Epigenetic modulation during hippocampal development (Review)

  • Authors:
    • Si‑Jing Fan
    • An‑Bang Sun
    • Lian Liu
  • View Affiliations

  • Published online on: October 18, 2018     https://doi.org/10.3892/br.2018.1160
  • Pages: 463-473
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The hippocampus is located in the limbic system and is vital in learning ability, memory formation and emotion regulation, and is associated with depression, epilepsy and mental retardation in an abnormal developmental situation. Several factors have been found to modulate the development of the hippocampus, and epigenetic modification have a crucial effect in this progress. The present review summarizes the epigenetic modifications, including DNA methylation, histone acetylation, and non‑coding RNAs, regulating all stages of hippocampal development, focusing on the growth of Ammon's horn and the dentate gyrus in humans and rodents. These modifications may significantly affect hippocampal development and health in addition to cognitive processes.

Introduction

The hippocampus is a part of the limbic system that is vital in learning ability, memory formation, and emotion regulation (13). It is also associated with the emergence of several neuropsychiatric disorders, including epilepsy, mental retardation and Alzheimers disease (AD) (4). During development of the rodent hippocampus, aberrant neuronal migration results in anomalous hippocampal lamination, neuronal differentiation disorders and neural circuit defects, leading to severe epilepsy syndrome following birth (5). Exposure to ethanol during fetal development can cause apoptosis of hippocampal neurons, leading to postnatal mental retardation (6). It has been demonstrated that the abnormal proliferation of neuronal cells and disordered differentiation of neurons appear in the dentate gyrus (DG) of offspring when pregnant rats are exposed to depleted uranium for a prolonged period; the neonatal rats exhibited depressive symptoms at 2 months postnatally (7). In advanced eukaryotes, epigenetic modifications exist in the normal developmental progress of eukaryotic cells, including DNA methylation, histone acetylation and non-coding RNAs. These major epigenetic modifications for the regulation of mammalian gene expression are crucial in ontogenesis and phenotypic transmission. Epigenetic regulation is defined as a genetic variation in gene expression in which the DNA sequence remains unaltered (8). Several epigenetic modifications affect the growth of the hippocampus therefore, the present review summarizes the various stages of human and rodent hippocampal development and discusses their effects from the perspective of DNA methylation, histone acetylation and non-coding RNAs.

Development of the hippocampus

Humans

The mammalian nervous system originates from its embryonic ectoderm, which differentiates into the neural tube and neural crest. The neural tube expands gradually and forms into a brain vesicle, following which the hippocampus appears in the prosencephalon vesicle. A previous study showed that the cortical hem, a region of the dorsomedial telencephalon, mediates hippocampal development by producing a bone morphogenetic protein and wingless-related signal (9). In the process of human neurogenesis, the neural tube forms in embryonic week (EW)3 and prosencephalon vesicles occur in EW5. The local parietal lobe then thickens to form the hippocampal primordium in EW6 (in EW14 the hippocampus disappears from the parietal lobe), and another hippocampal primordium derives from the temporal lobe in EW7 via a series of spins and folds to eventually form a complete hippocampus, of which the developmental hallmarks are shown in Table I (10). The hippocampal primordium is composed of neuroepithelium, and in EW8, the neuroepithelium is initiated to migrate and generate the sub granular zone (SGZ), the middle layer and the marginal layer, respectively (10). Between EW10 and EW11, the earliest granular cell layer (GCL) of the DG emerges from the middle hippocampal layer of the temporal lobe, which marks the beginning of DG development (2,10). The DG is a specific region of the hippocampus that possesses the capacity of neurogenesis in adulthood (1). A previous study showed that the ability of the DG to sustain neurogenesis is associated with the neurogenic niche, which is maintained in the SGZ (11). Another study traced the key markers of hippocampal neurogenesis to confirm that the neurogenic niche was formed in the hippocampus prior to the mammal reaching adulthood (12).

Table I.

Comparison of the hippocampal development process in humans and rodents.

Table I.

Comparison of the hippocampal development process in humans and rodents.

Author, dateHumansRatsMiceHippocampal development milestoneRefs.
Deng et al, 1996; Bayer, 1980; Berg et al, 2018EW6ED14ED10-11Differentiate into hippocampal primordia(10,22,30)
Xu et al, 2015; Deng et al, 1996; Radic et al, 2017; Berg et al, 2018EW10-11ED16ED12-14Appearance of DGCs(2,10,23,30)
Deng et al, 1996; Altman and Bayer, 1990; Bayer, 1980; Altman and Bayer, 1990; Nakahira and Yuasa, 2005EW15ED17-19ED14Earliest pyramidal cells emerging in Ammon's horn(10,24,26,27,31)
Xu et al, 2009; Deng et al, 1996; Rice and Barone, 2000; Radic et al, 2017EW15PD7PD1Peak DGC growth spurt(2,10,15,23)
Li et al, 2009; Humphrey, 1967; Berg et al, 2018EW30PD30PD14Generation of SGZ(1,14,30)

[i] EW, embryonic week; ED, embryonic day; PD, postnatal day.

In EW13 through EW14, the Ammons horn and the DG starts to fold, and the DG is narrowed into a U-shape; however, the CA1, CA2 and CA3 fields of Ammons horn are linearly arranged (13). The primal Ammons horn appears on EW14, and the boundary between the Ammons horn and the DG is divided by the hippocampal fissure. In EW15, the undifferentiated cells of the middle hippocampal layer begin to differentiate into hippocampal pyramidal cells, which gradually form the pyramidal layer of the CA1, CA2 and CA3 regions (10). The settling and generation of pyramidal cells occurs in opposing order between the CA1 and CA3 regions. The early-generated pyramidal cells are destined to settle in the late-forming stratum pyramidal of the CA3 region, whereas the late-generated pyramidal cells settle in the early-forming CA1 region (1). Between EW16 and EW18, the Ammons horn and the DG are stacked in the temporal lobe, the CA1-3 regions are curved, and the hilus of the DG is occupied by the CA4 region (13). In EW30, the hippocampal fissure disappears; subsequently, the Ammons horn and the DG already have a typical three-tier structure, which is the fundamental structure of the hippocampus (14).

Approximately 80% of dentate granule cells (DGCs) appear in the prenatal period of humans and reach peak proliferation in EW15 (10,15). Following birth, the proliferation of neural progenitor cells (NPCs) in the SGZ gradually evolve into DGCs, which is known for neurogenesis in the DG; these newly generated DGCs become increasingly mature by a complicated process and eventually settle in the GCL (3,16). The new neurons from neurogenesis establish a synaptic connection with the other neurons and are incorporated into the trisynaptic circuit [entorhinal cortex (EC)-DG-CA3-CA1-EC], which is formed in the prenatal hippocampus; it gives rise to corresponding neurological functions and is associated with the learning and memory functions of humans (1). The role of SGZ neurogenesis in emotional control has been investigated previously; it was found that stressors inhibit the proliferation of NPCs in SGZ and antidepressants exert an opposite effect (17). The volumes of the hippocampus and temporal lobe increase markedly in 2-year-old children, following which the rate of increase plateaus and the proportion of the hippocampus in the temporal lobe declines with age (18). The right-left asymmetry of the hippocampus and temporal lobe has been suggested in previous studies; as a statute, the right side is larger than the left side (19,20).

Rats

The neural tube of the rats takes shape between ED10.5 and ED11 (15,21). The hippocampal primordium of the rats is composed of neuroepithelium, which appears in the dorsomedial telencephalon by ED14 (22). On ED16, the incrassate hippocampal primordium is categorized into three forms: Primary dentate neuroepithelium, fimbria glioepithelium and ammonic neuroepithelium. These forms evolve into the DGCs, the glial cells of the hippocampal fimbria and the pyramidal neurons of Ammons horn, (1,11,22). The formation of DGCs in rats is divided into two phases: The first phase begins on ED16 and produces ~15% DGCs prior to birth; the second phase begins at birth when ~85% of the DGCs take shape, and peak proliferation occurs on postnatal day (PD)7 (2,23). The multiple migration transforms the primary dentate neuroepithelium cells into secondary dentate matrix or tertiary dentate matrix and SGZ, which further differentiates into DGCs sequentially (24). On ED18, the migration of primary dentate neuroepithelial cells form the secondary dentate matrix; these cells transplant into the DG fields following proliferation (25). There are two stages in the process of migration of the secondary dentate matrix cells to the DG fields. The first dentate migration develops into the shell of the GCL on ED20 (22), and the secondary dentate matrix disappears on PD5 (25); after 5 days, the first stage is complete (1). The second dentate migration is diffused in the polymorph layer of the DG, between PD3 and PD10 (25). Consequently, the tertiary dentate matrix begins to form, which evolves into the SGZ on PD20 through PD30 (1). Subsequently, the second stage is completed, however, the SGZ persists and generates DGCs during adolescence and adulthood. Furthermore, radial glial cells (RGCs) are crucial in the formation of DGs. The earliest RGCs appear in the neuroepithelium, and the radial glial fibers of RGCs originate from the ventricular zone (VZ) throughout the DG fields. In addition to the migration of RGCs from the VZ to the SGZ, the primary dentate neuroepithelium cells migrate along the fiber scaffold of RGCs and differentiate into DGCs. Whether antenatal or postpartum, RGCs are divided into primary and secondary RGCs, which affect the two production phases of DGCs as described above (2).

