Watch the epigenetics video from PBS. Begin your paper by defining epigenetics in your own words and discussing your reaction to the video. (you can choose whatever disease) Interview your family mem

Review Article Epigenetic Regulations in Neural Stem Cells and Neurological Diseases Hang Zhou, 1,2 Bin Wang, 2Hao Sun, 3Xingshun Xu ,1,2 and Yongxiang Wang 3 1Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China2Institute of Neuroscience, Soochow University, Suzhou, China3Department of Orthopedics, Clinical Medical School, Yangzhou University, Northern Jiangsu People ’s Hospital, Yangzhou 225001, China Correspondence should be addressed to Xingshun Xu; [email protected] and Yongxiang Wang; [email protected] Received 4 November 2017; Accepted 8 January 2018; Published 18 March 2018 Academic Editor:

Yujing Li Copyright © 2018 Hang Zhou et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Among the regulatory mechanisms of the renewal and di fferentiation of neural stem cells, recent evidences support that epigenetic modi fications such as DNA methylation, histone modi fication, and noncoding RNAs play critical roles in the regulation on the proliferation and di fferentiation of neural stem cells. In this review, we discussed recent advances of DNA modifi cations on the regulative mechanisms of neural stem cells. Among these epigenetic modifi cations, DNA 5-hydroxymethylcytosine (5hmC) modi fication is emerging as an important modulator on the proliferation and di fferentiation of neural stem cells. At the same time, Ten-eleven translocation (Tet) methylcytosine dioxygenases, the rate-limiting enzyme for the 5-hydroxymethylation reaction from 5-methylcytosine to 5-hydroxymethylcytosine, play a critical role in the tumorigenesis and the proliferation and di fferentiation of stem cells. The functions of 5hmC and TET proteins on neural stem cells and their roles in neurological diseases are discussed. 1. Introduction Human beings are developed from a fertilized egg into a complete individual; during the whole process, a series of precise regulations on the development are included, such as gene expression and gene silence [1], transcriptional regu- lation [2], posttranscriptional regulation [3], hormone regulation [4], chromosome behavior regulation [5], and apoptosis [6]. For these di fferent regulative pathways, their target cells are embryonic stem cells (ESCs). ESCs are totipo- tent stem cells that had a capability to proliferate and di ffer- entiate into appropriate lineages to form specialized cells and organs and play a central role in the developmental process [7]. Due to the powerful plasticity and potential of ESCs as a high potential cell replacement therapy for many diseases, stem cells are considered to have an appreciable translational prospect in the field of regenerative medicine [8]. Except for ESCs at the embryonic stage of the development, adult stem cells (ASCs) exist in di fferent tissues at the adult stage of the development [9]. ASCs are often in a resting state in individuals and exhibit di fferent potentials of regeneration and di fferentiation under pathological conditions or special incentives. Reynolds and Weiss first found that the neurons isolated from the striatum of the adult mouse brain could proliferate and di fferentiate in vitro with epidermal growth factors [9], indicating the existence of neural stem cells (NSCs) in the mature nervous system. They also demon- strated that NSC has the ability to self-renew and di fferenti- ate into other types of cells like neurons, astrocytes, and oligodendrocytes under many conditions such as growth fac- tors, neurotransmitters, hormones, injury, or environmental factors [9]. However, the renewal and di fferentiation ability of NSC is limited; in the process of aging or pathological conditions, neuronal cell loss is much more than newly gen- erated neurons and glial cells from NSCs, resulting in di ffer- ent neurological disorders including Alzheimer ’s disease [10], Parkinson ’s disease [11], Huntington ’s disease [12], neuroendocrine tumors [13], and ataxia [14]. Therefore, the regulation on the renewal and di fferentiation of NSCs or NSC transplantation therapy are considered an important Hindawi Stem Cells International Volume 2018, Article ID 6087143, 10 pages https://doi.org/10.1155/2018/6087143 therapeutic strategy for the treatment of these neurodegener- ative diseases.Among the regulatory mechanisms of the renewal and di fferentiation of NSCs, epigenetic modi fication plays a crit- ical role in monitoring the phase transition during individual development, maintaining the directional di fferentiation of stem cells, regulating the proliferation of speci fic cells, and controlling the process of di fferentiation [15, 16]. For exam- ple, in the process of umbilical cord mesenchymal stem cells (UMSCs) being di fferentiated to neural stem-like cells (uNSCLs), E1A-like inhibitor of di fferentiation 3 (EID3), an important member of EID gene family that has the main function of p300/CBP inhibitors (a transcriptional coactiva- tor) in response to cell transformation, growth arrest, or cell apoptosis, directly interacts with DNMT3A, a DNA methyl- transferase (DNMT) for DNA methylation, suggesting that DNA methylation may be involved the regulation of transdif- ferentiating from UMSCs to uNSCLs as a key mechanism in epigenetic regulation of stem cell reprogramming [17]. So far, epigenetic modifi cation is a hot topic in recent years. Except for DNA methylation, histone modi fication, micro-RNA, chromatin remodeling, and other epigenetic modi fication are found to play important roles in the regulation of stem cells [18]. In this article, we will review the recent advances of di fferent epigenetic modifi cations on NSCs, but mainly focus on the role of 5hmC as a new player in the regulation of the renewal and di fferentiation of ESCs or NSCs.

2. Recent Advances on Epigenetic Regulation on Stem Cells It is strongly believed that the basis of cell di fferentiation in ontogeny is based on the regulation of intracellular factors, while environmental factors also play a role as a main cause [19]. Epigenetic modi fications including methylation, acety- lation, ubiquitination, and phosphorylation on DNA, RNA, or proteins mediate the interaction between the environment and the organism [20]. Interestingly, recent evidences dem- onstrate that epigenetic modifi cation changes can be inher- ited to the next generation [21]. Here, we present a brief overview of current advances on epigenetic modi fications and NSCs.

2.1. DNA Methylation. The increasing evidences demonstrate that DNA methylation is involved in the proliferation and di fferentiation of stem cells [22]. DNA methylation prevents transcriptional factors from binding to promoters, such as Oct4 and Nanog, thereby limiting gene expression [23].

The process of DNA methylation is catalyzed by DNA meth- yltransferase, mainly DNMT1, DNMT3A, and DNMT3B.

DNMT3 enzyme is a de novo methyltransferase [24] and DNMT1 is mainly involved in the maintaining of DNA methylation in dividing somatic cells [25]. The deletion of DNMT3A in hematopoietic stem cells impaired the di fferen- tiation of transplanted hematopoietic stem cells and increased the level of hematopoietic stem cells in the bone marrow [22]. In skeletal muscle stem cells, the DNA methyl- ation of CpG dinucleotide in the promoter or enhancer region reduces gene expression of Pax7 and MyoD [26]. Similarly, Uhrf1 (ubiquitin-like PHD ring finger-1; also known as Np95) mainly interacts with DNMT1 to maintain DNA methylation in NSCs; the deletion of Uhrf1 in NSCs leads to increase the global DNA methylation and delayed neurodegeneration [27]. Recent evidences showed that Methyl CpG binding domain protein 1 (MBD1) is expressed in neural stem cells (aNSCs) of dentate gyrus of the adult hippocampus and maintains the integrity and stemness of NSC by inhibiting di fferentiation [28]. MBD1 and Methyl CpG binding protein 2 (Mecp2) belong to the methyl-CpG- binding protein family and play a key role to link DNA meth- ylation and transcriptional regulation on di fferentiation genes [29]. MBD1 de ficiency leads to the accumulation of undi fferentiated NSCs and impaired transition into the neu- ronal lineage [28]. DNA methylation is closely related to stem cell-related diseases. A recent study found that there are a large number of gene mutations of DNMT3A in acute myeloid leukemia which is a malignant tumor characterized by clonal stem cell proliferation and aberrant block in di ffer- entiation [30]. Fetal alcohol syndrome showed that alcohol exposure to cultured NSCs altered normal DNA methylation programming of key neural stem cell genes and retarded NSC migration and di fferentiation [31], supporting the role of aberrant patterns of DNA methylation in fetal neural devel- opment after embryonic alcohol exposure.

