze | Visual-spatial memory task. | Yes | Yes | Mouse Mouse Mouse Mouse+Ra t |
FXS ASD RTT FASD |
(Kazdoba et al., 2014) (Kwon et al., 2006) (Stearns et al., 2007) (Berman and Hannigan, 2000) |
|
Novel Location Test |
Preference for object in a novel location. | Yes | Yes | Mouse | FXS RTT |
(Guo et al., 2011; Guo et al., 2012; Zhao et al., 2005) (Stearns et al., 2007) | |
Sociability | Three-chamber sociability task | Preference for novel mouse over novel object. | Yes | ? | Mouse+Ra t Rat |
ASD FXS |
(Harony-Nicolas et al.,2017; Jones et al., 2011; Kwon et al., 2006; Sungur et al., 2017; Won et al., 2012) (Tian et al., 2017) |
Caged adult social interaction test |
Social preference test for novel mouse or wild-type mouse | ? | ? | Mouse | ASD | (Tabuchi et al., 2007) | |
General Cognition |
Novel Object Recognition |
Preference for novel object. |
Yes |
? |
Mouse Mouse Mouse |
ASD FXS RTT |
(Sungur et al., 2017) (Kazdoba et al., 2014; Li et al., 2016) (Stearns et al., 2007) |
Anxiety | Open field | Overall activity and anxiety | Yes | ? | Mouse | FXS | (Guo et al., 2011; Kazdoba et al., 2014) |
Dark/Light Box |
Anxiety | Yes | ? | Mouse | FXS | (Kazdoba et al., 2014) | |
Elevated plus maze | Anxiety | Yes | ? | Mouse | FXS | (Kazdoba et al., 2014) |
Note: RTT, Rett Syndrome; FXS, fragile X syndrome; FSAD, fetal alcohol spectrum disorders, ASD, autism spectrum disorders.
?, Insufficient information
2. Hippocampal phenotypes in human neurodevelopmental disorders
2.1. Autism and Autism Spectrum Disorder
Autism is a complex developmental disability that appears during the first few years of life54. Autism and its related disorders, collectively called autism spectrum disorder (ASD), affect 1 in 68 children in the United States (https://www.cdc.gov/mmwr/volumes/65/ss/ss6503a1.htm), with four times more boys than girls affected55. Although about 1000 genes are linked to ASDs, the exact causes are not fully understood. The hypothesis is that a combination of genetic and environmental factors are involved in the pathogenesis and severity of ASDs56. Non-invasive MRI has been a key technology for examining abnormalities in the brains of ASD patients, especially children29. A comprehensive literature review of early structural MRI studies shows increased total brain, parieto-temporal lobe, and cerebellar hemisphere volume in juvenile and adult males with ASDs, after adjusting for height, IQ, and intra-cranial volume57. Notably, however, only some studies observed a reduction in the volume of the hippocampus of individuals with an ASD58. The differences among these results could be due to the different ages of participants included and the relatively low sensitivity of methods used to measure the hippocampus. A more recent study revealed that the relative volume, but not the absolute volume, of the hippocampus is reduced in individuals between the ages of four and 18 who have an ASD, compared to matched controls59. Another study of mature adult men (40-64 years old) with ASD found a bilateral reduction in hippocampal volume 60. In addition to volume measurements, improved and innovative methods are being developed to study ASD patients. For example, Chaddad et al. used a novel MRI multi-scale image texture analysis to quantify the spatial heterogeneity of brain tissues, including hippocampal volume. They characterized the link between neuroanatomical regions and ASDs and found that five out of 31 prominent subcortical brain regions exhibit significant changes, including the right hippocampus, which is believed to be responsible for encoding spatial relationships61. In a parallel study, Chaddad et al. performed more detailed image texture analysis on the hippocampus and the amygdala and found that hippocampi are significantly different between those with an ASD and controls with regard to several texture features analyzed62. These findings indicate that hippocampal texture features could be used as biomarkers to diagnose and characterize ASD.
2.2. Fragile X syndrome (FXS)
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and the most common single-gene mutation associated with autism63,64. FXS results from CGG expansion in the 5’ untranslated region of the fragile X mental retardation 1 (FMR1) gene on the X chromosome, which leads to epigenetic silencing of the gene63,64. Since the identification of FMR1 as the causal gene for FXS in 199165, extensive research, based mainly on animal models, has explored the functions of fragile x mental retardation protein (FMRP) the protein encoded by FMR1 gene66-69. FMRP is an RNA-binding protein that regulates the translation of a number of mRNAs whose protein products are important for synaptic development, maintenance, and plasticity70,71. However, despite several research advances, how FMRP deficiency affects human development and the pathogenesis of FXS remains unclear, and there is currently no cure for this ND.
FXS patients exhibit extensive behavioral impairment in executive function, learning, short-term and working memory, and social ability, as well as heightened anxiety72. Many of these deficits are related to hippocampal functions and dysfunctions73. Despite advances in our understanding of the molecular changes in FXS, there is limited neuropathological information on FXS patients. To address this issue, Greco et al. analyzed the hippocampus and cerebellum in three men with FXS using histologic, immunochemical, and molecular techniques74. They found that individuals with FXS had dysmorphic, enlarged hippocampi, decreased cerebellar size, and preferential atrophy of vermal lobules VI to VII74. Another study, however, has found no change in the size of the hippocampus in human FXS75. The discrepancies among these studies may result from different methods used and different ages of the patients, similar to those seen in ASD studies. On the other hand, fMRI and behavioral assessments have revealed that individuals with FXS exhibit reduced hippocampal activation during visual memory encoding 76 and deficits in episodic memory 77. Both of these functions are dependent on the hippocampus. Therefore, the neurological and neuropsychiatric symptoms seen in FXS may, at least in part, be due to abnormal development and dysfunction of the hippocampus.
2.3. Rett syndrome (RTT)
Epigenetic mechanisms, including DNA methylation and histone modification, are known to play significant roles in brain development and function. They also mediate environmental impact on brain functions78-80. DNA methylation, mostly on CpG dinucleotides (mCG), helps regulate both neurodevelopment and adult neuroplasticity81-83. Methylated cytosines (mCs) are recognized by proteins with methyl-CpG-binding domains, including methyl-CpG binding protein 2(MeCP2)84,85. Spontaneous mutations in MeCP2 genes on the X-chromosome are found in 95% of classic RTT patients86,87. RTT is a progressive neurodevelopmental disorder that almost exclusively affects girls with an incidence of ~1/10,000 live female births88. RTT is characterized by an apparently normal development for the first 6–18 months of life followed by a gradual appearance of mental, neurological and physical symptoms89. RTT patients exhibit severe mental impairment, slowed growth, motor dysfunction and seizures90. Other symptoms include cardiac abnormalities and pronounced breathing disturbance91. MRI analysis has shown significantly reduced volume in both grey and white matters, including the hippocampus, in RTT, and patients with more severe symptoms exhibit more significant reductions92,93. Magnetic resonance spectroscopy (MRS) has found significant changes in metabolites, including N-acetyl aspartate (NAA), in the hippocampus of RTT patients94. Recently, a MeCP2-mutant monkey model has been created through genome editing. MRI analysis has shown that the size of the hippocampus, particularly the posterior parahippocampal gyrus, is reduced in these monkeys95, similar to what has been found in human patients. In addition, histological studies of human postmortem tissues have found that pyramidal neurons in the cortex and the hippocampus of RTT individuals have reduce dendritic complexity96-98. Chapleau et al showed hippocampal CA1 pyramidal neurons from postmortem female patients had reduced dendritic spine density compared to controls98. To confirm this result, they introduced RTT-mutant MeCP2 into hippocampal neurons of rat brain slices and found that expression of mutant MeCP2 leads to reduced dendritic spine density compared to wild type MeCP298. Hence, hippocampal deficits are apparent and severe in RTT and likely contribute to neurological and cognitive symptoms.
2.4. Down syndrome (DS)
Down syndrome, or trisomy 21, is the most common human aneuploidy condition leading to intellectual disability 99. Chromosome 21 is the smallest human chromosome100, which may explain why trisomy of this chromosome is compatible with life, and results in only mild-to moderate intellectual disability. DS patients exhibit cognitive impairment, motor dysfunction, increased seizure risk, and increased incidence of Alzheimer’s disease101,102. There have been a large number of MRI analyses for DS, and these studies have shown that DS patients have reduced volume of the hippocampus103. In addition, DS patients with dementia exhibit more severe reductions of hippocampus volume compared to DS patients without dementia103-105.
2.5. 15q syndromes
The human chromosome 15q11-q13 region is prone to copy number variations, which can be either deletions or duplications, that lead to several distinct neurodevelopmental disorders with some overlapping phenotypes106. In addition, part of this genomic region is imprinted and its haploinsufficiency leads to either Angelman syndrome (AS), if it is a maternal deletion, or Prader-Willi syndrome (PWS), if it is a paternal deletion107. Genetic changes in this 15q region lead to several characteristic NDs, and all of them exhibit hippocampal deficits to some extent, as discussed below.
Angelman syndrome, which affects 1 in 12,000-20,000 people, results mostly from maternal deficiency of UBE3A, a gene located on 15q11-q13108. Children with AS seem to develop normally during the first few months of life with clinical symptoms manifesting later in development109. Children with AS have delayed development, impaired speech, defective movement, and unique behavioral features including a happy demeanor and frequent laughing110. Microcephaly is a common feature of children with AS111. Therefore, the overall brain volume, including global white matter and gray matter (including the hippocampus), in children with AS is smaller compared to normal children112.
Prader-Willi syndrome (PWS) has a prevalence of around 1 in 15,000 births and affects males and females equally. It can result from either paternal genetic deletion of 15q11-q13 or maternal uniparental disomy or imprinting defects113. PWS patients exhibit profound early childhood-onset obesity caused by excessive and pathological overeating. They also show intellectual disability, including speech delay, learning disabilities (low or very low intelligence and deficits in adaptive behaviors without reference to etiology), and cognitive deficiencies (depressed general cognitive functioning or IQ, processing deficits, including problems with short-term memory, language processing deficits, and difficulty with higher order processing)114. Structural MRI studies have shown that PWS patients have reduced gray matter volume that encompasses several major brain regions, including the hippocampus115. Functional MRI studies found that PWS patients have reduced neural network connectivity in several cortical regions, including the prefrontal cortex and the hippocampus116. PWS is one of a very few well defined genetic causes of obesity. Therefore, food-induced brain changes have been studied extensively117. Studies have shown that several brain regions, including the medial prefrontal cortex, amygdala, and hippocampus are activated by food or either sight or smell of food, but there was a decreased response to food stimulus after eating118-120. It has been shown that PWS patients exhibit abnormal food stimuli-induced activity in these brain regions compared to typical individuals with normal body weight121. Another study comparing PWS versus non-PWS obese individuals discovered that PWS patients show hyperactivation in subcortical reward circuitry but hypoactivation in cortical regions including the hippocampus after eating122.
Patients with maternal copy number gains of 15q11-q13 are diagnosed with 15q duplication syndromes (Dup15q) and exhibit global developmental delay, intellectual disability, autistic phenotypes, and epilepsy106. The prevalence of Dup15q is estimated to be 1 in 30,000 with a sex ratio of 1:1123. MRI studies showed that Dup15q patients have increased hippocampus sclerosis, a lesion possibly caused by excitotoxicity124.