Between ED15 and ED17, the primary Ammons horn appears in the hippocampus of rats, and the hippocampal fissure divides the boundary between Ammons horn and DG (22). On ED17 through ED19, a large number of pyramidal cells are present in the middle layer of Ammons horn; the later-generated pyramidal cells are settled earlier in the CA1 region; however, the earlier-generated pyramidal cells are deposited into the CA3 region following formation of the GCL (24,26,27). This phenomenon is described as the opposite of the generation time and the settling time of the pyramidal cells in the Ammons horn. The development of the pyramidal stratum of CA1 is complete on ED20 (24,26). The formation of the shell of the GCL is the immediate cause for the pyramidal layer of CA1-3 to begin to turn inwards (1); between days E18 and E22, the CA3 extends into the hilus of the DG to transform into the CA4 field (the hilus) (22). Consecutively, the pyramidal cells in the middle layer of Ammons horn settle in the C-type pyramidal stratum, which disappears gradually with the thickening of the pyramidal stratum (24). Generally, the structure of the pyramidal layer in Ammons horn is almost complete during the embryonic period. Following birth, the arrangement of cells in the pyramidal layer gradually increases from the inside to the outside (28); the arrangement of cells increases from the outside to the inside in the GCL (29), and the pyramidal layer of Ammons horn thins due to a reduction in the number of cells and increased volumex(22). By contrast, the morphological development of the DG is mainly postnatal; the number of DGCs increase and exhibit a large volume (22).

Mice

The neural tube takes shape in mice between ED9 and ED9.5 (21). Subsequently, the hippocampal primordium emerges between ED10 and ED11 (30). The process involved in the morphological development of the DG is similar in rats and mice; however, there are differences in the time. The production of DGCs in the DG of mice can be divided into two phases: ~85% of the DGCs are generated in the second phase postnatally, and peak proliferation is observed on PD1 (23). The DGCs in rats are mature 4–6 days earlier than in mice, although the peak proliferation appears later (23). The secondary dentate matrix is formed on ED15, and the cells migrate to the DG field following proliferation (30). The rudimentary GCL shell is generated on ED18 by the first dentate migration (31). In the secondary dentate migration, the tertiary dentate matrix is also generated on ED18, and turns into the SGZ on PD14 (30). Furthermore, severe morphological defects of the DG and loss of the hippocampal fissure have been found in Nfib-deficient mice, demonstrating the vital function of RGCs in the formation of the DG (32). Between days P10 and P14, the secondary RGCs differentiate into astrocytes following formation of the radial glial fiber scaffold, which accompany GCL migration to the molecular layer of the DG (2).

On ED14, the primary Ammons horn appears in the hippocampus of mice (31). Subsequently, a large number of pyramidal cells is observed in the middle layer of Ammons horn (31,33), and the settlement process of the pyramidal cells in the CA1 and CA3 regions is consistent with that of rats (34). The pyramidal layer of Ammons horn is bent into type C on ED15, followed by the appearance of the CA4 field (35); on ED18, the middle layer disappears with complete settlement of the pyramidal cells in Ammons horn (33). Between PD10 and PD15, the abundance of pyramidal cells in Ammons horn increases rapidly; the structure of Ammons horn stabilizes in postnatal month (PM)3 (36).

The extreme of entorhinal fibers reaches the molecular layer of the DG in mice by ED17 and the axon of Cajal-Retzius cells in the DG act as scaffolds for neuronal projection in the EC, which is the starting signal of the trisynaptic circuit (1). At ~PW1, the neonatal DGCs extend the dendrites into the granular and molecular layers of the DG and project axons into the hilus of the DG (CA4 field), which receive the input of excitatory GABA-ergic neurons (16). The DGCs begin to receive Glu-ergic neurotransmitter by the performant pathway on PW3. With the completion of synapse integration at PW4, the transmission of GABA signals changes from excitability to inhibitory; until PM2, the structure and function of the newborn DGCs matures (1,16). Previous studies have shown that in PW3 in rodents, the regenerative capacity of the brain, particularly the DG of the hippocampus, is minimal (21). When the DG is damaged, the damaged neurons are not replaced, resulting in long-term memory defects (21).

Epigenetic modifications in hippocampal development

DNA methylation

DNA methylation refers to the conversion of cytosine to 5-methylcytosine (5mC), catalyzed by DNA methyltransferases (DNMTs) and acquisition of a methyl group from S-adenosylmethionine (37,38). Among the three types of DNMTs, DNMT1 and DNMT3 function as the maintenance methylase and nascent methylase, respectively; however, the role of DNMT2 remains to be fully elucidated (3840). The non-5-C-phosphate-G-3 (CpG) dinucleotide is localized and clustered close to the transcription regulation domain, which is the major site of DNA methylation in vertebrates; the methylation of this site causes inactivation of the corresponding gene (37,41). DNA demethylation is effectuated by two methods: Active demethylation is the translation of 5-hydroxymethylcytosine (5hmC) into 5mC under the action of 5mC hydroxylase (Ten-eleven translocation 1/2/3; TET1/2/3), and the passive demethylation is caused by the lack of DNMTs during cell replication (37). 5hmC is a relatively stable epigenetic marker persistent during the life cycle of neurons (37). The 5hmC preferentially binds to methyl-CpG-binding domain-3 (MBD3), which antagonizes the combination of methyl CpG binding protein 2 (MeCP2) with 5hmC. Consequently, the weak bond between DNMT1 and 5hmC decreases the effect of DNMT1 and promotes DNA demethylation; however, 5mC binds to MeCP2 and MBD1 (combination with MeCP2 precedes MBD1) and maintains the methylation status of DNA (37,42).

The mature sperm and eggs of humans and rodents are highly methylated, followed by genome-wide demethylation in the preimplantation embryo and the re-emergence of DNA methylation by embryo implantation (42,43). Studies have confirmed that the concentrations of 5mC and 5hmC increase during the proliferation and differentiation in the mouse neuroepithelial cells (44). The point at which 5mC surges coincide with the time at which neuroepithelial cells proliferate prior to differentiation; however, the 5hmC surge is in accordance with the differentiation of NPCs (44). Setoguchi et al showed that the differentiation of neuroepithelial cells in mice was closely associated with the methylation status of the glial fibrillary acidic protein (GFAP) gene (45). Until day E14.5, the neuroepithelial cells of mice start to differentiate into astrocytes, which previously evolved only into neurons (45). The CpG in the signal transducer and activator of transcription (STAT) protein binding site of the GFAP gene promoter is hypermethylated at an early developmental stage, which results in GFAP gene silencing. However, with demethylation during the developmental process, STAT protein binds to the promoter and leads to activation of the GFAP gene; gradually, the neuroepithelial cells evolve into astrocytes (45). Another study showed that neural stem cells (NSCs) in DNMT3a-deficient mice exhibit a low level of hypomethylation and a proliferation rate significantly higher than that of normal mice. In addition, the NSCs of DNMT3a-deficient mice differentiate into astrocytes in early development with increase in quantity (46). DNMT1 promotes the development of secondary RGCs by regulating the Reelin signaling pathway. Noguchi et al utilized the TAM-inducible Cre recombinase system to knock out the DNMT1 gene in mice NSCs at the onset of DG development; this resulted in the inability of NSCs to establish appropriate secondary radial gliosis scaffolds (47). Therefore, a subset of NPCs and granulosa cells migrate into the SGZ unsuccessfully, followed by accumulation in the molecular layer of the DG. Although NPCs and granulosa cells began to differentiate, they fail to enter the GCL and ultimately die. DNMT1-deficiency also promotes the differentiation of NSCs into astrocytes (47). The above-mentioned factors lead to a decrease in the volume of postnatal GCL.

