Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Apr 23.
Published in final edited form as: Prog Neurobiol. 2015 Jun 11;131:21–64. doi: 10.1016/j.pneurobio.2015.05.002

The epigenetics of aging and neurodegeneration

Roy Lardenoije a, Artemis Iatrou a, Gunter Kenis a, Konstantinos Kompotis b, Harry WM Steinbusch a, Diego Mastroeni a,c, Paul Coleman c, Cynthia A Lemere d, Patrick R Hof e, Daniel LA van den Hove a,f,#, Bart PF Rutten a,*,#
PMCID: PMC6477921  NIHMSID: NIHMS1012122  PMID: 26072273

Abstract

Epigenetics is a quickly growing field encompassing mechanisms regulating gene expression that do not involve changes in the genotype. Epigenetics is ofincreasing relevance to neuroscience, with epigenetic mechanisms being implicated in brain development and neuronal differentiation, as well as in more dynamic processes related to cognition. Epigenetic regulation covers multiple levels of gene expression; from direct modifications of the DNA and histone tails, regulating the level of transcription, to interactions with messenger RNAs, regulating the level of translation. Importantly, epigenetic dysregulation currently garners much attention as a pivotal player in aging and agerelated neurodegenerative disorders, such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease, where it may mediate interactions between genetic and environmental risk factors, or directly interact with disease-specific pathological factors. We review current knowledge about the major epigenetic mechanisms, including DNA methylation and DNA demethylation, chromatin remodeling and noncoding RNAs, as well as the involvement of these mechanisms in normal aging and in the pathophysiology of the most common neurodegenerative diseases. Additionally, we examine the current state of epigenetics-based therapeutic strategies for these diseases, which either aim to restore the epigenetic homeostasis or skew it to a favorable direction to counter disease pathology. Finally, methodological challenges of epigenetic investigations and future perspectives are discussed.

Keywords: Epigenetics, Aging, Neurodegeneration, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease

1. Epigenetics

Conrad Hal Waddington coined the term “epigenetics” in 1942, an event commonly viewed as the birth of epigenetics as it developed from a phenomenon to an immensely studied branch of science (Choudhuri, 2011). A merger between the terms “genetics” and “epigenesist”, the concept of epigenetics in a way represents the association of two views on development that have been clashing at least since the time of Hippocrates and Aristotle (Muller and Olsson, 2003). Hippocrates proposed what became known as the preformationist view of development; all parts of a mature organism are already present at the embryonic stage, albeit in a miniature stage, and they simply grow during development. Aristotle argued against this preformationist view, providing an alternative explanation that lies at the foundation of the epigenesis concept: embryonic development involves the formation of new parts. After numerous scientific discoveries these views evolved over the centuries. A contemporary preformationist would hold that all that is needed to generate a mature organism is its genetic code, whereas a supporter of epigenesis would argue that the genome only holds the information of building blocks - but that how these are put together depends on environmental influences. The contemporary perspective of “epigenetics” is that of the field of science that studies how changes in gene expression occur without changes in the DNA sequence (Choudhuri, 2011). Such changes can be induced by environmental factors, while some are more programmed, as seen during cell differentiation. As such, these epigenetic alterations can be highly stable, such as those resulting from genetic imprinting, or dynamic such as the epigenetic changes associated with memory. Many Epigenetic modifications can be inherited through mitosis and some have even found to be transgenerational (Handel et al., 2010; Hsieh and Eisch, 2010; Hu et al., 2012; Ma et al., 2010). Thus, whereas genetic alterations usually reflect permanent changes of the DNA sequence, epigenetic changes are mediated through processes that are in principle reversible (Henikoff and Matzke, 1997). While environmental influences can potentially alter the phenotype of an organism by interacting with and by acting on both the genome and epigenome (Liu et al., 2008), the reversible nature of epigenetic changes makes them more suitable as candidates for clinical interventions (Feinberg, 2008). Over the past decade there have been ample studies investigating the contributions of epigenetic modifications to aging and age-related neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD) and Huntington’s disease (HD). The epigenetic machinery covers multiple levels of control, including DNA methylation and hydroxymethylation, chromatin remodeling, and non-coding RNA (ncRNA) regulation (Adwan and Zawia, 2013). See Fig. 1 for a general overview of the epigenetic mechanisms discussed below.

Fig. 1.

Fig. 1.

The three levels of epigenetic regulation. The upper section summarizes DNA methylation and demethylation processes, the middle section summarizes the most important chromatin remodeling processes, and the bottom section summarizes non-coding RNA regulation. Abbreviations: 5-caC, 5-carboxylcytosine; 5-fC, 5-formylcytosine; 5-hmC, 5-hydroxymethylcytosine; 5-hmU, 5-hydroxymethyluracil; 5-mC, 5-methylcytosine; A, acetyl modification; AICDA, activation-induced cytidine deaminase; APOBEC, apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like protein; BER, base excision repair; C, cytosine; DNMT, DNA methyltransferase; H, histone; HDAC, histone deacetylase; HKDM, histone lysine demethylase; HKMT, histone lysine methyltransferase; HRDM, histone arginine demethylase; K, lysine; KAT, lysine acetyltransferase; lincRNA, large intergenic non-coding RNA; M, methyl modification; miRNA, micro RNA; MSK1, mitogen- and stress-activated protein kinase 1; ncRNA, non-coding RNA; P, phosphate modification; piRNA, piwi-interacting RNA; PPT1, protein phosphatase 1; PRMT, protein arginine methyltransferase; R, arginine; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine; snoRNA, small nucleolar RNA; TET, ten-eleven translocation; TDG, thymine DNA glycosylase; tiRNA, transcription initiation RNA; tRNA, transfer RNA.

1.1. DNA methylation

The best characterized epigenetic modification, DNA methyl-ation involves the addition of a methyl group at the 5 position on the pyrimidine ring of cytosines, creating 5-methylcytosine (5-mC) (Razin and Riggs, 1980). These modifications primarily occur at cytosine-phosphate-guanine (CpG) islands. Recently, however, non-CpG methylation has received increased attention (Guo et al., 2013). Apart from cytosines, there are also reports of guanine and adenine methylation, resulting in 7-methylguanine (7-mG) and 3- methyladenine, respectively (Thomas et al., 2013). In this review, however, DNA methylation refers exclusively to 5-mC, unless mentioned otherwise. Generally, DNA methylation is associated with transcriptional repression and is mostly found in heterochromatin (Miller and Sweatt, 2007), while the euchromatin typically contains low amounts of methylated DNA. Some genes, however, are suspected to show enhanced expression when hypermethy-lated (Silva et al., 2008). Additionally, DNA methylation within gene bodies (the transcribed portion of a gene) has been implicated in alternative splicing (Flores et al., 2012; Lyko et al., 2010). How DNA methylation exactly affects gene transcription is highly dependent on the location in or around the gene (Ziller et al., 2013). In promoter regions, methylated DNA can directly disrupt the transcriptional process by interfering with the binding of transcription factors (Klose and Bird, 2006). Additional repression can be established through the attraction of methyl-CpG-binding domain proteins (MBDs) and subsequent activation of the histone tail modifying machinery, leading ultimately to chromatin compaction (Portela and Esteller, 2010). How gene expression is enhanced through methylation of gene bodies remains unclear.

Although DNA methylation is the most stable epigenetic modification, the DNA methylation profile, or ‘methylome’, is highly dynamic (Bhattacharya et al., 1999; Levenson et al., 2006; Weaver et al., 2004). DNA methylation profiles are, at least partly, heritable, both after cell division, as well as in a transgenerational fashion (Bergman and Cedar, 2013; Guerrero-Bosagna and Skinner, 2012). Heritable DNA methylation needs to be copied to the newly synthesized DNA strand, a process that is referred to as maintenance DNA methylation. The addition of completely new DNA methylation marks is called de novo DNA methylation. DNA methyltransferases (DNMT) are responsible for maintenance and de novo DNA methylation (Mastroeni et al., 2010). There are four known types of DNMTs; DNMT1, DNMT2, DNMT3a and DNMT3b, all of which use S-adenosylmethionine (SAM) as the methyl donor (Klose and Bird, 2006; Mastroeni et al., 2010). Note, however, that DNMT2 was actually found to be a RNA methyltransferase (Jurkowski et al., 2008). Furthermore, there is another DNMT, DNMT3-like (DNMT3L), which exhibits no enzymatic activity, but detects unmethylated lysine 4 of histone H3 tails (H3K4) and recruits or activates DNMT3a (Ooi et al., 2007). Interestingly, the DNMT3B splice variants DNMT3B3 and DNMT3B4 are also inactive and play a regulatory role in de novo DNA methylation (Gordon et al., 2013). DNMT1 is the most common variant in somatic cells and primarily involved in maintenance DNA methylation, and DNMT3a and DNMT3b are responsible for de novo DNA methylation (Okano et al., 1999). DNMT3a and DNMT3b isoforms are expressed in a more cell-type-specific manner (Guo et al., 2013; Okano et al., 1999).

It is worth mentioning that the methyl donor of the DNMTs, SAM, is generated through a complex cycle and is the methyl donor of numerous additional transmethylation reactions (Mastroeni et al., 2011; Wang et al., 2013). This cycle starts with the conversion of tetrahydrofolate (THF) to 5,10-methylenetetrahydrofolate (MTHF) by vitamin B6-dependent serine hydroxymethyltransferase (SHMT), and the subsequent conversion of 5,10-MTHF to 5-MTHF by B2-dependent MTHF reductase (MTHFR). 5-MTHF acts as the methyl donor for the methylation of homocysteine (Hcy), producing methionine, by cobalamin-dependent methionine synthase (MetH). SAM is subsequently generated from methionine by methionine adenosyltransferase (MAT). During methyltransferase reactions SAM is converted to S-adenosylhomocysteine (SAH), which is further hydrolyzed to Hcy and adenosine by SAH hydrolase (SAHH). Folate serves as cardinal input for this cycle and the proper elimination of Hcy and adenosine is important to maintain homeostasis. For instance, global DNA hypomethylation could be induced through folate deficiency and high levels of Hcy (Mastroeni et al., 2011).

As DNA methylation can be relatively simple and robustly assessed using genomic DNA, it has been the primary focus of human epidemiological epigenetic research (Lunnon and Mill, 2013). Early investigations into DNA methylation showed its cardinal importance in the proliferation and differentiation of neural stem cells (Mattson, 2003). More recently, it has been established that DNA methylation is pivotal for synaptic plasticity, neuronal repair, neuronal survival, and learning and memory (Fan et al., 2001; Feng et al., 2010; Iskandar et al., 2010). Such dynamic processes are more dependent on de novo methylation, although the importance of maintenance DNA methylation should not be underestimated, as a loss of DNMT1 was shown to result in increased histone acetylation, a disruption of the nuclear organization and eventually cell death (Chan et al., 2001; Espada et al., 2007; Fan et al., 2001; Jackson et al., 2004; Milutinovic et al., 2004). Because these are factors disturbed in a neurodegenerative state, DNA methylation is a valid target when investigating neurodegeneration.

1.2. DNA demethylation

While DNA methylation is a well-established epigenetic mechanism, the existence of active DNA demethylation in animals has long been a point of controversy (Ooi and Bestor, 2008). Observations such as high levels of DNMTs in nondividing cells (Sharma et al., 2008) and a significant decrease in methylated DNA levels when DNA methylation is blocked (Levenson et al., 2006; Miller and Sweatt, 2007), despite the stability of the 5-mC mark, have led to an avid search for the players responsible for an active demethylation process. This search generated several mechanisms, including an RNA-dependent pathway by which the methyl group is removed from 5-mC, a pathway involving the nucleotide excision repair mechanism, and a base excision repair based pathway (Barreto et al., 2007; Bhattacharya et al., 1999; Gavin et al., 2013; Zhu et al., 2000a,b).

Although still a point of discussion, the base excision repair pathway is a prime candidate as the primary road to demethyla-tion in post-mitotic neurons (Gavin et al., 2013), which does not exclude the possibility of multiple overlapping demethylation pathways. DNA demethylation is thought to be initiated by the oxidation of 5-mC into 5-hydroxymethylcytosine (5-hmC) by the ten-eleven translocation (TET) proteins (Guo et al., 2011; Ito et al., 2011). There are 3 TET proteins, TET1, TET2 and TET3, which are differentially expressed and regulated throughout the body (Delatte and Fuks, 2013). Interestingly, in the last few years 5-hmC was shown to be an important epigenetic marker that is functionally different from 5-mC (van den Hove et al., 2012). While DNA hydroxymethylation is generally associated with increased gene activity, work by Jin et al. (2011b) indicates that this correlation does not always hold and depends on the location of 5-hmC in the gene and the CpG content. 5-hmC is present in most tissues and cell types, but is especially enriched in the brain (Globisch et al., 2010), with cerebellar Purkinje cells exhibiting some of the highest levels (Kriaucionis and Heintz, 2009). Furthermore, in the frontal cortex, 5-hmC is selectively enriched in promoter and intragenic regions (Jin et al., 2011b). Interestingly, 5-hmC levels are particularly low in stem cell-rich areas (Globisch et al., 2010; Orr et al., 2012). In addition to the formation of 5-hmC, it has recently been discovered that TET enzymes can further oxidize 5-hmC to 5-formylcytosine (5-fC), and 5-fC to 5- carboxylcytosine (5-caC) (Ito et al., 2011). Although it is generally accepted that 5-hmC is a functional epigenetic marker, such a role remains to be established for 5-fC and 5-caC (Raiber et al., 2012; Song et al., 2013). Apart from oxidation, 5-mC and 5-hmC can be deaminated instead, by either activation-induced cytidine deaminase (AICDA) or apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like protein (APOBEC), resulting in thymidine or 5-hydroxymethyluracil (5-hmU) (Guo et al., 2011; Popp et al., 2010). 5-caC, thymidine or 5-hmU present a mismatch in the base pairing of the DNA sequence (5-caC:G, T:G or 5-hmU:G, respectively). Such a mismatch can be detected and mended through the removal of the transformed bases by thymidine or uracil glycosylases (Cortellino et al., 2011; Guo et al., 2011; He et al., 2011; Matsubara et al., 2004). Alternatively, 5-fC and 5-caC can be changed back to cytosine by removal of formic acid or decarboxylation, respectively (Ito et al., 2011). In addition to the aforementioned enzymes, the growth arrest and DNA damage 45 (GADD45) proteins are crucially involved in directing the activity of these enzymes, thereby assisting in the localization of demethylation activity to specific gene promoters (Barreto et al., 2007; Ma et al., 2009; Rai et al., 2008; Schmitz et al., 2009). Clearly, the exact mechanisms underlying DNA demethylation remain to be elucidated.

1.3. Chromatin remodeling

Chromatin can be seen as a string ofnucleosomes, which mainly consist of DNA and the histones around which it is wrapped. There are five types of histone proteins; H2A, H2B, H3, and H4 forming the octameric core of the nucleosome, and H1 serving as a linker and stabilizer, binding to DNA among nucleosomes (Luger et al., 1997; Wang et al., 2013). The conformation of these histones largely determines the accessibility of the DNA for transcription, and can be adjusted through reversible modifications of their N-terminal tails. Such modifications include lysine (K), arginine (R) or histidine (H) methylation (Murray, 1964), K acetylation (Gershey et al., 1968), serine (S), threonine (T) or tyrosine (Y) phosphorylation (Kleinsmith et al., 1966), ubiquitination (Hunt and Dayhoff, 1977), adenosine diphosphate (ADP)-ribosylation (Ueda et al., 1975), crotonylation (Tan et al., 2011), hydroxylation (Houston et al., 2013), proline isomerization (Kouzarides, 2007) and K SUMOylation (Shiio and Eisenman, 2003), which together constitute the histone code. A specific state of the histone code may either lead to gene activation or silencing (Jenuwein and Allis, 2001). The endless possible combinations of the various modifications and target sites, allows the histone code for highly versatile fine tuning of gene expression, but is also critically involved in DNA repair and replication (Day and Sweatt, 2011). Owing to the attention that chromatin-modifying enzymes have received over the past years, many enzymes that had been identified as modifying histones, were later found to have many additional substrates. In view of this, Allis et al. (2007) have proposed an updated nomenclature that better reflects the full spectrum of functions of these enzymes. For instance, histone (lysine) acetyltransferases (HATs) were renamed to lysine acetyltrans- ferases (KATs). As, however, this new nomenclature has only sporadically been adopted, both old and new names will be stated to avoid confusion.

Acetyl coenzyme A serves as donor of the acetyl group, which is transferred to lysines of histone tails by KATs (Marmorstein and Roth, 2001). There are a multitude of proteins with KAT activity, which can be divided into five main groups, comprising KAT2A (or general control of amino acid synthesis [GCN] 5), KAT2B (or P300/ CBP-associated factor [PCAF]), KAT6–8, cyclic adenosine monophosphate (cAMP) response element-binding protein binding protein (CREBBP or CBP) and adenovirus early region 1A (E1A)-binding protein P300 (EP300 or P300) (Huynh and Casaccia, 2013).

Currently, there are 18 known histone deacetylases (HDACs) in humans, generally subdivided into four classes; class I (HDACs 1,2, 3 and 8), class IIa (HDACs 4, 5, 7 and 9), class IIb (HDACs 6 and 10), class III (sirtuins [SIRTs] 1,2, 3, 4, 5, 6 and 7) and class IV (HDAC11) (Dokmanovic et al., 2007). Although their name suggests that histones are the primary targets of HDACs, phylogenetic analysis indicates that de evolutionary development of HDACs preceded that of histones (Gregoretti et al., 2004). Indeed, over 50 nonhistone targets of HDACs have been identified, including proteins important for proliferation, migration, and cell death (Marks and Breslow, 2007; Minucci and Pelicci, 2006; Rosato and Grant, 2005). Thus, as for HATs, it was suggested that HDACs should be more appropriately referred to as lysine deacetylases (Xu et al., 2007). The different HDACs fulfill many different roles, either by affecting gene expression or by directly regulating protein functioning. Class I HDACs for instance, are thought to play a general role in cell survival and proliferation, whereas class II HDACs have a more tissue-specific role (Chang et al., 2006; Harms and Chen, 2007; Kim et al., 2007b; Laggeret al., 2002; Parra et al., 2007; Vega et al., 2004; Zhang et al., 2002). The SIRTs further differ from the others HDACs in that their activity is nicotine adenine dinucleotide (NAD+)-dependent, whereas the other classes require the presence of zinc. Not all SIRTs are even primarily deacetylases. This has led to the suggestion that SIRTs should be classified as deacylases, as opposed to deacetylases (Hirschey, 2011). Although class I and class IV HDACs are mainly nuclear, class Ila shuttles between the nucleus and cytoplasm and class llb is primarily cytoplasmic. The sirtuins of class lll are most varied in their localization, and can be either expressed in the nucleus (SIRTs 1, 2, 6 and 7), cytoplasm (SIRTs 1 and 2) or mitochondria (SIRTs 3, 4 and 5) (Michan and Sinclair, 2007). The expression of the different HDACs is also highly region- and cell-type-specific, for instance, while HDAC2 is expressed in most brain regions, it is predominantly active in mature neurons and weakly or not in progenitor and glial cells (Guan et al., 2009; MacDonald and Roskams, 2008).

Methylation of histone tails happens at K or R residues of H3 or H4, and is executed by histone lysine methyltransferases (HKMTs) and protein arginine methyltransferase (PRMT), respectively, whereas demethylation is performed by histone lysine demethy-lases (HKDMs) and histone arginine demethylases (HRDMs) (Chang et al., 2007; Habibi et al., 2011). The other histone modifications are less well characterized. Phosphorylation and dephosphorylation of histones is executed by protein kinases, such as mitogen- and stress-activated protein kinase (MSK)-1, and protein phosphatases, such as protein phosphatase (PPT) 1, respectively (Brami-Cherrier et al., 2009; Koshibu et al., 2009). Histone phosphorylation is dynamic in function; H3 phosphorylation, for instance, marks open chromatin during active gene expression, whereas during mitosis this marker associates with condensed chromatin (Sawicka and Seiser, 2012). Ubiquitination can either enhance or inhibit gene expression, whereas SUMOylation is primarily thought to suppress transcription (Habibi et al., 2011).

The actual effect and interplay between these modifications are complex, and depend on the type of histone protein and the specific amino acid that is modified, and a combination of certain modifications can even have a function that is different from that of these modifications in isolation (Bernstein et al., 2006; Greer and Shi, 2012; Hwang et al., 2013; Jenuwein and Allis, 2001; Sawicka and Seiser, 2012). For instance, acetylation of K 9 (H3K9ac) and 14 (H3K14ac) of H3, or tri-methylation at K 4 of H3 (H3K4me3), H3K36me, H3K79me, H3R2me, H3R17me, H3R26me and H4R3me are associated with gene activation, whereas H3K9me2/3, H3K27me3, H3R8me, H4K20me3 and H4R3me are generally associated with gene repression (Habibi et al., 2011 ). Interestingly, in case of histone methylation, the number of attached methyl groups also matters, as the previously mentioned H3K9me3, H3K27me3 and H4K20me3 marks that are associated with gene repression, all have monomethylated counterparts that are associated with gene activation (Barski et al., 2007). Of note, recent studies mapping histone tail modifications to genomic regions found that many transposable elements (TEs) are enriched with certain histone marks (Kondo and Issa, 2003; Martens et al., 2005; Pauler et al., 2009) and it has therefore been suggested that TEs may attract certain histone marks to induce heterochromatic and euchromatic states, or serve as boundary elements that prevent the propagation of such states (Huda and Jordan, 2009).

In addition to histone modifications, chromatin remodeling also occurs through adenosine triphosphate (ATP)-dependent multiprotein chromatin remodeling complexes. Four distinct remodeling complex families have been identified, including the Brg1/hBrm associated factor (BAF; previously known as switching defective/sucrose nonfermenting [SWI/SNF]), imitation SWI (ISWI), chromodomain, helicase, DNA-binding (CHD) and inositol requiring 80 (INO) families (Clapier and Cairns, 2009; Hargreaves and Crabtree, 2011; Vogel-Ciernia and Wood, 2014). These complexes, or remodelers, can bind nucleosomes, disrupt nucleosome-DNA binding, and then move, destabilize, eject or restructure them, using ATP hydrolysis as energy source. In doing this, remodelers can either induce transcriptional activation or repression, through the recruitment of coactivator or corepressor complexes. The different remodeling complexes are defined by their ATPase subunits, but variation in their remaining subunit composition, possibly altering the DNA and protein binding properties of the complex, allows for great diversity, leading to cell-type specific roles (Ho and Crabtree, 2010; Ronan et al., 2013). Furthermore, multiple functionally different versions of a complex may be present within one cell (Wang et al., 2004). The BAF complex, consisting of at least 15 subunits, is of particular interest, as it is the only chromatin remodeling complex with a neuron-specific subunit, BAF53b. The BAF complex is thought to play an important role in neuronal development and functioning, with unique subunit compositions in embryonic stem cells, neuronal progenitors and mature neurons (Vogel-Ciernia and Wood, 2014). The BAF53b subunit was shown to be important for dentritic development in vitro and long term memory in mice (Vogel-Ciernia et al., 2013). See the excellent review by Hargreaves and Crabtree (Hargreaves and Crabtree, 2011) fora detailed discussion of the different chromatin remodeling complexes.