2.2. Histone Modi fication. Histone modifi cation refers to the process of histone methylation, acetylation, phosphorylation, polyadenylation, ubiquitination, and ADP glycosylation under the action of related enzymes. Histone-mediated epi- genetic gene silencing is to remove acetyl groups from histone tails catalyzed by histone deacetylase (HDAC) enzymes and enhance the binding of histones to DNA and the aggregation of chromosomes, preventing transcription factors into the regulatory region [32]. HDAC1 is highly expressed in the oligodendrocyte di fferentiation period of the corpus callosum; HDAC inhibitors blocked oligodendro- cyte di fferentiation and cause demyelination in the corpus callosum of postnatal rats [33]. The recent study indicated that the Arf-p53 axis also might be involved in the regulation of histone acetylation on the proliferation and senescence of the neurospheres [34]. Histone demethylation is also an important histone mod- i fi cation. It has two families, LSD1 (Lysine-speci fic demethy- lase 1) and JmjC (a domain), to regulate the proliferation and di fferentiation of stem cells. Inhibiting the activity of LSD1 or knockdown of LSD1 expression leads to the decreased prolif- eration of neural stem cells [35]. In addition, LSD1 plays a crucial role in maintaining the silencing of several develop- mental genes in human embryonic stem cells by regulating the balance between H3K4 (lysine 4 on histone H3 protein) and H3K27 methylation in its regulatory region [36]. Thus, histone modi fications play a role in inducing NSC di fferenti- ation into neurons and glial lineages, but the mechanisms are still not clear.

2.3. Noncoding RNA. Noncoding RNAs (ncRNAs) are a class of RNA molecules that have no ability to translate into pro- teins but function as regulatory factors at transcriptional or 2 Stem Cells International posttranscriptional levels, including ribosomal RNAs (rRNAs), microRNAs (miRNAs), piwi-interacting RNAs (piRNAs), long noncoding RNAs (lncRNAs), and others [37]. These ncRNAs have shown to play distinct but also con- served roles in regulation of differentiation of NSCs [38–40].

Among di fferent ncRNAs, current evidences demonstrate that miRNAs play critical roles in the regulation of di fferen- tiation of NSCs. miRNAs are a group of small RNA mole- cules of 20–24 nucleotides widely found in eukaryotes.

They bind to target mRNAs to regulate their gene expression by promoting the degradation of target mRNAs. Similarly, microRNA is also involved in the regulation of NSC di fferen- tiation and proliferation dynamic homeostasis; for example, high levels of miR-184, which are inhibited by methyl-CpG binding protein 1, promote stem cell proliferation but inhibit adult neural stem/progenitor cell (aNSCs) di fferentiation [41]. MiR-145 directly regulates Nurr1 (a nuclear receptor) expression level, and overexpression of miR-145 inhibits the di fferentiation e ffect of BMP2; knockdown of miR-145 promoted the upregulation of Nurr1, resulting in the di ffer- entiation of NSCs into dopaminergic neurons [42]. Micro- RNA can regulate many factors such as CT4, SOX2, and KLF4 in embryonic stem cells that are the direct targets of miR-145. The deletion of miR-145 increases the expression of OCT4, SOX2, and KLF4 and further inhibits the di fferen- tiation of NSC [43]. Recent studies showed that aging process begin when hypothalamic stem cells that coexpress Sox2 and Bmi1 are ablated accompanying with substantial loss of hypothalamic cells; the injection of exosomal miRNA in the cerebrospinal fluid, greatly prevented the cell aging process [44]. Therefore, more and more evidences showed that ncRNAs like miRNA are involved in the regulation of di ffer- entiation of NSCs.

3. Ten-Eleven Translocation (Tet) Proteins- 5hmC Modification-Related Enzymes Tet family proteins are a group of α-ketoglutarate ( α-KG) and Fe2+ ‐dependent monooxygenase to catalyze the con- version of 5mC to 5hmC, concluding Tet1, Tet2, and Tet2 [45]. In 2009, Tahiliani et al. found that Tet1 catalyzes the reaction of 5mC to 5hmC [46]; thereafter, Tet2 and Tet3 have been found to have similar catalytic activity [47]. Although the main functions of the three enzymes are to oxidize 5mC to 5hmC, the distribution of the enzymes is di fferent. The expression of Tet1 protein in embryonic stem cells and ner- vous system is high [48–50]; Tet2 is widely distributed and relatively high in hematopoietic system; Tet3 is mainly expressed in colon, muscle tissues, and less in brain tissues [51]. The three Tet enzymes contain a structurally similar carboxyl terminal catalytic region, which catalyzes the syn- thesis of 5hmC activity [45]. The catalytic domain of Tet proteins has 3 metal ion (Fe2+) and α-KG binding site to enhance its catalytic activity [46]. Tet1 and Tet3 have an amino terminal CXXC zinc finger protein domain, whereas Tet2 lacks this structure and needs to be assisted by IDAX protein with similar functions [52]. The CXXC domain protein of Tet2 is encoded by a distinct gene IDAX. The IDAX CXXC domain binds DNA sequences containing unmethylated CpG dinucleotides, localizes to promoters and CpG islands in genomic DNA, and interacts directly with the catalytic domain of Tet2 [52]. IDAX (also known as CXXC4), a reported inhibitor of Wnt signaling, regulates Tet2 protein expression [53]. Unexpectedly, IDAX expres- sion results in caspase activation and Tet2 protein downreg- ulation in a manner that depends on DNA binding through the IDAX CXXC domain, suggesting that IDAX recruits Tet2 to DNA before degradation [52]. Notably, the IDAX- related protein CXXC5 resembles IDAX in inhibiting Wnt signaling [54]. Therefore, the distribution and structure of Tet enzymes determine the distribution of 5hmC modifi ca- tions in brain and their di fferent roles in di fferent diseases.

Tet1 knockout mice showed impaired hippocampal neuro- genesis resulting in learning and memory de ficiency [55].

Tet2 functional disruption or knockout in fluences hemato- poietic cell homeostasis and hematopoietic di fferentiation and promotes the development of myeloid malignancies [56]. Although either Tet1 or tet2 knockout mice are viable and fertile, Tet3 knockout mice are perinatally lethal [51].

These demonstrate the di fferent roles of Tet proteins in the di fferent tissues and in the devolvement of di fferent organs.