DNA methylation of the hippocampal Ammons horn (CA1-4) in mice shows a patterned progression. Between P15 and P17, the 5mC appears first and the cells multiply in the pyramidal and neuroepithelial layers of the hippocampus; in the subsequent hours of the first day, the progression of 5hmC is synchronized with cell migration, followed which 5mC and 5hmC continue to increase throughout the migration process (48). Until the pyramidal cells are colonized in the CA regions, the 5mC in the pyramidal cells are reduced, and the 5mC in the nucleus translates into the 5hmC, which activates the corresponding gene, allowing the pyramidal cells to differentiate further and mature. Here, the 5mC in the pyramidal cells of the pyramidal layer turn from the inside out into 5hmC, completing the inside-out maturity (48). By P17, 5mC and 5hmC continually persist in mature pyramidal cells that are aggregated in different regions of the nucleus (48). DNMT1 and DNMT3 maintain DNA methylation, regulate the division of neural progenitors and mature neurons, and are crucial in the synaptic plasticity of mature neurons. Feng et al showed that abnormalities in synaptic plasticity occur in the CA1 field of the DNMT1 and DNMT3 double-knockout mice hippocampus; thus, learning and memory disorders are also exhibited (49). In addition, the genome-wide analysis of NSCs in postnatal mice showed that DNMT3a catalyzes the methylation of a large number of proximal non-promoter sequences (50). The methylation of these DNMT3a-dependent proximal non-promoters facilitates the transcriptional expression of neurogenic genes by functional antagonism of polycomb inhibition in order to maintain the active chromatin status of the core developmental genes (50). The mRNA expression levels of DNMT (DNMT1, DNMT3a and DNMT3b) peak on PW1 and decrease with age in various brain structures of rats, however, global DNA methylation continues to increase in the hippocampus (51).

DNA methylation is involved in the development of hippocampal DG. During migration to the DG, the neuroepithelial cells are first presented at 5mC, followed by 5hmC, ac companied by the decrease of 5mC which converts to 5hmC (48). As Sex determining region Y-box 2 (Sox2) and Ki67 markers are lost in the cells and eventually differentiate into mature granule cells, the earlier granule cells that reached the DG region aggregate to form the GCL shell (48). Consecutively, with development of the DG matrix, a class of NPCs derived from the secondary DG and expressing Sox2 and Ki67 accumulate in the innermost layer of the DG to form the SGZ (48). The changes in NPC migration and differentiation and maturation of DNA methylation in the SGZ are similar to those described above, and neurogenesis in the adult SGZ also follows this pattern. The prenatal deletion of DNMT1 leads to the increased expression of cyclin-dependent protein kinase (CDK) inhibitors, p21 and p57, resulting in decreased proliferation of NSCs in the postnatal SGZ and impaired neurogenesis in the adult SGZ (47). The investigation of neurogenesis in the postnatal hippocampus of TET1-deficient mice indicates that TET1 deficiency results in a decrease in the number of NPCs in the SGZ (~45% decrease). In addition, the self-replication ability of NPCs decreases; however, the differentiation ability of NPCs is unaffected, as the deficiency of TET1 has its function replaced by TET2 and TET3, leading to the marginal increase of 5hmC in the NPC (52). Further investigations have demonstrated that the function of TET1 primarily maintains the hypomethylated state of specific genes to activate them transcriptionally (52). Larimore et al focused on the effect of MeCP2 on the terminal differentiation of rat hippocampal neurons, it was found that the overexpression of MECP2 increased axon length, and the number of dendrites and axons, whereas the knockdown of MECP2 only shortened dendrite length. This suggests that MeCP2 is an important factor in the terminal differentiation of hippocampal neurons (53).

It is suggested that the destruction of DNA methylation processes during human embryogenesis, triggering synaptic plasticity damage in hippocampus, leads to postnatal Autism and Rett syndrome (54). In addition, the reduction of neuropils in the hippocampus of patients with schizophrenia leads to structural changes in the hippocampus and the interference of synaptic plasticity and regenerative mechanisms, which have been shown to arise from the abnormal epigenetic regulation (55). Falkai et al reported that more severe synaptic plasticity damage was accompanied by more marked negative symptoms of schizophrenia (55). The downregulation of GABA-ergic gene expression caused by hypermethylation of the GABA-ergic promoter in the hippocampus has been confirmed as a characteristic pathological phenotype of schizophrenia (56). This is different from the static state of DNA methylation in the hippocampus in human health, autism and AD; there is extensive gene promoter hypermethylation in the hippocampus of patients with temporal lobe epilepsy (57). This may be a mechanism for persistent hyperexcitability in chronic epilepsy. Similarly, Blanco-Luquin et al showed that the downregulation of expression caused by hypermethylation of the phospholipase D family, member 3 gene is closely associated with late-onset AD (58). Studies have suggested that childhood neglect or abuse can cause methylation and histone modifications in the childs hippocampal glucocorticoid receptor promoter region, which reduces the childs hippocampal glucocorticoid receptor expression and increases the adulthood stress response leading to depression and anxiety (59,60). There is also a significant correlation between hypermethylation of the zinc finger and BTB domain containing 20 gene in the hippocampal CA1 region with major depressive disorder (61). A study showed that the overexpression of Dnmt3a2 in the hippocampus consolidated memory and improved anxiety and phobia (62).

Histone acetylation

The basic unit of chromatin is nucleosome consisting of DNA and histone (37). Histone modifications include methylation, phosphorylation, acetylation and ubiquitination. These modified histones recruit other proteins to bind to DNA in a synergistic or antagonistic manner at the transcriptional level. This mechanism regulates the transcription dynamically, and is termed as the histone code hypothesis (63). The most well-known histone modification is histone acetylation, wherein the acetyl group from acetyl-CoA is transferred to the N-terminus of the lysine residue in histone. Histone acetylation can reduce the electrostatic interaction between histones and DNA, altering the conformation of histones, which leads to the relaxation of chromatin structure and binds the transcription factors to corresponding sites; thus, the expression of these modified genes is activated (37,64). By contrast, when histones are deacetylated, the chromatin structure condenses and the transcription factors cannot combine with the corresponding sites, leading to expression of the genes being inhibited (37,64). Histone deacetylases (HDACs) are modulators of histone acetylation. Currently, HDAC-containing complexes are known to be exhibit two types of HDAC structures (HDAC1 and HDAC2); usually, histone deacetylation requires the involvement of two HDAC domains (65).

HDAC1 and HDAC2 have different expression characteristic functions in the development of the hippocampus. HDAC1 is expressed at a high level in proliferating NPCs but disappears following the differentiation of NPCs; however, it appears in glial cells persistently (66). By contrast, HDAC2 is expressed during the proliferation of NPCs, increases following the differentiation of NPCs into neurons and continues to emerge in mature neurons; however, it does not exist in the majority of glial cells (66). In the neurodevelopment of HDAC1 and HDAC2 double-knockout mice, the early nerve cells migrate normally and the proliferation of NPCs increases, although the NPCs do not differentiate into neuronal cells. On ED15.5, a large number of NPCs demonstrate apoptosis, and Montgomery et al found that the hippocampal structure was damaged, the cortex was disorganized and cerebellar foliation was lacking postnatally (67). The defect of HDAC1 or HDAC2 in mice does not cause variations in the overall anatomy of the brain (68). Although HDAC1-deficient mice can survive and multiply, HDAC2-deficient mice die within hours of birth, suggesting that HDAC2 is an essential histone deacetylase for survival and brain development (68).

The transcription factor Ski serves as a scaffolding protein to link HDAC, Sin3, cell type-specific transcription factors (69). The early hippocampal neuroepithelial cells of Ski-deficient mice differentiate into neurons, such that the number of proliferating neuroepithelial cells is reduced in early hippocampal development. The possible mechanism is that Ski acts as an inhibitor of the transforming growth factor-β pathway, which recruits Sin3/HDAC complexes to mothers against decapentaplegic (Smad)s, followed by inhibition of the entry of Smads into the nucleus and regulation of the transcription of target genes, which in turn, regulates the balance of proliferation and differentiation of neuroepithelial cells (69). Furthermore, Ski binds to MeCP2 and is involved in MeCP2-mediated transcriptional repression (70). Bromodomain- and PHD finger-containing protein 1 (BRPF1) is an activator of three types histone acetyltransferases, lysine acetyltransferase (KAT)6A, KATB and KAT7. The hippocampal neuroepithelial cells in BRPF1-deficient mice were reported to be significantly reduced and the distribution of RGCs was sustained in an abnormal state from embryonic to the postnatal stage, which significantly affected the migration of primary dentate neuroepithelium cells (71). Consecutively, the cell cycle of the migrating cells was abnormal, which led to an abnormal state of proliferation during migration and affected the formation of the SGZ as the number of NPCs in the SGZ decreased; the hippocampal anatomy in the adult mice suffered maldevelopment, particularly in the DG (71).