2.6. Fetal alcohol spectrum disorders (FASDs)
Fetal alcohol spectrum disorders (FASDs) is the umbrella term covering the full range of deficits resulting from prenatal exposure to alcohol125. The most severe form of FASD is fetal alcohol syndrome (FAS), which affects 0.5-2 per 1000 births in the US but varies across the world, with the highest reported rates in South Africa (60 per 1000 births)126,127. Individuals with FASDs exhibit abnormal appearance, short height, low body weight, small head size, poor coordination, low intelligence, behavior problems, and problems with hearing or seeing128. Surveys from the United States have found that about 19.6% of women drink alcohol while pregnant129. The drinkers reported significantly higher levels of depressed mood and more problems with alcohol than their counterparts who abstained129. Studies have shown that even low to moderate levels of exposure to alcohol can have detrimental effects130. The incidence of FASD is unclear because of the difficulty in identifying the affected individuals, but the prevalence is likely high. Individuals with FASD have a wide range of deficits130,131, including reduced IQ, cognitive impairments, and social and behavioral difficulties as well as predisposition to mental health issues130,131. Individuals with FASD typically do not complete their education and are often unemployed and many of them experience depression, increased suicide risk, and trouble with the legal system. Consequently, FASD imposes an enormous life-long burden on families and society132,133.
Neuroimaging analysis has identified significant abnormality in brain regions of individuals with FASD with overall reduction in brain volumes134. The deficits associated with FASD point to impairment of hippocampal functions135. MRI assays have shown reduced hippocampal volumes in individuals with FASD, which correlated with learning and memory deficits. Magnetic resonance spectroscopy (MRS) has also identified changes in metabolites in the hippocampus of FASD individuals134. A recent MRI analysis has confirmed that the sizes of all brain structures analyzed were reduced in individuals with FASD compared to controls, including gray and white matter of the cerebrum and cerebellum, and all deep gray matter, including the hippocampus, amygdala, thalamus, caudate, putamen, and pallidum136. Although alcohol exerts extensive effects on developing brains, these research findings indicate that its impact on the hippocampus likely contributes to the learning and memory deficits seen in FASD.
3. Hippocampal phenotypes in animal models for neurodevelopmental disorders
3.1. ASD
Alterations in neural network connectivity and memory function are frequently observed in autism patients and often involving the hippocampus137. However, the changes that occur during early brain development that lead to disrupted brain functions in ASD remain largely unclear. A large number of animal models for ASD have been developed and used for investigating the changes in developing brains138. Studies have shown that rodents exposed to valproic acid (VPA) during fetal development go on to exhibit many features of autism. Therefore, prenatal exposure to VPA has been used to establish model of autism in rodents to extensively study the underlying mechanisms behind autism. However, since different dosages and timing of prenatal VPA treatment may result in various phenotypes, interpretations of results should be made with caution139,140. Advancement in human genetics has led to the discovery of many genes associated with ASD, and based on these discoveries, over 1000 autism genetic mouse models and over 200 rat genetic models have been created. Simons Foundation Autism Research Initiative (SFARI) has identified 65 high–ranking candidate risk genes for ASD, with SHANK3 and PTEN among the high confidence genes (https://www.sfari.org/resource/sfari-gene/).
SHANK proteins are scaffolding proteins, located at excitatory synapses, and are crucial for proper synaptic development and function. The SHANK proteins SHANK1, 2, and 3 are encoded by separate genes in both human and mouse genomes141. Mutations in any of the three SHANK genes have been associated with autism142. Mutant mouse models have been generated to study the functions of these genes, and these mutant mice shared apparent hippocampal deficits. Hippocampal neurons of Shank1null mice exhibited altered postsynaptic density protein (PSD) composition and thinner PSDs, reduced size of dendritic spines, and decreased AMPAR-mediated excitatory synaptic transmissions143. In addition, Shank1null mice exhibit decreased reduced tonic firing rates of parvalbumin (PV) positive neurons in the hippocampus144. At the behavioral level, Shank1 null mice are defective in hippocampus-dependent contextual fear memory and object recognition memory145,146. SHANK1 protein is highly co-localized in PV-expressing fast-spiking inhibitory interneurons in the hippocampus. Lack of SHANK1 in these cells leads to a decrease in excitatory synaptic inputs and inhibitory synaptic outputs to pyramidal neurons at the functional level, together with molecular changes, including the downregulation of the postsynaptic proteins GKAP, PSD-95, and gephyrin144. Shank2 (also known as ProSAP1) null mice are extremely hyperactive and display profound autistic-like behavioral alterations including repetitive grooming as well as abnormalities in vocal and social behaviors147. At the cellular level, neurons in the hippocampus of Shank2 null mice exhibit reduced spine density and impaired NMDAR-dependent synaptic plasticity 147,148. Because SHANK3 has the strongest link to ASD, several mouse lines with distinct Shank3 gene mutations have been developed to help understand ASD-related neurobiology. These mouse models showed ASD-related behavioral phenotypes, including impaired social behavior, increased repetitive behaviors, motor deficits, as well as altered synaptic transmission and neuronal morphology in the hippocampus142. To better study ASD-related behaviors, Shank3-deficient rats have been created. These rats exhibit impaired long-term social recognition memory and attention, and reduced synaptic plasticity in the hippocampal-medial prefrontal cortex pathway149. The peptide hormone oxytocin has been shown to improve social memory, enhance social reward, and modulate social attention in human and non-human primates150-152,153, Harony-Nicolas et al utilized oxytocin to rescue social and attention deficits in Shank3 deficient rats149. Several studies have identified the downstream pathways and proteins associated with the SHANK3 network. It has been shown that Shank3 deletion impairs the function and synaptic localization of mGluR5 in several brain regions, including the hippocampus in mice154,155 and rats156. Cope et al has also assessed adult hippocampal neurogenesis in Shank3+/ΔC transgenic mice that have deletion of a Homer-binding region, a mutation also found in some humans with autism and found that both Shank3+/ΔC mice exhibit significant reduction in the number of radial glia-like neural stem cells and immature neurons in the DG of the ventral hippocampus157. Therefore, the phenotypes of animal models with SHANK protein mutations or deletions support the involvement of the hippocampus in the pathology of ASD.
The phosphatase and tensin homolog on chromosome ten (PTEN) gene is a tumor suppressor, and its mutation leads to many human cancers158. PTEN mutations are associated with a number of neurological conditions, including ASD, megalocephaly and epilepsy as well159. Homozygous deletion of PTEN in mice leads to embryonic lethality, and heterozygote mutant mice are prone to tumor formation160,161, demonstrating the important role of this protein in development. PTEN catalyzes dephosphorylation of phosphatidylinositol 3, 4, 5 trisphosphate, a critical cellular signaling molecule that controls many cellular pathways162. Extensive studies using mouse models have shown that PTEN-induced hyperactivation of the mammalian target of rapamycin (mTOR) pathway leads to abnormalities in neuronal proliferation, survival, growth and plasticity in the brain including in the hippocampus163. PTEN deletion in hippocampal DG and CA3 neurons leads to dysregulation of synaptic plasticity of DG granule neurons, which seems to precede visible morphological changes164. Several laboratories have investigated the role of PTEN in adult hippocampal neurogenesis. Conditional deletion of PTEN in Nestin-expressing cells in mice leads to increased proliferation, reduced differentiation and stem cell depletion 165. In addition, acute knockdown of PTEN in new neurons in the adult hippocampus leads to hyper-excitability166. Furthermore, although it is not known whether adult hippocampal neurogenesis regulates social interaction, neurogenesis during adolescence has been found to be important in developing social ability167. Interestingly, deletion of PTEN in new DG neurons born in 4-weeks old mice leads to impaired social interaction165, and is a core phenotype of autism. Therefore, juvenile neurogenesis may contribute to social impairment seen in ASD.
Neurexin (NRXN) and Neuroligin (NLGN) family proteins cooperatively function at synaptic terminals, and the absence of both is strongly associated with the occurrence of ASDs, especially NRXN1 and NLGN3 168-170. Mammals have three NRXN genes (NRXN1, NRXN2 and NRXN3), and all of them produce longer α- and shorter β- form of proteins through transcription from independent promoters171. Several different lines of NRXN knockout (KO) mice have been developed. α-NRXN triple KO mice die shortly after birth due to respiratory failure 172. β-NRXN triple KO mice are ~80% smaller in body weight, exhibit impairment in hippocampus-dependent contextual fear memory and reduction in excitatory neurotransmitter release in the hippocampus173,174. NRXN3 KO mice lacking both α- and β- forms show reduced AMPA receptor function in hippocampal neurons due to a deficit of NRXN3-mediated trans-synaptic regulation of postsynaptic AMPA receptors175.
There are four NLGNs in mammals176. The NGLN3 mutant (R451C allele) mouse model, carrying a single rare point mutation of NLGN3 was originally discovered in patients with ASDs. This mouse model shows impaired social interaction but enhanced spatial learning abilities with increased LTP and excitatory transmission in the hippocampal region 177,178. NLGN3 R451C mice also display impairment in tonic endocannabinoid signaling at inhibitory synapses formed between interneurons and hippocampal pyramidal neurons, and this endocannabinoid signaling may be essential for maintaining excitation and inhibition balance179. On the other hand, NLGN3 R704C knockin mice, which carry another autism mutation, show a selective reduction of AMPA receptor-mediated synaptic transmission in the hippocampus180. Mice deficient in other Neuroligins (NGLN 1, 2, 4) also exhibit hippocampal deficits, including synaptic plasticity changes in the CA regions and impaired hippocampal-dependent spatial learning172.
In addition to the genetic models created based on human ASD gene discoveries, phenotypic screening using inbred strains as part of the Jackson Lab Phenome project has identified BTBRT+tf/J (BTBR) mice as the strain that shows the strongest ASD-related phenotypes. These mice show impaired social interaction and communication, increased repetitive behaviors and learning and memory deficits138,181. Additional studies show that BTBR mice also have hippocampal deficits, including enhanced development of dendritic arbor of hippocampal CA1 pyramidal neurons 182, greater relative hippocampal volume 183,184 and increased local connectivity in the hippocampal commissure185. BTBR mice also show reduced GABAergic neurotransmission in the hippocampus. Low-dose benzodiazepine treatment, which increases inhibitory neurotransmission through activation of GABAA receptors, improves deficits in social interaction, repetitive behavior, and spatial learning186. Therefore, mouse models of ASD exhibit altered hippocampal functions and hippocampal associated behavioral deficits.
3.2. Fragile X syndrome (FXS)
Animal models for FXS have been developed in Drosophila, zebrafish, mice, and rats. Most FXS behavioral modeling has been done using mouse models with a large number of publications 187-190. In the most widely used mouse model (Fmr1 KO), exon 5 was interrupted by the positive selection marker gene neomycin (neo), resulting in a mutant Fmr1 mRNA, which fails to produce FMRP protein191. The Fmr1 KO mouse model has been extensively tested and compared with human FXS phenotypes. These mice exhibit several human FXS-related deficits but not others192. Several mouse behavioral tests have been used to assess hippocampus-dependent cognitive functions in Fmr1 KO mice. Although some studies have shown that Fmr1 KO mice are impaired in hippocampus-dependent contextual fear conditioning, delayed trace conditioning, Morris Watermaze, novel object recognition, and novel location tests, other laboratories have found no change or even opposite results (Figure 1, Table 1) (Please refer to Kazdoba et al for a comprehensive review on variabilities in behavioral deficits of Fmr1 KO mice192).