The functions of Tet proteins and its related phenotypes in rodent animals and diseases in human are summarized in Table 1. 5-Hydroxymethylcytosine (5hmC), the oxidative product of 5mC, was found in mammals with surprisingly high abun- dance in 2009 [46, 57]. Recent studies showed that 5mC is not the final chemical steps for gene silencing; Tet protein- associated DNA demethylation can transform 5-methyl cytosine (5mC) into 5-hydroxymethycytosine (5hmC), 5- formylcytosine (5fC), and 5-carbosycytosine (5caC), but 5fC and 5caC is much less than 5-hydroxymethylcytosine [58, 59]. Interestingly, for individual tissues, the levels of 5hmC, 5fC, and 5caC were not signi ficantly related; for example, although 5hmC is more abundant in mouse brains than in ESCs, the levels of 5fC and 5caC are less abundant [45]. 5fC and 5caC can be further removed by the base exci- sion repair (BER) pathway and thymine-DNA glycosylase [60]. This pattern suggests that the di fferent steps of demeth- ylation cycle are di fferent in di fferent tissues [61]. In addi- tion, Tet protein overexpression or depletion can increase or decrease the content of 5hmC, 5fC, and 5caC in the genome [61]. The discovery of Tet proteins speeds the explo- ration of the functions of 5hmC [46, 57]. Because of its important functions, 5hmC in DNA has been considered as the sixth base. More evidences show that demethylation by 5hmC regulates the proliferation of NSCs and neurogenesis [27]. Therefore, we further discuss the regulation of 5hmC on NSCs and related neurological diseases. 4. Tet Proteins and DNA 5hmC Modifications Are Involved in the Regulation on the Proliferation and Differentiation of NSCs The direction of cell di fferentiation is determined by the speci fic expression of tissue-speci fic genes, while DNA 5mC is involved in the regulation of gene expression and 3 Stem Cells International differentiation of cells in a speci fic direction [62 –64]. Previ- ous studies have shown that about 1.4% of CpG islands undergo a signi ficant remethylation process during the dif- ferentiation of embryonic stem cells into NSCs and NPCs [65]. The increasing line of evidences indicate that 5mC directly inhibits transcription factors to bind to DNA [23] or recruits MeCP2 and MBD to form a complex and further prevent gene transcriptions that relate to the di fferentiation of NSCs [66]. Therefore, DNA methylation plays an impor- tant role in neural cell di fferentiation. Apparently, as an important demethylation mechanism, DNA 5hmC modi fica- tion and Tet enzymes can be involved in the regulations of NSCs in theory. Recently, 5hmC has been found in the mammalian genome and has been shown to be about 10 times more abundant in neurons than in some peripheral nervous tissues [67]. This suggests that 5hmC may be a stable epigenetic marker involved in cell speci fic mechanisms to achieve its function in the brain. More and more evidences demon- strated that Tet enzymes and Tet-mediated 5hmC modi fica- tions are involved in the proliferation and di fferentiation of ESCs and NSCs [29, 58, 68, 69]. Hahn et al. found that the increase of 5hmC modifi cation in gene bodies is associated with genes important for neuronal functions during neuronal di fferentiation in mouse brain regions; however, gene activa- tion for neuronal di fferentiation is not related to substantial DNA demethylation [69]. At the same time, overexpression of Tet2 and Tet3 also promotes the progression of neuronal di fferentiation [69]. Similarly, in Sirt6-knockout ESCs, the expression of Oct4, Sox2, and Nanog (the downstream of Sirt6) is inhibited and the upregulation of Tet enzymes and the signi ficant increase of DNA 5hmC are found, resulting in ESC skewed development towards neuroectoderm [68].

This suggests that Sirt6-regulated ESC di fferentiation is in a Tet enzyme and 5hmC-dependent manner [68], supporting Hahn et al. ’s results. A recent study further demonstrates that 5hmC dynamics is correlated with the di fferentiation of aNSCs; however, Tet2 primarily contributes to 5hmC acqui- sition during the di fferentiation of aNSCs [58]. Therefore, these evidences support the critical role of 5hmC modi fica- tions in the di fferentiation of NSCs.

Tet proteins, as the important enzymes for the conver- sion of 5mC to 5hmC, also showed their functions on the proliferation/di fferentiation of NSCs. Tet1 depletion impairs hippocampal neurogenesis accompanied with poor learning and memory in mice; at the same time, Tet1 defi ciency results in reduced neural progenitor pool in adult subgranu- lar zone [55]. These results provided in vivo evidences that Table 1: Tet proteins and their functions.

Genes Distribution StructureFunctions of Tet enzymes Knockout phenotypes in rodents Related diseases in humans Tet1 Mainly in ESCs and nervous system [48]. Contains CXXC, Cys-rich, and DSBH domains (1) Abnormal hippocampal neurogenesis, with learning and memory fading [55].

(2) Antidepressive phenotypes [94] (3) Skews di fferentiation towards extraembryonic lineages in the teratoma [99]. (1) Acute leukemia [100].

(2) Gastric cancer ([101, 102], Deng, [103]).

(3) Breast cancer [104].

Tet2 Widely distributed and high in hematopoietic system [48]. Contains Cys-rich and DSBH domains without CXXC domain (1) Hematopoietic cell homeostasis and hematopoietic di fferentiation impairment, myeloid malignancies [56].

(2) Retinal neurons developmental failure in zebra fish [105]. (1) Polycythemia vera [106, 107].

(2) Primary myelo fibrosis [107].

(3) Myelodysplastic syndrome [106].

(4) Myeloproliferative neoplasm [108].

(5) Melanoma [109].

Tet3 Mainly in colon and muscle tissues, less in brain tissue [51]. Contains CXXC, Cys-rich, and DSBH domains (1) Developmental failure [72] and embryonic sublethality [110].

(2) Impaired di fferentiation and increased apoptosis [72].

(3) Fear extinction impairments in mice [111].

(4) Abnormal morphogenesis of retinal neurons in zebra fish [105].

(5) Abnormal neural di fferentiation and skewed toward cardiac mesodermal fate in mouse ESC [112]. NA Tet1/2 DKO Embryonic stage death and little normal growth [113] NA Tet1/3 DKO (1) Dendritic arborization inhibition in mice [114] (2) Holoprosencephaly [115]. NA Tet1/2/3 TKO Developmental disorders [116] NA CXXC: Cys-X-X-Cys domain; DSBH: double-stranded beta helix; DKO: double knockout; ESCs: embryonic stem cells; TKO: triple knockout; NA: not available.

4 Stem Cells International Tet1 deficiency in the central nervous system decrease the proliferation of adult NSCs in the hippocampal dentate gyrus. Moran-Crusio et al. showed that the depletion of Tet2 stimulates aNSCs proliferation but impairs the di ffer- entiation of aNSCs [56]. Tet2 interacted with the neuronal transcription activator Foxo3a, a member of the helix-turn- helix-like family proteins [70], and coregulated key genes involved in aNSC di fferentiation [58]. Moreover, Tet3 plays critical roles in neural progenitor cell maintenance [71] but is not required for NSC fate [72]. However, how Tet proteins interact with cofactors to regulate target genes responsible for the proliferation and di fferentiation of NSCs remains unclear. The possible regulative mechanisms are proposed in Figure 1.

5. Abnormal 5hmC and Neurological Diseases The growing evidences demonstrate that 5hmC has high abundance in the brain and play a critical role to in the main- tenance of normal neurodevelopment and functions of cen- tral nervous system. Thus, accumulating evidences showed that abnormal 5hmC modi fications are involved the patho- physiology of di fferent neurological diseases.

5.1. Alzheimer ’s Disease (AD). Alzheimer’s disease (AD) is one of the most common age-related neurodegenerative dis- orders in the central nervous system, characterized by pro- gressive cognitive decline and loss of neuronal cells [73].

The pathogenesis of AD has yet to be de fined, but there are evidences to support its genetic abnormalities, such as the mutations in β-amyloid precursor gene and presenilin1/2.

Previous study has shown that AD is associated with DNA methylation [74]. It has been found that levels of 5mC and DNMT in neurons are reduced in patients with AD [74].

At the same time, 5hmC level was reported to decrease in the hippocampal tissue of patients with AD [75]. However, a study has shown that brain 5mC and 5hmC levels increased in patients with AD [76]. The reasons for this inconsistency need to be further investigated. In APP-presenilin1 double transgenic mice, 5hmC abundance in di fferent brain regions showed di fferential response to the pathogenesis [77].