Histone H3 lysine 9 acetylation (H3K9Ac) is an epigenetic marker that exists in transcriptionally active chromatin. Qiao et al showed that H3K9Ac declined gradually within 4 days prior to the neural differentiation of human ESCs and increased with the differentiation of nerves on days 4–8 (72). In phase one, the addition of histone deacetylase inhibitors (HDACIs) resulted in enhanced pluripotency and decreased neural differentiation of ESCs. By contrast, the use of HDACIs on days 4–8 promoted neural differentiation, which indicates that H3K9Ac exerts different effects in various stages of ESC development (72). Another study revealed that the expression levels of H3K9Ac and H3K14Ac in the hippocampal CA3 region of hypoxic rats were decreased, causing the neurodegeneration of neurons and impaired neurotransmitter transmission (73). HDACIs can increase the survival of neonatal neurons in the mouse hippocampus at a specific postnatal time and enhance the neurogenesis in adult hippocampus (74). HDAC2 is extensively distributed in the pyramidal cells and the DGCs of hippocampal CA1 and CA3 regions of postnatal rats. Furthermore, with the development and maturation of nerve cells, the expression of HDAC2 is decreased gradually and transferred from the nucleus to the cytoplasm; until PW4, HDAC2 is maintained at a relatively low level (75). The level of histone acetylation in the hippocampus of mice overexpressing HDAC2 is decreased (acetylation levels of H4K12 and H4K5, but not H3K14), which was most pronounced in the pyramidal cells of Ammons horn. Subsequently, the dendritic spine density of pyramidal cells and DGCs in the CA1 field is significantly reduced (76). However, the density of dendritic spines in these regions of HDAC2-knockdown mice is significantly increased, suggesting that HDAC2 regulates the formation of hippocampal synapses and affects the functions of learning and memory (76).

In the early stage of AD, Gräff et al (77) found that HDAC2 was significantly elevated and accumulated gradually in the hippocampus CA1, and the degree of HDAC2 elevation was associated with the process of cognitive decline in the human brain. It concluded that the current treatment of AD has limitations and should consider treatment options to increase HDAC2. Subsequent rat experiments have shown that the use of HDACIs was effective against a range of neurodegenerative diseases (78). HDACIs can also treat abnormal behavioral symptoms in schizophrenic rats and reverse the structure of ventral hippocampal lesions (79). In addition, the increase of histone acetylation in the hippocampus can counter emotional stress and reduce the emotional behavior of depression (80). However, exposure to HDACIs of the embryonic stage hippocampus may cause autism-like symptoms following birth (81).

Non-coding RNAs

Non-coding RNAs mainly include two types: Long non-coding RNA (lncRNAs) and microRNAs (miRNAs) that effectuate in epigenetic mechanisms. The length of lncRNAs is >200 nucleotides. They do not encode proteins but affect the chromatin structure, chromatin transcription activity and stability, translation, post-transcriptional processing of mRNAs, and gene expression (82,83). miRNAs are a class of non-coding small RNAs that typically consist of ~20 nucleotides. miRNAs bind to mRNAs in a complementary pairing manner, resulting in the degradation of mRNAs or inhibition of their translation, thereby being involved in regulating gene expression (8486).

The Cre-loxP system was previously used to knock out the expression of Dicer and inhibit the formation of miRNAs in the development of the hippocampus of three Cre mouse lines. Li et al showed that the deficiency of miRNA leads to a significant decrease in the number of proliferating NPCs in the hippocampus, which increases apoptosis and promotes early differentiation into hippocampal neurons; however, their development and maturation is impaired (87). The deletion of miRNAs leads to a decrease in the number of NPCs in all hippocampal formations, and the apoptosis of NPCs is most pronounced in CA1 and the DG. The lack of miRNAs in the late stages give rise to prominent early differentiation of neurons in CA1 and the DG on ED10.5, CA3 is mainly affected by ED13.5, and the proliferation and apoptosis of CA3 and DG cells increase; subsequently, the learning and memory functions of mice are enhanced (87,88). In summary, miRNA deletions primarily affect the development of CA1 and DG at an early stage, and CA3 at a later period.

In the hippocampus of mice, lncRNAs are abundant, and as the expression of lncRNAs in the different regions of hippocampus has tissue-cell specificity, it is hypothesized that lncRNAs are involved in the differentiation of hippocampal NSCs (89). A study on the differentiation of induced pluripotent stem cells (IPS cells) into neurons showed that the lncRNA-HOTAIRM1 in the homeobox (HOX) genome was not expressed in IPS cells; however, the level of its transcripts increased 54.6-fold in early differentiated neurons. In addition, the expression of other lncRNAs in the HOX genome was also augmented significantly, indicating that lncRNAs are involved in regulating the differentiation of NSCs into neurons (90). Nigro et al demonstrated abnormal proliferation, morphological disorder in the RGCs of the telencephalon, and abnormal migration of neuroepithelial cells in the hippocampus in Dicer-deficient mice. Between days E16.5 and E18.5, the number of proliferating cells in the DG decreased significantly; however, by day P15, the DG structure of the deficient mice was severely disordered, and cell proliferation and neurogenesis were distinctly reduced in the SGZ (91). The re-expression of miRNA (miR)-30e and miR-181d in the telencephalon of deficient mice, led to the return of RGCs to the normal state. The underlying mechanism may be that miR-30e and miR-181d binds to the 3 UTR of the HtrA serine peptidase 1 gene, inhibiting its expression (91). Another study confirmed that the expression of miR-124 was upregulated by embryonic period methyl donor deficiency (vitamin B9 and vitamin B12) which downregulated Stat3 signaling, leading to atrophy of the hippocampal CA1 pyramidal layer and the DGL of rats (92). Monteleone et al (93) showed that prenatal stress can eliminate the alterations in the methylation pattern at three specific CpGs, which are rooted in two CpG islands of the glycoprotein M6A (gpm6a) gene in the hippocampus, significantly upregulating the expression of miR-133b in the hippocampus, with the subsequent specific interaction of miR-133b with gpm6a affecting mRNA expression and protein levels and function and leading to abnormal hippocampal synapse formation. miR-338-3p exerts a regulatory effect during the proliferation and development of DG neurons. With the maturation of DG neurons, the expression of miR-338-3p increases gradually and eventually reaches a peak in the NeuN+ mature neurons (94). The knockdown of miR-338-3p results in decreased dendritic branching, abnormal distribution, and increased length of dendritic spines in the hippocampal neurons, leading to the abnormal proliferation of hippocampal cells and tumorigenesis (94). By contrast, the overexpression of miR-338-3p leads to decreased hippocampal glioblastoma activity and increased apoptosis (94). miR-9-5p/3p has been shown to effectuate neuronal development and maturation (9597). The long-term potentiation (LTP) of the hippocampus was impaired in miR-9-3p-knockdown mice, following which the memory and learning function were affected postnatally (97). The intrinsic mechanism may be that the knockdown of miR-9-3p results in the overexpression of synapse-associated protein 97, which is void on synaptic transmission of mature neurons but damages LTP (97). The transcription factor cAMP response element-binding (CREB) is a key regulator of synaptic growth and refinement in mature neurons; however, there is no direct target between CREB and neuronal plasticity (98). CREB binds to the miR-132 promoter in the hippocampal neurons of mice and facilitates its expression, which inhibits the translation of p250GAP, thereby regulating changes in the dendritic morphology of neurons (98). miR-134 inhibits the translation of LIM domain kinase 1 and is involved in regulating neuronal dendritic spine morphology (99). The dendritic spines of neurons become smaller when miR-134 is overexpressed and larger with miR-134 knockdown (99). Subsequent studies have shown that miR-138 exerts a similar effect to miR-134 by regulating the expression of acyl-protein thioesterase 1 (100). Bond et al found that in lncRNA-Evf2-knockout mice, the number of GABA-ergic interneurons in the early postnatal period was significantly decreased in the Ammons horn and DG, and the number gradually returned to normal; however, the number of GABA-ergic pyramidal cells in the CA1 region were maintained at a low level, resulting in the impaired function of neural circuits in the hippocampus (101). Evf2 recruits distal-less homeobox 5 (DLX) and MeCP2 to Dlx5/6, and regulates the expression of Dlx5, Dlx6 and GAD67, consequently regulating the production of GABA interneurons (101). The lncRNA-Malat1 regulates the synaptic density between hippocampal neurons. Of note, lncRNA-Malat1-knockout decreases the expression of synaptic cell adhesion molecule 1 (SynCAM1) and downregulates the expression of neuroligin 1 (NLGN 1), resulting in decreased synaptic density between hippocampal neurons (102).

The lncRNA-Uc.173 originates from the transcribed ultraconservative region of the genome in humans and rodents. Nan et al demonstrated that lead induced the apoptosis of hippocampal neurons in developing and maturing rats. Additionally, the expression of lncRNA-Uc.173 in the lead-exposed cells decreased over a period, whereas the overexpression of lncRNA-Uc.173 significantly reduced the apoptosis of these cells, suggesting that lncRNA-Uc.173 regulates the genes involved in hippocampal neuronal apoptosis and maintains hippocampal development (103). In the hippocampus of adult mice, lncRNA-AK089514 is expressed in the GCL; lncRNA-BC051426 appears in the pyramidal layer of Ammons horn and lncRNA-AK037594 emerges in the GCL and the pyramidal layer of the CA3 region (89).