Figure 1.
Comparison of behavioral deficits, particularly hippocampus-dependent deficits, observed in human fragile X syndrome patients and those found in Fmr1 mutant mouse models.
Hippocampus-dependent behaviors are indicated with asterisks (*). Adult hippocampal neurogenesis-dependent behaviors are indicated with an arrow (→).
Both the hippocampus and isolated hippocampal neurons of rodents have been widely used to study FXS. Fmr1 KO mice show enhanced protein synthesis–dependent long-term depression of synaptic transmission in the hippocampus, which results from the alteration of mGluR signaling193-195. A consistent discovery is that the absence of FMRP leads to exaggerated mGluR5 signaling, enhanced protein synthesis, and changes in synaptic plasticity. Based on the mGluR5 theory196, a number of pharmacological preclinical studies have been done and show remarkable effects in correcting Fmr1 KO mice phenotypes through mGluR5 inhibition197-199. Subsequently, large clinical trials were carried out between 2008 and 2014, which aimed at blocking mGluR5200 using molecules such as two mGluR5 antagonists, basimglurant201 and mavoglurant202. Both hippocampal neurons isolated from neonatal Fmr1 KO mice and neurons in the hippocampus of Fmr1 KO mice exhibit the immature dendritic spine phenotypes shown by limited studies of human postmortem tissues 203-205. Meredith et al investigated whether abnormal spine morphology observed in the Fmr1 KO mouse is paralleled by changes in basic functional synaptic properties at three different developmental stages of hippocampal development. They observed that inhibition of mGluR5 rescued synaptic plasticity when treatment was done at 2 weeks but not at 1 week or 8-10 weeks of age, pointing to the importance of treatment timing for certain FXS phenotypes206.
Several laboratories, including our own, have investigated the role of FMRP in adult hippocampal neurogenesis and found that FMRP deficiency leads to changes in adult neurogenesis207-209. FMRP deficiency in adult hippocampal NSCs led to increased proliferation of adult NSC and altered NSC specification from neurons to astrocytes52,208. In addition, selective deletion of FMRP from adult NSCs and new neurons leads to impaired performance of trace conditioning, radial arm maze, novel object recognition, and novel location test which are tasks dependent on an intact hippocampus52,189. On the other hand, restoration of FMRP only in adult new neurons partially rescued some of these deficits, supporting an important role of FMRP in adult neurogenesis and cognition52. Using NSCs isolated from adult hippocampus, Guo et al showed that FMRP deficiency in adult NSCs leads to impaired Wnt signaling and inhibition of GSK-3-β, an inhibitor of Wnt signaling, can rescue both adult neurogenesis and hippocampus-dependent learning208,210. Recently, Li et al discovered that FMRP controls the activation of adult NSCs which is important for rebalancing NSC quiescence and activation189. We showed that FMRP maintains the NSC pool through regulation of the MDM2-P53 pathway. A small molecule, Nutlin-3, rescued both neurogenic and cognitive deficits in fragile X syndrome mice189. Subsequently, we have found that loss of FMRP depletes adult hippocampal NSCs and leads to cognitive impairment, both of which are associated with dysregulation of EP300, a histone acetyltranferase, and targeting of HDAC1, a histone deacetylase, by MDM2, leading to elevated histone H3 and H4 acetylation. Rebalancing histone acetylation with the small molecules Nutlin-3 or Curcumin inhibits MDM2 and EP300 and rescues both neurogenic and cognitive deficits in Fmr1 mutant mice211. Fmr1 KO rats have been generated using CRISPR/Cas9 technology and these rats have impaired long-term synaptic plasticity and hippocampus-dependent learning212. Therefore, hippocampal dysfunctions likely underscore the extensive cognitive deficits in FXS.
3.3. Rett syndrome (RTT)
Methyl-CpG binding protein 2 (MECP2) binds to methylated DNA in the genome leading to transcriptional changes. Therefore, it is an important reader and interpreter of DNA methylation across the genome213. Several mouse models of RTT have been created to resemble human conditions, Interestingly, complete MeCP2 deficiency (null mutant), partial MeCP2 deletions, point mutations, phosphorylation site mutations, or haploinsufficiency recapitulate many neurological and behavioral deficits of human RTT214-218. In addition, conditional mutant mouse models have been generated and used to demonstrate the function of MeCP2 in specific cell types at different developmental stages, as well as identify MeCP2 regulated targets in various brain regions and cell types219-223. Furthermore, these mouse studies show that MeCP2 mutations or deficiency in hippocampal neurons leads to extensive functional deficits, including impaired dendritic and spine development, altered excitation inhibitor balance and changed synaptic plasticity224-227. In addition, mice with MeCP2 deletion specifically in GABAergic neurons has impaired hippocampal LTP and exhibit repetitive behaviors and impaired hippocampal learning and memory228.
One of the most consistent behavioral phenotypes of MeCP2-deficient mice is impaired contextual fear conditioning, which is dependent on both the hippocampus and adult hippocampal neurogenesis229. Several studies have investigated the role of overall MeCP2 expression in regulating hippocampal neurogenesis230,231. Although MeCP2 null mice did not show impaired NSC proliferation or differentiation, new adult hippocampal neurons exhibit reduced dendritic complexity and spine density232. MeCP2 regulates maturation of adult born neurons by repressing several microRNAs231,233. In post-mitotic neurons, depolarization-induced Ca2+ influx through voltage-gated calcium channels (VGCCs) has been shown to trigger phosphorylation of MECP2 at serine 421 (S421)234,235, which is required for regulating synaptogenesis, dendritic morphology, synaptic scaling, long-term potentiation and spatial memory in the adult mouse brain236-238. However, it is unclear whether phosphorylation at any of the identified sites on MECP2 can be induced by signals other than neuronal activity in other cell types, and what functions MECP2 phosphorylation may have in those contexts. Li et al analyzed adult neurogenesis in mice with S421 mutation of MECP2 and found that S421 phosphorylation regulates the proliferation and neural differentiation of adult NPCs by controlling key factors of Notch signaling pathways239. This finding suggests that MECP2 has cell type-specific roles in adult neurogenesis. Deep brain stimulation of the hippocampus in symptomatic RTT mice restored hippocampal synaptic plasticity, and adult hippocampal neurogenesis, and rescued hippocampus-dependent contextual fear conditioning.240. Although it remains to be determined whether this treatment effect is associated with adult neurogenesis, these exciting results underscore the significance of hippocampal deficits and rescue in RTT.
3.4. Down syndrome
Several Down syndrome (DS) mouse models have been developed: Ts65Dn241 and Ts1Cj242 mice carry larger trisomic segments of mouse chromosome 16, which contains most regions orthologous to human chromosome 21. Ts2Cje243 mice carry a smaller segment without APP. Ts65Dn mice exhibit extensive cognitive deficits similar to humans with DS and therefore have been used extensively as a DS model244. Quantitative MRI studies have been used to show that Ts65Dn mice exhibit impaired cholinergic circuits, including those in the hippocampus245. Diffusional kurtosis imaging (DKI) analysis shows that Ts65Dn mice have changes in the hippocampus at all ages analyzed (2 months to 8 months)246. Ts65Dn mice exhibit excessive inhibition in hippocampal circuits and show severe impairment of LTP, which can be normalized by inhibition of GABAA receptors247. A study using four mouse models carrying different regions of human chromosome 21 showed that none of these strains exhibited all impairments seen in human individuals with DS, but some strains show reference memory, working memory, and episodic memory changes. Nevertheless, the volume of hippocampus and cerebellum are reduced in all strains248. Interestingly, Ts65Dn, Ts1Cje, and Ts2cje mice all exhibited reduced adult hippocampal neurogenesis249-252. Since either treatment with fluoxetine or physical exercise, known to enhance adult neurogenesis, can rescue learning memory deficits of Ts65Dn mice250-252, promoting adult hippocampal neurogenesis might be a potential therapeutic method for DS.
3.5. 15q syndromes
Mice with maternal deletion of UBE3A have been used to study Angelman syndrome (AS)253. These mice show increased oxidative stress in the hippocampus, as measured by MRI254. Impaired adult neurogenesis has also been found in an AS mouse model; this phenotype can be rescued with fluoxetine255,256. Conditional reinstatement of UBE3A showed that restoring UBE3A in various ages resulted in various levels of behavioral rescue257. To rescue anxiety, repetitive behavior and epilepsy phenotypes, UBE3A had to be reinstated early in development but hippocampal synaptic plasticity could be restored at any age, including adult (>8 weeks old)257. However, hippocampal-dependent neurogenesis or learning was not assessed in these mice. It will be interesting to determine whether the plasticity of the hippocampus is due to the presence of adult neurogenesis.
3.6. Fetal alcohol spectrum disorder (FASD)
Animal models for FASD have been created to study the pathogenesis, mechanism, and treatment involving neurodevelopmental abnormalities, neurobehavioral problems, learning difficulties, cognitive problems, intellectual disability, and sleep disorders and associated behavioral impairments258. Many neural and cognitive deficits of FASD are related to disruptions of hippocampal functions. Therefore, extensive research has been focused on the impact of FASD on the hippocampus259,260.
A large number of studies have assessed adult hippocampal neurogenesis in rodent models of FASD261. Fetal alcohol exposure appears to cause a reduction in the volume of the hippocampus, which is thought to result from either an overall reduction in cell proliferation or an increase in cell death, leading to impairments in memory functions262,263. Chronic prenatal alcohol exposure reduces Doublecortin (DCX)-positive cells, suggesting the rate of conversion of proliferative cells to immature neurons in the hippocampus is impaired264. Kajimoto et al utilized inducible nestin-CreERT2 transgenic mice to determine the stage-specific impact of prenatal alcohol exposure on the stepwise maturation of adult hippocampal progenitors265. Using a limited alcohol access “drinking-in-the-dark” model of FASD, the study confirmed previous findings that moderate prenatal alcohol exposure had no effect on adult neurogenesis under standard housing conditions, but it did abolish the neurogenic response to enriched environments (EE)265. The effect of alcohol on immune activation in the developing CNS has just begun to be investigated. Drew et al demonstrated for the first time that alcohol induces neuroinflammation with elevated levels of pro-inflammatory molecules and microglial activation in many brain regions266. Neuroinflammation is well known to inhibit adult hippocampal neurogenesis and hippocampus-dependent cognitive functions16. This study has revealed that the PPAR-c agonist pioglitazone suppresses alcohol-induced neuroinflammation, suggesting that pioglitazone, and perhaps other anti-inflammatory therapeutics, may be effective in the treatment of FASD266. Therefore, hippocampal deficits might be a therapeutic target for FASD.
4. Summary and perspectives
In this review, we hope to convey that hippocampal deficits are prevalent in both human patients with NDs and animal models of NDs. A comparison between human data and animal models allows us to investigate the causes and consequences of changes in hippocampal structures and functions and establish direct causal links between these hippocampal changes with hippocampus-dependent behavioral deficits. Several key questions remain to be addressed. Do hippocampal changes arise from the NDs or precede and predispose toward NDs? What cellular mechanisms underlie the overall increase or decrease in hippocampal volume? In addition, several considerations emerge from this review. How does the study of certain NDs with clear genetic causes, for example, RTT, FXS, and Angelman syndrome, inform our understanding of complex disorders with unclear genetic causes, such as FASD and autism? How can the study of NDs lead to breakthroughs in our understanding of the basic biological mechanisms regulating human development and in the prevention of NDs? Although the extensive use of animal models facilitates our understanding of human NDs, animal models are limited in how much they can reveal about some of the most fundamental aspects of development, genetics, pathology, and disease mechanisms that are unique to humans.