1.4. Non-coding RNAs

Until recently it was widely believed that most of the human genome consisted of so-called ‘junk’, or nonfunctional DNA. It was later revealed that almost the whole genome is transcribed, but that only about 2% is actually translated into proteins (Amaral et al., 2008). Most of the ‘junk’ actually is functional and is primarily involved in the regulation of gene expression, usually in the form of ncRNAs. There are many types of ncRNAs, including microRNAs (miRNAs), small interfering RNAs (siRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), small Cajal body-specific RNAs (scaRNAs), piwi-interacting RNAs (piRNAs), splice junction- associated RNAs (spliRNAs), small modulatory RNAs (smRNAs), repeat-associated small interfering RNAs (rasiRNAs), transcription initiation RNAs (tiRNAs), promoter-associated short RNAs (PASRs), transcription start site-associated RNAs (TSSa-RNAs), promoter upstream transcripts (PROMPTS), ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), and small double-stranded RNAs (dsRNAs) (Jady et al., 2004; Mattick, 2011; Preker et al., 2008; Schouten et al., 2012; Seila et al., 2008). These are small ncRNAs (sncRNAs), of <200 nucleotides, whereas there are also long ncRNAs (lncRNAs), which can exceed 100,000 nucleotides, often including TE-derived sequences that may confer specific protein or nucleic acid interacting properties (Johnson and Guigó, 2014). Examples of lncRNAs are intergenic ncRNAs (lincRNAs), natural antisense transcripts (NATs), ncRNA expansion repeats, promoter-associated RNAs (PARs), and enhancer RNAs (eRNAs) (Kapranov et al., 2007; Khalil et al., 2009; Kim et al., 2010b; Nakamori and Thornton, 2010; Werner, 2005). The sncRNAs fulfill various functions, including infrastructural (rRNAs, tRNAs and snRNAs) and regulatory roles (miRNAs, siRNAs, snoRNAs, piRNAs and spliRNAs), whereas the lncRNAs are primarily regulatory. Interestingly, lncRNAs are expressed in a highly cell-specific manner, may undergo alternative splicing, and may even have protein-coding isoforms (Cabili et al., 2011; Chooniedass-Kothari et al., 2004; Djebali et al., 2012; Johnsson et al., 2013). Alternatively, some mRNAs may function as trans-acting regulatory RNAs (Ashe et al., 1997; Dinger et al., 2011; Mercer et al., 2011). In terms of epigenetics, ncRNAs are cardinally involved in gene expression control, in the silencing of TEs, X-chromosome inactivation, alternative splicing, and DNA imprinting (Jeon et al., 2012; Lisch, 2012; Morrissy et al., 2011). Additionally, some lncRNAs have been proposed to direct epigenetic enzymes to their target sites (Khalil etal.,2009; Koziol andRinn, 2010; Mercer and Mattick, 2013), while others are thought to bind and sequester other epigenetic players, such as DNMTs and miRNAs, thereby hampering their activity (Di Ruscio et al., 2013; Johnsson et al., 2013; Mercer and Mattick, 2013).

The best characterized of the ncRNAs are miRNAs. These begin their lives as primary miRNAs, after which they are cleaved by ribonuclease type III Drosha to form precursor miRNAs (pre-miRNAs) (Han et al., 2009). These pre-miRNAs are then transported from the nucleus to the cytosol by Exportin-5, where Dicer makes the final adjustment to produce double-stranded mature miRNAs. Mature miRNAs span 21–25 nucleotides and regulate protein production through an RNA interference pathway, involving the association of one of the strands of the miRNA duplex with the RNA-induced silencing complex (RISC) (Ambros, 2004; Han et al., 2004). They interfere with gene expression through binding to messenger RNA (mRNA), usually to the 3’ untranslated region (UTR), which hampers the initiation of translation and affects mRNA stability. MiRNAs can potentially regulate the translation of multiple genes, whereas genes can also be regulated by several miRNAs, as the sequences of the miRNA and its target are not required to be a perfect match for interference to take effect (He and Hannon, 2004; Lim et al., 2005). While siRNAs are processed and function similarly, they require stricter matching with their target sequence when compared to miRNAs (Zeng et al., 2003). Apart from interacting with RISC, some miRNAs have been observed to promote mRNA translation and gene transcription, by binding to gene promoter regions (Eulalio et al., 2008; Fabian et al., 2010). There are numerous miRNAs, many of which are expressed according to strict spatial and temporal patterns. Currently, there are 1881 precursors and 2588 mature human miRNAs registered in the fast growing miRBase (The University of Manchester, 2014). While expressed throughout the whole human body, the brain is especially enriched in miRNAs, suggesting an important role for them in neuronal development, functioning, and aging (Gokey et al., 2012; Hu et al., 2011a). Their biological role has been further characterized, and reviewed by Santosh, and colleagues, as well as Qu and Adelson (Qu and Adelson, 2012; Santosh et al., 2014). Both the reviews extend beyond the aforementioned functions, and present the key role of ncRNA in RNA splicing, transcriptional, post-transcriptional, and translational regulation by either binding directly to transcriptional factors or by generating siRNAs, that consequently interact with the translational machinery.

1.5. Additional epigenetic mechanisms

DNA methylation, chromatin remodeling, and ncRNAs represent the best-studied epigenetic mechanisms, especially in relation to aging and neurodegenerative diseases. Epigenetic regulation goes much deeper, however, and also includes the rising subfields of RNA editing, RNA methylation, and mitochondrial epigenetics, which will be briefly touched upon in this section, but will not be further discussed in relation to aging and neurodegeneration due to the as of yet extremely limited findings in this respect. Clearly, more studies on the role of these additional layers of epigenetic regulation in aging and neurodegeneration are waranted.

1.5.1. RNA editing

The observation of discrepancies in genomic and cDNA sequences led to the discovery of RNA editing (Bass, 2002). The finding that RNA can be edited, a process that seems particularly important in the brain, adds another layer to the transcriptional and post-transcriptional regulation of gene expression. It has even been proposed that a dramatically increased RNA editing capacity was crucial for the evolution of the mammalian brain as it may function as a mechanism driving phenotypic adaptability, which ultimately led to the superior cognitive abilities of humans (Mattick, 2010; Mattick and Mehler, 2008). In support of this, roughly 35 times more RNA editing is observed in humans compared to mice. This surplus appears to be mainly directed to retrotransposed Alu elements that are primate-specific and constitute 10.5% of the human genome (Athanasiadis et al., 2004; Kim et al., 2004; Lander et al., 2001; Levanon et al., 2004; Umylny et al., 2007).

In contrast to RNA and DNA modifications, RNA editing involves a change in RNA sequence by deamination of either adenosine (A) or cytosine (C), to inosine (I) (Bass, 2002; Valente and Nishikura, 2005) or uracil (U) (Conticello, 2008; Navaratnam and Sarwar, 2006), respectively. A to I editing is performed by adenosine deaminases that act on RNA (ADARs), while C to U editing is carried out by APOBECs. APOBECs are related to AICDAs, which targets DNA and is pivotal in the generation of the immunoglobulin repertoire (Muramatsu et al., 1999). Although not much is known about the targeting, regulation and functional impact of ADARs and APOBECs, they are thought to be evolved from adenosine deaminases that act on tRNAs (ADATs) and thus bind doublestranded RNA regions, such as those seen in hairpin formations that are also present in tRNAs (Bass, 2002; Conticello, 2008).

ADAR1 and ADAR2 are ubiquitously expressed and they appear to be enriched in the brain, while the expression of ADAR3 seems to be restricted to the brain. A to I RNA editing has been observed in coding transcripts, for instance leading to changes in the amino acid sequence of glutamate and serotonin receptors. Most editing, however, happens to noncoding sequences, such as miRNAs (Blow et al., 2006; Kawahara et al., 2008; Nishikura, 2006) and transposon-derived repetitive sequences (Morse et al., 2002), suggesting that RNA editing not only directly affects gene expression, but also indirectly by regulating other epigenetic players (Mattick, 2010). APOBECs, together with overall RNA editing, appear to have undergone a substantial expansion over the course of evolution, with APOBEC3 being especially favored in humans with eight orthologs, compared to one in mice (Conticello et al., 2005; Navaratnam and Sarwar, 2006; Sawyer et al., 2004). Although it has been suggested that these enzymes have evolved to combat retrotransposons and endogenous retroviruses (Aguiar and Peterlin, 2008; Schumann, 2007), there is also evidence indicating that these elements actually have been harnessed as epigenetic regulators involved in growth and differentiation, including the generation of neuronal diversity (Coufal et al., 2009; Dunlap et al., 2006; Faulkner et al., 2009).

Recently, the implication of RNA editing in the etiopathogenesis and progression of neurodegenerative disorders as well as normal aging processes has gained momentum and the few available studies begin to elucidate this connection. The majority of these studies are focusing on aging. Sebastiani et al. observed that 5 SNPs in the ADAR encoding genes Adarb1 and Adarb2 are associated with extreme longevity in 4 independent human studies. The observation of the critical role of ADARs in aging was also verified in a Caenorhabditis elegans (C. elegans) model with loss of function of adr1 and adr2 (Adarb1 and Adarb2 orthologues), which had a 50% decrease in lifespan (Sebastiani et al., 2009). After this study a lot of RNA editing targets were discovered, such as gamma-aminobu-tyric acid receptor subunit alpha-2 (Gabra2), cytoplasmic FMR1-interacting protein 2 (Cyfip2), potassium channel gene (Kcnal), filamin a (Flna), bladder cancer associated protein (Blcap), golgi complex subunit 3 (Cog3), nuclear paraspeckle assembly transcript 1 and 2 (Neat1, Neat2), metastasis associated lung adenocarcinoma transcript 1 (Malat1) and phosphatidylserine decarboxylase-pseudogene 1 (Pisd-ps1) that are differentially edited in the aging murine and human brain. Among them Cyfip2 and Pisd-psl have gained considerable attention (Holmes et al., 2013; Nicholas et al., 2010; Stilling et al., 2014). Nicholas et al. demonstrated that A to I editing declines with age in humans, in a gene-specific manner, resulting in downregulation of an ADAR2 target gene, Cyfip2, which is responsible for synaptic maintenance (Nicholas et al., 2010). In the hippocampal formation of aged mice Stilling et al. showed that the altered RNA editing levels of Pisd-ps1 results in higher editing frequency with age and thus upregulation of gene expression (Stilling et al., 2014).

In case of AD, Akbarian et al. observed a decrease in the RNA editing levels of glutamate receptor 2 (GluR2) in the prefrontal cortex of AD human brains (Akbarian et al., 1995). Rechavi’s team following the aforementioned research line, examined the GluR2 Q/R RNA editing levels in the hippocampus of AD brains, which they found to be decreased in comparison to controls. Additionally, they showed lower GluR2 Q/R RNA editing in the hippocampus of APOEε4 carriers. The mRNA expression of ADARs was also investigated in this specific study; unexpectedly no differences were found in the hippocampus but a 37% decrease of ADAR2 mRNA expression was noticed in the caudate (Gaisler-Salomon et al., 2014). Finally, Akbarian et al. extended his study on HD where he also showed a decrease in GluR2 RNA editing levels in the striatum (Akbarian et al., 1995).

1.5.2. RNA methylation

Although the discovery of methylated RNA was done decades ago (Desrosiers et al., 1974; Rottman et al., 1974), over a hundred RNA nucleotide modifications have been identified across different organisms (Behm-Ansmant et al., 2011; Kellner et al., 2010). In eukaryotes the best-studied mRNA modifications are N6-methy-ladenosine (m6A) and 5-mC, which mainly occur at 3’ UTRs and stop codon sites (Meyer et al., 2012). As in DNA, 5-hmC has also been observed in RNA (Fu et al., 2014). m6A is the most prevalent mRNA modification in mammals and has also been observed in tRNAs, rRNAs and snoRNAs(Bringmannand Luhrmann, 1987; Choi and Busch, 1978; Epstein et al., 1980; Harada et al., 1980; Munns et al., 1977; Perry et al., 1975; Shimba et al., 1995; Tanaka and Weisblum, 1975). In humans the m6A modification shows high tissue specificity, with the highest levels occurring in the brain, in transcripts such as Bdnf, Dscam, Lis1 and Ube3a (Dominissini et al., 2012). In mice and humans m6A methyltransferase methyltrans- ferase-like protein 3 (METTL3) (MT-70) is responsible for the post- transcriptional m6A RNA modification (Bujnicki et al., 2002). Additionally, METTL14 and Wilm’s tumor-associated protein (WTAP) have been shown to interact with METTL3 and are thought to be additional components involved in RNA methylation (Ping et al., 2014; Wang et al., 2014b). Just as DNA methylation, the identification of m6A demethylases fat mass and obesity-associated protein (FTO) and AlkB, alkylation repair homolog 5 (E. coli) (ALKBH5)(Jia et al., 2011; Zheng et al., 2013), indicates that RNA methylation is a dynamic regulatory mechanism. FTO, a dioxygenase, demethylates RNA via a similar oxidation procedure as is employed by the TET enzymes that are thought to be involved in DNA demethylation, namely through the generation of intermediates N6-hydroxymethyladenosine (hm6A) and N6-for-myladenosine (f6A), before being reversed to A (Fu et al., 2013). Although these intermediates remain stable for several hours, no separate regulatory roles for hm6A and f6A have been reported yet. ALKBH5 is thought to remove m6A directly, without the generation of intermediates (Zheng et al., 2013). Although the exact regulatory functions of m6A RNA methylation remain to be elucidated, its main occurrence at 3’ UTRs and stop codons has been suggested to indicate a role in switching genes on or off (Chandola et al., 2014). Alternatively, the observation that players involved in m6A RNA methylation were located at splice sites suggests that the m6A RNA modification may modulate splicing (Meyer et al., 2012). More recent findings show a relation between the m6A modification and mRNA degradation, as m6A selectively binds human YTH domain family 2 (YTHDF2) proteins, which can bind and target mRNA to decay sites, such as processing bodies (P-bodies) (Wang et al., 2014a). Other members of the YTH domain family, YTHDF1 and YTHDF3 also selectively bind m6A modified RNA. Another study, however, indicated that m6A does not lead to RNA decay through the YTHDF2 pathway, but by interacting with miRNAs, and that the removal of m6A promotes human antigen R (HuR) binding, a protein that protects against RNA decay (Wang et al., 2014b). These studies suggest that the m6A mark may dynamically regulate mRNA lifetime.

An alternative pathway of RNA methylation involves the versatile regulatory ncRNAs snoRNAs, which can guide 2’-O-methylation and pseudo-uridylation of RNA transcripts, including mRNAs (Bachellerie et al., 2002). 2’-O-methylation is important for the functioning of certain rRNAs (Maxwell and Fournier, 1995), but also determines the guide strand and targeting specificity of siRNAs (Chen et al., 2008). Apart from their role in RNA modifications, snoRNAs can be further processed into snoRNA-derived small RNAs (sdRNAS), which are similar to miRNAs (Ender et al., 2008; Politz et al., 2009; Saraiya and Wang, 2008; Taft et al., 2009).

The pathway that most closely resembles DNA methylation involves DNMT2, which, despite its name, transfers methyl groups to cytosines in RNA (Jurkowski et al., 2008; Schaefer et al., 2009). Apart from tRNA, the exact substrates of DNMT2 still need to be identified. Nevertheless, DNMT2 has been implicated in brain development and retrotransposon silencing (Phalke et al., 2009; Rai et al., 2007). Other known RNA methylation modifications include Nl-methyladenosine (m1A) and Nl-methylguanine (mlG), which occur mainly in tRNAs and are thought to enhance tRNA stability, and mlG also decoding accuracy (Anderson et al., 2000; Bjork et al., 1989; Jackman et al., 2003; Saikia et al., 2010).

The only reported studies connecting RNA methylation to aging, as well as neurodegenerative disorders, were performed by Giordano et al. (2012) and Thomas et al. (2013), respectively. Bellizzi’s team studied the methylated cytosine residues in two mitochondrial genes, 12S and 16S rRNA and they showed that the methylation levels of 12S rRNA are decreased with age in males. Thomas et al., while attempting to develop a novel method for detecting trace amounts of 7-mG in biological samples, observed differential methylation patters in murine HD models and significantly increased levels of 7-mG in postmortem human HD brain samples.

1.5.3. Mitochondrial epigenetics

Apart from the nuclear genome, human cells can harbor thousands of copies of the mitochondrial genome. Both the nuclear and mitochondrial genome consist of DNA, but there are some striking differences (Byun and Baccarelli, 2014). The mitochondrial genome is only 16 kb long, contains 37 genes without introns and is much more prone to mutations than the nuclear genome. With respect to epigenetics, its regulation seems to be less complex, as mitochondrial DNA (mtDNA) is thought not to be wrapped around histones and not to contain CpG islands; the 435 CpG sites in the mitochondrial genome are almost evenly dispersed.

Over 40 years ago, methylated mtDNA was discovered in loaches, and it was shown there is DNMT activity in mitochondria that is independent from DNMT activity outside mitochondria (Kudriashova et al., 1976; Nass, 1973). Later, mtDNA methylation was also observed in humans (Shmookler Reis and Goldstein, 1983), and a mitochondrial DNMT, (mtDNMT1) was discovered (Shock et al., 2011). Of note, however, more recent studies cast doubt on the general notion that DNA methylation is the prime epigenetic mechanism at work in mitochondria. For instance, Hong et al. (2013) were unable to detect CpG methylation in the genome of human mitochondria, whereas Choi et al. (2011) report on the possible existence of mitochondrial histones, and Barrey et al. (2011) found miRNAs in mitochondria. Nevertheless, there are also many recent reports supporting the presence of methylated mtDNA and even hydroxymethylated mtDNA (Chestnut et al., 2011; Dzitoyeva et al., 2012; Iacobazzi et al., 2013; Manev et al., 2012), showing that it is not always located at CpG sites (Bellizzi et al., 2013; Sun et al., 2013), and that mtDNA methylation plays a role in mitochondrial gene regulation (Feng et al., 2012; Pirola et al., 2012). Clearly, these rapid developments within the field of mitochondrial epigenetics warrant further attention. In the recent study of Dzitoyeva et al. not only the hydroxymethylation of mtDNA was reported but they also demonstrated that solely the levels of hydroxymethylated mtDNA reduce with age in the frontal cortex of mice (Dzitoyeva et al., 2012). This decrease in 5-hmC is associated with an increase in complex I components (ND2, ND4, ND4L, ND5, ND6) in the same area. Furthermore, they observed region-specific differential expression of epigenetic players; the mRNA levels of TET2 and TET3, which are also responsible for the hydroxymethylation of mtDNA, are only increased in the cerebellum, whereas the mRNA levels of mtDNMT1 decrease solely in the frontal cortex.

1.6. Epigenetic processes are interdependent

The epigenetic processes of DNA (de)methylation, chromatin remodeling, and miRNAs do not act independently, but closely interact to form a complex, multilayered regulatory system that can dynamically fine-tune gene expression. DNA methylation stability in promoter regions, for instance, is enforced by methyl CpG binding protein (MeCP) 1, which also binds the nucleosome remodeling and histone deacetylase (NuRD) core and cyclin-dependent kinase 2 associated protein (CDK2AP) 1, forming a protein complex not only able to stabilize DNA methylation, but also to modify the histone code (Grewal and Jia, 2007; Zhang and Reinberg, 2001 ). MeCP1 is attracted to methylated DNA through its affinity for MBD2, which directly binds to DNA methylated at CpG sites. As such, DNA methylation and histone modifications act in concert to regulate gene expression, through interference with transcription factor binding and chromatin compaction (Klose and Bird, 2006). Another interesting interplay, between DNMT3a-dependent DNA methylation and Polycomb-group (PcG)-depen- dent H3K27me3 marks was discovered by Wu et al. (2010). They showed that DNMT3a activity at non-promoter regions correlated with increased expression of neurogenic genes, by interfering with PcG binding and H3K27me3-mediated gene repression. In contrast, DNMT3a activity at the promoter regions inhibited gene expression. Alternatively, MBD1 can antagonize H3K4me3, leading to chromatin compaction. DNA methylation can thus in a bottom-up fashion induce changes on the chromatin level (Liu et al., 2010a). The other way around is also possible, as exemplified by Detich et al. (2003). They showed that increases in H3 acetylation can induce DNA demethylation, and thereby gene expression in vitro. Conversely, HDAC activity is thought to inhibit gene expression through the induction of DNA methylation (Sun et al., 2007). There are additional complex interactions between miRNAs and other components of the epigenetic machinery. Where some miRNAs regulate the expression of proteins involved in epigenetic regulation, the expression of various miRNAs themselves is also subject to factors such as DNA methylation and histone modifications (Saito and Jones, 2006). For example, miRNA 184 (miR-184), involved in the regulation of proliferation and differentiation of neural stem cells, is surrounded by CpG islands, attracting MBD1, which can suppress its expression as described above (Liu et al., 2010a).

2. Aging

Before delving into the aberrant epigenetic processes associated with neurodegeneration, it is important to consider the epigenetic changes associated with normal aging and related hallmarks, such as oxidative stress, as these can already be quite dramatic. Bocklandt et al. (2011) for instance, devised a method to determine the age of an individual based on the methylation of specific sites in the ectodysplasin-A receptor-associated death domain (EDAR-ADD), target of myb1 (chicken)-like 1 (TOM1L1) and neuronal pentraxin II (NPTX2) genes. At these sites a linear correlation between methylation and age was observed, allowing for a prediction of age with an average accuracy of 5.2 years. Horvath later devised an even more accurate method to determine the ‘DNAm age’, based on the methylation status of 353 CpGs (Horvath, 2013). The DNAm age of Horvath has a chronological age correlation of about 0.96 and an error of 3.6 years and is applicable in many different tissue and cell types. Note, however, that epigenetic processes are not the only players involved in aging. According to the “free radical theory of ageing”, oxidative stress is thought to play an integral role in the aging process (Beckman and Ames, 1998). Oxidative stress refers to the generation of reactive oxygen species (ROS), which are damaging to proteins, nucleic acids and lipids and are known to also affect epigenetic players (Cencioni et al., 2013). Furthermore, as aging is the prime riskfactor of most neurodegenerative diseases, it is possible that age-related processes including epigenetic alterations and oxidative stress, facilitate the development of these illnesses.

2.1. DNA (de)methylation in aging

Early research established that DNA methylation plays a crucial role during development. Later studies identified aging to be a pivotal modulator of the epigenome. The DNAm age of Horvath offers some interesting insights in this respect (Horvath, 2013). Of the 353 CpG sites used to predict the DNAm age, 193 got hypermethylated and 160 got hypomethylated with age, and most are associated with genes involved in cell death and survival, cell growth and proliferation, organismal and tissue development, and cancer. Additionally, DNAm age shows a logarithmic relationship with chronological age until adulthood, and a linear relationship later in life, indicating that the epigenetic clock ‘ticks’ faster during growth and development. While highly accurate in most tissues, Horvath found that the DNAm age was consistently lower in tissues which may be renewed through the presence of stem cells, such as skeletal and heart muscle. It was, however, also observed that DNAm age does not reflect cellular senescence, as it highly correlated with chronological age in short and long lived cells, as well as immortal cells. As could be expected, embryonic stem cells appeared to have a DNAm age close to zero. Interestingly, the DNAm age of induced pluripotent stem cells did not differ significantly from that of embryonic stem cells. While the clock CpGs used for Horvath’s DNAm age are enriched in cancer genes, there are some important differences between normally aging and cancerous tissue. In general, cancer tissue exhibits an accelerated DNAm age. Due to the heterogeneity of cancer types, however, general statements about its use should be interpreted with caution, as for example thyroid cancer progression negatively correlates with age acceleration. Additionally, Horvath observed that an increased DNAm age may promote genomic stability, as he found in several cancer types a negative relation between DNAm age acceleration and somatic mutations. He proposes that cancer triggers a hypothetical epigenetic maintenance system that promotes genetic stability, a process that is dependent on P53, as mutations in the TP53 gene are associated with a lower DNAm age acceleration. Interestingly, in glioblastoma multiforme TP53 mutations appear to be associated with an increased DNAm age acceleration. While it thus seems that in general cancer is associated with an increased DNAm age profile and aging with global DNA hypermethylation, neurodegenerative diseases such as AD and PD are associated with global DNA hypomethylation (Mastroeni et al., 2010; Obeid et al., 2009). Note, however, that the DNAm age itself has not yet been assessed in neurodegenerative tissue. Nevertheless, despite having age as a common risk factor, cancer and age-related neurodegenerative diseases seem to involve (at least partly) different epigenetic dysregulation or compensatory mechanisms.