Further gene ontology analyses indicated that di fferential hydroxymethylation region- (DhMR-) associated genes are highly enriched in multiple signaling pathways involving neuronal development/di fferentiation [77], suggesting that DNA 5hmC modi fication is an epigenetic modifi er on neuro- genesis or NSC di fferentiation in aging or AD [78]. Interest- ingly, Tet1 is found to decrease in the hippocampus of patients with AD [79]. Tet1 knockout mice show impaired hippocampal neurogenesis as well as learning and memory defects [55, 80]. Therefore, Tet1 functions as a critical enzyme to regulate 5hmC modi fications on those genes related to the proliferation and di fferentiation of NSCs and further promotes neurogenesis in adult brains.

5.2. Huntington ’s Disease (HD). HD is an autosomal domi- nant disorder characterized by chorea, dystonia, slow and unexpected decline in cognitive function, and mental disor- ders [81]. At present, Huntington gene exon CAG repeats are considered as the major cause that leads to abnormal accumulation of the first amino acid polyglutamine in hun- tingtin proteins. Despite extensive research, the pathogenesis of neurodegeneration in HD is still unknown. ADORA2A gene encodes an adenylate A2A receptor, a G protein- coupled receptor that is highly expressed in the normal basal ganglia and is severely reduced in HD [82]. Recent studies have shown that HD results in an increase of 5mC expression and a decrease of 5hmC expression at the 5 ′ -UTR end of the ADORA2A gene compared with the same age group [83].

Except for the decreased of ADORA2A gene 5hmC modifi ca- tion, a signi ficant decrease of global 5hmC modifi cation is found in HD mice with 128 CAG repeats, indicating the involvement of 5hmC in the pathogenesis of HD and a novel epigenetic marker in HD [82]. Further 5hmC pro filing anal- ysis indicates that most genes with di fferentially hydroxy- methylated regions are highly related to the pathological changes in HD, suggesting that gene 5hmC modi fications are involved in the regulation of neurogenesis, neuronal function, and survival in HD brain [82]. Because previous studies have shown the abnormal neurogenesis in HD [84], aberrant epigenetic regulation on relevant genes may impair the neurogenesis in brains with HD. Recent study demon- strated that targeting histone modi fication to downregulate the key genes for the pathology of HD causes bene ficial e ff ects in a Drosophila model of HD [85]. Therefore, the modulation of 5hmC signature in HD may be an e ffective strategy to ameliorate the symptoms of HD.

5.3. Rett Syndrome. Rett syndrome is considered as an inher- ited disease characterized by progressive mental decline, autistic behavior, ataxia, and anxiety in the early life of those who suff er from the disease. The etiology and genetic pattern of this disease remain unknown. The primary cause of Rett syndrome is caused by methyl CpG binding protein 2 (MeCP2) gene mutations that result in loss of function of MeCP2 [86]. Because brains have the highest expression of MeCP2, MeCP2 functional de ficiency causes neurological diseases such as Rett syndrome [87]. Recent study showed th at MeCP2 was identi fied as the major 5hmc binding pro- tein in the brain to facilitate gene expression by organizing the chromatin [88]. Previous study showed a reverse correla- tion between MeCP2 and 5hmC level, suggesting that MeCP2 binds to 5mC blocking the conversion of 5mC to 5hmC [67]. MeCP2 mutations such as R133C (an MeCP2 residue mutated in Rett syndrome) preferentially abolish its binding ability to 5hmC and account for the role of 5hmC in the pathophysiology of Rett syndrome, supporting that 5hmC and MeCP2 constitute an epigenetic regulation com- plex to control cell di fferentiation or chromatin structure [88]. Recent studies have shown that MeCP2 is required for brain development and neuronal di fferentiation by inhibiting the ID1/Her2 (the zebra fish ortholog of mammalian Hes5) axis in zebra fish because genetic depletion of MeCP2 inhib- ited neuronal di fferentiation but its overexpression promoted neuronal di fferentiation [89]. However, it is still unclear whether the blocking of MeCP2 binding to 5hmC is respon- sible for neuronal di fferentiation in Rett syndrome, as awaits more investigations. 5 Stem Cells International 5.4. Major Depressive Disorders (MDD).The high morbidity and suicide of depression has become a major health concern in the world [90]. However, the pathogenesis of MDD remains unclear. So far, genetic and environmental factors are considered to interact and participate in the MDD, in which environmental factors mainly a ffect gene transcription and expression through epigenetic modi fication. DNA meth- ylation is considered a major epigenetic modi fication from environmental stress [91]. 5hmC functions as a new DNA demethylation mechanism, however, its role in depressive disorders is unclear. Epigenetic 5hmC modi fication, to some extent, provides a possible mechanism for explaining envi- ronmental factors that a ffect gene expression. Recent reports showed that patients with MDD had decreased gray matter volume and white matter integrity in the hippocampus [92]. In addition, Bansal et al. found structural changes in the cerebral cortex of patients with MDD, indicating that thickening of the cerebral cortex is a compensatory nerve growth response [93]. Recent studies have also provided evi- dence that Tet1 knockout showed antidepressive phenotypes by a ffecting neurogenesis in the hippocampus [94]. There- fore, Tet proteins-mediated 5hmC modi fications on depression-related genes are involved the regulation of neu- rogenesis in the mechanisms of MDD. 6. Conclusions Epigenetic modi fication is likely to be the collective response to changes in environmental factors as a means of cells or organisms to mitigate the adverse e ffects [95]. The dynamic changes of methylation (5mC) and demethylation (5hmC) in DNA could a ffect its structure as well as the functions of genes and further lead to di fferent kinds of diseases. Recent advances on 5hmC modi fication have demonstrated that Tet proteins and Tet-mediated 5hmC play important roles in the proliferation and di fferentiation of NSCs. However, it is unclear how Tet protein, Tet-interacting factors, and DNA 5hmC in target genes interplay and regulate the devol- vement of NSCs. These need more investigations in the future. Recently, DNA N6-Methyldeoxyadenosine (6 mA) is Fe2+ Fe2+ Fe2+ Tets Tets Tets TFs TFs TFs Motif Nucleus Promoter/TSS of genes Promoter/TSS of genes 5hmC binding proteins Environmental stimuli Proliferation and di erentiation of NSC Promote gene transcription -KG -KG -KG 5mC 5hmC Figure 1: Tet proteins and 5-hmC mediated regulation of NSC proliferation and di fferentiation. Under the conditions of environmental stimuli, some transcriptional factors (TFs) such as FOXO3a enhance the a ffinity to Tet proteins along with cofactors of Tet enzymes including α-KG and Fe 2+to form a functional complex. By binding to DNA motifs of the targeting genes, the TFs guide the Tet enzymes to catalyze the conversion of 5mC to 5hmC. Generation of 5hmC facilitates the recruitment of the 5hmC binding proteins or other factors to enhance the transcription of targeting genes, thereby regulating the proliferation and di fferentiation of NSCs. 6 Stem Cells International emerging as a new DNA modification and plays an important role in the regulation of the proliferation and di fferentiation of NSCs [96–98]. The interaction or crosstalking of DNA 5hmC modifi cation and 6 mA modifi cation will be an inter- esting topic. Considering the critical role of neuronal stem cells in the neurological diseases, targeting epigenetic regula- tion, especially on DNA 5hmC modi fication, is a promising strategy for the treatment of these neurological diseases.

Disclosure Hang Zhou and Bin Wang are co-fi rst authors.

Conflicts of Interest The authors declare that they have no confl icts of interest.

Acknowledgments This study was supported by the grants from National Key R&D Program of China (2017YFE0103700), Natural Science Foundation of Jiangsu Province (BK20141281), Special Foundation Project on the Prospective Study of Social Development in Jiangsu Province (BE2013911), Jiangsu Six Categories of Talent Summit Fund (WSW-133), Social Development of Science and Technology Research Project in Yangzhou (YZ2011082), and Jiangsu Province 333 talent Project (BRA2016159).

References [1] L. Bonetta,“Gene expression: an expression of interest, ” Nature, vol. 440, no. 7088, pp. 1233 –1237, 2006.