GABAergic inhibition shapes cortical-hippocampal neural activity, which is highly associated with important aspects of neural information processing267. GABA dysfunction has been implicated in many types of NDs, such as ASD186, DS247, RTT228 and FXS268. Disruption of hippocampal GABA function by intra-hippocampal infusion of subconvulsive doses of a GABA-A antagonist has recently been shown to disrupt attentional function and hippocampus-dependent place memory performance in rats269. The development of human pluripotent stem cells (hPSCs) and gene editing methods has greatly advanced our ability to study human diseases; however, whether these cell culture-based models are sufficiently complex to correctly mimic human brain development is unclear. Thus, how to best model NDs remains controversial. Since most NDs likely result from a combination of genetic, biological, psychosocial, and environmental risk factors, how do we evaluate the impact of these risk factors and develop treatments for NDs long past the developmental period? Despite the extensive hippocampal deficits observed in human NDs and the widespread observations of deficits in adult hippocampal neurogenesis in animal ND models, neurogenic changes in human NDs remain unexplored, and hippocampal neurogenesis-focused therapeutic interventions have yet to be developed. Much of our knowledge about hippocampal deficits is associative and superficial. We hope that with rapid technological advances in genomics, brain imaging, circuit-specific and cell type-specific manipulation techniques, and more refined and relevant behavioral analysis methods for both animal models and human patients, we can systematically tackle these questions soon.
Highlights.
The mammalian hippocampus has an important role in learning and cognition.
Hippocampal deficits are found in several human neurodevelopmental disorders,
Animal models for neurodevelopmental disorders exhibit hippocampal deficits.
Targeting the hippocampus helps to understand etiology and development treatment
Acknowledgements
We thank Dr. A. Chakravorty for editing. This work was supported by grants from the NIH (MH080434, MH07897, R21NS095632) John Merck Fund, and Brain Research Foundation to X.Z.; NIH (P30HD03352; U54 HD090256) to Waisman Center IDDRC; the Wisconsin Distinguished Undergraduate Student Award to M.E.S.
Footnotes
Conflict of Interest Statement
The authors declare no competing interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Hertz-Picciotto I, Schmidt RJ & Krakowiak P Understanding environmental contributions to autism: Causal concepts and the state of science. Autism Res 11, 554–586 (2018). [DOI] [PubMed] [Google Scholar]
- 2.Lyall K, et al. The Changing Epidemiology of Autism Spectrum Disorders. Annu Rev Public Health 38, 81–102 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wais PE Hippocampal Signals for Strong Memory When Associative Memory Is Available and When It Is Not. Hippocampus 21, 9–21 (2011). [DOI] [PubMed] [Google Scholar]
- 4.Maren S, Phan KL & Liberzon I The contextual brain: implications for fear conditioning, extinction and psychopathology. Nat Rev Neurosci 14, 417–428 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kesner RP & Rolls ET A computational theory of hippocampal function, and tests of the theory: new developments. Neurosci Biobehav Rev 48, 92–147 (2015). [DOI] [PubMed] [Google Scholar]
- 6.Soltesz I & Losonczy A CA1 pyramidal cell diversity enabling parallel information processing in the hippocampus. Nat Neurosci 21, 484–493 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sahay A, Wilson DA & Hen R Pattern separation: a common function for new neurons in hippocampus and olfactory bulb. Neuron 70, 582–588 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lagali PS, Corcoran CP & Picketts DJ Hippocampus development and function: role of epigenetic factors and implications for cognitive disease. Clin Genet 78, 321–333 (2010). [DOI] [PubMed] [Google Scholar]
- 9.Bast T & Feldon J Hippocampal modulation of sensorimotor processes. Prog Neurobiol 70, 319–345 (2003). [DOI] [PubMed] [Google Scholar]
- 10.Bannerman DM, et al. Regional dissociations within the hippocampus--memory and anxiety. Neurosci Biobehav Rev 28, 273–283 (2004). [DOI] [PubMed] [Google Scholar]
- 11.Okuyama T, Kitamura T, Roy DS, Itohara S & Tonegawa S Ventral CA1 neurons store social memory. Science 353, 1536–1541 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Xu C, et al. Distinct Hippocampal Pathways Mediate Dissociable Roles of Context in Memory Retrieval. Cell 167, 961–972 e916 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bast T The hippocampal learning-behavior translation and the functional significance of hippocampal dysfunction in schizophrenia. Curr Opin Neurobiol 21, 492–501 (2011). [DOI] [PubMed] [Google Scholar]
- 14.Takeuchi T, Duszkiewicz AJ & Morris RG The synaptic plasticity and memory hypothesis: encoding, storage and persistence. Philos Trans R Soc Lond B Biol Sci 369, 20130288 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Neves G, Cooke SF & Bliss TV Synaptic plasticity, memory and the hippocampus: a neural network approach to causality. Nat Rev Neurosci 9, 65–75 (2008). [DOI] [PubMed] [Google Scholar]
- 16.Kempermann G, Song H & Gage FH Neurogenesis in the Adult Hippocampus. Cold Spring Harb Perspect Biol 7, a018812 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Eriksson PS, et al. Neurogenesis in the adult human hippocampus. Nat Med 4, 1313–1317 (1998). [DOI] [PubMed] [Google Scholar]
- 18.Spalding KL, et al. Dynamics of hippocampal neurogenesis in adult humans. Cell 153, 1219–1227 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Boldrini M, et al. Human Hippocampal Neurogenesis Persists throughout Aging. Cell Stem Cell 22, 589–599 e585 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kempermann G, Kuhn HG & Gage FH More hippocampal neurons in adult mice living in an enriched environment. Nature 386, 493–495 (1997). [DOI] [PubMed] [Google Scholar]
- 21.Garthe A, Behr J & Kempermann G Adult-Generated Hippocampal Neurons Allow the Flexible Use of Spatially Precise Learning Strategies. PLoS One 4(2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kee N, Teixeira CM, Wang AH & Frankland PW Preferential incorporation of adult-generated granule cells into spatial memory networks in the dentate gyrus. Nat Neurosci 10, 355–362 (2007). [DOI] [PubMed] [Google Scholar]
- 23.Lacar B, Parylak SL, Vadodaria KC, Sarkar A & Gage FH Increasing the resolution of the adult neurogenesis picture. F1000Prime Rep 6, 8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fanselow MS & Dong HW Are the dorsal and ventral hippocampus functionally distinct structures? Neuron 65, 7–19 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jones KL The effects of alcohol on fetal development. Birth Defects Res C Embryo Today 93, 3–11 (2011). [DOI] [PubMed] [Google Scholar]
- 26.Samoilov M, et al. Acetylation of histones in neocortex and hippocampus of rats exposed to different modes of hypobaric hypoxia: Implications for brain hypoxic injury and tolerance. Acta Histochem 118, 80–89 (2016). [DOI] [PubMed] [Google Scholar]
- 27.Belluscio LM, Alberca CD, Pregi N & Canepa ET Altered gene expression in hippocampus and depressive-like behavior in young adult female mice by early protein malnutrition. Genes Brain Behav 15, 741–749 (2016). [DOI] [PubMed] [Google Scholar]
- 28.Sahin M & Sur M Genes, circuits, and precision therapies for autism and related neurodevelopmental disorders. Science 350(2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Geuze E, Vermetten E & Bremner JD MR-based in vivo hippocampal volumetrics: 2. Findings in neuropsychiatric disorders. Mol Psychiatry 10, 160–184 (2005). [DOI] [PubMed] [Google Scholar]
- 30.Kable JA & Mukherjee RA Neurodevelopmental disorder associated with prenatal exposure to alcohol (ND-PAE): A proposed diagnostic method of capturing the neurocognitive phenotype of FASD. Eur J Med Genet 60, 49–54 (2017). [DOI] [PubMed] [Google Scholar]
- 31.Martin GE, et al. Signaling of noncomprehension in communication breakdowns in fragile X syndrome, Down syndrome, and autism spectrum disorder. J Commun Disord 65, 22–34 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Barnes KV, et al. Anxiety-like behavior in Rett syndrome: characteristics and assessment by anxiety scales. J Neurodev Disord 7, 30 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jacob-Hirsch J, et al. Whole-genome sequencing reveals principles of brain retrotransposition in neurodevelopmental disorders. Cell Res 28, 187–203 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Durukan I, Kara K, Almbaideen M, Karaman D & Gul H Alexithymia, depression and anxiety in parents of children with neurodevelopmental disorder: Comparative study of autistic disorder, pervasive developmental disorder not otherwise specified and attention deficit-hyperactivity disorder. Pediatr Int 60, 247–253 (2017). [DOI] [PubMed] [Google Scholar]
- 35.Hansen BH, Oerbeck B, Skirbekk B, Petrovski BE & Kristensen H Neurodevelopmental disorders: prevalence and comorbidity in children referred to mental health services. Nord J Psychiatry, 1–7 (2018). [DOI] [PubMed] [Google Scholar]
- 36.Eisinger BE & Zhao X Identifying molecular mediators of environmentally enhanced neurogenesis. Cell Tissue Res 371, 7–21 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sun J, Ming GL & Song H Epigenetic regulation of neurogenesis in the adult mammalian brain. Eur J Neurosci 33, 1087–1093 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tu M, et al. Notch1 Signaling Activation Contributes to Adult Hippocampal Neurogenesis Following Traumatic Brain Injury. Med Sci Monit 23, 5480–5487 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ladron de Guevara-Miranda D, et al. Lysophosphatidic acid-induced increase in adult hippocampal neurogenesis facilitates the forgetting of cocaine-contextual memory. Addict Biol (2018). [DOI] [PubMed] [Google Scholar]
- 40.Hollands C, et al. Depletion of adult neurogenesis exacerbates cognitive deficits in Alzheimer's disease by compromising hippocampal inhibition. Mol Neurodegener 12, 64 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dupret D, et al. Spatial relational memory requires hippocampal adult neurogenesis. PLoS One 3, e1959 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hernandez-Rabaza V, et al. Inhibition of adult hippocampal neurogenesis disrupts contextual learning but spares spatial working memory, long-term conditional rule retention and spatial reversal. Neuroscience 159, 59–68 (2009). [DOI] [PubMed] [Google Scholar]
- 43.Lin MS, et al. alpha2-glycine receptors modulate adult hippocampal neurogenesis and spatial memory. Dev Neurobiol 77, 1430–1441 (2017). [DOI] [PubMed] [Google Scholar]
- 44.Jin JJ, et al. Age-dependent differences of treadmill exercise on spatial learning ability between young- and adult-age rats. J Exerc Rehabil 13, 381–386 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Poulose SM, Miller MG, Scott T & Shukitt-Hale B Nutritional Factors Affecting Adult Neurogenesis and Cognitive Function. Adv Nutr 8, 804–811 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Woost L, et al. Physical Exercise and Spatial Training: A Longitudinal Study of Effects on Cognition, Growth Factors, and Hippocampal Plasticity. Sci Rep 8, 4239 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chen L, Gao X, Zhao S, Hu W & Chen J The Small-Molecule TrkB Agonist 7, 8-Dihydroxyflavone Decreases Hippocampal Newborn Neuron Death After Traumatic Brain Injury. J Neuropathol Exp Neurol 74, 557–567 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhao S, Yu A, Wang X, Gao X & Chen J Post-Injury Treatment of 7,8- Dihydroxyflavone Promotes Neurogenesis in the Hippocampus of the Adult Mouse. J Neurotrauma 33, 2055–2064 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sahay A, et al. Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 472, 466–470 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lotan A, et al. Differential effects of chronic stress in young-adult and old female mice: cognitive-behavioral manifestations and neurobiological correlates. Mol Psychiatry 23, 1432–1445 (2017). [DOI] [PubMed] [Google Scholar]