Taking a more specific approach, Siegmund et al. (2007) investigated the DNA methylation status of 50 CpG islands associated with genes involved in brain growth and development in subjects of various ages, and they observed a robust and progressive increase in DNA methylation of multiple genes with age (Table 1). They also confirmed that a rise in DNA methylation typically results in a decline of corresponding mRNA levels. In addition, it was observed that DNMT3a was expressed across all ages, supporting the notion that DNA methylation can be dynamically altered throughout the lifespan. Interestingly, in relation to AD, the promoter region of the amyloid β precursor protein (APP) gene becomes hypomethylated with age (Tohgi et al., 1999). Additionally, binding sites for granulocyte chemotactic factor (GCF), known to repress CG-rich promoters and interaction sites for specificity factor (SP) 1, which enhances gene expression in the tau promoter, became hypo- and hypermethylated, respectively, with age, decreasing its overall expression. This finding suggests that certain age-related epigenetic changes might facilitate the development of AD. Although expressed across all ages, levels of DNMT3a and 5-mC actually increase with age in the dentate gyrus (DG), cornu ammonis (CA) 1–2, and CA3 regions of the mouse hippocampus (Chouliaras et al., 2011a, 2011b), which is in line with previous reports (Lopatina et al., 2002). An interesting study by Oliveira et al. (2012) showed that hippocampal levels of DNMT3a2, an isoform of DNMT3a, decrease with age and that this decrease correlated with age-related cognitive decline in mice. Importantly, experimental restoration of DNMT3a2 levels alleviated this age-related cognitive impairment. Additionally, Hernandez et al. (2011) investigated 27,000 CpG sites in brain samples of varying ages and detected a general positive correlation between age and methylation levels. In contrast, it was found that expression levels of DNMT1 decrease with aging in human fetal lung fibroblasts, which would be in support of reports of global DNA hypomethylation with aging and cell senescence, including non-coding regions and repetitive sequences in the blood (Bollati et al., 2009; Lopatina et al., 2002; Pan et al., 2013). Mazin (Mazin, 2009) put forward an interesting hypothesis, proposing a DNA methylation-dependent aging process. This model is based on the observation that methylation of cytosines may induce C>T mutations, which is suggested to result in age-related genome disintegration, and eventually cell apoptosis, organism aging and death. Due to the age-related increase in 5-mC>T transitions, this model predicts an age-related depletion of 5-mC.

Table 1.

Epigenetic dysregulation in aging: DNA (de)methylation.

Gene Regulation Epigenetic
modification
Observed in Sample
size
Approach Methods Reference

GABRA2 Methylation Human 125 Targeted BS, Methylight PCR Siegmund et al., 2007
GAD1 (promoter region) (50 loci)
HOXA1
NEUROD1
NEUROD2
PGR
STK11
SYK
IFN-γ ↑↑ 784 Targeted
(9 loci)
BS Madrigano et al., 2012
RUNX3 ↑↑ 26 Targeted
(7 loci)
BS, MS-PCR So et al., 2006
ERß ↑↑ 7–10 Targeted
(1 locus)
BS, MS-PCR,
Pyrosequencing, ChIP
Westberry et al., 2011
APP Methylation Human 16 Targeted BS Tohgi et al., 1999
GCF Methylation
(binding sites)
(18 loci)
SP1 ↑↑ Methylation
(interaction sites)
Protein/(Hydroxy)
Methylated base
Regulation Modification in Observed in Sample size Methods Reference

DNMT1 Expression Fibroblasts (fetal lung) - DNMT assay
ChIP, RT-PCR
Lopatina et al., 2002;
Pan et al., 2013
DNMT3a2 Mice (12 month old) 15 IHC Oliveira et al., 2012
DNMT3a Mouse Hippocampus 48 IHC Chouliaras et al., 2011a
5-mC (DG, CA1–2 and 3) Chouliaras et al., 2011b
5-hmC 4 HPLC Munzel et al., 2010;
Song et al., 2011b

indicates increased expression levels and

indicates decreased expression levels.

Abbreviations: 5-hmC, 5-hydroxymethylcytosine; 5-mC, 5-methylcytosine; APP, amyloid β precursor protein; CA, cornu ammonis; DG, dentate gyrus; DNMT, DNA methyltransferase; ER, estrogen receptor; GABRA2, γ-aminobutyric acid receptor subunit alpha-2; GAD1, glutamate decarboxylase 1; GCF, granulocyte chemotactic factor; HOXA1, homeobox protein A1; IFN-γ, interferon γ; NEUROD, neurogenic differentiation factor; PGR, progesterone receptor; RUNX3, runt-related transcription factor 3; SP1, specificity factor 1; STK11, serine/threonine kinase 11; SYK, spleen tyrosine kinase; BS, bisulfite sequencing; PCR, polymerase chain reaction; MS-PCR, methylation specific-PCR; RT-PCR, real time-PCR; ChIP; chromatin immunoprecipitation; IHC, immunohistochemistry; HPLC: high-performance liquid chromatography.

Note, however, that DNA methylation profiles are not only known to be different between different tissues, regions and cell types, but that these also seem to be differentially affected by the aging process (Brown et al., 2008; Ladd-Acosta et al., 2007; Thompson et al., 2010). An interesting study by Fraga et al. (2005), investigating DNA methylation and histone acetylation during the lifetime of monozygotic twins, illustrates that the epigenome not only changes with age, but also that differences in the epigenome might explain phenotypic disparity in genotypically identical individuals.

In addition to the age-related increases of DNMT3a and 5-mC levels, a significant age-related increase in 5-hmC levels was found in the DG, CA1–2, and CA3 regions of the mouse hippocampus (Chouliaras et al., 2012), which is in line with previous investigations into the spatial and temporal distribution of 5-hmC in the brain (Münzel et al., 2010; Song et al., 2011b). While some of the genes that exhibit age-related increases in 5-hmC levels are associated with age-related neurodegenerative diseases (Song et al., 2011b), further investigations are required to elucidate the functional consequences of these findings, taking into account the differential functions of the 5-mC and 5-hmC markers. Table 1 summarizes the age-related alterations regarding DNA (de)methylation.

2.2. Chromatin remodeling in aging

Apart from widespread changes in the neuronal DNA methyl-ation profile throughout the lifespan, the neuronal histone code also undergoes age-related alterations (Table 2). An example is the observation of lower levels of histone acetylation with aging in vitro (Ryan and Cristofalo, 1972), and an age-related progressive decline of H3 and H4 methylation (Thakur and Kanungo, 1981) and monoacetylated H4 levels, discovered in neurons from the rat cerebral cortex (Pina et al., 1988). Apart from detecting decreased levels of H3K9ac and increased levels of H3S10p (Kawakami et al., 2009), Nakamura et al. (2010) detected decreased acetylation of extranuclear proteins. In senescence-accelerated prone mouse 8 (SAMP8) brains it was shown that many histone marks are alterated with age (Wang et al., 2010a; Table 2). In rats, however, some of these markers were observed not to undergo significant age-related changes (Sarg et al., 2002). These not always compatible findings between species illustrate the necessity of translating results regarding epigenetic changes to the human situation. Apart from changes in specific histone methylation marks, the HKMTs polycomb repressive complex member Bmi1 (PRC1) and polycomb repressive complex member enhancer of zeste homolog (EZH) 2 (Drosophila) (PRC2) have been observed to decrease with cell senescence, a common, but limited, in vitro model of aging, while the HKDM jumonji domain containing (JMJD) 3 increased (Agger et al., 2009; Jung et al., 2010). The balance between PcG and JMJD3 gene expression is in turn thought to be regulated by HDAC activity.

Table 2.

Epigenetic dysregulation in aging: chromatin remodeling.

Chromatin
remodeling target
Regulation Epigenetic
modification
Observed in Sample size Methods Reference

H3 Methylation Rat cerebral cortex 12–18 WB Thakur and Kanungo, 1981
H3K9 Acetylation Rat (liver) N.S. WB Kawakami et al., 2009
H3K27 Methylation SAMP8 mice 6 Nano-LC, MALDI-TOF/ Wang et al., 2010a
H3K36 Tri-Methylation TOF MS, WB, IHC
H3K79 Tri-/Methylation
H3S10p Phosphorylation Rat cerebral cortex N.S. WB Kawakami et al., 2009
H4 Methylation Rat cerebral cortex 12–18 WB Thakur and Kanungo, 1981
Monoacetylation Pina et al., 1988
H4K12 Acetylation 16-months old mice 12 ChIP Seq-PCR, Peleg et al., 2010
HAT/HDAC assay
H4K16 Acetylation SIRT-1 transfected cell lines ChiP Pruitt et al., 2006
H4K20 Methylation SAMP8 mice 6 Nano-LC, MALDI-TOF/TOF
MS, WB, IHC
Wang et al., 2010a
H4K20 Tri-methylation Rats (Kidney, Liver) N.S. HPLC Sarg et al., 2002
Enzyme Regulation Modification in Observed in Sample size Methods Reference

HDAC2 Expression SOD mice 48 IHC Chouliaras et al., 2013b
SIRT1 Rat Brain, Senescent cells 24–45,– WB, IHC, RT-PCR Quintas et al., 2012; Sasaki et al.,2006; Sommer et al., 2006
PRC1, PRC2 hAD-MSCs, hUCB-MSCs ICC, WB, RT-PCR Jung et al., 2010

indicates increased expression levels,

indicates decreased expression levels, and

indicates altered expression, not further specified.

Abbreviations: H, histone; HDAC, histone deacetylase; JMJD3; jumonji domain containing 3, K, lysine; p, phosphate modification; PRC; polycomb repressive complex, S, serine; SAMP, senescence-accelerated prone mouse 8; SIRT, sirtuin; SOD, copper-zinc superoxide dismutase 1; hAD-MSC; human adipose tissue-derived mesenchymal stem cells, hUCB-MSC; human umbilical cord blood-derived MSCs N.S., not specified; WB, Western blot; Nano-LC, nano liquid chromatography; MALDI-TOF, matrix assisted laser desorption/ionization time-of-flight; TOF MS, time-of-flight mass spectrometry; ICC; immunocytochemistry, IHC, immunohistochemistry; ChIP Seq, chromatin immunoprecipitation sequencing; HPLC: high-performance liquid chromatography; RT-PCR, real time-PCR.

The finding that HDAC2 expression increases with age in the mouse hippocampus is in line with findings of decreased acetylation levels (Chouliaras et al., 2013b). Age-associated reduction in acetylated H4 is thought to reduce chromatin structural plasticity and may result in a decreased accessibility of the DNA for repairing enzymes and other regulatory factors (Perry and Chalkley, 1982; Pina et al., 1988). Other studies are more specific, pointing towards a role of deregulated H4K12ac in age-related memory impairment (Peleg et al., 2010), a negative influence of HDAC2 on synaptic plasticity and memory formation through the suppression of neuronal gene transcription (Fischer et al., 2007; Guan et al., 2009), and a dependence of histone acetylation on citrate levels (Wellen et al., 2009), which decline in the aging brain (Jiang et al., 2008). Furthermore, the KAT CREBBP is important for long-term memory formation and late-phase long-term potentiation in the hippocampus of mice (Alarcón et al., 2004; Korzus et al., 2004). Apart from histone acetylation, H3K4me3 (Gupta et al., 2010) and H3 phosphorylation (Chwang et al., 2006) are also involved in memory formation. The SIRT HDACs have also been implicated in aging. In contrast to HDAC2, SIRT1 levels were found to drop with age, a change not limited to the brain (Quintas et al., 2012; Sommer et al., 2006) and also observed in senescent cells (Sasaki et al., 2006). Reduced levels of SIRT1 have been associated with increased levels of H4K16ac in vitro (Pruitt et al., 2006). SIRT1 can in addition directly deacetylate the HKMT suppressor of variegation 3–9 homologue (SUV39H) 1, which increases the activityofSUV39H1 (Vaqueroetal., 2007).This HKMT is responsible for H3K9me3, which is important for the formation of facultative heterochromatin. Despite the association between senescence and decreased H3K9me3 levels, H3K9me3 is thought to accumulate in senescence-associated heterochromatin foci (SAHF), a form of facultative heterochromatin, which are thought to induce senescence through the repression of the pro-proliferation E2F transcription factor family (Narita et al., 2003; Ye et al., 2007). Alternatively, in C. elegans, it has been observed that sir-2.1, the ortholog of mammalian SIRTI, can extend lifespan through its product nicotinamide (Schmeisser et al., 2013). Nicotinamide can be methylated by nicotinamide-N-methyltrans-ferase-1, producing 1-methylnicotinamide, and 1-methylnicotina-mide in turn is processed by aldehyde oxidase gastrulation defective 3 (GAD-3) to generate hydrogen peroxide. This hydrogen peroxide is thought to play a mitohormetic role, inducing longevity (Ristow and Zarse, 2010). Reinstating SIRT levels, for instance through caloric restriction, has in addition been reported to increase lifespan in yeast, invertebrates, and vertebrates (Guarente and Picard, 2005; Rutten et al., 2010), and has been argued to facilitate healthy aging in humans, thereby slowing the development of age-related neurodegenerative diseases such as AD (Baur et al., 2006; Haigis and Sinclair, 2010).

2.3. Non-coding RNAs in aging

One of the first ncRNAs reported to affect the aging process was miRNA lin-4, whose expression was observed to modulate lifespan in C. elegans (Lee et al., 1993). In neurons of C. elegans, miR-71 promotes longevity through the dauer 16/forkhead box O (DAF-16/ FOXO) pathway, increasing resistance towards heat shock and oxidative stress (Boulias and Horvitz, 2012). Other studies found increased levels of some miRNAs with age, but did not detect any significantly downregulated miRNAs in mice (Maes et al., 2008; Table 3). In human blood mononuclear cells, however, various miRNAs were significantly decreased in older participants (Noren Hooten et al., 2010; Table 3 ). Altered expression of various miRNAs, has been linked to age-related cardiovascular problems (Boon et al., 2011, 2013; Menghini et al., 2009; Olivieri et al., 2013; Vasa- Nicotera et al., 2011; Table 3). Moreover, various members of the miR-17–92 cluster were reported to be downregulated in humans (Hackl et al., 2010; Table 3). Another study in human endothelial cells detected additional miRNAs affected by age (Vasa-Nicotera et al., 2011; Table 3). Increased ROS levels in human endothelial cells were observed to induce miR-200c and concomitant initiation of apoptosis and senescence (Magenta et al., 2011 ). Several studies have recently shown the importance of certain miRNAs specific to the aging brain and their roles in the development of neurodegenerative diseases (Hebert and De Strooper, 2009; Somel et al., 2010). In the cortex and cerebellum of humans, chimpanzees, and macaque monkeys, miR-144 was observed to be upregulated (Persengiev et al., 2011). This miRNA targets the ataxin-1 gene, which is critically involved in the development of spinocerebellar ataxia type 1, and its age-related dysregulation could thus facilitate the development of this disease. Li et al. (2011) forged a link between aberrant miRNA expression and age-related declines in mitochondrial respiration rates. They found 70 miRNAs to be upregulated in the aging mouse brain, 27 of which were implicated in the downregulation of mitochondrial complexes III, IV and F0F1-ATPase that are all pivotal to the oxidative phosphorylation process. Interestingly, in the SAMP8, a mouse model of accelerated aging, miR-16 was found to be dysregulated. This miRNA modulates AD-related APP protein expression and with age APP levels were shown to drastically increase in the hippocampus of SAMP8 mice, leading to the suggestion that this model might serve as a model for AD (Liu et al., 2012b). Table 3 provides an overview of some of the ncRNAs that undergo age-related changes.

Table 3.

Epigenetic dysregulation in aging: non-coding RNAs.

Regulation ncRNA Observed in Sample size Approach Methods Reference

miRNA lin-4 C. elegans N.S. Targeted RFLP mapping, Southern
Blot, Northern Blot
Lee et al., 1993
miR-34 C57BL/6J mice N.S. Genome-wide microRNA microarray Maes et al., 2008
miR-93
miR-2l4
miR-669c
miR-709
miR-24 Human (blood) 10-14 Targeted miRNome miRNA profiling Noren Hooten
miR-103 (800 miRNAs) et al., 2010
miR-107
miR-l28
miR-130a
miR-155
miR-221
miR-496
miR-1538
miR-29 C57BL/6J mice and human Mice: 4; Human: l09 Genome-wide microRNA microarray Boon et al., 2011
miR-34a Sirt1+/−, Ku80−/− and
miR-34a−/− mice
≥4 Targeted (570 miRNAs) PCR, lSH Boon et al., 2013
miR-l46a HUVEC - Genome-wide microRNA microarray Vasa-Nicotera et al., 2011
HUVEC, HAEC, HCAEC - Targeted (367 miRNAs) microRNA microarray, PCR Olivieri et al., 2013
miR-217 HUVEC, HAEC, HCAEC - Targeted (10 miRNAs) microRNA microarray, PCR,
Northern Blot
Menghini et al., 2009
miR-17 Human cell lines - Targeted (599 miRNAs) microRNA microarray, PCR Hackl et al., 2010
miR-l9c
miR-20a
miR-106a
miR-l46a HUVEC - Genome-wide microRNA microarray Vasa-Nicotera et al., 2011
miR-26a
miR-181a
miR-221
miR-200b
miR-200c Human cell lines - Targeted
(1 miRNA)
WB Magenta et al., 2011
miR-144 Human (Cortex and Cerebellum)
Chimpanzee
Macaque monkey
8
4
5
Genome-wide microRNA microarray Persengiev et al., 2011
miR-l6 SAMP8 mouse N.S. Targeted
(7 miRNAs)
PCR, ISH, IHC Liu et al., 2012b

indicates increased expression levels,

indicates decreased expression levels, and

indicates altered expression, not further specified.

Abbreviations: C57BL/6J mice, C57 black 6 inbred mouse strain; C. elegans, Caenorhabditis elegans; miR, micro RNA; SAMP8 mouse, senescence-accelerated prone mouse 8; HUVEC, human umbilical vein endothelial cells; HAEC, human aortic endothelial cells; HCAEC, human coronary artery endothelial cells; N.S., not specified; RFLP, Restriction fragment length polymorphism; PCR, polymerase chain reaction; ISH: in situ hybridization; WB, Western blot; IHC, immunohistochemistry.

3. Neurodegeneration

Neurodegenerative diseases typically involve a progressive loss of neuronal integrity and function, followed by neuronal death. Depending on where in the brain the loss of integrity and neuronal loss occur, various functional disabilities may arise and which gradually worsen as the neurodegeneration spreads. The underlying cause and localization of the neurodegenerative processes, however, often vary between different neurodegenerative disorders. Some of the most common include AD, PD and HD, but also amyotrophic lateral sclerosis and prion diseases are well studied forms of neurodegeneration (Coppede et al., 2006). Multiple sclerosis is more recently also being investigated as a neurodegenerative disease (Trapp and Nave, 2008). The exact etiology of most neurodegenerative diseases is far from clear, while in some cases, such as for HD (Bates et al., 2006), it is clear that the origin is largely genetic, for others, including sporadic AD and PD, the link between genetics and disease development is much more complex, possibly involving gene-gene and gene-environment interactions (Chouliaras et al., 2010a,b; Migliore and Coppede, 2009). Numerous studies have, where genetics did not give simple answers, investigated other possible instigators, of which epigenetic mechanisms seem to be most promising. Although it remains to be elucidated whether dysfunctional epigenetic machinery plays a causal role, it has been critically implicated in various neurodegenerative processes. Additionally, environmental factors enjoy much attention as either direct modulators of disease development, or indirect via genetic or epigenetic pathways (Babenko et al., 2012; Chouliaras et al., 2010a,b; Jaenisch and Bird, 2003).

3.1. Alzheimer’s disease

The most ubiquitous neurodegenerative disorder and form of dementia is AD, with an estimated worldwide prevalence of over 35 million cases (Selkoe, 2012). Mainly characterized by cognitive decline, late-stage AD concomitantly involves progressive motor aberrancies, mood instabilities and other behavioral and physical abnormalities (Bediou et al., 2009; Budson and Solomon, 2012). Although most of these symptoms arise as a result of cortical degeneration, others are due to degeneration of subcortical or autonomic function-related areas. It should be noted, however, that AD pathology does not equally affect the whole brain, as certain brain areas and cell types are specifically vulnerable to AD pathology (Hardy, 2006). Among the areas mainly affected by degeneration in AD are the frontal cortex, temporal and parietal lobes, including the hippocampus and entorhinal cortex (EC), and the cingulate gyrus, whereas the cerebellum is largely spared (Wenk, 2003). Interestingly, there is some evidence indicating that, while the cerebellum is mostly spared, the Purkinje cells are specifically targeted by AD pathology (Fukutani et al., 1996). Despite numerous pre-clinical and clinical trials for AD treatments, only basic symptom management therapies are currently Food and Drug Administration-approved (some acetylcholinesterase inhibitors and an N-methyl-D-aspartic acid [NMDA] receptor antagonist), which cannot halt, or slow down the progressive neurodegeneration and the associated decline of memory, cognitive and executive functions. Apart from being a scourge among the elderly and the relatives of patients, dementia also incurs a tremendous socioeconomic burden; amounting to an estimated $200 billion in 2013 in the United States of America alone (Alzheimer’s Association, 2013).