Partial miRNAs can target key enzymes in the regulation of epigenetic mechanisms, termed epigenetic-miRNA (epi-miRNA). Epi-miRNA can inhibit the expression of DNMT by binding to the 3′-UTR of DNMT mRNA, which in turn affects the genome-wide methylation pattern (104). miR-290, a type of epi-miRNA, affects the differentiation of embryonic stem cells by controlling the de novo DNA methylation of embryonic stem cells, and differentiation is inhibited when miR-290 is deficient (105). Epi-miRNA inhibits DNA methyltransferase by regulating B cell-specific Mo MLV insertion site-1 (BMI-1) and enhancer of zeste homolog-2 (EZH2), promoting the senescence of pluripotent stem cells (106). Epi-miRNAs (miR-125 and miR-181) act as modulators of chromobox 7, mediate the expression of the polycomb genes and are key in establishing embryonic stem cell pluripotency (107). Epi-miRNA can target Bmi-1 to regulate the expression of polycomb genes (108). The downregulation of miR-128 has been shown to result in the upregulation of Bmi-1, which induces the abnormal proliferation of brain cells (109). Ezh2, which interferes with the expression of polycomb genes, is regulated by Epi-miRNA. The overexpression of miR-137 inhibits the appearance of Ezh2 and promotes neurogenesis in the hippocampal SGZ (110). HDACs are also regulated by Epi-miRNA, miR-134 mediates the post-transcriptional modification of CREB, leading to increased activation of the NAD-dependent deacetylase Sir2, which disrupts the plasticity of hippocampal synapses and leads to memory impairment (111). Zovoilis et al (112) showed that the expression of miRNA 34c was increased in the hippocampus of AD mice, and that miR-34c was able to activate the NAD-dependent deacetylase sirtuin 1 and cause memory impairment.

Miller-Delaney et al (57) indicated that there are multiple miRNA differential methylation events in the human hippocampus in temporal lobe epilepsy, leading to the upregulation and downregulation of corresponding miRNA expression; in particular, the upregulation of miR-876-3p and miR-193a-5p is most valuable, suggesting that miRNA dysregulation in the hippocampus causes epilepsy. It has been found that miRNAs cause Glu-ergic and GABA-ergic synaptic dysfunction of the hippocampus by regulating fragile X mental retardation protein, leading to fragile X syndrome epilepsy (113). In addition, lncRNA-BC1 and lncRNA-Evf2 were reported to act on Glu-ergic and GABA-ergic interneurons, respectively, and the excited interneurons induced epilepsy when lncRNA-BC1 and lncRNA-Evf2 were absent (101,114). Impaired synaptic plasticity in AD is associated with the dysregulation of specific miRNAs. Hu et al showed that miR-34c was significantly increased in amyloid-β (Aβ) accumulation in hippocampal neurons, whereas decreased miR-34c improved Aβ-induced synaptic failure and memory deficits (115). Furthermore, deletions on chromosome 22q11.2 caused by the upregulation of miR-185 in the hippocampus is an important genetic risk factor for schizophrenia, which indicates that miR-185 can be used as a novel target for the development of therapeutic drugs (116). A study on the schizophrenia risk gene MIR137 in rat hippocampal neurons indicated that MIR137 is key in the pathophysiological progression of schizophrenia as a hippocampal gene network node (117).

Conclusions

Humans and rodents exhibit marked similarity in the basic process of the development of the hippocampus. However, a marked difference is observed in the generation time of the DGC; ~80% of the human DGC is formed at the embryonic stage, whereas ~85% of the rodent DGC appears postnatally. The RGCs, including cell migration scaffolds, are crucial in the development of the hippocampal DG in humans and rodents.

Epigenetic inheritance is important in various stages of development of the hippocampus, and affects the proliferation, differentiation, migration and maturation of nerve cells. DNA methylation, histone acetylation and non-coding RNAs significantly influence the formation of synapses in the hippocampal neurons; this affects the construction of hippocampal neural circuits, and postnatal learning and memory function. Notably, future investigations are likely to considerably affect the analysis of clinically relevant disease mechanisms in addition to clinical diagnosis and treatment. DNA methylation, histone modifications and non-coding RNAs may act on the same gene or different aspects of the same mechanism of action, for example, the negative correlation between DNA methylation and histone tagging (H3K4me3) (118); histone modifications and lncRNAs regulate the Sox gene (119,120), and lncRNAs and miRNAs act on mRNAs differently to affect gene expression. These phenomena suggest the occurrence of interactions between different epigenetic regulatory mechanisms to produce synergistic or antagonistic effects in the same regulatory process. With an increasing number of epigenetic mechanisms revealed, further correlations can be identified to provide in-depth insight into the role and significance of epigenetic modifications in the development of the hippocampus.

Acknowledgements

Not applicable.

Funding

This study was supported by the Nature Science Foundation of Hubei Province (grant no. 2018CFB667), the Foundation of Health and Family Planning Commission of Hubei Province (grant no. WJ2016-YZ-01) and the Yangtze Youth Science and Technology Innovation Team Funds (grant no. 2016cqt04).

Availability of data and materials

Not applicable.

Authors contributions

SJF searched the related literature and wrote the manuscript. ABS aided with the literature search and edited the manuscript. LL drafted the manuscript and revised it critically for important intellectual content. All authors aided to prepare the manuscript and approved the final version to be published.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

DG

dentate gyrus

PM

postnatal month

EW

embryonic week

SGZ

subventricular zone

GCL

granular cell layer

GD

gestational day

NPCs

neural progenitor cells

ED

embryonic day

RGCs

radial glial cells

VZ

ventricular zone

DNMTs

DNA methyltransferases

MBD3

methyl-CpG-binding domain-3

MeCP2

methyl CpG binding protein 2

GFAP

glial fibrillary acidic protein

HDACs

histone deacetylases

ESCs

embryonic stem cells

lncRNA

long non-coding RNA

miRNA

microRNA

IPS cells

induced pluripotent stem cells

LTP

long-term potentiation

APT1

acyl-protein thioesterase 1

SynCAM1

synaptic cell adhesion molecule 1

NLGN1

neuroligin 1

References

1 

Li Y, Mu Y and Gage FH: Development of neural circuits in the adult hippocampus. Curr Top Dev Biol. 87:149–174. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Xu L, Tang X, Wang Y, Xu H and Fan X: Radial glia, the keystone of the development of the hippocampal dentate gyrus. Mol Neurobiol. 51:131–141. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Zhao C, Deng W and Gage FH: Mechanisms and functional implications of adult neurogenesis. Cell. 132:645–660. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Khalaf-Nazzal R and Francis F: Hippocampal development-old and new findings. Neuroscience. 248:225–242. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Stouffer MA, Golden JA and Francis F: Neuronal migration disorders: Focus on the cytoskeleton and epilepsy. Neurobiol Dis. 92:18–45. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Burke MW, Ptito M, Ervin FR and Palmour RM: Hippocampal neuron populations are reduced in vervet monkeys with fetal alcohol exposure. Dev Psychobiol. 57:470–485. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Legrand M, Lam S, Anselme I, Gloaguen C, Ibanez C, Eriksson P, Lestaevel P and Dinocourt C: Exposure to depleted uranium during development affects neuronal differentiation in the hippocampal dentate gyrus and induces depressive-like behavior in offspring. Neurotoxicology. 57:153–162. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Inbar-Feigenberg M, Choufani S, Butcher DT, Roifman M and Weksberg R: Basic concepts of epigenetics. Fertil Steril. 99:607–615. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Caronia-Brown G, Yoshida M, Gulden F, Assimacopoulos S and Grove EA: The cortical hem regulates the size and patterning of neocortex. Development. 141:2855–2865. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Deng JB, Cai Y, Sun XJ, Zhang Y, Su M and Song QX: Morphological study of human embryo development in the Hippocampus-I. General observation of structure. Shenjing Jiepouxue Zazhi. (1):1–9. 1996.(In Chinese).