- 51.Lee H, et al. DISC1-mediated dysregulation of adult hippocampal neurogenesis in rats. Front Syst Neurosci 9, 93 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Guo W, et al. Ablation of Fmrp in adult neural stem cells disrupts hippocampus-dependent learning. Nat Med 17, 559–565 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yun S, Reynolds RP, Masiulis I & Eisch AJ Re-evaluating the link between neuropsychiatric disorders and dysregulated adult neurogenesis. Nat Med 22, 1239–1247 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Estes A, et al. Behavioral, cognitive, and adaptive development in infants with autism spectrum disorder in the first 2 years of life. J Neurodev Disord 7, 24 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Autism, Developmental Disabilities Monitoring Network Surveillance Year Principal, I., Centers for Disease, C. & Prevention. Prevalence of autism spectrum disorders--Autism and Developmental Disabilities Monitoring Network, 14 sites, United States, 2008. MMWR Surveill Summ 61, 1–19 (2012). [PubMed] [Google Scholar]
- 56.Hallmayer J, et al. Genetic Heritability and Shared Environmental Factors Among Twin Pairs With Autism. Arch Gen Psychiat 68, 1095–1102 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Jumah F, Ghannam M, Jaber M, Adeeb N & Tubbs RS Neuroanatomical variation in autism spectrum disorder: A comprehensive review. Clin Anat 29, 454–465 (2016). [DOI] [PubMed] [Google Scholar]
- 58.Brambilla P, et al. Brain anatomy and development in autism: review of structural MRI studies. Brain Res Bull 61, 557–569 (2003). [DOI] [PubMed] [Google Scholar]
- 59.Sussman D, et al. The autism puzzle: Diffuse but not pervasive neuroanatomical abnormalities in children with ASD. Neuroimage Clin 8, 170–179 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Braden BB, et al. Executive function and functional and structural brain differences in middle-age adults with autism spectrum disorder. Autism Res 10, 1945–1959 (2017). [DOI] [PubMed] [Google Scholar]
- 61.Chaddad A, Desrosiers C, Hassan L & Tanougast C Hippocampus and amygdala radiomic biomarkers for the study of autism spectrum disorder. BMC Neurosci 18, 52 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chaddad A, Desrosiers C & Toews M Multi-scale radiomic analysis of sub-cortical regions in MRI related to autism, gender and age. Sci Rep 7, 45639 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang T, Bray SM & Warren ST New perspectives on the biology of fragile X syndrome. Curr Opin Genet Dev 22, 256–263 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bagni C, Tassone F, Neri G & Hagerman R Fragile X syndrome: causes, diagnosis, mechanisms, and therapeutics. Journal of Clinical Investigation 122, 4314–4322 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Verkerk AJ, et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 65, 905–914 (1991). [DOI] [PubMed] [Google Scholar]
- 66.Santos AR, Kanellopoulos AK & Bagni C Learning and behavioral deficits associated with the absence of the fragile X mental retardation protein: what a fly and mouse model can teach us. Learn Mem 21, 543–555 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Myrick LK, et al. Independent role for presynaptic FMRP revealed by an FMR1 missense mutation associated with intellectual disability and seizures. Proc Natl Acad Sci U S A 112, 949–956 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Smidak R, et al. Reduced Levels of the Synaptic Functional Regulator FMRP in Dentate Gyrus of the Aging Sprague-Dawley Rat. Front Aging Neurosci 9, 384 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bechara EG, et al. A novel function for fragile X mental retardation protein in translational activation. PLoS Biol 7, e16 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bassell GJ & Warren ST Fragile X syndrome: loss of local mRNA regulation alters synaptic development and function. Neuron 60, 201–214 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Pfeiffer BE & Huber KM The state of synapses in fragile X syndrome. Neuroscientist 15, 549–567 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Saldarriaga W, et al. Fragile X syndrome. Colomb Med (Cali) 45, 190–198 (2014). [PMC free article] [PubMed] [Google Scholar]
- 73.Bostrom C, et al. Hippocampal dysfunction and cognitive impairment in Fragile-X Syndrome. Neurosci Biobehav Rev 68, 563–574 (2016). [DOI] [PubMed] [Google Scholar]
- 74.Greco CM, et al. Neuropathologic features in the hippocampus and cerebellum of three older men with fragile X syndrome. Mol Autism 2, 2 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Jakala P, et al. Fragile-X: neuropsychological test performance, CGG triplet repeat lengths, and hippocampal volumes. J Clin Invest 100, 331–338 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Greicius MD, Boyett-Anderson JM, Menon V & Reiss AL Reduced basal forebrain and hippocampal activation during memory encoding in girls with fragile X syndrome. Neuroreport 15, 1579–1583 (2004). [DOI] [PubMed] [Google Scholar]
- 77.de von Flindt R, Bybel B, Chudley AE & Lopes F Short-term memory and cognitive variability in adult fragile X females. Am J Med Genet 38, 488–492 (1991). [DOI] [PubMed] [Google Scholar]
- 78.Yam KY, Naninck EF, Schmidt MV, Lucassen PJ & Korosi A Early-life adversity programs emotional functions and the neuroendocrine stress system: the contribution of nutrition, metabolic hormones and epigenetic mechanisms. Stress 18, 328–342 (2015). [DOI] [PubMed] [Google Scholar]
- 79.Yeshurun S & Hannan AJ Transgenerational epigenetic influences of paternal environmental exposures on brain function and predisposition to psychiatric disorders. Mol Psychiatry (2018). [DOI] [PubMed] [Google Scholar]
- 80.Wilson ME & Sengoku T Developmental regulation of neuronal genes by DNA methylation: environmental influences. Int J Dev Neurosci 31, 448–451 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Chen Y, Damayanti NP, Irudayaraj J, Dunn K & Zhou FC Diversity of two forms of DNA methylation in the brain. Front Genet 5, 46 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Feng J, et al. Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat Neurosci 13, 423–430 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gallegos DA, Chan U, Chen LF & West AE Chromatin Regulation of Neuronal Maturation and Plasticity. Trends Neurosci 41, 311–324 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ho KL, et al. MeCP2 binding to DNA depends upon hydration at methyl-CpG. Mol Cell 29, 525–531 (2008). [DOI] [PubMed] [Google Scholar]
- 85.Wakefield RI, et al. The solution structure of the domain from MeCP2 that binds to methylated DNA. J Mol Biol 291, 1055–1065 (1999). [DOI] [PubMed] [Google Scholar]
- 86.Amir RE, et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet 23, 185–188 (1999). [DOI] [PubMed] [Google Scholar]
- 87.Shahbazian MD & Zoghbi HY Molecular genetics of Rett syndrome and clinical spectrum of MECP2 mutations. Curr Opin Neurol 14, 171–176 (2001). [DOI] [PubMed] [Google Scholar]
- 88.Leonard H, Cobb S & Downs J Clinical and biological progress over 50 years in Rett syndrome. Nat Rev Neurol 13, 37–51 (2017). [DOI] [PubMed] [Google Scholar]
- 89.Hagberg B, Aicardi J, Dias K & Ramos O 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 (1983). [DOI] [PubMed] [Google Scholar]
- 90.Pohodich AE & Zoghbi HY Rett syndrome: disruption of epigenetic control of postnatal neurological functions. Hum Mol Genet 24, R10–16 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Katz DM, Dutschmann M, Ramirez JM & Hilaire G Breathing disorders in Rett syndrome: progressive neurochemical dysfunction in the respiratory network after birth. Respir Physiol Neurobiol 168, 101–108 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Reiss AL, et al. Neuroanatomy of Rett syndrome: a volumetric imaging study. Ann Neurol 34, 227–234 (1993). [DOI] [PubMed] [Google Scholar]
- 93.Carter JC, et al. Selective cerebral volume reduction in Rett syndrome: a multiple-approach MR imaging study. AJNR Am J Neuroradiol 29, 436–441 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Naidu S, et al. Neuroimaging studies in Rett syndrome. Brain Dev 23 Suppl 1, S62–71 (2001). [DOI] [PubMed] [Google Scholar]
- 95.Chen YC, et al. Modeling Rett Syndrome Using TALEN-Edited MECP2 Mutant Cynomolgus Monkeys. Cell 169, 945–955 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Belichenko PV, Oldfors A, Hagberg B & Dahlstrom A Rett syndrome: 3-D confocal microscopy of cortical pyramidal dendrites and afferents. Neuroreport 5, 1509–1513 (1994). [PubMed] [Google Scholar]
- 97.Armstrong D, Dunn JK, Antalffy B & Trivedi R Selective dendritic alterations in the cortex of Rett syndrome. J Neuropathol Exp Neurol 54, 195–201 (1995). [DOI] [PubMed] [Google Scholar]
- 98.Chapleau CA, et al. Dendritic spine pathologies in hippocampal pyramidal neurons from Rett syndrome brain and after expression of Rett-associated MECP2 mutations. Neurobiol Dis 35, 219–233 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Asim A, Kumar A, Muthuswamy S, Jain S & Agarwal S "Down syndrome: an insight of the disease". J Biomed Sci 22, 41 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hattori M, et al. The DNA sequence of human chromosome 21. Nature 405, 311–319 (2000). [DOI] [PubMed] [Google Scholar]
- 101.Antonarakis SE Down syndrome and the complexity of genome dosage imbalance. Nat Rev Genet 18, 147–163 (2017). [DOI] [PubMed] [Google Scholar]
- 102.Choong XY, Tosh JL, Pulford LJ & Fisher EM Dissecting Alzheimer disease in Down syndrome using mouse models. Front Behav Neurosci 9, 268 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Smigielska-Kuzia J, et al. A volumetric magnetic resonance imaging study of brain structures in children with Down syndrome. Neurol Neurochir Pol 45, 363–369 (2011). [DOI] [PubMed] [Google Scholar]
- 104.Pearlson GD, et al. MRI brain changes in subjects with Down syndrome with and without dementia. Dev Med Child Neurol 40, 326–334 (1998). [PubMed] [Google Scholar]
- 105.Prasher VP, Barber PC, West R & Glenholmes P The role of magnetic resonance imaging in the diagnosis of Alzheimer disease in adults with Down syndrome. Arch Neurol 53, 1310–1313 (1996). [DOI] [PubMed] [Google Scholar]
- 106.Finucane BM, et al. 15q Duplication Syndrome and Related Disorders in GeneReviews((R)) (eds. Adam MP, et al.) (Seattle (WA), 1993). [Google Scholar]
- 107.Kalsner L & Chamberlain SJ Prader-Willi, Angelman, and 15q11-q13 Duplication Syndromes. Pediatr Clin North Am 62, 587–606 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Buiting K, Williams C & Horsthemke B Angelman syndrome - insights into a rare neurogenetic disorder. Nat Rev Neurol 12, 584–593 (2016). [DOI] [PubMed] [Google Scholar]
- 109.Williams CA Neurological aspects of the Angelman syndrome. Brain Dev 27, 88–94 (2005). [DOI] [PubMed] [Google Scholar]