AD is a complex, multifaceted disorder, involving dysregulated homeostasis on various fronts, including energy metabolism, inflammation, and cell cycle control (Mastroeni et al., 2010), likely resulting from a complex interplay between genetic, epigenetic and environmental factors (Coppede et al., 2006; Mastroeni et al., 2011). Despite much research into the pathophysiology of AD, including amyloid β (Aβ) and phosphorylated tau proteins (Tiraboschi et al., 2004), its exact etiology remains to be elucidated (Mill, 2011). Aβ, which exists in monomeric, oligomeric, and aggregated forms (senile plaques), is the product of APP cleavage by the β- and γ-secretases (Citron et al., 1995; Shoji et al., 1992). APP cleavage by γ-secretases can result in either Aβ40 or Aβ42, of which Aβ42 is thought to be especially neurotoxic. APP can also be cleaved by α-secretases such as a disintegrin and metalloproteases domain 10 (ADAM10) and tumor necrosis factor alpha (TNF-α) converting enzyme (TACE), but that this cleavage does not result in Aβ, but generates APPs-α, which is thought to be neuroprotective (Groen, 2010). One of the most prominent theories of AD pathology is the amyloid hypothesis, which states that Aβ is responsible for initiating the pathogenic pathway that leads to neurodegeneration and dementia in AD. Generally, this theory proposes that neurodegeneration is the result of impaired Aβ homeostasis, which leads to aberrant calcium homeostasis, triggering - and sensitizing cells to -damaging processes, including excitotoxicity and the formation of neurofibrillary tangles (NFTs) (Hardy and Higgins, 1992; Mattson etal., 1992; Selkoe, 1993). This hypothesis is applicable especially to early onset familial types of AD (fAD), which have a much more evident genetic component than the far more common late onset sporadic AD (sAD) (Tanzi, 2012). Mutations in the APP gene and the presenilin (PS) genes PS1 and PS2, have been observed in fAD cases (Czechetal.,2000; DeStrooperetal., 1998; Goateetal., 1991; Hardy and Higgins, 1992; Migliore and Coppede, 2009; Sherrington et al., 1995; Tanzi et al., 1996). PS1 and PS2 are γ-secretase-associated proteins involved in the generation of Aβ from APP, and PS mutations are able to bias this process towards Aβ42 production, the 42 amino acid-long Ab isoform that is more prone to aggregate then the shorter Aβ40 isoform, by either increasing Aβ42 production, or lowering Aβ40 production (Lemere et al., 1996). This relation has been corroborated by the detection of elevated Aβ42 levels in the blood and brains of fAD cases with PS mutations (Czech et al., 2000). Their major impact on disease development has led to the widespread use of mutant forms of the APP and PS genes to generate animal models of AD (see (Brasnjevic et al., 2013)). Although some mutations in the PS and APP genes seem to play a large role in disease development in fAD cases, most of the sAD susceptibility genes, including the risk factor with the highest population-attributable risk, the ε4 allele of the apolipoprotein E (APOE) gene and those identified through genome-wide association studies (ATP-binding cassette, sub-family A [ABC1], member 7 [ABCA7], clusterin [CLU], complement component receptor 1 [CR1], cluster of differentiation 33 [CD33], phosphatidylinositol-binding clathrin assembly protein [PICALM], membrane-spanning 4-domains, subfamily A, member 6A [MS4A6A], membrane-spanning 4-domains, subfamily A, member 4E [MS4A4E], cluster of differentiation 2-associated protein [CD2AP]) have a relatively minor influence on AD progression when altered (Cacabelos, 2005, 2007; Slooter et al., 1998). Moreover, despite the robust association with sAD of some of these common sequence variants, it remains largely unknown how they influence the development and course of sAD (Harold et al., 2009; Hollingworth et al., 2011; Naj et al., 2011; Sleegers et al., 2010). The same applies to the rare mutations recently discovered in the triggering receptor expressed on myeloid cells 2 (TREM2) gene, although they confer a much larger increase in risk to develop sAD than the common sequence variants (Guerreiro et al., 2013; Jonsson et al., 2013; Neumann and Daly, 2013). Although these genetic risk factors may be informative in screening for populations at risk to develop sAD, it has not yet been discovered how they exactly affect AD development (Slooter et al., 1998). Most is known about the involvement of the major risk factor APOEε4. For instance, increased levels of brain APOEε4 mRNA in AD cases, compared to controls with the same allele, were detected (Yamagata et al., 2001). Interestingly, the APOEε3 allele is thought to protect against Ab neurotoxicity (Caesar and Gandy, 2012). Additionally, a study with a transgenic mouse model of AD expressing human APOE isoforms indicated that different APOE alleles might influence clearing soluble Ab from the brain (Castellano et al., 2011). This is in line with evidence indicating that sAD is characterized by an inability to clear Aβ from the brain and not an increase Aβ production (Mawuenyega et al., 2010). A similar effect is suggested for the CLU gene, another important risk gene associated with sAD, implicating it in the aggregation and clearance of Ab, thereby mainly influencing age of onset and progression (Yu and Tan, 2012). Apart from the gene, CLU levels were shown to be elevated in the cerebrospinal fluid and brains of AD patients and CLU plasma levels were associated with several AD hallmarks (Schrijvers et al., 2011; Thambisetty et al., 2010).

Besides an abnormal Aβ homeostasis, dysfunctional tau has also been pointed out as a pivotal player in AD pathology. Tau, a microtubule-associated protein that promotes microtubule assembly (Weingarten et al., 1975), becomes hyperphosphorylated in AD. This causes it to dissociate from microtubules and aggregate, which induces cytoskeletal disorganization, neuronal dysfunction and cell death (Lee et al., 2001; Lovestone and Reynolds, 1997). This pathological process of aggregation is thought to play a part in the neurodegeneration and memory deficits as seen in AD (Alonso et al., 1997; Iqbal et al., 2009). Interestingly, while a similar process occurs in other tauopathies, diseases involving pathological tau aggregation, these generally involve mutations of the tau encoding microtubule-associated protein tau (MAPT) gene, whereas such mutations are usually not found in AD cases (Klafki et al., 2006; Lee et al., 2001). Mitochondrial abnormalities have also been investigated as contributors to AD pathogenesis, mainly in relation to energy imbalances and increased reactive oxygen species (Khairallah and Kassem, 2011).

Aβ and tau have long been the direct focus of treatment strategies, involving potential aggregation inhibitors, immunotherapy, and enzyme modulators (Hardy and Selkoe, 2002). More recently, however, while the epigenetic involvement in neurodegeneration is being explored, the epigenetic machinery has become an attractive target for novel intervention strategies. That minor aberrancies in the epigenetic machinery can have widespread consequences on gene expression, combined with the sporadic and complex nature of AD, has led to a recent interest in the role of epigenetic factors in the etiology of AD (Lahiri et al., 2009; Mastroeni et al., 2011).

3.2. Parkinson’s disease

PD is the second most common progressive neurodegenerative disorder, affecting the dopaminergic neurons of the midbrain substantia nigra. Because the dopaminergic projections from the substantia nigra are crucially involved in the initiation of motor events, PD is mainly known for symptoms such as tremor, rigidity, bradykinesia, and gait disturbances (Jankovic, 2008). These motor disturbances are, however, complemented by psychiatric symptoms, autonomic impairments, sleep disturbances, and cognitive dysfunctions, including dementia, that are intrinsic to the disease pathology and may even precede the motor symptoms (Aarsland et al., 1999; Naimark et al., 1996; Riedel et al., 2008). These nonmotor symptoms are related to imbalances in other neurotransmitter systems, including serotonergic, noradrenergic, and cholinergic malfunctions (Francis and Perry, 2007). Furthermore, cognitive impairments in PD are generally accompanied by the occurrence of Lewy bodies in brain areas including the midbrain and cortex (van de Berg et al., 2012). Lewy bodies are cytoplasmic protein aggregates, consisting mainly of α-synuclein, parkin, and ubiquitin (Jellinger, 2009). Exactly what part Lewy bodies play in PD pathophysiology warrants additional investigation. As with AD, PD exists as a familial (fPD) and a sporadic (sPD) variant, of which the former is again much rarer. Synuclein alpha (SNCA), the gene encoding the presynaptic protein α-synuclein, is one of the cardinal risk genes for PD; increased expression of only this gene (through point mutations and multiplications) can already induce familial Parkinsonian syndromes (Singleton et al., 2003; Thomas and Beal, 2011). In addition to SNCA, MAPT, Parkinson disease 16 (PARK16) and leucine-rich repeat kinase 2 (LRRK2) are also indicated as risk genes, with SNCA and MAPT SNPs conferring the highest risk (Edwards et al., 2010; Simon-Sanchez et al., 2009). Although genetic predisposition remains a high risk factor for sPD, age and environmental variations are also thought to be highly influential (Ammal Kaidery et al., 2013; Houlden and Singleton, 2012; Veldman et al., 1998), with factors such as a rural environment increasing the risk to develop PD, while smoking and the consumption of coffee decrease the risk (de Lau and Breteler, 2006). Additionally, the development of sPD has often been linked with exposure to environmental toxins, of which 1- methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) has the most prominent link to developing PD, leading to its widespread use to induce PD-like symptoms in animal models (Fukuda, 2001; Kopin, 1987). A causal role in the development of PD of most other toxins, however, remains a point of controversy (Franco et al., 2010). Nevertheless, evidence is accumulating pointing towards a cardinal role of the epigenetic machinery in mediating the effect of chronic environmental exposures on alterations in gene expression that can lead to the development of late-onset neurodegenerative diseases (Kanthasamy et al., 2012). At least for some genes a mechanism of DNA methylation-induced allelic skewing is proposed as the underlying mechanism of how an epigenetic process can modulate the interaction between genotype and environment. DNA methylation-induced allelic skewing is the process by which the paternal and maternal alleles are differentially methylated, leading to the preferential expression of either one.

3.3. Huntington’s disease

In contrast to AD and PD, HD is primarily a genetic, autosomal dominant neurologic disorder, with the sporadic variant being rarer. When symptoms start to occur progress can be fast and will result in death, with no treatment options currently available to change this devastating process (Ryu et al., 2005). The most characteristic symptom of HD is chorea, but other prominent symptoms include cognitive deterioration and psychiatric disturbances. It is known that HD pathology is ignited by an expansion of a cytosine-adenine-guanine (CAG) repeat section, coding for glutamine, in the coding region of the Huntingtin (HTT) gene on chromosome 4p16.3 (MacDonald et al., 1993). Note that the familial and sporadic variants thus have the same genetic origin. A CAG repeat number of 36 units leads to the development of HD and sporadic cases are caused by de novo mutations that increase the repeat number to above the critical number, with high repeat numbers leading to a younger age of onset (Kremer et al., 1994; Myers et al., 1993). The primary risk factor for developing HD is thus having family members with HD, or members with a high CAG repeat number. The expansion results in a dysfunctional HTT protein, which has been shown to disrupt transcription via multiple pathways (Okazawa, 2003; Sugars and Rubinsztein, 2003). It remains, however, unclear exactly how the production of mutant HTT leads to the lethal neurodegeneration associated with HD (Thomas, 2006). Curiously, HD neurodegeneration is very region and cell-type specific, mainly affecting the medium-sized spiny neurons of the neostriatal nuclei, caudate nucleus and putamen, explaining the grave motor symptoms (Ferrante et al., 1985,1986,1987,1991; Graveland etal., 1985; Kowall etal., 1987; Lee et al., 2013a; Vonsattel et al., 1985). Despite the specificity of neurodegeneration in HD, the HTT protein can be found in neurons throughout the whole brain (Ross, 1997). Wildtype HTT is mainly situated in the cytoplasm, its exact function, however, remains elusive, with proposed roles in intracellular transport, autophagy, transcription, mitochondrial functioning and signal transduction (Cattaneo, 2003; Mangiarini et al., 1996; Nucifora et al., 2001; Ross and Poirier, 2004). Nevertheless, HTT is critical for survival, as complete deletion of the HTT gene results on nonviable offspring (Zhang et al., 2003). Mutant HTT was shown to impair fast axonal transport, destabilize microtubules, and through its interactions with a multitude of proteins it disrupts important cellular pathways leading to hampered proteolysis, mitochondrial dysfunction, oxidative damage, inflammatory reactions, excitotoxicity and induction of apoptosis (Beal and Ferrante, 2004; Szebenyi et al., 2003; Trushina et al., 2004). Additionally, evidence indicates that mutant HTT has a widespread impact on gene expression, through interactions with specific transcription factors (Li et al., 2002), interference with the core transcriptional machinery and posttranscriptional modifications of histones, skewing the chromatin towards a more condensed state (Thomas et al., 2008).

4. DNA (de)methylation in neurodegeneration

4.1. DNA (de)methylation in Alzheimer’s disease

Early epigenetic investigations related to AD by West et al. (1995) focused on DNA methylation, reporting an AD-specific hypomethylation of the APP gene promoter region in a single patient. This was confirmed by another study and linked to elevated Ab levels (Tohgi et al., 1999). A later study with a larger sample was, however, unable to replicate this finding, nor find any other significant AD-related abnormalities in MAPT, APP and PS1 methylation (Barrachina and Ferrer, 2009). Others also did not find significant AD-related methylation changes in the APP promoter Barrachina et al. did report the presence of low and high methylated CpG sites in and close to the APP promoter region, as did Fuso et al. for the APP, PS and BASE genes (Fuso et al., 2005). Conversely, Brohede et al. (2010) observed no methylation at the investigated CpG site of the APP gene in a small sample of fAD patients, in all brain areas investigated, including the frontal cortex, parietal cortex, temporal cortex and cerebellum, concluding that APP is not transcriptionally regulated by methylation. All in all, these studies provide inconclusive evidence of whether APP methylation is involved in AD, raising the need for studies clearly separating between sAD and fAD, investigating multiple CpG sites and ideally also differentiate between cell types instead of using homogenates of whole regions. Wang et al. (2008b) observed a high interindividual variance in promoter methylation of the PS1, APOE, MTHFR, and DNMT1 genes, and a particularly marked epigenetic drift in AD cases.

A finding relevant not only to global DNA methylation, but also for many other biochemical pathways, is a severe AD-associated reduction of SAM (up to 85%) and its demethylated metabolite SAH (up to 79%) in several neocortical areas, the hippocampus and putamen (Morrison et al., 1996). Additionally, cerebrospinal fluid levels of folate and SAM, and levels of SAM in the frontal cortex, occipital cortex, temporal cortex, putamen and hippocampus, were found to be decreased in AD cases (Bottiglieri etal., 1990; Morrison et al., 1996; Serot et al., 2001), concomitant with an increase in brain SAH levels (Kennedy et al., 2004). Accordingly, lower serum folate levels and increased plasma Hcy levels were observed in sAD patients versus controls (Coppede et al., 2012). Cell culture work has indicated that increased Hcy levels can be linked to enhanced tau hyperphosphorylation and subsequent NFT formation (Sontag et al., 2007), which may be the result of the inhibitory effect of Hcy on methyltransferases, thereby preventing the methylation of protein phosphatase 2A (PP2A), which is required for its proper activation. PP2A can dephosphorylate phosphorylated tau and its decreased activity thus promotes the hyperphosphorylation of tau. In both mouse Neuro-2a (N2a) cells expressing human mutant APP and transgenic mice expressing human mutant PS1 and APP, PP2A was also found to be hypomethylated, resulting in elevated tau phosphorylation (Zhou et al., 2008). Furthermore, antagonizing folate with methotrexate in rat primary neuron cultures heightened phosphorylated tau, APP and BACE levels (Yoon et al., 2007). Interestingly, hypomethylated PP2A, but not normally methylated PP2A colocalized with hyperphosphorylated tau in the hippocampus of rats and AD cases (Zhang et al., 2008).

The apparent importance of folate and vitamins B12 and B6 in maintaining SAM levels has stimulated investigations into the potentially protective effects of supplementing these vitamins to counteract cognitive decline and possibly the onset of dementia (Cacciapuoti, 2013). In vitro folate deprivation was able to induce global DNA hypomethylation, leading to an increased expression of BACE and PS1, but unaltered TACE, ADAM10 and APP expression (Fuso et al., 2005). SAM supplementation successfully restored the folate deficiency-induced abnormalities. In a follow-up study, mutant human APP transgenic mice deprived of folate, vitamin B12 and vitamin B6 (Fuso et al., 2008), showed increased SAH to SAM ratios and increased PS1 and BACE levels, thus corroborating the in vitro findings. These increases in PS1 and BACE expression were paired with elevated Aβ aggregation, early appearance of intraneuronal Ab and mild spatial learning and memory impairments. In a similar study, it was later shown that SAM supplementation was also able to remedy the vitamin B deficiency-induced detrimental effects in mice, resulting in a reduction in PS1 and BACE1 expression, amyloid production, tau phosphorylation, and subsequent enhanced spatial memory (Fuso et al., 2012). Vitamin B deficiency induced hypomethylation of CpG sites near the PS1 promoter, indicating that PS1 expression is indeed regulated by methylation (Fuso et al., 2011). Another group also found benefical effects of dietary SAM supplementation in the 3xTg-AD mouse model (Lee et al., 2012). Additionally, a vitamin/nutriceutical formulation including folate and vitamin B was shown to delay the progression of dementia in a small sample of early stage (Chan et al., 2008), and moderate to late stage AD (Remington et al., 2009).

Observations of an overall reduction in DNA methylation in AD patients are in line with these findings and further stress the importance of DNA methylation in AD (Mastroeni et al., 2010, 2011; Wang et al., 2008b). Interestingly, despite this AD-associated global DNA hypomethylation, specific loci of the MTHFR gene, which is crucial for SAM synthesis, were found to be hypermethy-lated, in both postmortem prefrontal cortex and peripheral lymphocyte samples of AD patients (Wang et al., 2008b).

Studies focusing on the hippocampus, one of the brain areas early affected by AD and aging, have observed that levels of 5-mC (Chouliaras et al., 2011b) and DNMT3a (Chouliaras et al., 2011a) increase with age in mice, whereas these levels are significantly decreased in the hippocampus of AD patients (Chouliaras et al., 2013a).

Siegmund et al. (2007) found an increase in the methylation of sorbin and v-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog homology 3 domain containing 3 (SORBS3) and a decrease in the methylation of S100 calcium-binding protein A2 (S100A2) in AD subjects, compared to controls of 60 years and older. Interestingly, although a progressive increase in SORBS3 and decrease in S100A2 methylation is normal with aging, this process is accelerated in AD. SORBS3 encodes a cell adhesion molecule and the product of S100A2 is observed in corpora amylacea, which are a hallmark frequently found in human brain aging and, in greater numbers, of neurodegenerative diseases (Hoyaux et al., 2000). A decline in SORBS3 expression might have a hand in the synaptic abnormalities associated with AD (Urdinguio et al., 2009). Intriguingly, recent epigenome-wide association studies identified another gene that encodes an adaptor protein and its methylation signature is highly associated with AD pathology. More specifically, a differential cortex-specific hypermethylated region of ankyrin 1(ANK1) was found to be associated with the early stages as well as the progression of AD neuropathology (De Jager et al., 2014; Lunnon et al., 2014).

Remarkably, Aβ has also been implicated as a trigger of epigenetic changes. Chen et al. (2009) found that Aβ induces global DNA hypomethylation, while promoting hypermethylation of NEP, a gene that encodes neprilysin. Neprilysin is one of the enzymes involved in Aβ degradation and its expression is known to decrease with aging and AD. This finding indicates that Aβ is able to induce a vicious cycle that depends on epigenetic processes and favors Aβ deposition. Other regulatory players may further enforce this cycle, for instance TNF-α and cysteine-dependent aspartate-directed protease (caspase)-3 were found to increase Aβ production, and they are increasingly expressed in response to hypomethylation (Muerkoster et al., 2008; Sommer et al., 2009; Wilson, 2008; Xiong et al., 2008).

Tau gene expression is also subject to complex epigenetic regulation, involving differentially methylated binding sites for transcription factors. It was found that with age, the activator- binding site for transcription factor SP1 became hypermethylated in the tau gene promoter region, whereas the repressor-binding site for GCF was hypomethylated in the human cerebral cortex (Tohgi et al., 1999), which might be relevant to AD and other age-related tauopathies. This points toward an age-related decrease in tau expression, which has indeed been detected in the human frontal cortex and hippocampus, but this did not correlated with NFT pathology (Mukaetova-Ladinska et al., 1996).

The APOE gene promoter has a low CpG count and generally exhibits low levels of DNA methylation. There is, however, a CpG island located at the 3’ end that is usually heavily methylated, and which contains the sequence of the APOE ε4-haplotype, the prime genetic risk factor for sAD (Wang et al., 2008b). It has been suggested that the £4 allele might disturb the epigenetic regulation of the APOE gene, as this allele is associated with a C>T transition, preventing this site from being methylated (Wang et al., 2008b). The CLUgene is more clearly regulated by epigenetic mechanisms, as its promoter regions contain a CpG island, the demethylation of which after 5- aza-2’-deoxycytidine (decitabine; DAC) treatment was shown to enhance CLU expression in cancer cell lines (Rauhala et al., 2008). A similar demethylating treatment in addition to the administration of HDAC inhibitors (HDACIs) has also been observed to increase CLU expression and secretion in human neurons and retinal pigment epithelial cells (Nuutinen et al., 2005; Suuronen et al., 2007).

There is increasing evidence of disturbed cell-cycle control and subsequent induction of apoptosis in degenerating AD neurons and, although not directly investigated, many of the proteins involved in these processes that have been shown to be upregulated in these neurons and are also known to be regulated through DNA methylation (Jee et al., 2005; Moreira et al., 2009; Muerkoster et al., 2008; Robertson and Jones, 1998; Tschöp and Engeland, 2007). In addition to genes involved in cell-cycle control, the promoter regions of cyclooxygenase-2 (COX-2) and nuclear transcription factor kappa B (NF-κB) were found to be hypo-methylated, while the promoter regions of brain-derived neuro-trophic factor (BDNF) and cAMP response element-binding protein (CREB) were hypermethylated in the frontal cortex of AD patients (Rao et al., 2012).

Bollati et al. (2011) specifically investigated blood for the methylation status of repetitive elements, including Arthrobacter luteus elements (Alu), long interspersed element 1 (LINE-1) and satellite-α (SAT-α), which comprise a large portion of the human genome and are known to contain large numbers of CpG sites. Interestingly, they found that LINE-1 methylation was increased in AD patients and that within the AD group enhanced LINE-1 methylation was associated with a better cognitive performance.

Although not as well studied in relation to AD as DNA methylation, the DNA demethylation process is receiving increased attention. As for 5-mC, 5-hmC levels were also found to be greatly decreased in the hippocampus of AD patients (Chouliaras et al., 2013a). This is in line with previous findings indicating a global DNA hypomethylation in EC NFT-bearing neurons of AD patients (Mastroeni et al., 2010). Additionally, it was found that global 5- hmC levels were decreased in the EC and cerebellum of AD subjects, while no significant disease-related changes in 5-mC, 5-fC and 5-caC were detected (Condliffe et al., 2014). In contrast, levels of 5-mC and 5-hmC were immunohistochemically found to be increased in the mid frontal gyrus and mid temporal gyrus of AD patients and positively correlated with Aβ, NFT, and ubiquitin load (Coppieters et al., 2014). This study included a cell-type specific analysis and found that 5-hmC and 5-mC were mainly present in Neuronal Nuclei (NeuN; a neuronal marker)-positive cells, with glial fibrillary acidic protein (GFAP; an astrocyte marker)-positive cells and ionized calcium-binding adapter molecule 1 (1BA1; a microglial/ macrophage maker)-positive cells only presenting with weak or no immunoreactivity. This latter study is in line with findings from Bradley-Whitman and Lovell (Bradley-Whitman and Lovell, 2013), who observed increased levels of TET1, 5-mC and 5-hmC in the hippocampus and parahippocampal gyrus in subjects with preclinical and late-stage AD. In addition, it was found that 5-fC and 5-caC levels were significantly decreased. Another study detected global hypermethylation in the frontal cortex of AD patients (Rao et al., 2012). Whether global 5-mC and 5-hmC levels are thus decreased or increased in AD remains to be conclusively determined. Possible factors contributing to the discordant findings have been suggested and include differences in the brain regions studied, tissue processing, and detection methods and protocols (Coppieters et al., 2014). An additional factor that could influence readings is whether a cell type-specific analysis is conducted, or different cell types are grouped together. Considering the uncertainty regarding global 5-mC and 5-hmC changes in relation to AD it might be a bit too early to speculate about the consequences of such changes. Nevertheless, Coppieters et al. (2014), who detected a global DNA hypermethylation and hyperhydroxymethy-lation argue that these changes may facilitate cell death, as the methylation of cytosines is thought to enhance the mutation rate of these cytosines and this increased mutation rate could facilitate the loss of neurons in AD. There are, however, also studies indicating that DNA hypomethylation leads to neuronal degeneration (Fan et al., 2001; Hutnick et al., 2009), suggesting that no simple conclusions can be made from observations of globally increased or decreased DNA methylation levels.