[2] X. Li and M. G. Rosenfeld, “Transcription: origins of licens- ing control, ”Nature, vol. 427, no. 6976, pp. 687-688, 2004.

[3] Z. Shi, K. Fujii, K. M. Kovary et al., “Heterogeneous ribo- somes preferentially translate distinct subpools of mRNAs genome-wide, ”Molecular Cell , vol. 67, no. 1, pp. 71–83.e7, 2017.

[4] S. E. Dwi Putra, C. Reichetzeder, M. Meixner, K. Liere, T. Slowinski, and B. Hocher, “DNA methylation of the gluco- corticoid receptor gene promoter in the placenta is associated with blood pressure regulation in human pregnancy, ”Journal of Hypertension , vol. 35, no. 11, pp. 2276 –2286, 2017.

[5] S. Kimmins and P. Sassone-Corsi, “Chromatin remodelling and epigenetic features of germ cells, ”Nature, vol. 434, no. 7033, pp. 583–589, 2005.

[6] G. I. Evan and K. H. Vousden, “Proliferation, cell cycle and apoptosis in cancer, ”Nature, vol. 411, no. 6835, pp. 342 – 348, 2001.

[7] A. Chhabra, “Derivation of human induced pluripotent stem cell (iPSC) lines and mechanism of pluripotency: historical perspective and recent advances, ”Stem Cell Reviews and Reports , vol. 13, no. 6, pp. 757 –773, 2017.

[8] G. Caocci, M. G. Oro fino, A. Vacca et al., “Long-term survival of beta thalassemia major patients treated with hematopoietic stem cell transplantation compared with survival with con- ventional treatment, ”American Journal of Hematology , vol. 92, no. 12, pp. 1303 –1310, 2017.

[9] B. A. Reynolds and S. Weiss, “Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system, ”Science , vol. 255, no. 5052, pp. 1707 –1710, 1992.

[10] S. E. Marsh, S. T. Yeung, M. Torres et al., “HuCNS-SC human NSCs fail to di fferentiate, form ectopic clusters, and provide no cognitive bene fits in a transgenic model of Alzheimer ’s disease, ”Stem Cell Reports, vol. 8, no. 2, pp. 235 –248, 2017.

[11] Y. H. Rhee, T. H. Kim, A. Y. Jo et al., “LIN28A enhances the therapeutic potential of cultured neural stem cells in a Parkinson ’s disease model, ”Brain , vol. 139, no. 10, pp. 2722 –2739, 2016.

[12] K. Wiatr, W. J. Szlachcic, M. Trzeciak, M. Figlerowicz, and M. Figiel, “Huntington disease as a neurodevelopmental dis- order and early signs of the disease in stem cells, ”Molecular Neurobiology , pp. 1–21, 2017, inpress.

[13] A. Ramsawhook, L. Lewis, B. Coyle, and A. Ruzov, “Medullo- blastoma and ependymoma cells display increased levels of 5-carboxylcytosine and elevated TET1 expression, ”Clinical Epigenetics , vol. 9, no. 1, 2017.

[14] J. Winkelmann, L. Lin, B. Schormair et al., “Mutations in DNMT1 cause autosomal dominant cerebellar ataxia, deaf- ness and narcolepsy, ”Human Molecular Genetics , vol. 21, no. 10, pp. 2205 –2210, 2012.

[15] L. Q. Cheng, Z. Q. Zhang, H. Z. Chen, and D. P. Liu, “Epige- netic regulation in cell senescence, ”Journal of Molecular Medicine , vol. 95, no. 12, pp. 1257 –1268, 2017.

[16] Y. C. Sun, Y. Y. Wang, W. Ge, S. F. Cheng, P. W. Dyce, and W. Shen, “Epigenetic regulation during the di fferentiation of stem cells to germ cells, ”Oncotarget , vol. 8, no. 34, pp. 57836 –57844, 2017.

[17] L. Luo, W. J. Chen, J. Q. Yin, and R. X. Xu, “EID3 directly associates with DNMT3A during transdi fferentiation of human umbilical cord mesenchymal stem cells to NPC-like cells, ”Scienti fic Reports , vol. 7, article 40463, 2017.

[18] W. Feng, M. A. Khan, P. Bellvis et al., “The chromatin remo- deler CHD7 regulates adult neurogenesis via activation of SoxC transcription factors, ”Cell Stem Cell , vol. 13, no. 1, pp. 62–72, 2013.

[19] M. Strigini and F. Leulier, “The role of the microbial environ- ment in drosophila post-embryonic development, ”Develop- mental and Comparative Immunology , vol.

64, pp. 39–52, 2016.

[20] N. Kuzumaki, D. Ikegami, R. Tamura et al., “Hippocampal epigenetic modifi cation at the brain-derived neurotrophic factor gene induced by an enriched environment, ”Hippo- campus , vol. 21, no. 2, pp. 127 –132, 2011.

[21] F. Zenk, E. Loeser, R. Schiavo, F. Kilpert, O. Bogdanovic, and N. Iovino, “Germ line-inherited H3K27me3 restricts enhancer function during maternal-to-zygotic transition, ” Science , vol. 357, no. 6347, pp. 212 –216, 2017.

[22] G. A. Challen, D. Sun, M. Jeong et al., “Dnmt3a is essential for hematopoietic stem cell di fferentiation, ”Nature Genetics , vol. 44, no. 1, pp. 23 –31, 2011.

[23] N. S. Christophersen and K. Helin, “Epigenetic control of embryonic stem cell fate, ”The Journal of Experimental Medicine , vol. 207, no. 11, pp. 2287 –2295, 2010.

[24] M. Kaneda, M. Okano, K. Hata et al., “Essential role for de novo DNA methyltransferase Dnmt3a in paternal and mater- nal imprinting, ”Nature, vol. 429, no. 6994, pp. 900–903, 2004.

[25] Y. Cai, H. C. Tsai, R. C. Yen et al., “Critical threshold levels of DNA methyltransferase 1 are required to maintain DNA 7 Stem Cells International methylation across the genome in human cancer cells,” Genome Research , vol. 27, no. 4, pp. 533 –544, 2017.

[26] R. C. Laker and J. G. Ryall, “DNA methylation in skeletal muscle stem cell speci fication, proliferation, and di fferenti- ation, ”Stem Cells International , vol. 2016, Article ID 5725927, 9 pages, 2016.

[27] V. Ramesh, E. Bayam, F. M. Cernilogar et al., “Loss of Uhrf1 in neural stem cells leads to activation of retroviral elements and delayed neurodegeneration, ”Genes & Development , vol. 30, no. 19, pp. 2199 –2212, 2016.

[28] E. M. Jobe, Y. Gao, B. E. Eisinger et al., “Methyl-CpG- binding protein MBD1 regulates neuronal lineage commit- ment through maintaining adult neural stem cell identity, ” The Journal of Neuroscience , vol. 37, no. 3, pp. 523–536, 2017.

[29] V. R. Liyanage, R. M. Zachariah, J. R. Davie, and M. Rastegar, “Ethanol deregulates Mecp2/MeCP2 in di fferentiating neural stem cells via interplay between 5-methylcytosine and 5- hydroxymethylcytosine at the Mecp2 regulatory elements,” Experimental Neurology , vol. 265, pp. 102–117, 2015.

[30] S. F. Chaudry and T. J. Chevassut, “Epigenetic Guardian: a review of the DNA methyltransferase DNMT3A in acute myeloid Leukaemia and clonal Haematopoiesis, ”BioMed Research International , vol. 2017, Article ID 5473197, 13 pages, 2017.