- 110.Clayton-Smith J & Laan L Angelman syndrome: a review of the clinical and genetic aspects. J Med Genet 40, 87–95 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tan WH, et al. Angelman syndrome: Mutations influence features in early childhood. Am J Med Genet A 155A, 81–90 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Aghakhanyan G, et al. From Cortical and Subcortical Grey Matter Abnormalities to Neurobehavioral Phenotype of Angelman Syndrome: A Voxel-Based Morphometry Study. PLoS One 11, e0162817 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Butler MG Prader-Willi syndrome: current understanding of cause and diagnosis. Am J Med Genet 35, 319–332 (1990). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cataletto M, Angulo M, Hertz G & Whitman B Prader-Willi syndrome: A primer for clinicians. Int J Pediatr Endocrinol 2011, 12 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Honea RA, et al. The neuroanatomy of genetic subtype differences in Prader-Willi syndrome. Am J Med Genet B Neuropsychiatr Genet 159B, 243–253 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang Y, et al. Altered functional brain networks in Prader-Willi syndrome. NMR Biomed 26, 622–629 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Herrera BM & Lindgren CM The genetics of obesity. Curr Diab Rep 10, 498–505 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Guthoff M, et al. Insulin modulates food-related activity in the central nervous system. J Clin Endocrinol Metab 95, 748–755 (2010). [DOI] [PubMed] [Google Scholar]
- 119.Hsu TM, et al. A hippocampus to prefrontal cortex neural pathway inhibits food motivation through glucagon-like peptide-1 signaling. Mol Psychiatry 23, 1555–1565 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Land BB, et al. Medial prefrontal D1 dopamine neurons control food intake. Nat Neurosci 17, 248–253 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Holsen LM, et al. Neural mechanisms underlying hyperphagia in Prader-Willi syndrome. Obesity (Silver Spring) 14, 1028–1037 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Holsen LM, et al. Importance of reward and prefrontal circuitry in hunger and satiety: Prader-Willi syndrome vs simple obesity. Int J Obes (Lond) 36, 638–647 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Battaglia A The inv dup (15) or idic (15) syndrome (Tetrasomy 15q). Orphanet J Rare Dis 3, 30 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Boronat S, Mehan WA, Shaaya EA, Thibert RL & Caruso P Hippocampal abnormalities in magnetic resonance imaging (MRI) of 15q duplication syndromes. J Child Neurol 30, 333–338 (2015). [DOI] [PubMed] [Google Scholar]
- 125.Jones KL & Smith DW Recognition of the fetal alcohol syndrome in early infancy. Lancet 302, 999–1001 (1973). [DOI] [PubMed] [Google Scholar]
- 126.Roozen S, et al. Worldwide Prevalence of Fetal Alcohol Spectrum Disorders: A Systematic Literature Review Including Meta-Analysis. Alcohol Clin Exp Res 40, 18–32 (2016). [DOI] [PubMed] [Google Scholar]
- 127.Popova S, Lange S, Bekmuradov D, Mihic A & Rehm J Fetal alcohol spectrum disorder prevalence estimates in correctional systems: a systematic literature review. Can J Public Health 102, 336–340 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Mattson SN, Crocker N & Nguyen TT Fetal alcohol spectrum disorders: neuropsychological and behavioral features. Neuropsychol Rev 21, 81–101 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Meschke LL, Holl JA & Messelt S Assessing the risk of fetal alcohol syndrome: understanding substance use among pregnant women. Neurotoxicol Teratol 25, 667–674 (2003). [DOI] [PubMed] [Google Scholar]
- 130.Glass L, Ware AL & Mattson SN Neurobehavioral, neurologic, and neuroimaging characteristics of fetal alcohol spectrum disorders. Handb Clin Neurol 125, 435–462 (2014). [DOI] [PubMed] [Google Scholar]
- 131.Chokroborty-Hoque A, Alberry B & Singh SM Exploring the complexity of intellectual disability in fetal alcohol spectrum disorders. Front Pediatr 2, 90 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lupton C, Burd L & Harwood R Cost of fetal alcohol spectrum disorders. Am J Med Genet C Semin Med Genet 127C, 42–50 (2004). [DOI] [PubMed] [Google Scholar]
- 133.Stade B, et al. The burden of prenatal exposure to alcohol: revised measurement of cost. Can J Clin Pharmacol 16, e91–102 (2009). [PubMed] [Google Scholar]
- 134.Norman AL, Crocker N, Mattson SN & Riley EP Neuroimaging and fetal alcohol spectrum disorders. Dev Disabil Res Rev 15, 209–217 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lewis CE, et al. Verbal learning and memory impairment in children with fetal alcohol spectrum disorders. Alcohol Clin Exp Res 39, 724–732 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhou D, et al. Preserved cortical asymmetry despite thinner cortex in children and adolescents with prenatal alcohol exposure and associated conditions. Hum Brain Mapp 39, 72–88 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ha S, Sohn IJ, Kim N, Sim HJ & Cheon KA Characteristics of Brains in Autism Spectrum Disorder: Structure, Function and Connectivity across the Lifespan. Exp Neurobiol 24, 273–284 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ellegood J & Crawley JN Behavioral and Neuroanatomical Phenotypes in Mouse Models of Autism. Neurotherapeutics 12, 521–533 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nicolini C & Fahnestock M The valproic acid-induced rodent model of autism. Exp Neurol 299, 217–227 (2018). [DOI] [PubMed] [Google Scholar]
- 140.Roullet FI, Lai JK & Foster JA In utero exposure to valproic acid and autism--a current review of clinical and animal studies. Neurotoxicol Teratol 36, 47–56 (2013). [DOI] [PubMed] [Google Scholar]
- 141.Naisbitt S, et al. Shank, a novel family of postsynaptic density proteins that binds to the NMDA receptor/PSD-95/GKAP complex and cortactin. Neuron 23, 569–582 (1999). [DOI] [PubMed] [Google Scholar]
- 142.Monteiro P & Feng G SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat Rev Neurosci 18, 147–157 (2017). [DOI] [PubMed] [Google Scholar]
- 143.Hung AY, et al. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci 28, 1697–1708 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Mao W, et al. Shank1 regulates excitatory synaptic transmission in mouse hippocampal parvalbumin-expressing inhibitory interneurons. Eur J Neurosci 41, 1025–1035 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sungur AO, Schwarting RKW & Wohr M Behavioral phenotypes and neurobiological mechanisms in the Shank1 mouse model for autism spectrum disorder: A translational perspective. Behav Brain Res (2017). [DOI] [PubMed] [Google Scholar]
- 146.Sungur AO, et al. Aberrant cognitive phenotypes and altered hippocampal BDNF expression related to epigenetic modifications in mice lacking the post-synaptic scaffolding protein SHANK1: Implications for autism spectrum disorder. Hippocampus 27, 906–919 (2017). [DOI] [PubMed] [Google Scholar]
- 147.Won H, et al. Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature 486, 261–265 (2012). [DOI] [PubMed] [Google Scholar]
- 148.Schmeisser MJ, et al. Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/Shank2. Nature 486, 256–260 (2012). [DOI] [PubMed] [Google Scholar]
- 149.Harony-Nicolas H, et al. Oxytocin improves behavioral and electrophysiological deficits in a novel Shank3-deficient rat. Elife 6(2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Parr LA, et al. Effects of chronic oxytocin on attention to dynamic facial expressions in infant macaques. Psychoneuroendocrinology 74, 149–157 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Guastella AJ & MacLeod C A critical review of the influence of oxytocin nasal spray on social cognition in humans: evidence and future directions. Horm Behav 61, 410–418 (2012). [DOI] [PubMed] [Google Scholar]
- 152.Chang SWC, Barter JW, Ebitz RB, Watson KK & Platt ML Inhaled oxytocin amplifies both vicarious reinforcement and self reinforcement in rhesus macaques (Macaca mulatta). Proc Natl Acad Sci U S A 109, 959–964 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Guastella AJ & Hickie IB Oxytocin Treatment, Circuitry, and Autism: A Critical Review of the Literature Placing Oxytocin Into the Autism Context. Biol Psychiatry 79, 234–242 (2016). [DOI] [PubMed] [Google Scholar]
- 154.Wang X, et al. Altered mGluR5-Homer scaffolds and corticostriatal connectivity in a Shank3 complete knockout model of autism. Nat Commun 7, 11459 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Vicidomini C, et al. Pharmacological enhancement of mGlu5 receptors rescues behavioral deficits in SHANK3 knock-out mice. Mol Psychiatry 22, 689–702 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Verpelli C, et al. Importance of Shank3 protein in regulating metabotropic glutamate receptor 5 (mGluR5) expression and signaling at synapses. J Biol Chem 286, 34839–34850 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Cope EC, et al. Immature Neurons and Radial Glia, But Not Astrocytes or Microglia, Are Altered in Adult Cntnap2 and Shank3 Mice, Models of Autism. eNeuro 3(2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Abdulkareem IH & Blair M Phosphatase and tensin homologue deleted on chromosome 10. Niger Med J 54, 79–86 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zhou J & Parada LF PTEN signaling in autism spectrum disorders. Curr Opin Neurobiol 22, 873–879 (2012). [DOI] [PubMed] [Google Scholar]
- 160.Suzuki A, et al. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr Biol 8, 1169–1178 (1998). [DOI] [PubMed] [Google Scholar]
- 161.Wang SI, Parsons R & Ittmann M Homozygous deletion of the PTEN tumor suppressor gene in a subset of prostate adenocarcinomas. Clin Cancer Res 4, 811–815 (1998). [PubMed] [Google Scholar]
- 162.Chalhoub N & Baker SJ PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol 4, 127–150 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Garcia-Junco-Clemente P & Golshani P PTEN: A master regulator of neuronal structure, function, and plasticity. Commun Integr Biol 7, e28358 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Takeuchi K, et al. Dysregulation of synaptic plasticity precedes appearance of morphological defects in a Pten conditional knockout mouse model of autism. Proc Natl Acad Sci U S A 110, 4738–4743 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Amiri A, et al. Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnormalities and alters neurogenesis. J Neurosci 32, 5880–5890 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Williams MR, DeSpenza T Jr., Li M, Gulledge AT & Luikart BW Hyperactivity of newborn Pten knock-out neurons results from increased excitatory synaptic drive. J Neurosci 35, 943–959 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Wei L, Meaney MJ, Duman RS & Kaffman A Affiliative behavior requires juvenile, but not adult neurogenesis. J Neurosci 31, 14335–14345 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Bourgeron T A synaptic trek to autism. Curr Opin Neurobiol 19, 231–234 (2009). [DOI] [PubMed] [Google Scholar]
- 169.Rothwell PE, et al. Autism-associated neuroligin-3 mutations commonly impair striatal circuits to boost repetitive behaviors. Cell 158, 198–212 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Jamain S, et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat Genet 34, 27–29 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Tabuchi K & Sudhof TC Structure and evolution of neurexin genes: Insight into the mechanism of alternative splicing. Genomics 79, 849–859 (2002). [DOI] [PubMed] [Google Scholar]
- 172.Baig DN, Yanagawa T & Tabuchi K Distortion of the normal function of synaptic cell adhesion molecules by genetic variants as a risk for autism spectrum disorders. Brain Res Bull 129, 82–90 (2017). [DOI] [PubMed] [Google Scholar]