Münzel et al. (2010) observed an age-related increase in 5-hmC levels, which seemed to be especially prominent in genes associated with neurodegeneration. Another finding indicating DNA demethylation to play a role in the development of AD is a single nucleotide polymorphism (SNP) in the TET1 gene that was associated with sAD (Morgan et al., 2008). See Table 4 for an overview of the aberrant DNA (de)methylation in AD.

Table 4.

Epigenetic dysregulation in Alzheimer’s disease: DNA (de)methylation.

Gene Regulation Epigenetic modification Observed in Sample size Approach Methods Reference

APP Methylation (APP gene promoter region) AD patients 16

1
Targeted
(18 loci)
Targeted
(1 locus)
BS

Southern Blot
Tohgi et al., 1999

West et al., 1995
Methylation (CpG sites in and
close to the promoter region)
AD patients AD Stages I to II (n=17),
AD Stages III to IV (n=15),
AD Stages V to VI (n=12)
Targeted
(6 loci)
BS Barrachina and Ferrer,
2009, Fuso et al., 2005
PS Methylation (CpG sites in and
close to the promoter region)
SK-N-SH and
SK-N-BE cell lines
- Targeted
(5 loci)
HPLC/Nucleic
acid analysis
Fuso et al., 2005
BACE Methylation (CpG sites in and
close to the promoter region)
SK-N-SH and
SK-N-BE cell lines
- Targeted
(5 loci)
HPLC/Nucleic
acid analysis
Expression due to folate
deptivation-induced global
DNA hypomethylation
SK-N-SH and
SK-N-BE cell lines
- Targeted
(5 loci)
HPLC/Nucleic
acid analysis
PS1 Methylation (promoter region) AD patients 24 Targeted
(12 loci)
BS Wang et al., 2008b
Expression due to folate
deptivation -induced global
DNA hypomethylation
SK-N-SH and
SK-N-BE cell lines
- Targeted
(5 loci)
HPLC/Nucleic
acid analysis
Fuso et al., 2005
APOE Methylation (promoter
region)
AD patients 24 Targeted
(12 loci)
BS, MALDI Wang et al., 2008b
MTHFR Methylation (promoter
region)
AD patients
(prefrontal cortex,
peripheral
lymphocytes)
24 Targeted
(12 loci)
BS
Methylation
DNMT1 Methylation (promoter
region)
AD patients 24 Targeted
(12 loci)
BS
PP2A Inhibition of methylation due
to increase of Hγ levels
N2a cells, AD
mouse model
(APP/PS1)
-, N.S. Targeted
(1 locus)
IHC Zhou et al., 2008
SORBS3
S100A2

Methylation AD patients 18 Targeted
(50 loci)
BS, MS-PCR Siegmund et al., 2007
NEP Methylation CEC cell line
exposed to Aβ
- Targeted
(1 locus)
MS-PCR Chen et al., 2009
SP1 Methylation (activator
binding site)
Human
(cerebral cortex)
16 Targeted
(18 loci)
BS Tohgi et al., 1999
GCF Methylation (repressor
binding site)
Human
(cerebral cortex)
16 Targeted
(18 loci)
BS Tohgi et al., 1999
APOE Methylation (CpG island
located at the 3’ end)
AD patients
(postmortem)
24 Targeted
(12 loci)
BS Wang et al., 2008b
COX-2 Methylation (promoter
region)
AD patients
(frontal cortex)
10 Targeted
(8 loci)
Promoter
region-specific
CpG methylation
assay
Rao et al., 2012
NF-kB Methylation (promoter
region)
BDNF Methylation (promoter
region)
CREB Methylation (promoter region)
LINE-1 Methylation AD patients (blood) 43 Targeted
(3 loci)
BS, PCR,
pyrosequencing
Bollati et al., 2011
ANK1 ↑↑ Methylation AD patients (EC, superior
temporal cortex, PFC)
62, 122, 144 Genome-wide 450K BeadChip Lunnon et al., 2014
Protein/(hydroxy)
methylated base
Regulation Modification in Observed in Sample size Methods Reference

5-hmC Expression AD mouse model (APP/PS1; hippocampus)
AD patients (EC, cerebellum)
10
10
IHC
IHC
Chouliaras et al., 2013a
Chouliaras et al., 2013a
5-mC, 5-hmC AD patients (MGF, MTG, hippocampus) 7 late-stage AD IHC Bradley-Whitman and
5 preclinical AD Lovell, 2013
13 MGF 29 MTG IHC Coppieters et al., 2014
5-fC, 5-caC AD patients (hippocampus) 7 late-stage AD IHC Bradley-Whitman and
5 preclinical AD Lovell, 2013
DNMT3a AD mouse model (APP/PS1; hippocampus) 10 IHC Chouliaras et al., 2013a
AD patients 10 IHC Chouliaras et al., 2013a
DNMT3a2 Mice (12 months old) 15 IHC Oliveira et al., 2012
TET1 AD patients (hippocampus) 7 late-stage AD IHC Bradley-Whitman and
5 preclinical AD Lovell, 2013

indicates increased expression levels,

indicates decreased expression levels, and

indicates altered expression, not further specified.

Abbreviations: 450K BeadChip, Illumina Infinium HumanMethylation 450K BeadChip; 5-caC, 5-carboxylcytosine; 5-fC, 5-formylcytosine; 5-hmC, 5-hydroxymethylcytosine; 5-mC, 5-methylcytosine; AD, Alzheimer’s disease; APOE, apolipoprotein E; APP, amyloid β precursor protein; BACE, β-secretase; BDNF, brain-derived neurotrophic factor; CEC, cerebral endothelial cell, COX-2, cyclooxygenase-2; CpG, cytosine-phosphate-guanine; CREB, cyclic adenosine monophosphate response element-bindingprotein; DNMT, DNA methyltransferase; EC, entorhinal cortex; GCF, granulocyte chemotactic factor; LINE-1,longinterspersed element 1; ANK1,ankyrin 1; MALDI, matrix-assisted laserdesorption/ionization; MFG, middle frontal gyrus; MTG, middle temporal gyrus; MTHFR, 5,10-methylenetetrahydrofolate reductase; NEP, neprilysin; NF-κB, nuclear transcription factor kappa B; PP2A, protein phosphatase 2A; PS, presenilin; S100A2, S100 calcium-binding protein A2; SORBS3, sorbin and v-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog homology 3 domain containing3; SP1, specificity factor 1; TET1, ten-eleven translocation 1; SK-N-SH, SK-N-BE, human neuroblastoma cell lines; N2A, neuro-2a cell line; CEC, circulating endothelial cells; N.S., not specified; BS, bisulfite sequencing; HPLC: high-performance liquid chromatography; IHC, immunohistochemistry; MS-PCR: methylation specific-PCR.

4.2. DNA (de)methylation in Parkinson’s disease

Obeid et al. (2009) explored the relation between the methylation potential, represented by the SAM/SAH ratio, and cognitive performance in PD patients, and found that a higher methylation potential correlated with better cognitive capabilities. In addition, it was found that α-synuclein can associate with DNMT1, sequestering it in the cytoplasm, resulting in global DNA hypomethylation. This property of α-synuclein was not only found in PD cases, but also in dementia with Lewy bodies and a transgenic mouse model expressing human a-synuclein (Desplats et al., 2011). Because α-synuclein can also be observed in AD (Trojanowski et al., 1998), this mechanism might also contribute to the global DNA hypomethylation observed there. In vitro overexpression of DNMT1, as well as in transgenic mice, was able to normalize the nuclear localization of DNMT1.Jowaed et al. (2010) specifically investigated methylation of human SNCA and showed that expression of this gene is regulated through methylation of the first intron. Interestingly, a negative correlation between SNCA intron 1 methylation and SNCA expression has also been identified, and that SNCA methylation is decreased in the substantia nigra, putamen, and cortex of sPD patients (Jowaed et al., 2010; Matsumoto et al., 2010). Another study, investigating the high-resolution methylome of Lewy body disease cases, including PD, found, however, no overall differences in SNCA intron 1 methylation (de Boni et al., 2011). Although this study reported some differences at the single CpG level, it signifies that the extent of erroneous DNA methylation in PD warrants additional research efforts. Apart from SNCA, however, additional genes, including PARK16, glycoprotein (transmembrane) nmb (GPNMB) and syntaxin 1B (STX1B) have also reported to be differentially methylated in PD (Plagnol et al., 2011). A very recent EWAS in blood from PD patients, using a discovery and replication cohort, identified additional differentially methylated genes, with the most reliable differentially methylated CpGs being located in the Fanconi anemia, complementation group C (FANCC) and tankyrase, telomeric repeat binding factor 1 (TRF1)-interacting ankyrin-related ADP-ribose polymerase 2 (TNKS2) genes (Moore et al., 2014). How this aberrant DNA methylation exactly affects gene expression and ultimately influences PD pathology remains to be unveiled.

Interestingly, although a mutation in parkin has been associated with a juvenile form of PD, deviant methylation patterns in the promoter of this gene have been observed in myelogenous leukemia and acute lymphoblastic leukemia, but not PD (Agirre et al., 2006; Cai et al., 2011). Similar observations were made for deubiquitinating enzyme ubiquitin carboxy-terminal hydrolase L1 (UCHL1), a gene associated with PD, and ATPase type 13A2 (ATP13A2,) which causes a recessive form of Parkinsonism, failing to establish a relation between abnormal promoter methylation and PD, although the promoter of UCHL1 was found to be hypermethylated in cancer (Barrachina and Ferrer, 2009; Coppede, 2012; Thomas and Beal, 2011). Table 5 summarizes the findings regarding dysregulated DNA (de)methylation in PD.

Table 5.

Epigenetic dysregulation in Parkinson’s disease: DNA (de)methylation.

Gene Regulation Epigenetic modification Observed in Sample size Approach Methods Reference

SNCA Methylation sPD patients (SN,
Putamen, Cortex)
6, 14 Targeted
(1 locus)
BS Jowaed et al., 2010
11 Targeted
(1 locus)
BS Matsumoto et al., 2010
PARK16 Methylation PD patients 12.386 Genome-wide methQTL Plagnol et al., 2011
GPNMB Methylation PD patients
STX1B Methylation PD patients
FANCC Methylation PD patients 30 Genome-wide Infinium Moore et al., 2014
TNKS2

indicates decreased expression levels and

indicates altered expression, not further specified.

Abbreviations: FANCC, fanconi anemia group C; GPNMB, glycoprotein (transmembrane) nmb; PARK16, Parkinson disease 16; (s)PD, (sporadic) Parkinson’s disease; SN, substantia nigra; SNCA, synuclein alpha; sPD, sporadic PD; STX1B, syntaxin 1B; TNKS2, tankyrase 2; BS, bisulfite sequencing; GWA: genome-wide association; methQTL, methylation quantitative trait locusc.

4.3. DNA (de)methylation in Huntington’s disease

DNA methylation states have been investigated in transgenic models, and to a lesser extend in HD patients (Ng et al., 2013; Villar-Menendez et al., 2013; Wood, 2013; Table 6). Promoter regions of genes important for neurogenesis were found to be hypermethylated in the presence of mutant HTT (Ng et al., 2013; Table 6). Although these findings need to be replicated in HD patients, reduced hippocampal neurogenesis might partially underlie the cognitive impairments seen in HD (Lee et al., 2013a). Decreased expression of the adenosine A2a receptor in HD patients is also epigenetically regulated. In both HD patients and transgenic mice adenosine A2a receptor expression was observed to be downregulated (Wood, 2013). However, in patients this was associated with increased 5’ UTR DNA methylation, whereas in the mouse model with decreased 5’ UTR DNA hydroxymethylation of the adenosine A2a receptor gene (ADORA2A). This finding indicates that epigenetic regulation might differ between species and illustrates the importance of replicating findings in human cases. The widely neglected 7-mG form of DNA methylation, which also occurs in RNA, was found to be disturbed in HD mouse models and patients, in both the nucleus and cytoplasm, the latter primarily reflecting methylated RNA (Thomas et al., 2013).

Table 6.

Epigenetic dysregulation in Huntington’s disease: DNA (de)methylation.

Gene Regulation Epigenetic modification Observed in Sample size Approach Methods Reference

Ap-1 ↑↑ Methylation of Mouse striatal neurons - Targeted RRBS, MeDIP-Seq, Ng et al., 2013
Sox2 ↑↑ promoter region overexpressing HTT (10 loci) ChIP-Seq Assay
Pax6 ↑↑
Nes ↑↑
7-mG DNA and RNA
methylation
HD mouse models
(R6/2, CAG140 Kl mice)
HD patients
10

N.S.
ECD/HPLC Thomas et al., 2013

indicates increased expression levels and

indicates altered expression, not further specified.

Abbreviations: 7-mG, 7-methylguanine; Ap−1, activator protein 1; CAG140 KI mice, transgenic mouse model overexpressing human HTT with 140 cytosine-adenine-guanine repeats; HD, Huntington’s disease; HTT, Huntingtin; Nes, nestin; Pax6, paired box 6; R6/2 mice, transgenic mouse model overexpressing exon 1 of human HTT with an expanded cytosine-adenine-guanine repeat length; Sox2, sex-determining region Y (SRY)-box 2; UTR, untranslated region; N.S., not specified; RRBS, reduced representation bisulfite sequencing; MeDIP-Seq, methylated DNA immunoprecipitation sequencing; ECD, electrochemical detection; HPLC, high-performance liquid chromatography; ChIP-Seq, chromatin immunoprecipitation sequencing.

5. Chromatin remodeling in neurodegeneration

5.1. Chromatin remodeling in Alzheimer’s disease

Going from the DNA to the chromatin level, additional epigenetic dysregulation can be observed in AD (Table 7). Histone acetylation was found to be drastically decreased in the temporal lobe of AD patients when compared to aged controls (Zhang et al., 2012), but also in animal models of AD (Graff et al., 2011). The importance of gene-specific investigations apart from global changes in epigenetic markers is exemplified by the observation of increased H3 acetylation at the promoter region of the BACE1 gene in AD patients (Marques et al., 2012). The increase in H3 acetylation enhanced promoter accessibility and subsequent gene expression. Importantly, it was found that indirectly enhancing histone acetylation through chronic treatment with HDACIs was able to reverse cognitive deficits in double transgenic mice overexpressing human APP isoform 695 with the double KM670/671NL Swedish mutation (APPswe) and the human PS1 deleted in exon 9 mutation (PS1ΔE9) (APPswe/PS1ΔE9 mice) (Kilgore et al., 2009). The mechanism of action of HDACI treatment might be related to the finding that dysregulation of H4K12ac is implicated in mediating cognitive impairment exhibited seen in aged mice, impairments which were alleviated through HDACI administration (Peleg et al., 2010). Another study using transgenic APP/PS1 mice observed diminished acetylation of H4 and linked this to memory impairments, which could be alleviated through trichostatin A (TSA), an HDACI, administration (Francis et al., 2009). Decreased histone acetylation is in line with the discovery of elevated nuclear translocation of EP300 interacting inhibitor of differentiation 1 (EID1) in cortical neurons of AD subjects (Liu etal., 2012a). EID1 inhibits EP300 and CREBBP, important KATs, and the overexpression of EID1 in mice resulted in learning and memory impairments thought to be the result of this inhibition. In the triple transgenic mouse model of AD (3xTg-AD mice) CREBBP expression was also decreased, while overexpression of CREBBP elevated BDNF levels and restored memory function in this AD model (Caccamo et al., 2010). Additionally, expression of a truncated inhibitory form of EP300 impaired memory in transgenic mice (Oliveira et al., 2011; Oliveira et al., 2007). Curiously, while knockout of KAT2B resulted in memory impairments in mice (Maurice et al., 2008), such mice are resistant to the neurotoxic effects of Ab injected into the lateral ventricles in another study (Duclot et al.,2010).

Table 7.

Epigenetic dysregulation in Alzheimer’s disease: chromatin remodeling.

Chromatin Remodelling Regulation Epigenetic Observed in Sample Methods Reference
Target modification size

Histones (Globally) Acetylation AD patients 5–6 Targeted Proteomics
assay
Zhang et al., 2012
H3 Acetylation (BACEl
promoter region)
AD patients 31 FAlRE/ChIP Marques et al., 2012
Acetylation AD mouse model (3xTg-AD
mice; 4 months of age)
6 IF Walker et al., 2013
AD mouse model (Tg2576 mice;
PFC)
N.S. N.S. Lithner et al., 2009
Phosphorylation AD mouse model (Tg2576 mice; PFC)
AD patients (Hippocampus)
Methylation AD mouse model (Tg2576 mice;
PFC)
Methylation AD mouse model (Tg2576 mice; Striatum)
H3K9 Tri-methylation AD monozygotic twin (temporal
cortex and hippocampus)
l IHC Ryu et al., 2008
H4 Acetylation AD mouse model (APP/PS1) 4 WB Francis et al., 2009
Acetylation AD mouse model (3xTg-AD
mice; 4 months of age)
6 IF Walker et al., 2013
Acetylation AD mouse model (Tg2576 mice;
CA1)
N.S. N.S. Lithner et al., 2009
H2AX (S1S9) Phoshorylation AD patients (Hippocampus and
Astrocytes)
13 ICC Myung et al., 2008
HDAC2 Expression AD mouse models (CK-p25,
5xFAD, Cdk5cKO mice;
Hippocampus and PFC)
AD patients (Hippocampus and EC)
6–9
4–8
IHC, WB, Co-IP,
ChIP, PCR
Graff et al., 2012
H2BKS Acetylation AD mouse models (CK-p25,
5xFAD, Cdk5cKO mice;
Hippocampus and PFC)
6–9
HSKl4 Acetylation AD mouse models (CK-p25, 6–9
H4KS Acetylation 5xFAD, Cdk5cKO mice;
Hippocampus and PFC)
H4Kl2 Acetylation Mice (16 month old) 4–5 ChIP, PCR,
HAT/HDAC assay
Peleg et al., 2010
SlRTl Expression AD patients (parietal cortex) l9 WB, ISH Julien et al., 2009
Expression AD mouse model (3xTg-AD
mice)
N.S.

indicates increased expression levels,

indicates decreased expression levels, and

indicates altered expression, not further specified.

Abbreviations: 3xTg-AD mice, triple transgenic mouse model of AD; 5xFAD, transgenic mouse model overexpressing mutant human APP (695) with the Swedish (K670N and M671L), Florida (I716V), and London (V717I) mutations, and mutant human presenilin 1 with the M146L and L286V familial AD mutations; AD, Alzheimer’s disease; BACE1, β-secretase 1; CA, cornu ammonis; Cdk5 KO mice; cyclin-dependent kinase 5 knock-out mouse model; CK-p25 mice, transgenic mouse model overexpressing p25 under control of an inducible calcium/ calmodulin-dependent protein kinase II α promoter; EC, entorhinal cortex; H, histone; HDAC, histone deacetylase; K, lysine; PFC, prefrontal cortex; S, serine; SIRT, sirtuin; Tg2576 mice, transgenic mice overexpressing human APP isoform 695 with the double KM670/671NL Swedish mutation; N.S., not specified; FAIRE, formaldehyde-assisted isolation of regulation; IF: immunofluorescence; ICC, immunocytochemistry; Co-IP, protein complex immunoprecipitation; IHC, immunohistochemistry; ChIP, chromatin immunoprecipitation; PCR, polymerase chain reaction; WB, Western blot; ISH: in situ hybridization.

Conversely, cultured neurons from 3xTg-AD mice and non-transgenic controls, harvested at different ages, revealed an increased H3 and H4 acetylation levels from an age of 4 months, which is before the onset of memory impairments in this model of AD (Walker et al., 2013). With normal aging, H3 acetylation levels seem to remain unchanged, whereas H4 acetylation levels decreased, but administration of Aβ to the non-transgenic neurons increased acetylation levels. The repressive H3K9 mark in these same neurons increased with age in both the transgenic and non-transgenic neurons, but was more prominent in the transgenic cells. This later finding has been corroborated in humans; comparing two monozygotic twins discordant for AD it was found that the one with AD exhibited higher levels of H3K9me3 in the temporal cortex and hippocampus (Ryu et al., 2008). Using transgenic mice overexpressing APPswe (Tg2576 mice), increased H3 acetylation levels were found in the prefrontal cortex, as well as increased H4 acetylation levels in the CA1 region of the hippocampus (Lithner et al., 2009). Additionally, they also reported elevated levels of H3 phosphorylation and methylation in the prefrontal cortex, but decreased H3 methylation in the striatum.

While the use of non-selective HDACIs is a promising strategy for the treatment of cognitive problems, it might be even better to target the specific HDACs that induce the memory problems. Currently, HDAC2 is a prime suspect (Graff et al., 2012; Guan et al., 2009). Especially the group of Graff and Tsai has contributed significantly in this respect, starting with their detection of increased levels of HDAC2 in the hippocampus and prefrontal cortex of a mouse model of AD, while levels of the related HDAC1 and HDAC3 were not affected. Note, however, that recently it was reported that MS-275 treatment, an HDACI that favors HDAC1, was able to partially alleviate behavioral deficits, neuroinflammation and plaque load in transgenic mice overexpressing APPswe and human PS1 with the L166P mutation, line 21 (APP/PS1–21 mice) (Zhang and Schluesener, 2013), and that HDAC3 inhibition enhanced long-term memory formation in the C57 black 6 inbred mouse strain (C57BL/6 mice) (McQuown et al., 2011). To study the effects of this HDAC2 dysregulation at the gene level, Graff et al. (2012) focused on genes involved in learning, memory and synaptic plasticity that were previously shown to be down-regulated in the AD brain and found that HDAC2 was significantly enriched at the promoter and coding regions of these genes, in their mouse model. In addition, they found several acetylation marks, associated with neuroplasticity, to be hypoacetylated (Table 7). Subsequently, it was shown that increased localization of HDAC2 to the investigated genes and hypoacetylation negatively correlated with RNA polymerase II (RNAPII) binding and mRNA expression. Interestingly, knock-down of HDAC2 ameliorated the cognitive problems and aberrant synaptic plasticity. It was then investigated how HDAC2 could be induced in AD, by testing in vitro the effect of the AD associated neurotoxic stimuli hydrogen peroxide and Aβ in primary hippocampal neurons. Both stimuli were found to enhance HDAC2 mRNA level through activation of glucocorticoid receptor 1 (GR1). Importantly, HDAC2 was also investigated in human postmortem brain samples from AD patients revealing that in actual AD cases HDAC2 was markedly increased in the hippocampus and EC. Already at Braak stages I and II HDAC2 levels were found to be significantly elevated in hippocampal CA1 field and the EC, indicating that increased HDAC2 activity might be involved in the early stages of AD. In addition to HDAC2, HDAC6 levels were found to be significantly higher in the hippocampus of AD cases when compared to controls (Ding et al., 2008). Interestingly, HDAC6 is thought to interact with tau, affecting its phosphorylation and aggregation (Simões-Pires et al., 2013). HDAC6 has been suggested to make tau vulnerable to phosphorylation through deacetylation, a finding relevant to tauopathies in general (Cook et al., 2014). HDAC6 also indirectly affects tau clearance through deacetylation of chaperone protein heat shock protein 90 (HSP90), which affects its drive towards refolding or degradation (Cook et al., 2012). It has been reported that tau can actually act as a HDAC6 inhibitor (Perez et al., 2009). Accordingly, in a mouse model for AD, reduction of HDAC6 levels mitigated learning and memory problems (Govindarajan et al., 2013). Studies on HDAC6 suggest that the role of HDACs in neurodegeneration might not solely depend on the deacetylation of histones, but also on the deacetylation of other targets, such as a-tubulin in the case of HDAC6 (Govindarajan et al., 2013; Simoes-Pires et al., 2013; Xiong et al., 2013). The same holds true for KATs, as the KAT human immunodeficiency virus type 1 transactivating protein interactive protein (TIP60/KAT5), the proposed counterpart of HDAC6 (Fischer, 2014) that has been associated with microtubule acetylation (Sarthi and Elefant, 2011), has been shown to guard against Aβ toxicity (Pirooznia et al., 2012). KAT5 in addition regulates the expression of genes involved in apoptosis (Pirooznia et al., 2012), axonal transport (Johnson et al., 2013) and DNA damage control (Kaidi and Jackson, 2013), and was found to interact with the amyloid precursor protein intracellular domain (Muller et al., 2013).