11 

Li G and Pleasure SJ: Morphogenesis of the dentate gyrus: What we are learning from mouse mutants. Dev Neurosci. 27:93–99. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Nicola Z, Fabel K and Kempermann G: Development of the adult neurogenic niche in the hippocampus of mice. Front Neuroanat. 9:532015. View Article : Google Scholar : PubMed/NCBI

13 

Kier EL, Kim JH, Fulbright RK and Bronen RA: Embryology of the human fetal hippocampus: MR imaging, anatomy, and histology. AJNR Am J Neuroradiol. 18:525–532. 1997.PubMed/NCBI

14 

Humphrey T: The development of the human hippocampal fissure. J Anat. 101:655–676. 1967.PubMed/NCBI

15 

Rice D and Barone S Jr: Critical periods of vulnerability for the developing nervous system: Evidence from humans and animal models. Environ Health Perspect. 108 Suppl 3:511–533. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Deng W, Aimone JB and Gage FH: New neurons and new memories: How does adult hippocampal neurogenesis affect learning and memory? Nat Rev Neurosci. 11:339–350. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Warner-Schmidt JL and Duman RS: Hippocampal neurogenesis: Opposing effects of stress and antidepressant treatment. Hippocampus. 16:239–249. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Utsunomiya H, Takano K, Okazaki M and Mitsudome A: Development of the temporal lobe in infants and children: Analysis by MR-based volumetry. AJNR Am J Neuroradiol. 20:717–723. 1999.PubMed/NCBI

19 

Woolard AA and Heckers S: Anatomical and functional correlates of human hippocampal volume asymmetry. Psychiatry Res. 201:48–53. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Rogers BP, Sheffield JM, Luksik AS and Heckers S: Systematic error in hippocampal volume asymmetry measurement is minimal with a manual segmentation protocol. Front Neurosci. 6:1792012. View Article : Google Scholar : PubMed/NCBI

21 

Semple BD, Blomgren K, Gimlin K, Ferriero DM and Noble-Haeusslein LJ: Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol. 106–107, 1-16. 2013.PubMed/NCBI

22 

Bayer SA: Development of the hippocampal region in the rat. II. Morphogenesis during embryonic and early postnatal life. J Comp Neurol. 190:115–134. 1980. View Article : Google Scholar : PubMed/NCBI

23 

Radic T, Frieß L, Vijikumar A, Jungenitz T, Deller T and Schwarzacher SW: Differential postnatal expression of neuronal maturation markers in the dentate gyrus of mice and rats. Front Neuroanat. 11:1042017. View Article : Google Scholar : PubMed/NCBI

24 

Altman J and Bayer SA: Mosaic organization of the hippocampal neuroepithelium and the multiple germinal sources of dentate granule cells. J Comp Neurol. 301:325–342. 1990. View Article : Google Scholar : PubMed/NCBI

25 

Altman J and Bayer SA: Migration and distribution of two populations of hippocampal granule cell precursors during the perinatal and postnatal periods. J Comp Neurol. 301:365–381. 1990. View Article : Google Scholar : PubMed/NCBI

26 

Bayer SA: Development of the hippocampal region in the rat. I. Neurogenesis examined with 3H-thymidine autoradiography. J Comp Neurol. 190:87–114. 1980. View Article : Google Scholar : PubMed/NCBI

27 

Altman J and Bayer SA: Prolonged sojourn of developing pyramidal cells in the intermediate zone of the hippocampus and their settling in the stratum pyramidale. J Comp Neurol. 301:343–364. 1990. View Article : Google Scholar : PubMed/NCBI

28 

Schlessinger AR, Cowan WM and Swanson LW: The time of origin of neurons in Ammons horn and the associated retrohippocampal fields. Anat Embryol (Berl). 154:153–173. 1978. View Article : Google Scholar : PubMed/NCBI

29 

Eriksson PS, Perfilieva E, Björk-Eriksson T, Alborn AM, Nordborg C, Peterson DA and Gage FH: Neurogenesis in the adult human hippocampus. Nat Med. 4:1313–1317. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Berg DA, Bond AM, Ming GL and Song H: Radial glial cells in the adult dentate gyrus: What are they and where do they come from? F1000 Res. 7:2772018. View Article : Google Scholar

31 

Nakahira E and Yuasa S: Neuronal generation, migration, and differentiation in the mouse hippocampal primoridium as revealed by enhanced green fluorescent protein gene transfer by means of in utero electroporation. J Comp Neurol. 483:329–340. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Barry G, Piper M, Lindwall C, Moldrich R, Mason S, Little E, Sarkar A, Tole S, Gronostajski RM and Richards LJ: Specific glial populations regulate hippocampal morphogenesis. J Neurosci. 28:12328–12340. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Angevine JB Jr: Time of neuron origin in the hippocampal region. An autoradiographic study in the mouse. Exp Neurol Suppl. (Suppl 2):1–70. 1965.

34 

Hayashi K, Kubo K, Kitazawa A and Nakajima K: Cellular dynamics of neuronal migration in the hippocampus. Front Neurosci. 9:1352015. View Article : Google Scholar : PubMed/NCBI

35 

Reznikov KY: Cell proliferation and cytogenesis in the mouse hippocampus. Adv Anat Embryol Cell Biol. 122:1–74. 1991. View Article : Google Scholar : PubMed/NCBI

36 

Pérez Delgado MM, Serrano Aguilar PG, Castañeyra Perdomo A and Ferres Torres R: Postnatal development of the Ammons horn (CA1 and CA3 fields). A karyometric and topographic study. Histol Histopathol. 9:715–721. 1994.PubMed/NCBI

37 

Podobinska M, Szablowska-Gadomska I, Augustyniak J, Sandvig I, Sandvig A and Buzanska L: Epigenetic modulation of stem cells in neurodevelopment: The role of methylation and acetylation. Front Cell Neurosci. 11:232017. View Article : Google Scholar : PubMed/NCBI

38 

Moore LD, Le T and Fan G: DNA methylation and its basic function. Neuropsychopharmacology. 38:23–38. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Uysal F, Akkoyunlu G and Ozturk S: Dynamic expression of DNA methyltransferases (DNMTs) in oocytes and early embryos. Biochimie. 116:103–113. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Jeltsch A and Jurkowska RZ: New concepts in DNA methylation. Trends Biochem Sci. 39:310–318. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Jones PA: Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Reik W, Dean W and Walter J: Epigenetic reprogramming in mammalian development. Science. 293:1089–1093. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Denomme MM and Mann MR: Maternal control of genomic imprint maintenance. Reprod Biomed Online. 27:629–636. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Chen Y, Damayanti NP, Irudayaraj J, Dunn K and Zhou FC: Diversity of two forms of DNA methylation in the brain. Front Genet. 5:462014. View Article : Google Scholar : PubMed/NCBI

45 

Setoguchi H, Namihira M, Kohyama J, Asano H, Sanosaka T and Nakashima K: Methyl-CpG binding proteins are involved in restricting differentiation plasticity in neurons. J Neurosci Res. 84:969–979. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Wu Z, Huang K, Yu J, Le T, Namihira M, Liu Y, Zhang J, Xue Z, Cheng L and Fan G: Dnmt3a regulates both proliferation and differentiation of mouse neural stem cells. J Neurosci Res. 90:1883–1891. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Noguchi H, Murao N, Kimura A, Matsuda T, Namihira M and Nakashima K: DNA Methyltransferase 1 Is Indispensable for Development of the Hippocampal Dentate Gyrus. J Neurosci. 36:6050–6068. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Chen Y, Ozturk NC and Zhou FC: DNA methylation program in developing hippocampus and its alteration by alcohol. PLoS One. 8:e605032013. View Article : Google Scholar : PubMed/NCBI

49 

Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, Silva AJ and Fan G: Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat Neurosci. 13:423–430. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Wu H, Coskun V, Tao J, Xie W, Ge W, Yoshikawa K, Li E, Zhang Y and Sun YE: Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science. 329:444–448. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Simmons RK, Stringfellow SA, Glover ME, Wagle AA and Clinton SM: DNA methylation markers in the postnatal developing rat brain. Brain Res. 1533:26–36. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Zhang RR, Cui QY, Murai K, Lim YC, Smith ZD, Jin S, Ye P, Rosa L, Lee YK, Wu HP, et al: Tet1 regulates adult hippocampal neurogenesis and cognition. Cell Stem Cell. 13:237–245. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Larimore JL, Chapleau CA, Kudo S, Theibert A, Percy AK and Pozzo-Miller L: Bdnf overexpression in hippocampal neurons prevents dendritic atrophy caused by Rett-associated MECP2 mutations. Neurobiol Dis. 34:199–211. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Zoghbi HY: Postnatal neurodevelopmental disorders: Meeting at the synapse? Science. 302:826–830. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Falkai P, Reich-Erkelenz D, Malchow B, Schmitt A and Majtenyi K: Brain development before onset of the first psychotic episode and during outcome of schizophrenia. Fortschr Neurol Psychiatr. 81:260–264. 2013.(In German). PubMed/NCBI