- 173.Wang H Endocannabinoid Mediates Excitatory Synaptic Function of beta-Neurexins. Commentary: beta-Neurexins Control Neural Circuits by Regulating Synaptic Endocannabinoid Signaling. Front Neurosci 10, 203 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Anderson GR, et al. beta-Neurexins Control Neural Circuits by Regulating Synaptic Endocannabinoid Signaling. Cell 162, 593–606 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Aoto J, Foldy C, Ilcus SM, Tabuchi K & Sudhof TC Distinct circuit-dependent functions of presynaptic neurexin-3 at GABAergic and glutamatergic synapses. Nat Neurosci 18, 997–1007 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Varoqueaux F, et al. Neuroligins determine synapse maturation and function. Neuron 51, 741–754 (2006). [DOI] [PubMed] [Google Scholar]
- 177.Tabuchi K, et al. A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 318, 71–76 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Etherton M, et al. Autism-linked neuroligin-3 R451C mutation differentially alters hippocampal and cortical synaptic function. Proc Natl Acad Sci U S A 108, 13764–13769 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Foldy C, Malenka RC & Sudhof TC Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron 78, 498–509 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Etherton MR, Tabuchi K, Sharma M, Ko J & Sudhof TC An autism-associated point mutation in the neuroligin cytoplasmic tail selectively impairs AMPA receptor-mediated synaptic transmission in hippocampus. Embo J 30, 2908–2919 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Meyza KZ, et al. The BTBR T+ tf/J mouse model for autism spectrum disorders-in search of biomarkers. Behav Brain Res 251, 25–34 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Cheng N, Alshammari F, Hughes E, Khanbabaei M & Rho JM Dendritic overgrowth and elevated ERK signaling during neonatal development in a mouse model of autism. PLoS One 12, e0179409 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Ellegood J, Babineau BA, Henkelman RM, Lerch JP & Crawley JN Neuroanatomical analysis of the BTBR mouse model of autism using magnetic resonance imaging and diffusion tensor imaging. Neuroimage 70, 288–300 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Dodero L, et al. Neuroimaging evidence of major morpho-anatomical and functional abnormalities in the BTBR T+TF/J mouse model of autism. PLoS One 8, e76655 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Sforazzini F, et al. Altered functional connectivity networks in acallosal and socially impaired BTBR mice. Brain Struct Funct 221, 941–954 (2016). [DOI] [PubMed] [Google Scholar]
- 186.Han S, Tai C, Jones CJ, Scheuer T & Catterall WA Enhancement of inhibitory neurotransmission by GABAA receptors having alpha2,3-subunits ameliorates behavioral deficits in a mouse model of autism. Neuron 81, 1282–1289 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Monyak RE, et al. Insulin signaling misregulation underlies circadian and cognitive deficits in a Drosophila fragile X model. Mol Psychiatry 22, 1140–1148 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Wu YJ, et al. Fragile X Mental Retardation-1 Knockout Zebrafish Shows Precocious Development in Social Behavior. Zebrafish 14, 438–443 (2017). [DOI] [PubMed] [Google Scholar]
- 189.Li Y, et al. MDM2 inhibition rescues neurogenic and cognitive deficits in a mouse model of fragile X syndrome. Sci Transl Med 8, 336ra361 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Till SM, et al. Conserved hippocampal cellular pathophysiology but distinct behavioural deficits in a new rat model of FXS. Hum Mol Genet 24, 5977–5984 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Consortium TD-BFX Fmr1 knockout mice: a model to study fragile X mental retardation. The Dutch-Belgian Fragile X Consortium. Cell 78, 23–33 (1994). [PubMed] [Google Scholar]
- 192.Kazdoba TM, Leach PT, Silverman JL & Crawley JN Modeling fragile X syndrome in the Fmr1 knockout mouse. Intractable Rare Dis Res 3, 118–133 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Huber KM, Gallagher SM, Warren ST & Bear MF Altered synaptic plasticity in a mouse model of fragile X mental retardation. Proc Natl Acad Sci U S A 99, 7746–7750 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Aschrafi A, Cunningham BA, Edelman GM & Vanderklish PW The fragile X mental retardation protein and group I metabotropic glutamate receptors regulate levels of mRNA granules in brain. Proc Natl Acad Sci U S A 102, 2180–2185 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Antar LN, Afroz R, Dictenberg JB, Carroll RC & Bassell GJ Metabotropic glutamate receptor activation regulates fragile x mental retardation protein and FMR1 mRNA localization differentially in dendrites and at synapses. J Neurosci 24, 2648–2655 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Bear MF, Huber KM & Warren ST The mGluR theory of fragile X mental retardation. Trends Neurosci 27, 370–377 (2004). [DOI] [PubMed] [Google Scholar]
- 197.Michalon A, et al. Chronic pharmacological mGlu5 inhibition corrects fragile X in adult mice. Neuron 74, 49–56 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Gasparini F, et al. 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology 38, 1493–1503 (1999). [DOI] [PubMed] [Google Scholar]
- 199.Lindemann L, et al. CTEP: a novel, potent, long-acting, and orally bioavailable metabotropic glutamate receptor 5 inhibitor. J Pharmacol Exp Ther 339, 474–486 (2011). [DOI] [PubMed] [Google Scholar]
- 200.Berry-Kravis EM, et al. Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov 17, 280–299 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Lindemann L, et al. Pharmacology of basimglurant (RO4917523, RG7090), a unique metabotropic glutamate receptor 5 negative allosteric modulator in clinical development for depression. J Pharmacol Exp Ther 353, 213–233 (2015). [DOI] [PubMed] [Google Scholar]
- 202.Vranesic I, et al. AFQ056/mavoglurant, a novel clinically effective mGluR5 antagonist: identification, SAR and pharmacological characterization. Bioorg Med Chem 22, 5790–5803 (2014). [DOI] [PubMed] [Google Scholar]
- 203.Rudelli RD, et al. Adult fragile X syndrome. Clinico-neuropathologic findings. Acta Neuropathol 67, 289–295 (1985). [DOI] [PubMed] [Google Scholar]
- 204.Hinton VJ, Brown WT, Wisniewski K & Rudelli RD Analysis of neocortex in three males with the fragile X syndrome. Am J Med Genet 41, 289–294 (1991). [DOI] [PubMed] [Google Scholar]
- 205.Irwin SA, et al. 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 (2001). [DOI] [PubMed] [Google Scholar]
- 206.Meredith RM, de Jong R & Mansvelder HD Functional rescue of excitatory synaptic transmission in the developing hippocampus in Fmr1-KO mouse. Neurobiol Dis 41, 104–110 (2011). [DOI] [PubMed] [Google Scholar]
- 207.Eadie BD, et al. Fmr1 knockout mice show reduced anxiety and alterations in neurogenesis that are specific to the ventral dentate gyrus. Neurobiol Dis 36, 361–373 (2009). [DOI] [PubMed] [Google Scholar]
- 208.Luo Y, et al. Fragile x mental retardation protein regulates proliferation and differentiation of adult neural stem/progenitor cells. PLoS Genet 6, e1000898 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Scotto-Lomassese S, et al. Fragile X mental retardation protein regulates new neuron differentiation in the adult olfactory bulb. J Neurosci 31, 2205–2215 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Guo W, et al. Inhibition of GSK3beta improves hippocampus-dependent learning and rescues neurogenesis in a mouse model of fragile X syndrome. Hum Mol Genet 21, 681–691 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Li Y, et al. Reducing histone acetylation rescues cognitive deficits in a mouse model of Fragile X syndrome. Nat Commun 9, 2494 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Tian Y, et al. Loss of FMRP Impaired Hippocampal Long-Term Plasticity and Spatial Learning in Rats. Front Mol Neurosci 10, 269 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Clouaire T & Stancheva I Methyl-CpG binding proteins: specialized transcriptional repressors or structural components of chromatin? Cell Mol Life Sci 65, 1509–1522 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Lombardi LM, Baker SA & Zoghbi HY MECP2 disorders: from the clinic to mice and back. J Clin Invest 125, 2914–2923 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Chen RZ, Akbarian S, Tudor M & Jaenisch R Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat Genet 27, 327–331 (2001). [DOI] [PubMed] [Google Scholar]
- 216.Guy J, Hendrich B, Holmes M, Martin JE & Bird A A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat Genet 27, 322–326 (2001). [DOI] [PubMed] [Google Scholar]
- 217.Goffin D, et al. Rett syndrome mutation MeCP2 T158A disrupts DNA binding, protein stability and ERP responses. Nat Neurosci 15, 274–283 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Shahbazian M, et al. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3. Neuron 35, 243–254 (2002). [DOI] [PubMed] [Google Scholar]
- 219.Adachi M, Autry AE, Covington HE & Monteggia LM MeCP2-Mediated Transcription Repression in the Basolateral Amygdala May Underlie Heightened Anxiety in a Mouse Model of Rett Syndrome. Journal of Neuroscience 29, 4218–4227 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Fyffe SL, et al. Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response to stress. Neuron 59, 947–958 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Lioy DT, et al. A role for glia in the progression of Rett's syndrome. Nature 475, 497–500 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Samaco RC, et al. Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities. Proc Natl Acad Sci U S A 106, 21966–21971 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Ward CS, et al. MeCP2 Is Critical within HoxB1-Derived Tissues of Mice for Normal Lifespan. Journal of Neuroscience 31, 10359–10370 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Ito H, Morishita R & Nagata KI Autism spectrum disorder-associated genes and the development of dentate granule cells. Med Mol Morphol 50, 123–129 (2017). [DOI] [PubMed] [Google Scholar]
- 225.Xu X, Miller EC & Pozzo-Miller L Dendritic spine dysgenesis in Rett syndrome. Front Neuroanat 8, 97 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Monteggia LM & Kavalali ET Rett syndrome and the impact of MeCP2 associated transcriptional mechanisms on neurotransmission. Biol Psychiatry 65, 204–210 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Shepherd GM & Katz DM Synaptic microcircuit dysfunction in genetic models of neurodevelopmental disorders: focus on Mecp2 and Met. Curr Opin Neurobiol 21, 827–833 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Chao HT, et al. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 468, 263–269 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Samaco RC, et al. Female Mecp2(+/−) mice display robust behavioral deficits on two different genetic backgrounds providing a framework for pre-clinical studies. Hum Mol Genet 22, 96–109 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Tsujimura K, Abematsu M, Kohyama J, Namihira M & Nakashima K Neuronal differentiation of neural precursor cells is promoted by the methyl-CpG-binding protein MeCP2. Exp Neurol 219, 104–111 (2009). [DOI] [PubMed] [Google Scholar]