Not all HDACs have a detrimental effect on learning and memory, as inhibition of the class IIa HDACs HDAC4 and HDAC5 impair these processes (Agis-Balboa et al., 2013; Kim et al., 2012). Moreover, SIRT1, also an HDAC, was found to be decreased in the parietal cortex of AD patients (Julien et al., 2009). SIRT1 has been linked to neurogenesis, DNA repair, apoptosis, cell stress responses, and various other vital signaling pathways (Morris, 2013). SIRT1 expression is suggested to be beneficial in case of AD (Kim et al., 2007a), as it induces ADAM10 expression, an α-secretase that can cleave APP without producing Aβ (Donmez et al., 2010). Additionally, SIRT1 is able to deacetylate tau and its deficiency in AD is thus thought to enhance tau expression and pathology (Julien et al., 2009; Min et al., 2010). Note that this is in conflict with more recent findings regarding HDAC6, which is thought to increase tau pathology through deacetylation of tau, as stated above (Cook et al., 2014), although the SIRT1 study investigated global tau acetylation, whereas the HDAC6 study specifically investigated the acetylation of KXGS motifs. Nevertheless, enhancing SIRT1 expression attenuated axonal neurodegeneration and microglia-dependent Aβ toxicity (Araki et al., 2004; Chen et al., 2005; Kim et al., 2007a). Interestingly, SIRT1 was found to be upregulated in AD mouse models, but which might by a defense mechanism (Kim et al., 2007a; Morris, 2013), although it was found to be decreased in AD patients (Julien et al., 2009). Histone modification abnormalities in AD also include histone phosphorylation, as H3 phosphorylation was found to be increased in the frontal cortex of AD patients (Rao et al., 2012). Phosphorylation of histone protein H2A member X (H2AX) at S139, a marker of DNA damage, was shown to be increased in the AD hippocampus, but specifically in astrocytes (Myung et al., 2008).

Accumulating evidence indicates that dysfunctional protein localization might be a chief player in the incapacitation of the epigenetic machinery in AD, and possibly in neurodegeneration in general. Ogawa et al. (2003) made some fundamental observations in this respect. As some neurons in AD erroneously exhibit signs of cell cycle activation, they investigated H3S10 phosphorylation, a histone modification critical for chromosome compaction during cell division. Strikingly, it was not only found that H3 phosphorylation was increased in hippocampal AD neurons, but also that this epigenetic marker was abnormally restricted to the cytoplasm in these neurons. In addition, it has also been shown that the mitogen-activated protein kinase (MAPK) pathway involved in the phosphorylation of H3 is upregulated in degeneration vulnerable neurons in AD (Hyman et al., 1994; Perry et al., 1999; Zhu et al., 2001 ). Furthermore, the presence of high levels of histones in the cytoplasm of neurons in the HD brain (Iqbal et al., 1974) suggests that incapacitated nuclear transport might be a common denominator for neurodegenerative processes. In support of this, Mastroeni et al. (2013) found that Aβ could reduce rat sarcoma (Ras)-related nuclear protein (RAN) expression, a pivotal player in nucleocytoplasmic transport. As an apparent result, they observed DNMT1 and RNAPII to be erroneously sequestered in the cytoplasm of neurons from AD patients.

Histone 1 ADP-ribosylation has not been directly investigated in relation to AD, but the observations that a loss of poly[ADP]-ribose polymerase 1 (PARP-1) induces memory problems in mice (Fontan-Lozano et al., 2010) and that a dysregulation of PARP-1 is associated with amyloid pathology and sAD (Abeti et al., 2011; Liu et al., 2010b; Strosznajder et al., 2012) suggests that ADP-ribosylation might be a relevant target for future studies. Although various histone methylation marks and histone methylation and demethylation enzymes have been linked to cognitive functioning in mice and humans (shortly reviewed in Fischer, 2014), no links with AD have been firmly identified yet.

5.2. Chromatin remodeling in Parkinson’s disease

α-Synuclein normally localizes to the nucleus and presynaptic nerve terminals, but increased nuclear targeting is neurotoxic, possibly contributing to PD-related neurodegeneration (Kontopou-los et al., 2006). This nuclear toxicity of α-synuclein is supported by the finding that fPD a-synuclein mutations A30P and A53T result in an increased nuclear targeting of α-synuclein. Kontopoulos et al. (2006) found that nuclear toxicity of α-synuclein might be the result of direct binding of α-synuclein to histones, reducing the levels of acetylated histone H3 and acetylation in general in cultured cells through interactions with S1RT2. In cell cultures and transgenic flies, it was further shown that a rescue of α-synuclein toxicity could be achieved through HDACIs (Outeiro et al., 2007; St Laurent et al., 2013). Similar findings were found after exposure to oxidative stress, which induces the relocation of α-synuclein to the nucleus, where it subsequently binds to the peroxisome proliferator receptor gamma coactivator-1 alpha (PGC1-α) promoter element (Siddiqui et al., 2012). This binding of α-synuclein causes histone deacetylation, lowering PGC1-α expression, which is deleterious for mitochondrial functioning. Interestingly, levels of PGC1-α were significantly reduced in post-mortem substantia nigra neurons of PD patients (Zheng et al., 2010).

Curiously, not only does α-synuclein interact with the epigenetic machinery, but the KAT EP300 interacts with protein aggregation in Lewy bodies. A specific domain of EP300, reminiscent of prion-like domains, was found to serve as a potential interaction site for misfolded proteins, such as α-synuclein found in Lewy bodies, and enhance their aggregation (Kirilyuk et al., 2012). Conversely, α-synuclein was found to have neuroprotective actions via its interactions with EP300 and NF-κB, downregulating the proapoptotic protein kinase Cδ (PKCδ) (Jin et al., 2011a).

In PD patients, most of the aforementioned findings regarding the involvement of histone modifications still need to be replicated, but there is a report of an fPD case with a heterozygous A53 T SNCA mutation, in which the affected allele was epigeneti-cally silenced through histone modifications and the normal allele displayed expression levels exceeding those of two normal alleles in controls (Voutsinas et al., 2010).

Previously, the mechanism of DNA methylation-induced allelic skewing was described as a mediator between the genotype and environment. Histone modifications, however, are the most common epigenetic modality affected by environmental toxins such as pesticides, herbicides and industrial agents (Ammal Kaidery et al., 2013). MPTP, for instance, has been shown to lower H3K4me3 levels in the striatum of mice and non-human primates (Nicholas et al., 2008). Interestingly, H3K4me3 levels could be restored through chronic L-3,4-dihydroxy-phenylalanine (L-DOPA) treatment. Additionally, the herbicide paraquat and the insecticide dieldrin, which have both been associated with the development of PD, were found to affect histone acetylation, with exposure to paraquat increasing H3 acetylation and hampering overall HDAC activity, and exposure to dieldrin increasing H3 and H4 acetylation, in N27 dopaminergic cells (Song et al., 2010, 2011a). Dieldrin induces apoptosis in neurons and is thought to enhance histone acetylation through its inhibitory interaction with the proteasome system, leading to the build-up of CREBBP, an important KAT. Administration of the KAT inhibitor anacardic acid in a mouse model exposed to dieldrin, decreased histone acetylation and apoptosis, suggesting that the neurotoxic effect of dieldrin leading to apoptosis might be the result of detrimental histone acetylation (Song et al., 2010). See Table 8 for an overview of the aberrant chromatin remodeling seen in PD.

Table 8.

Epigenetic dysregulation in Parkinson’s disease: chromatin remodeling.

Chromatin remodeling target Regulation Epigenetic modification Observed in Sample size Methods Reference

Histones (Globally) Deacetylation via binding of α-
synuclein to PGC1-α promoter element
PD patients (SN) 16 GWES Zheng et al., 2010
H3 Acetylation via the interaction of
α-synuclein with SIRT1
α-Synuclein-transfected
SH-SY5Y cells
PD Drosophila model
-

N.S.
WB

WB
Kontopoulos et al., 2006
Acetylation due to Paraquat and/
or Dieldrin exposure
N27 dopaminergic cells - WB Song et al., 2010, 2011a
H3K4 Tri-methylation due to MPTP-
induced toxicity
Mice (striatum)
Macaque monkeys (striatum)
5
18
WB Nicholas et al., 2008
H4 Acetylation due to Dieldrin exposure N27 dopaminergic cells - WB Song et al., 2011a
Heterozygous A53T
SNCA mutation
Epigenetically silenced affected
allele via histone modifications
fPD patient 1 PCR Voutsinas et al., 2010

indicates increased expression levels,

indicates decreased expression levels, and

indicates a genetic mutation affecting epigenetic regulation.

Abbreviations: fPD, familial PD; H, histone; K, lysine; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; PD, Parkinson’s disease; PGC1-α, peroxisome proliferator receptor gamma coactivator-1 alpha; SH-SY5Y cells, human neuroblastoma cell line; SIRT1, sirtuin 1; SN, substantia nigra; SNCA, synuclein alpha; N.S., not specified; GWES, genome-wide epistasis studies; WB, Western blot; PCR, polymerase chain reaction.

5.3. Chromatin remodeling in Huntington’s disease

In general, HD is associated with hypoacetylated and hyper-methylated histones (Ferrante et al., 2004; Jenuwein and Allis, 2001; Suzuki and Bird, 2008; Table 9). The mechanism underlying histone hypoacetylation has been fairly well characterized and is thought to center around the deleterious interaction between CREBBP and mutant HTT (Alarcón et al., 2004; Korzus et al., 2004). The polyglutamine section of mutant HTT is thought to physically interact and sequester CREBBP, hampering its KAT activity (Lee et al., 2013a). Besides its KAT activity, CREBBP has additional integral functions in the regulation of transcription, interacting with various transcription factors and the RNAPII complex. Sequestration of CREBBP by mutant HTT thus disrupts transcription at multiple levels (Ferrante et al., 2004; Gardian et al., 2005; McFarland et al., 2012; Ryu et al., 2006; Sadri-Vakili et al., 2007). Interestingly, a study using transgenic mice expressing a form of CREBBP without KAT activity found that this modification specifically affected the consolidation of short-term memory into long-term memory, leaving short-term memory unaffected (Korzus et al., 2004). A similar study with inactive EP300, a homolog of CREBBP, found long-term recognition and contextual fear memory to be impaired (Oliveira et al., 2007).

Table 9.

Epigenetic dysregulation in Huntington’s disease: chromatin remodeling.

Chromatin remodeling target Regulation Epigenetic modification Observed in Sample size Methods Reference

Histones (Globally) Acetylation HD mouse model
(R6/2 mouse model)
N.S. WB Ferrante et al., 2004
H3K9 ↑↑ Tri-Methylation HD mouse model (R6/2 10 RT-PCR, Histone Ryu et al., 2006
HD mice; striatal neurons) Methylation Assay
HD patients (striatum) 6

indicates increased expression levels and

indicates a genetic mutation affecting epigenetic regulation.

Abbreviations: H, histone; HD, Huntington’s disease; K, lysine; R6/2 mice, transgenic mouse model overexpressing exon 1 of human HTT with an expanded cytosine-adenine-guanine repeat length; N.S., not specified; WB, Western blot; RT- PCR, real-time PCR.

It has been proposed that disruption of CREBBP functioning by mutant HTT is also indirectly responsible for the induction of histone hypermethylation and the subsequent formation of large abnormal heterochromatin domains (Lee et al., 2008). CREBBP is normally thought to repress the expression of Drosophila Su(var)3–9 and enhancer of zeste proteins (SET) domain, bifurcated 1 (SETDB1), a gene encoding the HKMT SETDB1 that methylates H3K9. Due to the shutdown of CREBBP by mutant HTT, the repression of SETDB1 is released and SETDB1 levels increase, subsequently resulting in H3K9 hypermethylation. This mechanism is corroborated by observations of increased levels of SETDB1 and H3K9me3 in striatal neurons of both transgenic HD mice and HD cases (Ryu et al., 2006). Additionally, H3K9me3 induced chromatin remodeling has been directly associated with altered gene expression profiles in HD (Lee et al., 2008, 2013b; Stack et al., 2007). Among the genes thought to be affected by this aberrant chromatin condensation is cholinergic receptor, muscarinic 1 (CHRM1) (Lee et al., 2013b). Decreased expression of CHRM1 has been proposed to induce synaptic dysfunction and CHRM1 levels are indeed lowered in the HD striatum (Calabresi et al., 2000; Cha et al., 1998). Deregulation of striatal cholinergic signaling has been identified as a pivotal factor in the pathophysiology of HD, especially affecting medium spiny neurons (Wang et al., 2006).

6. Non-coding RNAs in neurodegeneration

6.1. Non-coding RNAs and Alzheimer’s disease

In addition to DNA methylation and chromatin remodeling, ncRNAs, and especially miRNAs, have more recently been identified as possible contributors to AD pathology (Sonntag, 2010; Table 10). Interestingly, miRNA profiling studies have found several miRNAs to be upregulated in peripheral blood mononuclear cells of AD patients (Schipper et al., 2007). Apart from the blood, many brain region-specific imbalances in miRNA expression have been identified in relation to AD (for review see van den Hove et al., 2014), including those with candidate binding sites in the 3’ UTRs of BACE, PS1 and APP. More specifically, miR-16, −17, −20a, - 101, −106a, −106b, −107, −124, −137, −147, −153, −195, −323–3p, - 520c, −644, −655 and let-7 are thought to regulate APP metabolism and Ab production (Bicchiet al., 2013; Delay et al., 2011; Fanet al., 2010; Hebert et al., 2009; Liang et al., 2012; Liu et al., 2012b; Long and Lahiri, 2011; Niwa et al., 2008; Patel et al., 2008). MiR-16 overexpression was found to reduce APP levels in SAMP8 mice (Liu et al., 2012b). In human neurons, miR-106a, −153 and −520c were found to target APP mRNA, downregulating APP and Ab levels (Long et al., 2012; Patel et al., 2008). Others, however, could not corroborate the involvement of miR-106a and miR-520c in the regulation of APP expression (Delay et al., 2011; Hebert et al., 2009). Inhibiting miR-101 in hippocampal neurons proved to decrease APP expression and Aβ load, indicating a possible detrimental role of the miRNA in AD (Long and Lahiri, 2011). Conversely, miR-124, a miRNA involved in adult neuronal differentiation (Cheng et al., 2009), is reported to be down-regulated in some AD patients (Smith et al., 2011a). MiR-124 is thought to, together with polypyrimidine tract binding protein 1 (PTBP1), modulate the alternative splicing of APP exons 7 and 8. Additionally, miR-124, but also miR-9, −29a/b-1, −29c, −107, - 195, −298, −328 and −485–5p, affect Aβ indirectly by modulating BACE1 translation (Fang et al., 2012; Hebert and De Strooper, 2009; Hebert et al., 2008; Zhu et al., 2012). In addition, in SAMP8 mice miR-195 expression was found to be decreased, whereas BACE1 levels were heightened (Zhu et al., 2012). The involvement of all these miRNAs in AD might, however, not be a general phenomenon. For instance, the miR-29a/b-1 cluster was found to be lowered in the anterior temporal cortex of sAD patients, coupled with high BACE1 protein levels, but only in approximately 30% of the examined cases (Hebert et al., 2008). In a transgenic AD mouse model miR-29c was observed to be highly expressed and was found to hamper BACE1 expression (Zong et al., 2011). Levels of miR-107 were found to be lowered in the temporal cortex of AD cases, which was suggested to facilitate AD progression as a result of diminished BACE1 repression (Wang et al., 2008c, 2011, 2010c). MiR-195, −298, and −328 also reduce Ab production by inhibiting BACE1 translation (Boissonneault et al., 2009; Zhu et al., 2012). Interestingly, while most of the miRNAs affecting BACE1 expression repress translation by binding to the 3’ UTR of its mRNA, miR-485–5p represses BACE1 by binding to the open reading frame in exon 6 (Faghihi et al., 2010). The involvement of post-transcriptional regulation of BACE1 is further supported by the observation that in AD brains BACE1 protein levels are increased, whereas mRNA levels remain unchanged (Hebert and De Strooper, 2009). Serine palmitoyltransferase (SPT) is an enzyme crucial for ceramide synthesis, which is thought to facilitate Aβ production. MiR-9, −29a/b-1, −137 and −181c negatively modulate SPT production and their levels were lowered in the frontal cortex of AD patients (Geekiyanage and Chan, 2011). MiR-137 is known to additionally promote proliferation of neural stem cells through the inhibition of differentiation and dendrite formation (Smrt et al., 2010; Szulwach et al., 2010).

Table 10.

Epigenetic dysregulation in Alzheimer’s disease: non-coding RNAs.

Regulation ncRNA Observed in Sample size Approach Methods Reference

miR-124 AD patients 11 Targeted WB, RT-PCR Smith et al., 2011a
miR-195 AD mouse model (SAMP8) N.S. Targeted
(5 miRNAs)
RT-PCR Zhu et al., 2012
miR-29a/b-1 sAD patients
(anterior temporal cortex)
34 Genome-wide miRNA microarray Hebert et al., 2008
↑↑ miR-29c AD mouse model (APPswe/PSΔE9) 6 Targeted RT-PCR Zong et al., 2011
miR-107 AD patients 7, 10 Genome-wide miRNA microarray,
rna22 algorithm
Wang et al., 2008c, 2011
AD mouse model (Tg19959 mice) 7 Targeted RT-PCR Yao et al., 2000
miR-106b AD patients (anterior temporal cortex) 19 Targeted
(200 miRNAs)
miRNA microarray,
Northern blot
Hebert et al., 2009
miR-9, miR-181c AD patients 15 (Brain)
10 (cSF)
Targeted
(48 miRNAs)
PCR Cogswell et al., 2008
miR-132 AD patients (hippocampus,
cerebellum, medial frontal gyrus)
6 Targeted
(15 miRNAs)
Northern Blot Sethi and Lukiw, 2009
miR-15 Dicer cKO mice 8–10 Targeted
(200 miRNAs)
miRNA microarray Hebert et al., 2010
miR-26a AD patients 15 (Brain)
10 (csf)
Targeted
(48 miRNAs)
PCR Cogswell et al., 2008
miR-101 HeLa, HEK293T, U373, SK-N-SH,
PC12 cells
- Targeted
(1 miRNA)
PCR Long and Lahiri, 2011
miR-125b AD patients (hippocampus,
cerebellum, medial frontal gyrus)
6 Targeted
(15 miRNAs)
Northern Blot Sethi and Lukiw, 2009
BC200 AD patients (hippocampus,
superior frontal gyrus)
12 Targeted
(2 miRNAs)
Northern Blot, ISH Mus et al., 2007
BC200 AD patients (temporal neocortex) 12 Targeted
(2 miRNAs)
Northern Blot, ISH Mus et al., 2007
miR-103 AD mouse model (Tg19959 mice) 7 Targeted
(2 miRNAs)
RT-PCR Yao et al., 2000
BACE1-AS AD patients 2 Targeted
(48 probes)
High-throughput
sequencing, ECA
Faghihi et al., 2010
ncRNA 17a AD patients 11 Targeted
(5 probes)
RT-PCR Massone et al., 2011

indicates increased expression levels,

indicates decreased expression levels, and

indicates altered expression, not further specified.

Abbreviations: AD, Alzheimer’s disease; BACE1-AS, β-secretase 1-antisense; BC200, brain cytoplasmic RNA 200; Dicer cKO mice, conditional Dicer1 knock-out mouse model; HeLa cells, immortalized cell line established from cervical cancer cells of Henrietta Lacks; miR, micro RNA; miR-29c transgenic mice, transgenic mouse model overexpressing miR-29c; ncRNA, non-coding RNA; sAD, sporadic AD; SAMP8 mouse, senescence-accelerated prone mouse 8; Tg19959 mice, transgenic mouse model overexpressing mutant human APP with the double KM670/671NL Swedish and V717F Indiana mutations; N.S., not specified; CSF, cerebrospinal fluid; PCR, polymerase chain reaction; RT-PCR, real time-PCR; WB, Western blot; ISH, in situ hybridization; ECA, enzyme complementation assay.

MiRNAs can thus affect Aβ production, but Aβ can also affect the expression of some miRNAs in vitro, for example inducing miR- 106b expression (Wang et al., 2010b) but repressing miR-9 and miR-181c (Schonrock et al., 2012). Curiously, Hebert et al. (2009) found miR-106b to be downregulated in the anterior temporal cortex of AD brains. Furthermore, miR-106 was reported to not only directly bind to and inhibit the translation of APP, but also affect APP trafficking and Aβ clearance. Additionally, by regulating the ATP-binding cassette, sub-family A (ABC1), member 1 (ABCA1), which transports cholesterol, it is thought to influence BACE and γ-secretase functioning. ABCA1 expression in the hippocampus has been positively correlated with cognitive impairments in AD (Akram et al., 2010). Normally, miR-106b is thought to promote neurogenesis through its regulation of the insulin-like growth factor 1 (IGF1) pathway (Brett et al., 2011). MiR-9 has been reported to be a pivotal player in the differentiation and migration of neural stem cells (Delaloy et al., 2010; Zhao et al., 2009).

Furthermore, while there are generally no AD-associated mutations in tau, miR-15, −16, −132, and −497 are thought to regulate tau expression and might play a role in AD. In example, a decrease in miR-132 is suggested to mediate the alternative splicing of tau exon 10, through a lowered repression of polypyrimidine tract-binding protein 2 (PTBP2), which hampers physiological phosphorylation of tau (Hebert et al., 2012; Smith et al., 2011b). Alternative splicing of tau influences whether it contains 3 or 4 microtubule-binding repeats (3R-tau and 4R-tau, respectively) (Liu and Gong, 2008). Furthermore, changes in the 3R:4R tau ratio are thought to be related to neurodegeneration (Caffrey et al., 2006). Apart from miR-132, miR-9, −124 and −137 have also been reported to affect the 3R:4R tau ratio. MiR-212 and miR-454 have also been implicated in NFT pathology in AD (Cogswell et al., 2008; Wang et al., 2011). Note that dysregulated miRNA expression in relation to tau is probably not unique for AD and likely also occurs in other tauopathies. For instance, miR-132 was found to be downregulated in progressive supranuclear palsy and frontotemporal lobar degeneration (Hebert et al., 2013; Smith et al., 2011b).