56 

Matrisciano F, Tueting P, Dalal I, Kadriu B, Grayson DR, Davis JM, Nicoletti F and Guidotti A: Epigenetic modifications of GABAergic interneurons are associated with the schizophrenia-like phenotype induced by prenatal stress in mice. Neuropharmacology. 68:184–194. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Miller-Delaney SF, Bryan K, Das S, McKiernan RC, Bray IM, Reynolds JP, Gwinn R, Stallings RL and Henshall DC: Differential DNA methylation profiles of coding and non-coding genes define hippocampal sclerosis in human temporal lobe epilepsy. Brain. 138:616–631. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Blanco-Luquin I, Altuna M, Sánchez-Ruiz de Gordoa J, Urdánoz-Casado A, Roldán M, Cámara M, Zelaya V, Erro ME, Echavarri C and Mendioroz M: PLD3 epigenetic changes in the hippocampus of Alzheimers disease. Clin Epigenetics. 10:1162018. View Article : Google Scholar : PubMed/NCBI

59 

Zhang TY, Labonté B, Wen XL, Turecki G and Meaney MJ: Epigenetic mechanisms for the early environmental regulation of hippocampal glucocorticoid receptor gene expression in rodents and humans. Neuropsychopharmacology. 38:111–123. 2013. View Article : Google Scholar : PubMed/NCBI

60 

McClelland S, Korosi A, Cope J, Ivy A and Baram TZ: Emerging roles of epigenetic mechanisms in the enduring effects of early-life stress and experience on learning and memory. Neurobiol Learn Mem. 96:79–88. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Davies MN, Krause L, Bell JT, Gao F, Ward KJ, Wu H, Lu H, Liu Y, Tsai PC, Collier DA, et al; UK Brain Expression Consortium, . Hypermethylation in the ZBTB20 gene is associated with major depressive disorder. Genome Biol. 15:R562014. View Article : Google Scholar : PubMed/NCBI

62 

Oliveira AM, Hemstedt TJ, Freitag HE and Bading H: Dnmt3a2: A hub for enhancing cognitive functions. Mol Psychiatry. 21:1130–1136. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Fischle W, Mootz HD and Schwarzer D: Synthetic histone code. Curr Opin Chem Biol. 28:131–140. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Zentner GE and Henikoff S: Regulation of nucleosome dynamics by histone modifications. Nat Struct Mol Biol. 20:259–266. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Zhang Y, Gilquin B, Khochbin S and Matthias P: Two catalytic domains are required for protein deacetylation. J Biol Chem. 281:2401–2404. 2006. View Article : Google Scholar : PubMed/NCBI

66 

MacDonald JL and Roskams AJ: Histone deacetylases 1 and 2 are expressed at distinct stages of neuro-glial development. Dev Dyn. 237:2256–2267. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Montgomery RL, Hsieh J, Barbosa AC, Richardson JA and Olson EN: Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci USA. 106:7876–7881. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Hagelkruys A, Lagger S, Krahmer J, Leopoldi A, Artaker M, Pusch O, Zezula J, Weissmann S, Xie Y, Schöfer C, et al: A single allele of Hdac2 but not Hdac1 is sufficient for normal mouse brain development in the absence of its paralog. Development. 141:604–616. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Baranek C and Atanasoski S: Modulating epigenetic mechanisms: The diverse functions of Ski during cortical development. Epigenetics. 7:676–679. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Kokura K, Kaul SC, Wadhwa R, Nomura T, Khan MM, Shinagawa T, Yasukawa T, Colmenares C and Ishii S: The Ski protein family is required for MeCP2-mediated transcriptional repression. J Biol Chem. 276:34115–34121. 2001. View Article : Google Scholar : PubMed/NCBI

71 

You L, Yan K, Zou J, Zhao H, Bertos NR, Park M, Wang E and Yang XJ: The lysine acetyltransferase activator Brpf1 governs dentate gyrus development through neural stem cells and progenitors. PLoS Genet. 11:e10050342015. View Article : Google Scholar : PubMed/NCBI

72 

Qiao Y, Wang R, Yang X, Tang K and Jing N: Dual roles of histone H3 lysine 9 acetylation in human embryonic stem cell pluripotency and neural differentiation. J Biol Chem. 290:2508–2520. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Biswal S, Das D, Barhwal K, Kumar A, Nag TC, Thakur MK, Hota SK and Kumar B: Epigenetic regulation of SNAP25 prevents progressive glutamate excitotoxicty in hypoxic CA3 neurons. Mol Neurobiol. 54:6133–6147. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Kim BW, Yang S, Lee CH and Son H: A critical time window for the survival of neural progenitor cells by HDAC inhibitors in the hippocampus. Mol Cells. 31:159–164. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Hou N, Gong M, Bi Y, Zhang Y, Tan B, Liu Y, Wei X, Chen J and Li T: Spatiotemporal expression of HDAC2 during the postnatal development of the rat hippocampus. Int J Med Sci. 11:788–795. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, Nieland TJ, Zhou Y, Wang X, Mazitschek R, et al: HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 459:55–60. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Gräff J, Rei D, Guan JS, Wang WY, Seo J, Hennig KM, Nieland TJ, Fass DM, Kao PF, Kahn M, et al: An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature. 483:222–226. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Sewal AS, Patzke H, Perez EJ, Park P, Lehrmann E, Zhang Y, Becker KG, Fletcher BR, Long JM and Rapp PR: Experience modulates the effects of histone deacetylase inhibitors on gene and protein expression in the hippocampus: Impaired plasticity in aging. J Neurosci. 35:11729–11742. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Sandner G, Host L, Angst MJ, Guiberteau T, Guignard B and Zwiller J: The HDAC inhibitor phenylbutyrate reverses effects of neonatal ventral hippocampal lesion in rats. Front Psychiatry. 1:1532011. View Article : Google Scholar : PubMed/NCBI

80 

Tsuji M, Miyagawa K and Takeda H: Epigenetic regulation of resistance to emotional stress: Possible involvement of 5-HT1A receptor-mediated histone acetylation. J Pharmacol Sci. 125:347–354. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Contestabile A and Sintoni S: Histone acetylation in neurodevelopment. Curr Pharm Des. 19:5043–5050. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Marchese FP and Huarte M: Long non-coding RNAs and chromatin modifiers: Their place in the epigenetic code. Epigenetics. 9:21–26. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Li X, Wu Z, Fu X and Han W: lncRNAs: Insights into their function and mechanics in underlying disorders. Mutat Res Rev Mutat Res. 762:1–21. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Chuang JC and Jones PA: Epigenetics and microRNAs. Pediatr Res. 61:24R–29R. 2007. View Article : Google Scholar : PubMed/NCBI

85 

Wang Z, Yao H, Lin S, Zhu X, Shen Z, Lu G, Poon WS, Xie D, Lin MC and Kung HF: Transcriptional and epigenetic regulation of human microRNAs. Cancer Lett. 331:1–10. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Tammen SA, Friso S and Choi SW: Epigenetics: The link between nature and nurture. Mol Aspects Med. 34:753–764. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Li Q, Bian S, Hong J, Kawase-Koga Y, Zhu E, Zheng Y, Yang L and Sun T: Timing specific requirement of microRNA function is essential for embryonic and postnatal hippocampal development. PLoS One. 6:e260002011. View Article : Google Scholar : PubMed/NCBI

88 

Konopka W, Kiryk A, Novak M, Herwerth M, Parkitna JR, Wawrzyniak M, Kowarsch A, Michaluk P, Dzwonek J, Arnsperger T, et al: MicroRNA loss enhances learning and memory in mice. J Neurosci. 30:14835–14842. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Mercer TR, Dinger ME, Sunkin SM, Mehler MF and Mattick JS: Specific expression of long noncoding RNAs in the mouse brain. Proc Natl Acad Sci USA. 105:716–721. 2008. View Article : Google Scholar : PubMed/NCBI

90 

Lin M, Pedrosa E, Shah A, Hrabovsky A, Maqbool S, Zheng D and Lachman HM: RNA-Seq of human neurons derived from iPS cells reveals candidate long non-coding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS One. 6:e233562011. View Article : Google Scholar : PubMed/NCBI

91 

Nigro A, Menon R, Bergamaschi A, Clovis YM, Baldi A, Ehrmann M, Comi G, De Pietri Tonelli D, Farina C, Martino G, et al: miR-30e and miR-181d control radial glia cell proliferation via HtrA1 modulation. Cell Death Dis. 3:e3602012. View Article : Google Scholar : PubMed/NCBI

92 

Kerek R, Geoffroy A, Bison A, Martin N, Akchiche N, Pourié G, Helle D, Guéant JL, Bossenmeyer-Pourié C and Daval JL: Early methyl donor deficiency may induce persistent brain defects by reducing Stat3 signaling targeted by miR-124. Cell Death Dis. 4:e7552013. View Article : Google Scholar : PubMed/NCBI