[31] F. C. Zhou, Y. Balaraman, M. Teng, Y. Liu, R. P. Singh, and K. P. Nephew, “Alcohol alters DNA methylation patterns and inhibits neural stem cell di fferentiation, ”Alcoholism, Clinical and Experimental Research , vol. 35, no. 4, pp. 735– 746, 2011.

[32] B. Srinageshwar, P. Maiti, G. L. Dunbar, and J. Rossignol, “Role of epigenetics in stem cell proliferation and di fferentia- tion: implications for treating neurodegenerative diseases, ” International Journal of Molecular Sciences , vol. 17, no. 2, p. 199, 2016.

[33] S. B. Foti, A. Chou, A. D. Moll, and A. J. Roskams, “HDAC inhibitors dysregulate neural stem cell activity in the postna- tal mouse brain, ”International Journal of Developmental Neuroscience , vol. 31, no. 6, pp. 434 –447, 2013.

[34] R. Soriano-Canton, A. Perez-Villalba, J. M. Morante-Redolat et al., “Regulation of the p19(Arf )/p53 pathway by histone acetylation underlies neural stem cell behavior in senescence- prone SAMP8 mice, ”Aging Cell , vol. 14, no. 3, pp. 453– 462, 2015.

[35] G. Sun, K. Alzayady, R. Stewart et al., “Histone demethylase LSD1 regulates neural stem cell proliferation, ”Molecular and Cellular Biology , vol. 30, no. 8, pp. 1997 –2005, 2010.

[36] A. Adamo, B. Sese, S. Boue et al., “LSD1 regulates the balance between self-renewal and di fferentiation in human embry- onic stem cells, ”Nature Cell Biology , vol. 13, no. 6, pp. 652 – 659, 2011.

[37] S. Bian and T. Sun, “Functions of noncoding RNAs in neural development and neurological diseases, ”Molecular Neurobi- ology, vol. 44, no. 3, pp. 359 –373, 2011.

[38] F. Gao, Y. F. Zhang, Z. P. Zhang et al., “miR-342-5p regulates neural stem cell proliferation and di fferentiation downstream to notch signaling in mice, ”Stem Cell Reports , vol. 8, no. 4, pp. 1032 –1045, 2017.

[39] M. A. Lopez-Ramirez and S. Nicoli, “Role of miRNAs and epigenetics in neural stem cell fate determination, ”Epige- netics , vol. 9, no. 1, pp. 90–100, 2014. [40] A. L. Morgado, C. M. Rodrigues, and S. Sola, “MicroRNA- 145 regulates neural stem cell di fferentiation through the Sox2-Lin28/let-7 signaling pathway, ”Stem Cells , vol. 34, no. 5, pp. 1386 –1395, 2016.

[41] C. Liu, Z. Q. Teng, N. J. Santistevan et al., “Epigenetic regulation of miR-184 by MBD1 governs neural stem cell proliferation and di fferentiation, ”Cell Stem Cell , vol. 6, no. 5, pp. 433 –444, 2010.

[42] W. Yan, Z. Y. Chen, J. Q. Chen, and H. M. Chen, “BMP2 promotes the di fferentiation of neural stem cells into dopami- nergic neurons in vitro via miR-145-mediated upregulation of Nurr1 expression, ”American Journal of Translational Research , vol. 8, no. 9, pp. 3689 –3699, 2016.

[43] N. Xu, T. Papagiannakopoulos, G. Pan, J. A. Thomson, and K. S. Kosik, “ MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells, ”Cell , vol. 137, no. 4, pp. 647 –658, 2009.

[44] Y. Zhang, M. S. Kim, B. Jia et al., “Hypothalamic stem cells control ageing speed partly through exosomal miRNAs, ” Nature, vol. 548, no. 7665, pp. 52 –57, 2017.

[45] X. Wu and Y. Zhang, “TET-mediated active DNA demethyl- ation: mechanism, function and beyond, ”Nature Reviews.

Genetics , vol. 18, no. 9, pp. 517 –534, 2017.

[46] M. Tahiliani, K. P. Koh, Y. Shen et al., “Conversion of 5- methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1, ”Science , vol. 324, no. 5929, pp. 930–935, 2009.

[47] S. Ito, A. C. D'Alessio, O. V. Taranova, K. Hong, L. C. Sowers, and Y. Zhang, “Role of Tet proteins in 5mC to 5hmC conver- sion, ES-cell self-renewal and inner cell mass speci fication, ” Nature, vol. 466, no. 7310, pp. 1129 –1133, 2010.

[48] K. P. Koh, A. Yabuuchi, S. Rao et al., “Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage speci fi- cation in mouse embryonic stem cells, ”Cell Stem Cell , vol. 8, no. 2, pp. 200 –213, 2011.

[49] K. Williams, J. Christensen, M. T. Pedersen et al., “TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity, ”Nature, vol. 473, no. 7347, pp. 343 – 348, 2011.

[50] Y. Xu, F. Wu, L. Tan et al., “Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells, ”Molecular Cell , vol. 42, no. 4, pp. 451–464, 2011.

[51] T. P. Gu, F. Guo, H. Yang et al., “The role of Tet3 DNA diox- ygenase in epigenetic reprogramming by oocytes, ”Nature , vol. 477, no. 7366, pp. 606–610, 2011.

[52] M. Ko, J. An, H. S. Bandukwala et al., “Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX, ”Nature, vol. 497, no. 7447, pp. 122 – 126, 2013.

[53] S. Hino, S. Kishida, T. Michiue et al., “Inhibition of the Wnt signaling pathway by Idax, a novel Dvl-binding protein, ” Molecular and Cellular Biology , vol. 21, no. 1, pp. 330–342, 2001.

[54] S. Knappskog, L. M. Myklebust, C. Busch et al., “RINF (CXXC5) is overexpressed in solid tumors and is an unfavor- able prognostic factor in breast cancer, ”Annals of Oncology , vol. 22, no. 10, pp. 2208 –2215, 2011.

[55] R. R. Zhang, Q. Y. Cui, K. Murai et al., “Tet1 regulates adult hippocampal neurogenesis and cognition, ”Cell Stem Cell , vol. 13, no. 2, pp. 237 –245, 2013. 8 Stem Cells International [56] K. Moran-Crusio, L. Reavie, A. Shih et al.,“Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation, ”Cancer Cell , vol. 20, no. 1, pp. 11 –24, 2011.

[57] S. Kriaucionis and N. Heintz, “The nuclear DNA base 5- hydroxymethylcytosine is present in Purkinje neurons and the brain, ”Science , vol. 324, no. 5929, pp. 929-930, 2009.

[58] X. Li, B. Yao, L. Chen et al., “Ten-eleven translocation 2 inter- acts with forkhead box O3 and regulates adult neurogenesis, ” Nature Communications , vol. 8, article 15903, 2017.

[59] L. M. Wheldon, A. Abakir, Z. Ferjentsik et al., “Transient accumulation of 5-carboxylcytosine indicates involvement of active demethylation in lineage speci fication of neural stem cells, ”Cell Reports , vol. 7, no. 5, pp. 1353 –1361, 2014.

[60] L. C. Lewis, P. C. Lo, J. M. Foster et al., “Dynamics of 5-carboxylcytosine during hepatic di fferentiation: potential general role for active demethylation by DNA repair in line- age speci fication, ”Epigenetics , vol. 12, no. 4, pp. 277 –286, 2017.

[61] S. Ito, L. Shen, Q. Dai et al., “Tet proteins can convert 5- methylcytosine to 5-formylcytosine and 5-carboxylcytosine, ” Science , vol. 333, no. 6047, pp. 1300 –1303, 2011.

[62] B. E. Bernstein, A. Meissner, and E. S. Lander, “The mamma- lian epigenome, ”Cell , vol. 128, no. 4, pp. 669–681, 2007.