Phosphorylation of tau is performed by extracellular signal-regulated kinase 1 (ERK1), which in turn is regulated by members of the miR-16 family (miR-15, −16, −195 and −495), of which miR- 15 was found to be downregulated in AD (Hebert et al., 2012). Tau can also be phosphorylated by glycogen synthase kinase 3 beta (GSK-3β), which has been implication in Aβ and NFT formation, and has been reported to be negatively regulated by miR-26a, a miRNA that is dysregulated in AD (Cogswell et al., 2008; Mohamed et al., 2010). As stated above, SIRTI negatively regulates tau expression, while miR-9, −34c and −181c, however, have been shown to (in their turn) inhibit S1RT1 production, thereby enhancing tau production in AD (Schonrock et al., 2012; Zovoilis et al., 2011). MiR-128 has been suggested to affect tau clearance, through its regulation of cochaperone B-cell chronic lymphocytic leukemia (CLL)/lymphoma 2 (BCL2)-associated athanogene 2 (BAG2), and has been reported to be altered in AD (Carrettiero et al., 2009; Lukiw, 2007). There is additional indirect evidence for the involvement of miRNAs in the regulation of tau metabolism, as studies knocking out Dicer, which is crucial for miRNA processing, observed increased hyperphosphorylation of tau, alternate splicing of tau and neurodegeneration (Bilen et al., 2006; Hebert et al., 2010).

Next to miRNAs impacting on Aβ and tau metabolism, various miRNAs that were found to be dysregulated in AD also affect other pathological hallmarks of AD. MiR-146a, is for instance a regulator of inflammatory processes through its interaction with interleukin-1 receptor-associated kinase 1 (1RAK1) that is upregulated in AD brains (Cui et al., 2010; Taganov et al., 2006). In addition to 1RAK1, miR-146a was reported to bind to the 3’ UTR of complement factor H, a suppressor of inflammation which is downregulated in AD (Lukiw et al., 2008). Another regulator of inflammation is miR- 101, which normally inhibits COX-2, but its levels were shown to be lowered in AD, whereas levels of COX-2 were increased (Long and Lahiri, 2011). MiR-132 and miR-125b have been linked to synaptic plasticity, and miR-132 was lower in the hippocampus, cerebellum and medial frontal gyrus of AD patients, whereas miR- 125b levels were higher in these areas (Sethi and Lukiw, 2009). Brain cytoplasmic RNA 200 (BC200) was initially reported to be decreased in the temporal neocortex of AD cases (Lukiw et al., 1992), but later studies reported increased BC200 levels in the hippocampus and superior frontal gyrus, but erroneously located in the neuronal soma (Mus et al., 2007). BC200 is thought to enhance long-term synaptic plasticity by interacting with protein synthesis in postsynaptic microdomains. In transgenic mice overexpressing a combination of APPswe and human APP with the V717F Indiana mutation (APPind; Tg19959 mice) miR-103 and miR-107 were found to be decreased, which was linked to increased cofilin expression (Yao et al., 2010). Cofilin is a pivotal player in cytoskeletal integrity and is thought to influence microtubule stability, neuronal transport and synaptic functioning (Minamide et al., 2000).

Compared to miRNAs, evidence for the involvement of other ncRNAs in AD pathology is sparse. RNA polymerase 111 (RNAP111)-dependent ncRNA neuroblastoma differentiation marker 29 (NDM29) was found to facilitate the production and secretion of Ab by influencing APP processing (Massone et al., 2012), whereas the lncRNA BACE1-antisense (BACE1-AS) positively affects BACE1 expression (Hebert and De Strooper, 2009). BACE1-AS has a length of about 2 kb and is transcribed from the DNA strand complementary to the BACE1 gene (Faghihi et al., 2008). 1t is thought to enhance the stability of BACE1 mRNA, facilitating BACE1 protein production. Interestingly, BACE1-AS transcription is enhanced in response to Ab exposure, initiating a vicious cycle, as its positive effects on BACE1 expression in turn enhances Ab production. In both AD patients and Tg19959 mice, BACE1-AS was indeed found to be overexpressed. Although only confirmed for the nonconventional miR-485–5p, evidence suggests that the binding of BACE1-AS to BACE1 mRNA enhances mRNA stability by competing with miRNA binding (Faghihi et al., 2010). The ncRNA 17a has been observed to promote Ab secretion and accumulation and is elevated in the cerebral cortex of AD cases, which is thought to be the result of inflammatory factors (Massone et al., 2011).

6.2. Non-coding RNAs and Parkinson’s disease

Apart from epigenetic transcriptional regulation of SNCA, some miRNAs have been identified that regulate its function on a translational level. One of these is miR-7, which negatively regulates α-synuclein expression through binding to the 3’ UTR of α-synuclein mRNA and is mainly expressed in neurons (Junn et al., 2009). Through its suppression of α-synuclein, including cytotoxic mutant forms, it is thought to have a neuroprotective role in PD. Interestingly, miR-7 levels were shown to be decreased in vitro and in animal models after exposure to the toxic metabolite of MPTP, 1-methyl-4-phenyl-pyridinium ion (MPP+), increasing α-synuclein expression. Downregulation of miR-7 might thus, at least in part, explain how MPTP induces PD-like pathology.

Another miRNA, miR-153, represses α-synuclein production both at a mRNA and protein level (Doxakis, 2010). Indirectly, miR-433 has also been implicated in SNCA expression, via its regulation of the fibroblast growth factor 20 (FGF20). FGF20 expression has been positively correlated with α-synuclein expression, and a 3 UTR SNP (rs1270208) has been linked to an increased risk to develop PD. This SNP interferes with miR-433 binding, increasing FGF20 expression (Wang et al., 2008a). Conversely, α-synuclein has been shown to affect the expression levels of certain miRNAs in in vivo models were α-synuclein was overexpressed. Levels of various miRNAs were affected in a transgenic mouse model overexpressing human A30P α-synuclein (Gillardon et al., 2008; Table 11). In a transgenic C. elegans model expressing human α- synuclein, alterations in levels of 12 miRNAs were found (Asikainen et al., 2010). The significance for the human situation, however, remains to be elucidated as the human orthologs of these miRNA remain to be identified.

Table 11.

Epigenetic dysregulation in Parkinson’s disease: non-coding RNAs.

Regulation ncRNA Observed in Sample size Approach Methods Reference

miR-7 HEK293T, SH-SY5Y, NS20Y cell lines - Targeted
(2 miRNAs)
PCR Junn et al., 2009
miR-10a Mouse model 3 Targeted miRNA expression profiling Gillardon et al., 2008
miR-10b (overexpressing (266 miRNAs) (microfluidic chips)
miR-132 A30P α-synuclein)
miR-212
miR-495
miR-205 sPD patients 8 Targeted
(1 miRNA)
PCR Cho et al., 2013
miR-1 Healthy individuals and 8 Targeted PCR Margis et al., 2011
miR-22* untreated PD patients (85 miRNAs)
miR-29
miR-16–2* Treated and untreated 7 treated
miR-26a2* PD patients 8 untreated
miR-30a

indicates decreased expression levels and

indicates altered expression, not further specified.

Abbreviations: (s)PD, (sporadic) Parkinson’s disease; HEK293T cells, human embryonic kidney cell line; miR, micro RNA; NS20Y, mouse cholinergic neuroblastoma cell line; PD, Parkinson’s disease; SH-SY5Y cells, human neuroblastoma cell line; PCR, polymerase chain reaction.

In addition to SNCA, the expression of LRRK2, a gene implicated in both fPD and sPD, is also regulated by miRNAs. MiR-205 targets the 3’ UTR of LRRK2 mRNA and was found to be downregulated in sPD cases in which LRRK2 protein levels were increased, whereas miR- 205 was able to mitigate the aberrant neurite growth induced by LRRK2 mutation R1411G in vitro (Cho et al., 2013). Conversely, mutant LRRK2 (I1915T or G2019S) was observed to inhibit the actions of let-7 and miR-184*. These miRNAs regulate E2F transcription factor 1 (E2F1) and differentiation regulated transcription factor protein (DP) levels, transcription factors associated with cell cycle regulation and cell survival. LRRK2 thus induces E2F1 and DP expression, which is associated with reduced dopaminergic neuron numbers and locomotor activity in Drosophila, effects that have also been linked to mutant LRRK2 (Gehrke et al., 2010). Overexpression of let-7 or miR-184* reversed the deleterious effects of mutant LRRK2 expression. Note, however, that let-7b was also found to progressively inhibit neural stem cell proliferation in the subventricular zone with age (Nishino et al., 2008). Interestingly, the disruption of let-7 and miR-184* activity by mutant LRRK2 is thought to be an indirect effect, as the increased activity of mutant LRRK2 increases the phosphorylation of eukaryotic translation initiation factor 4E binding protein (4E-BP). 4E-BP interacts with Argonaute 2, a pivotal constituent of the RISC, which in turn is required for proper let-7 and miR-184* functioning (Imai et al., 2008). The negative regulation of these miRNAs by LRRK2 thus depends on gain of function mutations, such as I1915T and G2019S (Imai et al., 2008; Smith et al., 2006). Indeed, mutant LRRK2 without enzymatic activity does not affect miRNA repression (Gehrke et al., 2010). Additionally, LRRK2 might also affect Dicer, another protein integral to the RNA interference (RNAi) pathway, as knocking down LRRK2 was able to attenuate some of the pathology in the Drosophila model related to decreased Dicer activity.

MiRNA profiling of PD brains at different stages of the disease pointed towards a miR-34b/c downregulation, mainly at the early premotor stages (1–3) (Minones-Moyano et al., 2011). MiR-34b/c is thought to modulate mitochondrial functioning via its modulation of DJ-1 and parkin, proteins that have both been associated with fPD. In the blood, comparing healthy individuals with untreated PD patients, miR-1, −22*, and −29 were found to be differentially expressed, while miR-16–2*, −26a2*, and −30a were differentially expressed comparing treated and untreated PD patients (Margis et al., 2011). Table 11 contains the most important findings regarding ncRNAs associated with PD.

6.3. Non-coding RNAs and Huntingon’s disease

In accordance with the widespread dysregulation of gene expression, the expression of miRNAs are also affected in HD (Table 12). In HD models and patients neuronal miRNA expression was found to be decreased in general, resulting in an upregulation of their target mRNAs (Han et al., 2004; Johnson et al., 2008; Lee et al., 2011; Table 12). In addition, it was observed that mutant HTT expression decreased miR-125b and miR-150 expression (Ghose et al., 2011). These miRNAs have P53 among their targets, which is known to repress xsNF-κB and miR-146a expression. Further interactions between P53 and mutant HTT mediate nuclear and mitochondrial damage in HD models and patients (Bae et al., 2005).

Table 12.

Epigenetic dysregulation in Huntington’s disease: non-coding RNAs.

Regulation ncRNA Observed in Sample size Approach Methods Reference

miR-22 HD murine models [YAC12 12 (mouse model) Targeted miRNA microassay Lee et al., 2011
miR-29c and R6/2 mouse model, 18 (rat model) (567 miRNAs)
miR-125b 3NP-induced rat model; Striatum]
miR-128
miR-132
miR-139
miR-146a
miR-150
miR-218
miR-222
miR-344
miR-674
Drosha

indicates increased expression levels and

indicates decreased expression levels.

Abbreviations: FVB-Tg(YAC128)53Hay/J mice, transgenic mouse model generated with a yeast artificial chromosome (YAC) containing the human Huntingtin gene with 128 cytosine-adenine-guanine repeats; HD, Huntington’s disease; miR(NA), micro RNA.

7. Epigenetic-based diagnostics and therapies

The available treatment strategies for most progressive neurodegenerative diseases only provide symptomatic relief, stressing the need to develop innovative, realistic therapeutic approaches that can effectively modulate the disease process. The factor common to all of the conditions discussed in this review is their neurodegenerative nature. Treatments providing a general neuroprotective effect could thus potentially be beneficial for any of them. Among such treatments HDAC and DNMT inhibitors represent interesting options to act upon the epigenetic machinery. These are already used in the treatment of other disorders such as epilepsy and cancer (Xu et al., 2012). The versatile and reversible nature of epigenetic changes makes epigenetic mechanisms ideal targets for the development of efficient, novel treatment strategies (Coppede, 2014). The adverse role of HDAC2 in memory facilitation has, for instance, led to the investigation of HDACIs as a potential treatment for memory impairment, for example in AD (Guan et al., 2009).

7.1. Strategies targeting DNA methylation

Neurodegenerative disorders may involve a dysregulated SAM metabolism, resulting in global DNA hypomethylation, as well as the hypermethylation of some crucial genes. It is thus not surprising that strategies aiming to increase or decrease DNA methylation have been investigated. Enhancing DNA methylation can be achieved by boosting SAM metabolism, for example through the administration of SAM itself, and by vitamin B12 and folate supplementation that was shown to be effective (Durga et al., 2007; Haan et al., 2007; Scarpa et al., 2003). Reducing the levels of methylated DNA can be accomplished with DNA demethylating agents, such as DAC (Wang et al., 2013). However, these treatment options are highly unspecific, which may, especially in the case of DNA demethylating compounds, result in considerable adverse effects. Apart from therapies targeting DNA methylation, it has also been suggested that differential genomic and mtDNA methylation patterns may serve as diagnostic biomarkers (Devall et al., 2014; How Kit et al., 2012).

7.1.1. Alzheimer’s disease

Scarpa et al. (2003) argued that the Hcy accumulation often seen in AD might be an indication for an abnormal SAM metabolism. The resulting decrement in SAM levels could explain a global decrease in DNA methylation, which in turn could lead to an overexpression of multiple genes, including ones involved in AD pathology. lnterestingly, in vitro SAM administration led to a repression of PS1 gene expression and Aβ production. Accordingly, folate and vitamin B12 supplementation have been found to enhance cognitive functioning and slow the development of dementia (Durga et al., 2007; Haan et al., 2007). There are, however, also other studies that could not detect a positive effect of folate and vitamin B12 supplementation (Malouf et al., 2005; McMahon et al., 2006), and it has been reported that folic acid supplementation, in addition to other side-effects, might exacerbate neuropathology in patients with low vitamin B12 levels (Campbell, 1996). The observation that some crucial genes are hypermethylated in AD has led to the suggestion that the DNA demethylating agent DAC could be used to restore normal expression levels of these genes. AD, however, is also associated with general hypomethylation and due to the non-specific nature of DAC it might in fact cause more harm than good (Wang et al., 2013).

7.1.2. Parkinson’s disease

Similarly to AD, a disturbed SAM metabolism has also been associated with PD, and decreased methylation was linked to cognitive decline (Obeid et al., 2009). A viable option to counteract this decline would be to increase the levels of SAM, through administration of methionine, choline, folates or vitamin B12, among other possibilities (Xu et al., 2012).

7.2. Strategies targeting chromatin modifications

One of the most promising epigenetics-based treatment options in relation to neurodegeneration are HDACls. There are many HDACls, which can be subdivided into four classes, including short-chain fatty acids, hydroxamic acids, epoxyketones and benzamides. Of these, sodium butyrate (SB) has received most of the attention for clinical use. The bioavailability of SB in the central nervous system has been characterized and is well tolerated in animals and in humans due to its low toxicity (Daniel et al., 1989; Egorin et al., 1999; Miller et al., 1987). Chen et al. (2006) investigated the short-chain fatty acid valproate (valproic acid, VPA), a drug used as a mood stabilizer and anti-epileptic that was found to be an HDACl. VPA is thought to enhance H3 acetylation indirectly, possibly through the recruitment of the KAT EP300 (Marinova et al., 2009). This study found that VPA exerts a neurotrophic effect, involving the repression of pro-inflammatory factors released by microglia and a stimulation of neurotropic factor expression, including glial cell line-derived neurotrophic factor (GDNF) and BDNF, by astrocytes. VPA may thus represent a viable treatment option to counteract neurodegeneration. Comparable effects have been attributed to other HDACls, including TSA, suberoylanilide hydroxamic acid (SAHA) and SB, as well as MS-275 and apicidin, which specifically inhibit class l HDACs (Chen et al., 2012; Kidd and Schneider, 2010; Leng et al., 2010; Marinova et al., 2011; 2009; Wu et al., 2008). Some HDACls, such as 4-phenylbutyrate (4PBA), VPA, and urocortin, might also exert some of their neuroprotective effects independent of their effects on HDACs (Huang et al., 2011; Roy et al., 2012; Zhou et al., 2011).

The use of HDACls in the treatment of neurodegenerative diseases is thus promising and deserves much attention. However, several issues, especially concerning the non-specific action of most tested HDACls, must be overcome for HDACls to be ready for clinical use. For example, as some HDACls are already being used in cancer therapy, it was observed that they induce cell death and cell-cycle arrest, which has also been reported to affect neurons (Brahe et al., 2005; Marks, 2010; Marks and Xu, 2009; Salminen et al., 1998). HDACls have additionally been observed to disturb the immune system (Kelly-Sell et al., 2012; Rossi et al., 2012). It has thus been found that targeting specific HDACs would be more preferable over the more general HDACls. Some examples of specific HDACls are tubacin, a selective HDAC6 inhibitor, and suramin, a selective S1RT1 and S1RT2 inhibitor (Haggarty et al., 2003; Trapp et al., 2007).

7.2.1. Alzheimer’s disease

A decrease in BDNF expression, a pivotal player in memory processes (Yamada et al., 2002), has been implicated as an early marker in the development of AD (Walker et al., 2013) and TSA treatment has been shown to enhance BDNF expression in vitro significantly, possibly through restoring BDNF promoter histone acetylation levels (lshimaru et al., 2010; Tian et al., 2010). Another HDAC1, VPA, can counter Aβ production in human embryonic kidney 293 (HEK293) cells expressing APPswe isoform 751 and in a transgenic mouse model overexpressing APPind (PDAPP mice) (Su et al., 2004). Using a transgenic mouse model with a 7-fold overexpression of APPswe (APP23 mice), this decrease in Aβ was shown to be due to an inhibition of GSK-3β-mediated γ-secretase cleavage of APP by VPA, which was also found to improve behavioral impairments (Qing et al., 2008). Another HDACl, (4PBA), was shown to reverse learning and memory problems in Tg2576 AD model mice, without affecting Aβ levels, but decreasing tau phosphorylation (Ricobaraza et al., 2009). This was accompanied by increases in GSK-3β, histone acetylation, as well as ionotropic glutamate receptor 1 (GluR1), postsynaptic density protein 95 (PSD95) and microtubule-associated protein 2 (MAP2), the later three being involved in synaptic plasticity (Ricobaraza et al., 2009). A subsequent study using the same mouse model showed that 4PBA elevated intraneuronal Aβ clearance, paired with an increase in plasticity-related proteins and subsequent restoring of dendritic spine densities in the hippocampus (Ricobaraza et al., 2012). Treatment in mice with the HDACl SAHA achieved an increase in H4K12 acetylation levels and accordingly restored expression levels of genes associated with learning (Peleg et al., 2010). VPA and SAHA were also reported to restore CLU expression in vitro (Nuutinen et al., 2010). Effective VPA, SB, and SAHA treatment in AD models has additionally been linked to elevating H4 acetylation levels and alleviation of memory deficits (Kilgore et al., 2009). lnterestingly, although VPA, SB, and SAHA by elevating H4 acetylation are likely to generally affect gene expression, the HDACl TSA was found to specifically enhance expression of those genes involved in memory consolidation (Vecsey et al., 2007). Curiously, inhibition of SIRTs, the class Ill HDACs, with nicotinamide was observed to restore cognitive impairments in 3xTg-AD mice, by indirectly promoting microtubule stability, which is affected by hyperphosphorylated tau in AD (Green et al., 2008). Recently, Forum Pharmaceuticals compound 0334 (FRM-0334), a class I HDACI specifically designed to cross the blood-brain barrier (BBB) was developed, addressing the problem of BBB permeability (Arrowsmith et al., 2012). FRM-0334 is one the first HDACI that is specifically being tested for the treatment of AD, with most others having an approved indication in cancer treatment (Arrowsmith et al., 2012). In relation to specific HDAC inhibition, the selective HDAC6 tubacin has been reported to affect tau phosphorylation in vitro (Ding et al., 2008). In addition to HDACIs, KAT agonists are being developed (Chatterjee et al., 2013) and it has also been suggested that targeting HKMTs and HKDMs may prove to be a viable treatment strategy for AD (Fischer, 2014).

7.2.2. Parkinson’s disease

As stated above, preventing histone deacetylation may alleviate memory problems, such as those associated with AD (Guan et al., 2009). Similar approaches in PD models suggest that HDAC inhibition could be neuroprotective. In in vitro and Drosophila models the HDACIs SB and SAHA attenuated α-synuclein-induced toxic effects (Kontopoulos et al., 2006), illustrating the prominent role of disrupted histone acetylation in the neurotoxic effects of α-synuclein, caused by its direct binding to histones. TSA was able to rescue mitochondrial fragmentation and cell death induced by MPP+ in human neuroblastoma cells (Zhu et al., 2014). Similar results were obtained when inhibiting the HDAC SIRT2 with 2-cyano-3-(5-(2,5-dichlorophenyl)-2-furanyl)-N-5-quinolinyl-2-propenamide (AGK2) (Outeiro et al., 2007). Additionally, pretreatment with VPA has been shown to protect midbrain dopaminergic neurons from inflammation and a-synuclein-induced neurotoxicity (Chen etal., 2006,2007; Kidd and Schneider, 2011; Pengetal.,2005).

Currently, one of the main treatments for PD is the dopamine precursor L-DOPA, which provides some symptom alleviation. Although not intended as such, chronic L-DOPA treatment was observed to induce epigenetic alterations. Specifically, the development of L-DOPA-induced dyskinesia presented with decreased H3K4me3 levels, whereas L-DOPA induced hyperkinesia was associated with decreased acetylation levels of H4K5, H4K8, H4K12 and H4K16, in the striatum of animal models (Nicholas et al., 2008). Additionally, it was shown that L-DOPA-induced dyskinesia paralleled H3 phosphoacetylation, suggesting that the inhibition of striatal H3 phosphoacetylation when using L-DOPA might prevent the development of dyskinesia (Darmopil et al., 2009).