93 

Monteleone MC, Adrover E, Pallarés ME, Antonelli MC, Frasch AC and Brocco MA: Prenatal stress changes the glycoprotein GPM6A gene expression and induces epigenetic changes in rat offspring brain. Epigenetics. 9:152–160. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Howe JR VI, Li ES, Streeter SE, Rahme GJ, Chipumuro E, Russo GB, Litzky JF, Hills LB, Rodgers KR, Skelton PD, et al: MiR-338-3p regulates neuronal maturation and suppresses glioblastoma proliferation. PLoS One. 12:e01776612017. View Article : Google Scholar : PubMed/NCBI

95 

Shibata M, Nakao H, Kiyonari H, Abe T and Aizawa S: MicroRNA-9 regulates neurogenesis in mouse telencephalon by targeting multiple transcription factors. J Neurosci. 31:3407–3422. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Delaloy C, Liu L, Lee JA, Su H, Shen F, Yang GY, Young WL, Ivey KN and Gao FB: MicroRNA-9 coordinates proliferation and migration of human embryonic stem cell-derived neural progenitors. Cell Stem Cell. 6:323–335. 2010. View Article : Google Scholar : PubMed/NCBI

97 

Sim SE, Lim CS, Kim JI, Seo D, Chun H, Yu NK, Lee J, Kang SJ, Ko HG, Choi JH, et al: The Brain-Enriched MicroRNA miR-9-3p Regulates Synaptic Plasticity and Memory. J Neurosci. 36:8641–8652. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Wayman GA, Davare M, Ando H, Fortin D, Varlamova O, Cheng HY, Marks D, Obrietan K, Soderling TR, Goodman RH, et al: An activity-regulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc Natl Acad Sci USA. 105:9093–9098. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M and Greenberg ME: A brain-specific microRNA regulates dendritic spine development. Nature. 439:283–289. 2006. View Article : Google Scholar : PubMed/NCBI

100 

Siegel G, Obernosterer G, Fiore R, Oehmen M, Bicker S, Christensen M, Khudayberdiev S, Leuschner PF, Busch CJ, Kane C, et al: A functional screen implicates microRNA-138-dependent regulation of the depalmitoylation enzyme APT1 in dendritic spine morphogenesis. Nat Cell Biol. 11:705–716. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Bond AM, Vangompel MJ, Sametsky EA, Clark MF, Savage JC, Disterhoft JF and Kohtz JD: Balanced gene regulation by an embryonic brain ncRNA is critical for adult hippocampal GABA circuitry. Nat Neurosci. 12:1020–1027. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Bernard D, Prasanth KV, Tripathi V, Colasse S, Nakamura T, Xuan Z, Zhang MQ, Sedel F, Jourdren L, Coulpier F, et al: A long nuclear-retained non-coding RNA regulates synaptogenesis by modulating gene expression. EMBO J. 29:3082–3093. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Nan A, Zhou X, Chen L, Liu M, Zhang N, Zhang L, Luo Y, Liu Z, Dai L and Jiang Y: A transcribed ultraconserved noncoding RNA, Uc.173, is a key molecule for the inhibition of lead-induced neuronal apoptosis. Oncotarget. 7:112–124. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Chhabra R: miRNA and methylation: A multifaceted liaison. ChemBioChem. 16:195–203. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Sinkkonen L, Hugenschmidt T, Berninger P, Gaidatzis D, Mohn F, Artus-Revel CG, Zavolan M, Svoboda P and Filipowicz W: MicroRNAs control de novo DNA methylation through regulation of transcriptional repressors in mouse embryonic stem cells. Nat Struct Mol Biol. 15:259–267. 2008. View Article : Google Scholar : PubMed/NCBI

106 

So AY, Jung JW, Lee S, Kim HS and Kang KS: DNA methyltransferase controls stem cell aging by regulating BMI1 and EZH2 through microRNAs. PLoS One. 6:e195032011. View Article : Google Scholar : PubMed/NCBI

107 

OLoghlen A, Muñoz-Cabello AM, Gaspar-Maia A, Wu HA, Banito A, Kunowska N, Racek T, Pemberton HN, Beolchi P, Lavial F, et al: MicroRNA regulation of Cbx7 mediates a switch of Polycomb orthologs during ESC differentiation. Cell Stem Cell. 10:33–46. 2012. View Article : Google Scholar : PubMed/NCBI

108 

Jobe EM, McQuate AL and Zhao X: Crosstalk among Epigenetic Pathways Regulates Neurogenesis. Front Neurosci. 6:592012. View Article : Google Scholar : PubMed/NCBI

109 

Cui JG, Zhao Y, Sethi P, Li YY, Mahta A, Culicchia F and Lukiw WJ: Micro-RNA-128 (miRNA-128) down-regulation in glioblastoma targets ARP5 (ANGPTL6), Bmi-1 and E2F-3a, key regulators of brain cell proliferation. J Neurooncol. 98:297–304. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Szulwach KE, Li X, Smrt RD, Li Y, Luo Y, Lin L, Santistevan NJ, Li W, Zhao X and Jin P: Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol. 189:127–141. 2010. View Article : Google Scholar : PubMed/NCBI

111 

Gao J, Wang WY, Mao YW, Gräff J, Guan JS, Pan L, Mak G, Kim D, Su SC and Tsai LH: A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature. 466:1105–1109. 2010. View Article : Google Scholar : PubMed/NCBI

112 

Zovoilis A, Agbemenyah HY, Agis-Balboa RC, Stilling RM, Edbauer D, Rao P, Farinelli L, Delalle I, Schmitt A, Falkai P, et al: microRNA-34c is a novel target to treat dementias. EMBO J. 30:4299–4308. 2011. View Article : Google Scholar : PubMed/NCBI

113 

Qureshi IA and Mehler MF: Epigenetic mechanisms underlying human epileptic disorders and the process of epileptogenesis. Neurobiol Dis. 39:53–60. 2010. View Article : Google Scholar : PubMed/NCBI

114 

Zhong J, Chuang SC, Bianchi R, Zhao W, Lee H, Fenton AA, Wong RK and Tiedge H: BC1 regulation of metabotropic glutamate receptor-mediated neuronal excitability. J Neurosci. 29:9977–9986. 2009. View Article : Google Scholar : PubMed/NCBI

115 

Hu S, Wang H, Chen K, Cheng P, Gao S, Liu J, Li X and Sun X: MicroRNA-34c downregulation ameliorates amyloid-β-induced synaptic failure and memory deficits by targeting VAMP2. J Alzheimers Dis. 48:673–686. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Wesseling H, Xu B, Want EJ, Holmes E, Guest PC, Karayiorgou M, Gogos JA and Bahn S: System-based proteomic and metabonomic analysis of the Df(16)A+/- mouse identifies potential miR-185 targets and molecular pathway alterations. Mol Psychiatry. 22:384–395. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Olde Loohuis NF, Nadif Kasri N, Glennon JC, van Bokhoven H, Hébert SS, Kaplan BB, Martens GJ and Aschrafi A: The schizophrenia risk gene MIR137 acts as a hippocampal gene network node orchestrating the expression of genes relevant to nervous system development and function. Prog Neuropsychopharmacol Biol Psychiatry. 73:109–118. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong CT, Low HM, Kin Sung KW, Rigoutsos I, Loring J, et al: Dynamic changes in the human methylome during differentiation. Genome Res. 20:320–331. 2010. View Article : Google Scholar : PubMed/NCBI

119 

Amaral PP, Neyt C, Wilkins SJ, Askarian-Amiri ME, Sunkin SM, Perkins AC and Mattick JS: Complex architecture and regulated expression of the Sox2ot locus during vertebrate development. RNA. 15:2013–2027. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Amador-Arjona A, Cimadamore F, Huang CT, Wright R, Lewis S, Gage FH and Terskikh AV: SOX2 primes the epigenetic landscape in neural precursors enabling proper gene activation during hippocampal neurogenesis. Proc Natl Acad Sci USA. 112:E1936–E1945. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 9 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan SJ, Sun AB and Liu L: Epigenetic modulation during hippocampal development (Review). Biomed Rep 9: 463-473, 2018
APA
Fan, S., Sun, A., & Liu, L. (2018). Epigenetic modulation during hippocampal development (Review). Biomedical Reports, 9, 463-473. https://doi.org/10.3892/br.2018.1160
MLA
Fan, S., Sun, A., Liu, L."Epigenetic modulation during hippocampal development (Review)". Biomedical Reports 9.6 (2018): 463-473.
Chicago
Fan, S., Sun, A., Liu, L."Epigenetic modulation during hippocampal development (Review)". Biomedical Reports 9, no. 6 (2018): 463-473. https://doi.org/10.3892/br.2018.1160