[63] M. Bibikova, L. C. Laurent, B. Ren, J. F. Loring, and J. B. Fan, “Unraveling epigenetic regulation in embryonic stem cells, ” Cell Stem Cell , vol. 2, no. 2, pp. 123 –134, 2008.

[64] D. Thomas and M. Kansara, “Epigenetic modifi cations in osteogenic di fferentiation and transformation, ”Journal of Cellular Biochemistry, vol. 98, no. 4, pp. 757 –769, 2006.

[65] S. Barrand and P. Collas, “Chromatin states of core pluripotency-associated genes in pluripotent, multipotent and di fferentiated cells, ”Biochemical and Biophysical Research Communications , vol. 391, no. 1, pp. 762 –767, 2010.

[66] N. Ballas, C. Grunseich, D. D. Lu, J. C. Speh, and G. Mandel, “ REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis, ”Cell , vol. 121, no. 4, pp. 645 –657, 2005.

[67] K. E. Szulwach, X. Li, Y. Li et al., “5-hmC-mediated epigenetic dynamics during postnatal neurodevelopment and aging, ” Nature Neuroscience , vol. 14, no. 12, pp. 1607 –1616, 2011.

[68] J. P. Etchegaray, L. Chavez, Y. Huang et al., “The histone dea- cetylase SIRT6 controls embryonic stem cell fate via TET- mediated production of 5-hydroxymethylcytosine, ”Nature Cell Biology , vol. 17, no. 5, pp. 545 –557, 2015.

[69] M. A. Hahn, R. Qiu, X. Wu et al., “Dynamics of 5- hydroxymethylcytosine and chromatin marks in mammalian neurogenesis, ”Cell Reports , vol. 3, no. 2, pp. 291 –300, 2013.

[70] S. Hannenhalli and K. H. Kaestner, “The evolution of Fox genes and their role in development and disease, ”Nature Reviews Genetics , vol. 10, no. 4, pp. 233 –240, 2009.

[71] R. Bose, S. Spulber, P. Kilian et al., “Tet3 mediates stable glucocorticoid-induced alterations in DNA methylation and Dnmt3a/Dkk1 expression in neural progenitors,” Cell Death & Disease , vol. 6, no. 6, article e1793, 2015.

[72] T. Li, D. Yang, J. Li, Y. Tang, J. Yang, and W. Le, “Critical role of Tet3 in neural progenitor cell maintenance and terminal di fferentiation, ”Molecular Neurobiology , vol. 51, no. 1, pp. 142 –154, 2015.

[73] A. Iatrou, G. Kenis, B. P. Rutten, K. Lunnon, and D. L. van den Hove, “Epigenetic dysregulation of brainstem nuclei in the pathogenesis of Alzheimer ’s disease: looking in the correct place at the right time?, ”Cellular and Molecular Life Sciences , vol. 74, no. 3, pp. 509 –523, 2017.

[74] S. W. Bihaqi, A. Schumacher, B. Maloney, D. K. Lahiri, and N. H. Zawia, “Do epigenetic pathways initiate late onset Alzheimer disease (LOAD): towards a new paradigm, ”Cur- rent Alzheimer Research , vol. 9, no. 5, pp. 574–588, 2012.

[75] L. Chouliaras, D. Mastroeni, E. Delvaux et al., “Consistent decrease in global DNA methylation and hydroxymethyla- tion in the hippocampus of Alzheimer’ s disease patients,” Neurobiology of Aging , vol. 34, no. 9, pp. 2091 –2099, 2013.

[76] J. S. Rao, V. L. Keleshian, S. Klein, and S. I. Rapoport, “Epige- netic modi fications in frontal cortex from Alzheimer ’s disease and bipolar disorder patients, ”Translational Psychiatry, vol. 2, no. 7, article e132, 2012.

[77] L. Shu, W. Sun, L. Li et al., “Genome-wide alteration of 5-hydroxymenthylcytosine in a mouse model of Alzheimer’ s disease, ”BMC Genomics, vol. 17, no. 1, 2016.

[78] A. I. Bernstein, Y. Lin, R. C. Street et al., “5-Hydroxymethyla- tion-associated epigenetic modi fiers of Alzheimer ’s disease modulate Tau-induced neurotoxicity, ”Human Molecular Genetics , vol. 25, no. 12, pp. 2437 –2450, 2016.

[79] M. A. Bradley-Whitman and M. A. Lovell, “Epigenetic changes in the progression of Alzheimer ’s disease, ”Mecha- nisms of Ageing and Development , vol. 134, no. 10, pp. 486– 495, 2013.

[80] A. Rudenko, M. M. Dawlaty, J. Seo et al., “Tet1 is critical for neuronal activity-regulated gene expression and memory extinction, ”Neuron, vol. 79, no. 6, pp. 1109 –1122, 2013.

[81] S. Horvath, P. Langfelder, S. Kwak et al., “Huntington ’s dis- ease accelerates epigenetic aging of human brain and disrupts DNA methylation levels, ”Aging , vol. 8, no. 7, pp. 1485 –1512, 2016.

[82] F. Wang, Y. Yang, X. Lin et al., “Genome-wide loss of 5-hmC is a novel epigenetic feature of Huntington ’s disease, ”Human Molecular Genetics , vol. 22, no. 18, pp. 3641 –3653, 2013.

[83] I. Villar-Menendez, M. Blanch, S. Tyebji et al., “Increased 5-methylcytosine and decreased 5-hydroxymethylcytosine levels are associated with reduced striatal A2AR levels in Huntington ’s disease, ”Neuromolecular Medicine , vol. 15, no. 2, pp. 295 –309, 2013.

[84] B. Winner, Z. Kohl, and F. H. Gage, “Neurodegenerative disease and adult neurogenesis, ”The European Journal of Neuroscience , vol. 33, no. 6, pp. 1139 –1151, 2011.

[85] M. Vashishtha, C. W. Ng, F. Yildirim et al., “Targeting H3K4 trimethylation in Huntington disease, ”Proceedings of the National Academy of Sciences of the United States of America , vol. 110, no. 32, pp. E3027 –E3036, 2013.

[86] R. E. Amir, I. B. Van den Veyver, M. Wan, C. Q. Tran, U. Francke, and H. Y. Zoghbi, “Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG- binding protein 2, ”Nature Genetics , vol. 23, no. 2, pp. 185 – 188, 1999.

[87] M. D. Shahbazian, B. Antalff y, D. L. Armstrong, and H. Y.

Zoghbi, “Insight into Rett syndrome: MeCP2 levels display tissue- and cell-speci ficdi fferences and correlate with neuro- nal maturation,” Human Molecular Genetics , vol. 11, no. 2, pp. 115–124, 2002.

[88] M. Mellen, P. Ayata, S. Dewell, S. Kriaucionis, and N. Heintz, “MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system, ”Cell , vol. 151, no. 7, pp. 1417 –1430, 2012. 9 Stem Cells International [89] H. Gao, Y. Bu, Q. Wu et al.,“Mecp2 regulates neural cell dif- ferentiation by suppressing the Id1 to Her2 axis in zebra fish, ” Journal of Cell Science , vol. 128, no. 12, pp. 2340 –2350, 2015.

[90] K. Smith, “Mental health: a world of depression, ”Nature, vol. 515, no. 7526, p. 181, 2014.

[91] E. Pishva, B. P. F. Rutten, and D. van den Hove, “DNA methylation in major depressive disorder, ”Advances in Experimental Medicine and Biology , vol. 978, pp. 185–196, 2017.

[92] X. Shen, L. M. Reus, S. R. Cox et al., “Subcortical volume and white matter integrity abnormalities in major depressive disorder: findings from UK Biobank imaging data, ”Scienti fic Reports , vol. 7, no. 1, p. 5547, 2017.