- 231.Szulwach KE, et al. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol 189, 127–141 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Smrt RD, et al. Mecp2 deficiency leads to delayed maturation and altered gene expression in hippocampal neurons. Neurobiol Dis 27, 77–89 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Gao Y, et al. Inhibition of miR-15a Promotes BDNF Expression and Rescues Dendritic Maturation Deficits in MeCP2-Deficient Neurons. Stem Cells 33, 1618–1629 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Chen WG, et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 302, 885–889 (2003). [DOI] [PubMed] [Google Scholar]
- 235.Zhou Z, et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 52, 255–269 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Li H, Zhong X, Chau KF, Williams EC & Chang Q Loss of activity-induced phosphorylation of MeCP2 enhances synaptogenesis, LTP and spatial memory. Nat Neurosci 14, 1001–1008 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Cohen S, et al. Genome-wide activity-dependent MeCP2 phosphorylation regulates nervous system development and function. Neuron 72, 72–85 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Zhong XF, Li HD & Chang Q MeCP2 Phosphorylation Is Required for Modulating Synaptic Scaling through mGluR5. Journal of Neuroscience 32, 12841–12847 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Li H, et al. Cell cycle-linked MeCP2 phosphorylation modulates adult neurogenesis involving the Notch signalling pathway. Nat Commun 5, 5601 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Hao S, et al. Forniceal deep brain stimulation rescues hippocampal memory in Rett syndrome mice. Nature 526, 430–434 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Davisson MT, Schmidt C & Akeson EC Segmental trisomy of murine chromosome 16: a new model system for studying Down syndrome. Prog Clin Biol Res 360, 263–280 (1990). [PubMed] [Google Scholar]
- 242.Sago H, et al. Ts1Cje, a partial trisomy 16 mouse model for Down syndrome, exhibits learning and behavioral abnormalities. Proc Natl Acad Sci U S A 95, 6256–6261 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Villar AJ, et al. Identification and characterization of a new Down syndrome model, Ts[Rb(12.1716)]2Cje, resulting from a spontaneous Robertsonian fusion between T(171)65Dn and mouse chromosome 12. Mamm Genome 16, 79–90 (2005). [DOI] [PubMed] [Google Scholar]
- 244.Holtzman DM, et al. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc Natl Acad Sci U S A 93, 13333–13338 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Chen Y, et al. In vivo MRI identifies cholinergic circuitry deficits in a Down syndrome model. Neurobiol Aging 30, 1453–1465 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Nie X, et al. Evidence of altered age-related brain cytoarchitecture in mouse models of down syndrome: a diffusional kurtosis imaging study. Magn Reson Imaging 33, 437–447 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Kleschevnikov AM, et al. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J Neurosci 24, 8153–8160 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Roubertoux PL, et al. Differential Brain, Cognitive and Motor Profiles Associated with Partial Trisomy. Modeling Down Syndrome in Mice. Behav Genet 47, 305–322 (2017). [DOI] [PubMed] [Google Scholar]
- 249.Ishihara K, et al. Enlarged brain ventricles and impaired neurogenesis in the Ts1Cje and Ts2Cje mouse models of Down syndrome. Cereb Cortex 20, 1131–1143 (2010). [DOI] [PubMed] [Google Scholar]
- 250.Llorens-Martin MV, et al. Effects of voluntary physical exercise on adult hippocampal neurogenesis and behavior of Ts65Dn mice, a model of Down syndrome. Neuroscience 171, 1228–1240 (2010). [DOI] [PubMed] [Google Scholar]
- 251.Bianchi P, et al. Early pharmacotherapy restores neurogenesis and cognitive performance in the Ts65Dn mouse model for Down syndrome. J Neurosci 30, 8769–8779 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Clark S, Schwalbe J, Stasko MR, Yarowsky PJ & Costa AC Fluoxetine rescues deficient neurogenesis in hippocampus of the Ts65Dn mouse model for Down syndrome. Exp Neurol 200, 256–261 (2006). [DOI] [PubMed] [Google Scholar]
- 253.Born HA, et al. Strain-dependence of the Angelman Syndrome phenotypes in Ube3a maternal deficiency mice. Sci Rep 7, 8451 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Berkowitz BA, et al. In vivo imaging of prodromal hippocampus CA1 subfield oxidative stress in models of Alzheimer disease and Angelman syndrome. Faseb J 31, 4179–4186 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Godavarthi SK, Dey P, Sharma A & Jana NR Impaired adult hippocampal neurogenesis and its partial reversal by chronic treatment of fluoxetine in a mouse model of Angelman syndrome. Biochem Biophys Res Commun 464, 1196–1201 (2015). [DOI] [PubMed] [Google Scholar]
- 256.Mardirossian S, Rampon C, Salvert D, Fort P & Sarda N Impaired hippocampal plasticity and altered neurogenesis in adult Ube3a maternal deficient mouse model for Angelman syndrome. Exp Neurol 220, 341–348 (2009). [DOI] [PubMed] [Google Scholar]
- 257.Silva-Santos S, et al. Ube3a reinstatement identifies distinct developmental windows in a murine Angelman syndrome model. J Clin Invest 125, 2069–2076 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Sulik KK Fetal alcohol spectrum disorder: pathogenesis and mechanisms. Handb Clin Neurol 125, 463–475 (2014). [DOI] [PubMed] [Google Scholar]
- 259.Fontaine CJ, Patten AR, Sickmann HM, Helfer JL & Christie BR Effects of prenatal alcohol exposure on hippocampal synaptic plasticity: Sex, age and methodological considerations. Neurosci Biobehav Rev 64, 12–34 (2016). [DOI] [PubMed] [Google Scholar]
- 260.Alfonso-Loeches S & Guerri C Molecular and behavioral aspects of the actions of alcohol on the adult and developing brain. Crit Rev Clin Lab Sci 48, 19–47 (2011). [DOI] [PubMed] [Google Scholar]
- 261.Boschen KE & Klintsova AY Neurotrophins in the Brain: Interaction With Alcohol Exposure During Development. Vitam Horm 104, 197–242 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Klintsova AY, et al. Persistent impairment of hippocampal neurogenesis in young adult rats following early postnatal alcohol exposure. Alcohol Clin Exp Res 31, 2073–2082 (2007). [DOI] [PubMed] [Google Scholar]
- 263.Gil-Mohapel J, Boehme F, Kainer L & Christie BR Hippocampal cell loss and neurogenesis after fetal alcohol exposure: insights from different rodent models. Brain Res Rev 64, 283–303 (2010). [DOI] [PubMed] [Google Scholar]
- 264.Olateju OI, Spocter MA, Patzke N, Ihunwo AO & Manger PR Hippocampal neurogenesis in the C57BL/6J mice at early adulthood following prenatal alcohol exposure. Metab Brain Dis 33, 397–410 (2018). [DOI] [PubMed] [Google Scholar]
- 265.Kajimoto K, Allan A & Cunningham LA Fate analysis of adult hippocampal progenitors in a murine model of fetal alcohol spectrum disorder (FASD). PLoS One 8, e73788 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Drew PD, Johnson JW, Douglas JC, Phelan KD & Kane CJ Pioglitazone blocks ethanol induction of microglial activation and immune responses in the hippocampus, cerebellum, and cerebral cortex in a mouse model of fetal alcohol spectrum disorders. Alcohol Clin Exp Res 39, 445–454 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Bast T, Pezze M & McGarrity S Cognitive deficits caused by prefrontal cortical and hippocampal neural disinhibition. Br J Pharmacol 174, 3211–3225 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Kang JY, et al. Deficits in the activity of presynaptic gamma-aminobutyric acid type B receptors contribute to altered neuronal excitability in fragile X syndrome. J Biol Chem 292, 6621–6632 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.McGarrity S, Mason R, Fone KC, Pezze M & Bast T Hippocampal Neural Disinhibition Causes Attentional and Memory Deficits. Cereb Cortex 27, 4447–4462 (2017). [DOI] [PubMed] [Google Scholar]
- Berman RF, and Hannigan JH (2000). Effects of prenatal alcohol exposure on the hippocampus: spatial behavior, electrophysiology, and neuroanatomy. Hippocampus 10, 94–110. [DOI] [PubMed] [Google Scholar]
- Guo W, Allan AM, Zong R, Zhang L, Johnson EB, Schaller EG, Murthy AC, Goggin SL, Eisch AJ, Oostra BA, et al. (2011). Ablation of Fmrp in adult neural stem cells disrupts hippocampus-dependent learning. Nature medicine 17, 559–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo W, Murthy AC, Zhang L, Johnson EB, Schaller EG, Allan AM, and Zhao X (2012). Inhibition of GSK3beta improves hippocampus-dependent learning and rescues neurogenesis in a mouse model of fragile X syndrome. Human molecular genetics 21, 681–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harony-Nicolas H, Kay M, Hoffmann JD, Klein ME, Bozdagi-Gunal O, Riad M, Daskalakis NP, Sonar S, Castillo PE, Hof PR, et al. (2017). Oxytocin improves behavioral and electrophysiological deficits in a novel Shank3-deficient rat. eLife 6 2017 January 31;6. pii: e18904. doi: 10.7554/eLife.18904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houle K, Abdi M, and Clabough EBD (2017). Acute ethanol exposure during late mouse neurodevelopment results in long-term deficits in memory retrieval, but not in social responsiveness. Brain and behavior 7, e00636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones CA, Watson DJ, and Fone KC (2011). Animal models of schizophrenia. British journal of pharmacology 164, 1162–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kazdoba TM, Leach PT, Silverman JL, and Crawley JN (2014). Modeling fragile X syndrome in the Fmr1 knockout mouse. Intractable & rare diseases research 3, 118–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon CH, Luikart BW, Powell CM, Zhou J, Matheny SA, Zhang W, Li Y, Baker SJ, and Parada LF (2006). Pten regulates neuronal arborization and social interaction in mice. Neuron 50, 377–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Stockton ME, Bhuiyan I, Eisinger BE, Gao Y, Miller JL, Bhattacharyya A, and Zhao X (2016). MDM2 inhibition rescues neurogenic and cognitive deficits in a mouse model of fragile X syndrome. Science translational medicine 8, 336ra361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stearns NA, Schaevitz LR, Bowling H, Nag N, Berger UV, and Berger-Sweeney J (2007). Behavioral and anatomical abnormalities in Mecp2 mutant mice: a model for Rett syndrome. Neuroscience 146, 907–921. [DOI] [PubMed] [Google Scholar]
- Sungur AO, Jochner MCE, Harb H, Kilic A, Garn H, Schwarting RKW, and Wohr M (2017). Aberrant cognitive phenotypes and altered hippocampal BDNF expression related to epigenetic modifications in mice lacking the post-synaptic scaffolding protein SHANK1: Implications for autism spectrum disorder. Hippocampus 27, 906–919. [DOI] [PubMed] [Google Scholar]
- Tabuchi K, Blundell J, Etherton MR, Hammer RE, Liu X, Powell CM, and Sudhof TC (2007). A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 318, 71–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian Y, Yang C, Shang S, Cai Y, Deng X, Zhang J, Shao F, Zhu D, Liu Y, Chen G, et al. (2017). Loss of FMRP Impaired Hippocampal Long-Term Plasticity and Spatial Learning in Rats. Frontiers in molecular neuroscience 10, 269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Won H, Lee HR, Gee HY, Mah W, Kim JI, Lee J, Ha S, Chung C, Jung ES, Cho YS, et al. (2012). Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature 486, 261–265. [DOI] [PubMed] [Google Scholar]
- Zhao MG, Toyoda H, Ko SW, Ding HK, Wu LJ, and Zhuo M (2005). Deficits in trace fear memory and long-term potentiation in a mouse model for fragile X syndrome. The Journal of neuroscience : the official journal of the Society for Neuroscience 25, 7385–7392. [DOI] [PMC free article] [PubMed] [Google Scholar]