[93] R. Bansal, D. J. Hellerstein, and B. S. Peterson, “Evidence for neuroplastic compensation in the cerebral cortex of persons with depressive illness, ”Molecular Psychiatry, vol. 23, no. 2, pp. 375 –383, 2018.

[94] J. Feng, C. J. Pena, I. Purushothaman et al., “Tet1 in nucleus accumbens opposes depression- and anxiety-like behaviors, ” Neuropsychopharmacology , vol. 42, no. 8, pp. 1657 –1669, 2017.

[95] M. Munzel, D. Globisch, and T. Carell, “5-Hydroxymethylcy- tosine, the sixth base of the genome, ”Angewandte Chemie (International Ed. in English) , vol. 50, no. 29, pp. 6460– 6468, 2011.

[96] Y. Fu, G. Z. Luo, K. Chen et al., “N6-methyldeoxyadenosine marks active transcription start sites in Chlamydomonas,” Cell , vol. 161, no. 4, pp. 879 –892, 2015.

[97] B. Yao, Y. Cheng, Z. Wang et al., “DNA N6-methyladenine is dynamically regulated in the mouse brain following environ- mental stress, ”Nature Communications , vol. 8, no. 1, p. 1122, 2017.

[98] K. J. Yoon, F. R. Ringeling, C. Vissers et al., “Temporal con- trol of mammalian cortical neurogenesis by m 6A methyla- tion, ”Cell , vol. 171, no. 4, pp. 877–889.e17, 2017.

[99] M. M. Dawlaty, K. Ganz, B. E. Powell et al., “Tet1 is dispens- able for maintaining pluripotency and its loss is compatible with embryonic and postnatal development, ”Cell Stem Cell , vol. 9, no. 2, pp. 166 –175, 2011.

[100] X. Jiang, C. Hu, K. Ferchen et al., “Targeted inhibition of STAT/TET1 axis as a therapeutic strategy for acute myeloid leukemia, ”Nature Communications , vol. 8, no. 1, p. 2099, 2017.

[101] H. L. Fu, Y. Ma, L. G. Lu et al., “TET1 exerts its tumor suppressor function by interacting with p53-EZH2 pathway in gastric cancer, ”Journal of Biomedical Nanotechnology , vol. 10, no. 7, pp. 1217 –1230, 2014.

[102] Q. Yang, K. Wu, M. Ji et al., “Decreased 5- hydroxymethylcytosine (5-hmC) is an independent poor prognostic factor in gastric cancer patients, ”Journal of Bio- medical Nanotechnology , vol. 9, no. 9, pp. 1607–1616, 2013.

[103] M. Deng, R. Zhang, Z. He et al., “TET-mediated sequestration of miR-26 drives EZH2 expression and gastric carcinogene- sis, ”Cancer Research , vol. 77, no. 22, pp. 6069 –6082, 2017.

[104] M. Sun, C. X. Song, H. Huang et al., “HMGA2/TET1/ HOXA9 signaling pathway regulates breast cancer growth and metastasis, ”Proceedings of the National Academy of Sciences of the United States of America , vol. 110, no. 24, pp. 9920 –9925, 2013.

[105] P. Seritrakul and J. M. Gross, “Tet-mediated DNA hydroxy- methylation regulates retinal neurogenesis by modulating cell-extrinsic signaling pathways,” PLoS Genetics, vol. 13, no. 9, article e1006987, 2017.

[106] J. S. Ha, D. S. Jeon, J. R. Kim, N. H. Ryoo, and J. S. Suh, “Anal- ysis of the Ten-Eleven Translocation 2 (TET2) gene mutation in myeloproliferative neoplasms, ”Annals of Clinical and Laboratory Science , vol. 44, no. 2, pp. 173 –179, 2014.

[107] P. Jain, S. Verstovsek, W. Wang et al., “DNMT3A, TET2, and JAK2 mutations in polycythemia vera following long-term remission of secondary acute myeloid leukemia,” Leukemia & Lymphoma , vol. 57, no. 8, pp. 1969 –1973, 2016.

[108] E. Chen, R. K. Schneider, L. J. Breyfogle et al., “Distinct eff ects of concomitant Jak2V617F expression and Tet2 loss in mice promote disease progression in myeloproliferative neoplasms,” Blood, vol. 125, no. 2, pp. 327–335, 2015.

[109] C. G. Lian, Y. Xu, C. Ceol et al., “Loss of 5- hydroxymethylcytosine is an epigenetic hallmark of mela- noma, ”Cell , vol. 150, no. 6, pp. 1135 –1146, 2012.

[110] A. Inoue, L. Shen, S. Matoba, and Y. Zhang, “Haploinsu ffi- ciency, but not defective paternal 5mC oxidation, accounts for the developmental defects of maternal Tet3 knockouts, ” Cell Reports , vol. 10, no. 4, pp. 463 –470, 2015.

[111] X. Li, W. Wei, Q. Y. Zhao et al., “Neocortical Tet3-mediated accumulation of 5-hydroxymethylcytosine promotes rapid behavioral adaptation, ”Proceedings of the National Academy of Sciences of the United States of America , vol. 111, no. 19, pp. 7120 –7125, 2014.

[112] X. Li, X. Yue, W. A. Pastor et al., “Tet proteins infl uence the balance between neuroectodermal and mesodermal fate choice by inhibiting Wnt signaling, ”Proceedings of the National Academy of Sciences of the United States of America , vol. 113, no. 51, pp. E8267 –E8276, 2016.

[113] M. M. Dawlaty, A. Breiling, T. Le et al., “Combined de ficiency of Tet1 and Tet2 causes epigenetic abnormalities but is com- patible with postnatal development, ”Developmental Cell , vol. 24, no. 3, pp. 310 –323, 2013.

[114] X. Zhu, D. Girardo, E. E. Govek et al., “Role of Tet1/3 genes and chromatin remodeling genes in cerebellar circuit forma- tion, ”Neuron, vol. 89, no. 1, pp. 100 –112, 2016.

[115] J. Kang, M. Lienhard, W. A. Pastor et al., “Simultaneous dele- tion of the methylcytosine oxidases Tet1 and Tet3 increases transcriptome variability in early embryogenesis, ”Proceed- ings of the National Academy of Sciences of the United States of America , vol. 112, no. 31, pp. E4236 –E4245, 2015.

[116] H. Q. Dai, B. A. Wang, L. Yang et al., “TET-mediated DNA demethylation controls gastrulation by regulating Lefty- Nodal signalling, ”Nature, vol. 538, no. 7626, pp. 528 –532, 2016. 10 Stem Cells International Submit your manuscripts athttps://www.hindawi.com Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Anatomy Research International Peptides International Journal of Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Hindawi Publishing Corporation http://www.hindawi.com International Journal of Volume 2014 Zoology Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Molecular Biology International Genomics International Journal of Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 The Scienti pc World Journal Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Hindawi Publishing Corporation http://www.hindawi.comVolume 2014Bioinformatics Advances in Marine Biology Journal of Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 Hindawi Publishing Corporation http://www.hindawi.comVolume 2014Signal TransductionJournal of Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 BioMed Research International Evolutionary BiologyInternational Journal of Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 Biochemistr y Research International ArchaeaHindawi Publishing Corporation http://www.hindawi.comVolume 2014 Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 Genetics Research International Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Advances in Vir ol og y Hindawi Publishing Corporationhttp://www.hindawi.com Nucleic Acids Journal of Volume 2014 Stem Cells International Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 Enzyme Research Hindawi Publishing Corporation http://www.hindawi.comVolume 2014 International Journal of Microbiology Copyright ofStem CellsInternational isthe property ofHindawi Limitedanditscontent may not becopied oremailed tomultiple sitesorposted toalistserv without thecopyright holder's express writtenpermission. However,usersmayprint, download, oremail articles for individual use.