7.2.3. Huntington’s disease

In HD, reversing the reduced expression of crucial genes due to histone hypoacetylation has been attempted through the application of HDACIs, showing promising results, both in terms of neuropathology and motor symptoms (Ferrante et al., 2004; Gardian et al., 2005; Igarashi et al., 2003; McFarland et al., 2012; Sadri-Vakili et al., 2007; Steffan et al., 2001; Sugai et al., 2004; Thomas et al., 2008). HDACIs improved memory and behavior in CREBBP deficiency or KAT deletion animal models (Alarcon et al., 2004; Korzus et al., 2004; Wood et al., 2006). Additionally, in an in vitro model based on the administration of toxic polyglutamine, a model that also exhibits histone hypoacetylation, HDACIs were able to mitigate the toxic effects of polyglutamine (McCampbell et al., 2001). As in PD, SAHA, and SB also were effective in transgenic HD mice (Hockly et al., 2003). HDAC inhibition, either through SAHA or SB administration, or HDAC2 knock-out, improved memory deficits in mice (Mielcarek et al., 2011). SB-treated transgenic mice overexpressing exon 1 of human HTT with an expanded CAG repeat length (R6/2 mice), however showed improved motor performance and decreased neuropathology, and survived significantly longer than non-treated mice (Ferrante et al., 2003). Alternatively, 4PBA may represent a promising candidate treatment for HD, as it is already Food and Drug Administration-approved and data about pharmacokinetics, toxicity, and dosing are available. Although 4PBA itself has no inhibitory effect on HDACs, its metabolite phenylacetate does, in addition to having a high bioavailability in the brain (Dasgupta et al., 2003). As with SB, treatment with 4PBA improved motor symptoms and neuropathology in a transgenic HD mouse model (Gardian et al., 2005). Unfortunately, a multicenter, double-blind, placebo-controlled clinical trial of 4PBA to determine safety and tolerability in HD, patients showed that its efficacy was very low, necessitating the use of high doses (Ebbel et al., 2010; Hogarth et al., 2007). Therefore, although promising in animal models, the use of 4PBA in the treatment of HD is not optimal. A novel HDACI, the pimelic diphenylamide HDACI 4b, has also shown to be effective in R6/2 mice, improving the HD-related transcriptional abnormalities, including H3 acetylation and mRNA levels, and behavioral phenotype (Thomas et al., 2008). Additionally, in a different HD model, that expresses the first 171 amino acids of HTT with 82 CAG repeats at a relatively low steady-state level (N171–82Q mice), HDACI 4b enhanced body weight, motor function and cognitive performance, which may be mediated by modulatory effects of HDACI 4b on post-translational mechanisms, such as protein phosphorylation and ubiquitination (Jia et al., 2012a). Accordingly, activation of inhibitor of kappaB kinase (IKK) by HDACI 4b enhanced phosphorylation and acetylation of HTT, and subsequent clearance effected by the ubiquitin-proteasomal and autophagy systems. The selectivity of HDACI 4b to inhibit class I and class II HDACs, and restore proper gene expression, was also explored in various HD models, including mice, flies, and cells (Jia et al., 2012b). Targeted inhibition of HDAC1 and HDAC3 was observed to mitigate mutant HTT-induced degeneration of the eyes and brain in Drosophila, and subdued some of the metabolic defects seen in STHdhQ111 mutant HTT knock-in striatal cells. In addition to HDACI 4b, some additional compounds were tested, revealing that one of the, compound 136, could effectively inhibit HDAC3 and restore proper gene expression in HD models. Although the exact targeting mechanisms remain elusive, HDACIs upregulate prosurvival genes selectively, while downregulating pro-death genes (Hu et al., 2011b).

Apart from drugs targeting HDACs, DNA-binding drugs have also received some attention in the context of HD. These efforts are mainly focused on the DNA intercalating anthracyclines, such as mithramycin A and chromomycin A3, which were isolated from Streptomyces argillaceus and Streptomyces griseus, respectively (Blanco et al., 1996; Chakrabarti et al., 2000; Prado et al., 1999). Mithramycin A and chromomycin A3 inhibit the replication and translation processes in cells, processes that are especially indispensable to tumors. Mithramycin A has already been used to treat Paget’s disease, hypercalcemia in malignancy, and various types of cancer (Ralston, 1994; Ryan, 1977; Kennedy, 1972).These DNA intercalating agents specifically block the binding of transcription activators and repressors that bind to GC-rich regions of gene promoters, thereby affecting gene expression (Ralston, 1994; Hagen et al., 1995; Majello et al., 1995). Their interference with transcription factors SP1 and SP3 are thought to be neuroprotective, as these induce detrimental responses after oxidative stress and DNA damage (Chatterjee et al., 2001). In R6/2 mice, mithramycin A was found to reduce clinical and neuro- pathological symptoms, as well as significantly increase survival rate, probably via the reduction of pericentromeric heterochromatin condensation through an epigenetic mechanism (Ferrante et al., 2004; Ryu et al., 2006). Mithramycin Acan repress the HKMT SETDB1 and thereby reverse the H3K9 hypermethylation seen in R6/2 mice (Ferrante et al., 2004). The effects of chromomycin A3 have been investigated in both N171–82Qand R6/2 mice, showing that it can beneficially tip the methylation-acetylation balance at H3K9 in favor of acetylation, reactivating the chromatin and improving the HD phenotype (Stack et al., 2007). Despite already being used as chemotherapy in cancer, mithramycin A and chromomycin A3 are not well-suited for chronic use, which would be required for HD treatment, due to their relatively high, dose-dependent, toxicity in humans. Nevertheless, they may serve as a template in the development of less toxic DNA-binding compounds to treat HD.

7.3. Strategies targeting non-coding RNAs

Due to their relatively high specificity, miRNAs have been investigated as potential therapeutic targets for the treatment of neurodegenerative disorders. Alternatively, miRNA mimics, miRNA precursor analogs, and anti-miRNAs could also be employed to restore miRNA homeostasis in such conditions (Junn and Mouradian, 2012). Although these RNA-based strategies are specific, a major obstacle, as with HDACIs, remains access and distribution to the brain. For instance, simple intravenous administration of anti-miRNAs conjugated to cholesterol molecules (“antagomirs”), while showing promise, failed to cross the BBB (Krutzfeldt et al., 2005). Additionally, although cholesterol facilitates cell entry, it might induce undesirable side effects (Junn and Mouradian, 2012). More invasive, direct injections into the ventricles may represent an effective way of circumventing the BBB to enhance the performance of such treatments (Kuhn et al., 2010; Yu et al., 2012). Packaging siRNAs into exosomes has been suggested as a less invasive strategy to pass the BBB (Alvarez-Erviti et al., 2011; Lakhal et al., 2013).

7.3.1. Alzheimer’s disease

Suggested miRNA targets for the treatment of AD include miR-124 and miR-195, which, when increased, could lower the levels of BACE1 and subsequently Aβ (Fang et al.,2012; Zhu et al., 2012). Alternatively, miR-323–3p, which is associated with inflammatory responses, has been proposed as a target for therapy in AD (Xu et al., 2014). Apart from being promising treatment targets, miRNA levels have also been investigated as potential diagnostic and prognostic markers for AD. For instance, Schipperetal. (2007) investigated miRNA expression in blood mononuclear cells of mild sAD patients, finding miR-34a and miR-181b to be upregulated in these patients. Although it remains to be elucidated whether these miRNAs play a significant role in AD pathology, they might serve as valuable prognostic biomarkers, especially as they can be relatively easily measured in the blood. Identifying changes in miRNA expression in very early, non- symptomatic stages of AD will substantially enhance AD diagnostic and treatment efficacy.

7.3.2. Huntington’s disease

Because HD is caused by the expression of mutant HTT, directly targeting its mRNA through RNAi is an attractive treatment strategy (Hu et al., 2009; Lombardi et al., 2009; Zhang and Friedlander, 2011). Due to the cardinal role of normal HTT in neuronal survival and functioning, it is crucial that such a treatment specifically target only mutant HTT. Choosing for adeno-associated virus short hairpin RNA (shRNA)-mediated RNAi, Harper et al. (2005) were able to improve motor function and neuropathology in transgenic N171–82Q mice. Subsequently, studies using adenovirus-shRNA, lentivirus-shRNA, adeno-associ-ated virus-miRNA, or cholesterol-conjugated siRNA were successful in downregulating mutant HTT, reducing aggregates and improving motor functions and neuropathology (Boudreau et al., 2009; DiFiglia et al., 2007; Drouet et al., 2009; Franich et al., 2008; Huang et al., 2007; Machida et al., 2006; McBride et al., 2008; Rodriguez-Lebron et al., 2005). Interestingly, chemically modified single-stranded siRNAs (ss-siRNAs) with mismatched bases have a 100-times higher mutant HTT targeting efficacy when compared to unmodified RNA, as tested in an HD mouse model expressing one mutant HTT copy with 150 CAG repeats and a normal HTT copy with 7 CAG repeats (HdhQ150/Q7 mice) after intraventricular infusion (Yu et al., 2012). This increased potency likely stems from the ability of these ss-siRNAs to distinguish mutant from normal HTT optimally, in collaboration with RISC, in a similar fashion as miRNAs.

8. Discussion and future perspectives

Epigenetic dysregulation currently garners much attention as a potentially pivotal player in aging and age-related neurodegenerative disorders, mediating interactions between genetic and environmental risk factors, or directly interacting with disease-specific pathological factors. Despite the profound differences in the epigenetic aberrancies, some similar patters begin to emerge and key-player molecules arise and build bridges between the seemingly diverse psychopathophysiology of neurodegenerative diseases, such as AD, PD, and HD. For instance, careful consideration of the (de)methylation dysregulations reveals a differential methylation pattern in genes that are accounted for the genetic predisposition of AD and PD; namely APP, PS1, BACE, APOE for AD and SNCA, PARKIN16 for PD. Moreover, there is derailled histone acetylation in all three diseases discussed and more specifically in AD and PD, a genome wide deacetylation of histones is observed. The various modifications on histone 3 are another common factor of these diseases that cannot be overlooked and especially the upregulated tri-methylation of H3K9 in both AD and HD. Finally, the deviant expression of specific ncRNAs in all the three discussed diseases posits their key-player role in their pathophysiology. Briefly, the differential expression of miR-132 and miR-29 is a common observation not only among all three age-related neurodegenerative disorders but also normal aging. MiR-22, miR-26a and miR-125 also present a differential expression pattern that is common in these diseases.

Even though the epigenetic research over more neurodegenerative disorders is expanding, their common points remain rather faint and sporadic, impeding the advancement towards innovative therapeutic strategies targeting neurodegeneration in general, instead of disease-specific processes. This notion stems from the fact that large, empirical and broad studies are rare, with most investigations using only small samples with low statistical power, focusing on very specific tissues, cell types, or genes, and looking only at one or a few epigenetic modifications (Lunnon and Mill, 2013). This substantial heterogeneity in research makes it hard to draw concrete conclusions about the exact involvement of epigenetics in neurodegeneration, stressing the need for studies with larger samples sizes, longitudinal designs with repeated sampling schemes, study designs with tissue and cell-specific analyses-but not just one type at a time-the inclusion of multiple epigenetic markers and levels, and genome-wide approaches. Although epigenome-wide association studies are performed, it should be noted that the Illumina 450k Methylation Beadchip array, which is the most commonly used platform for such studies, does not cover the complete methylome. Although this array covers most CpG-rich promoters, it may miss important phenotypically relevant variations in the methylome. Recent investigations have stressed the importance of DNA methylation at non-promoter and CpG-poor sites (Davies et al., 2012; Hansen et al., 2011; Lister et al., 2009). On a similar note, microarray- based transcriptome analyses are limited to known exons and transcripts (Guffanti et al., 2014). For a whole transcriptome approach, including known and potentially novel ncRNAs, strategies based on next-generation sequencing should be employed, complemented with proper bioinformatic analyses. When compared to proteins, a much larger proportion of the human genome is transcribed into ncRNAs (Amaral et al., 2008). Nevertheless, due to their codon-bias, open reading frames and strong sequence conservation, protein genes can be detected more reliably than ncRNAs (Raasch et al., 2010) Raasch et al. (2010 have therefore proposed a procedure combining multiple ncRNA identification strategies for increased sensitivity, but which is limited in its use for large genomes due to its high computational requirements.

An additional caveat of many published studies on epigenetics is the specificity of the detection techniques used. In the case of DNA modifications this is partly the result of the discovery of novel modifications. DNA methylation can be detected with techniques such as those based on sodium bisulfite sequencing or methyl-ation-sensitive restriction enzyme cleavage. With the discovery of DNA hydroxymethylation, however, it was found that these methods cannot distinguish between methylated and hydroxy-methylated DNA (Ito et al., 2011). By a method of quantitative subtraction, oxidative bisulfate sequencing can be used to identify DNA methylation and hydroxymethylation in parallel. This procedure involves the oxidation of 5-hmC to 5-fC and subsequently to uracil. 5-fC, however, has recently been observed to play a role in epigenetic priming, and thus has an independent function from 5-mC and 5-hmC (Song et al., 2013). Epigenetic priming of 5-fC occurs mainly at poised enhancer sequences and is thought to activate these sites, possibly through the recruitment of transcriptional coactivator EP300. To specifically detect 5-fC, Song et al. (2013) have described two methods, one of which has a single-base resolution and is also based on bisulfite sequencing. To detect 5-fC, this chemically assisted bisulfite sequencing method uses hydroxylamine protection of 5-fC to prevent it from bisulfite-mediated deamination and reduction to 5-hmC. The genomic location of 5-fC can then be determined by comparing hydroxylamine-treated bisulfite sequencing with traditional bisulfite sequencing. Sequencing of one of the various epigenetic DNA modifications should thus not be done without taking into account the other, functionally different, DNA modifications.

Although epigenomic profiling provides valuable gene-specific information, the input material for profiling studies often consists of tissue homogenates. Investigations into the regional and cellular specific effects of diseases illustrate that certain regions and cell types are often differentially affected and using homogenates may thus prevent the proper detection of potentially crucial epigenetic changes that only occur in a limited number of cells (Blalock et al., 2011). Indeed, in the healthy brain region-specific differentially methylated regions can be distinguished (Davies et al., 2012; Ladd-Acosta et al., 2007). Even when using a homogenate of a specific brain region of interest, different cell types could still give interfering read-outs, for instance when considering the widely different levels of 5-mC and 5-hmC between cerebellar Purkinje and granule cells (Kriaucionis and Heintz, 2009). Although attempts are being made to investigate cell-specific epigenetic profiles these studies are few and are mostly limited to DNA methylation (Guo et al., 2013). Interestingly, Guintivano et al. (2013) have developed a model to correct DNA methylation patterns for cellular heterogeneity in the brain. Additionally, various methods to isolate cells of interest are nowadays available, including density gradients (Whittemore et al., 1993), laser capture microdissection (Suarez-Quian et al., 1999), fluorescence-activated cell sorting (Uchida et al., 2000) and magnetic affinity cell sorting (Yu et al., 2004). These methods have only been sparingly used for epigenetic studies and need to be validated for this purpose. 1t has been suggested that the isolation processes themselves could already influence gene expression (Lunnon and Mill, 2013).

When looking at the potentially high variability of epigenetic markers across different tissue and cell types it may thus be worth investing in novel techniques such as CLARITY (Chung et al., 2013) and fluorescent in situ RNA sequencing (Lee et al., 2014) to determine the regional distribution of epigenetic markers and how this may result in regional differences in RNA and protein expression. To complicate matters further, there is increasing evidence that mitochondrial gene expression is also epigenetically regulated, the investigation of which presents a whole new set of challenges (Devall et al., 2014).

For molecular studies of the human brain most investigations depend on post-mortem tissue donated by patients. Apart from possible influences of cell isolation techniques on epigenetics markers, various peri- and post-mortem factors, such as postmortem interval, are known to affect tissue components, including RNA, and which could thus potentially affect epigenetic analyses (Barton et al., 1993; Stan et al., 2006). Such factors are thus most likely to influence ncRNA quality, but are in addition likely to compromise chromatin structure and possibly some DNA modifications. DNA methylation, however, is thought to be relatively stable and thereby represents one of the more reliable epigenetic markers when analyzing post-mortem tissue (Pidsley and Mill, 2011).

Although many epigenetic changes are associated with aging and neurodegeneration, it remains unclear whether they are integral to the aging and neurodegenerative processes, or are an epiphenome-non; the result of other factors such as increased oxidative stress. Investigating causality with respect to epigenetic alterations is challenging in epidemiological studies and especially in studies relying on post-mortem human tissue (Mill and Heijmans, 2013; Pidsley and Mill, 2011). Epigenetic alterations identified through the comparison of epigenetic profiles of post-mortem tissue between disease states and controls could be a combination of disease instigating alterations, but also epigenetic changes that are secondary to disease pathology (Relton and Davey Smith, 2012) as well as changes that are an effect of medication (Boks et al., 2012). Thus, when disease related epigenetic alterations are identified in epigenome-wide association studies, a major issue is to determine whether such changes actually played a role in the etiopathogenesis of the disease. An approach to overcome this hurdle would be to compare post-mortem brain samples of subjects with varying stages of disease severity and including samples from preclinical, possibly prodromal stages of the disease (Lunnon and Mill, 2013). Control samples should be very carefully selected, as for example amyloid plaques, a pathological hallmark of AD, also occur in subjects without any overt symptoms of the disease. Additionally, comparisons between familial and sporadic cases could help in the identification of causal epigenetic alternations and those that might be the secondary result of genetic mutations. Disease-specific epigenetic changes could in addition be identified by comparing patients with the target disease, with patients with similar diseases, such as frontotemporal lobar degeneration, and dementia with Lewy bodies when looking at AD, PD, and HD.

Alternatively, determining the exact role of epigenetic alterations in progressive, age-related neurodegenerative diseases could be achieved through the longitudinal assessment of the epigenome, starting with individuals in a preclinical stage of the disease. However, assessing the epigenome of living individuals is only achievable in easily accessible tissues, such as peripheral blood. Although robust disease associated epigenetic markers in the blood have great potential as diagnostic and prognostic markers, thorough comparisons between such markers in the blood and brain should be made before their relevance to the disease process can be established. Although many tissue-specific differentially methylated regions related to tissue-specific gene expression can be identified, an important study by Davies et al. (2012) indicates that at least some inter-individual methylomic variation is represented in both brain and blood. Blood sampling could be used to investigate epigenetic markers in the brain in such cases. In addition to DNA methylation, chromatin status and ncRNAs in peripheral mononuclear cells have been identified as potential diagnostic markers for brain-related conditions (Pasinetti et al., 2012; Sharma, 2012). Currently, for AD, PD, and HD, it is largely unknown whether epigenetic alterations relevant to the disease process are present in the blood. Nevertheless, in the case of AD, some changes in the blood transcriptome reflect disease-related changes in the brain (Lunnon et al., 2012).

Animal models could potentially be used to determine the relationships between disease-associated epigenetic markers in the brain and those in blood. Additionally, the epigenetic effects of specific environmental factors, such as medication, can be investigated in isolation from other possible confounding factors (Lunnon and Mill, 2013). Presently, the most used models are transgenic mouse models that express mutated human genes associated with familial disease forms (German and Eisch, 2004). Overexpressing human mutant APP in mice may, however, result in unwanted side-effects as these models will likely also have elevated levels of APP-related products such as C-terminal fragment-β/α and amyloid precursor protein intracellular domain (Saito et al., 2014). As Aβ plays an important role in AD, the increased presence of these additional APP-related products and APP itself may limited the usefulness of such models. Saito et al. (2014) have recently circumvented these problems of APP overexpression by directly manipulating the mouse App gene, inducing fAD-related mutations that selectively enhance Aβ production and the Aβ42 to Aβ40 ratio, without affecting APP expression. Nevertheless, there are so far only few animal models of the more common late-onset sporadic forms of AD and PD. Some examples of sAD models are those based on APOE (Raber et al., 1998), or specific environmental/pharmacological interventions such as colchicine (Kumar et al., 2007), cholesterol (Sparks et al., 1994), or inhibition of the neuronal insulin receptor (Hoyer et al., 2000). In case of sPD, models based on toxins are mainly used, such as those using the MPTP neurotoxin, which induces a permanent PD-like syndrome (Przedborski and Vila, 2003).

Using rodents to model diseases that occur mainly at the end of the life-span is attractive as they age relatively quickly. Rodent physiology, however, might prove to be too different to allow for the generation of a true model of sporadic late-onset neurodegenerative diseases. Moreover, as there are no natural counterparts of most of these diseases in rodents the successful generation of a true model depends on the available knowledge about the disease, which is in the case of sporadic late-onset neurodegenerative diseases very limited. Therefore, other model organisms may be more suitable, including non-human primates, which can naturally develop limited AD-like pathology (Podlisny et al., 1991), and in vitro models. Especially human primary cultures and induced pluripotent stem cells (iPSCs) represent highly promising alternatives to animal models (lsrael et al., 2012; Wojda and Kuznicki, 2013). A number of methods have been described to generate iPSCs from easily accessible fibroblasts that can be differentiated into neurons, or induce neural progenitor-like cells (iNPCs) directly (Qiang et al., 2011; Takahashi et al., 2007; Tian et al., 2013; Verma and Verma, 2011). However, these methods have not been fully optimized yet and involve procedures that induce widespread epigenetic alterations (Kim et al., 2010a).

To map the sequence of events leading to the development of complex diseases fully, epigenomic data should not be investigated in isolation, but should be complemented with other modalities, including genomic, transcriptomic and proteomic data (Meaburn et al., 2010). As such, through the integration of genetic and epigenetic approaches (Mill, 2011), non-coding genetic variation might be found to influence gene expression through epigenetic mechanisms. Such integrated data may also help in determining where in the etiopathogenesis of complex neurodegenerative conditions epigenetic players start to play a role. lntegrated knowledge may additionally help to reveal whether therapeutic strategies targeting epigenetic mechanisms should have a general mode of action, aiming at, for example, DNA methylation at large (Szyf, 2014), or a more targeted approach, for example changing the DNA methylation status of a specific DNA sequence (de Groote et al., 2012).

Concluding, although it is clear that various levels of epigenetic regulation, including DNA and chromatin modifications, and ncRNAs, are affected during aging, AD, PD, and HD, it remains to be elucidated exactly how these epigenetic processes fit into the etiopathogenesis of these disorders and whether they play a causal role. Such knowledge is crucial for the exploration of novel therapeutic avenues, which are sorely needed to combat the devastating neurodegenerative diseases.

Acknowledgements

We thank T. Vaessen for helping with the figures. Funds have been provided by the lnternationale Stichting Alzheimer Onder-zoek (lSAO) grants 07551 and 11532 (D.L.A.vdH.), by the lSAO grants 09552 and 13515, and the Netherlands Organization for Scientific Research (NWO), grant 916.11.086 (Veni Award) (B.P.F.R.), the Maastricht University Medical Centre+ (Koostra Talent Fellowship) (R.L.), and the National lnstitutes of Health grant P50 AG005138 (P.R.H.).

Abbreviations:

3-mA

3-methyladenine

3R-tau

tau with 3 microtubule-binding repeats

3xTg-AD

triple transgenic mouse model of AD

4PBA

4-phenylbutyrate

4E-BP

eukaryotic translation initiation factor 4E-binding protein

4R-tau

tau with 4 microtubule-binding repeats

5-caC

5-carboxylcytosine

5-fC

5-formylcytosine

5-hmC

5- hydroxymethylcytosine

5-hmU

5-hydroxymethyluracil

5-mC

5-methylcytosine

7-mG

7-methylguanine

A

adenosine

amyloid β protein

ABCA1

ATP-binding cassette, subfamily A (ABC1), member 1

ABCA7

ATP-binding cassette, subfamily A (ABC1), member 7

ac

acetylation (as in H3K9ac)

AD

Alzheimer’s disease

ADAM10

a disintegrin and metalloproteases domain 10

ADAR

adenosine deaminases that act on RNA

ADAT

adenosine deaminases that act on tRNAs

ADP

adenosine diphosphate

AGK2

2-cyano-3-(5-(2,5-dichlorophenyl)-2-furanyl)-N-5-quinolinyl-2-propenamide

AICDA

activation-induced cytidine deaminase

ALKBH5

AlkBalkylation repair homolog 5 (E. coli)

Alu

Arthrobacter luteus elements

ANK1

ankyrin 1

antagomirs

anti-miRNAs conjugated to cholesterol molecules

APOBEC

apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like protein

APOE

apolipoprotein E

APP

amyloid β precursor protein

APP23 mice

transgenic mouse model with a 7-fold overexpression of APPswe

APP/PS1–21 mice

transgenic mice overexpressing APPswe and human PS1 with the L166P mutation, line 21

APPind

human APP with the V717F Indiana mutation

APPswe

human APP isoform 695 with the double KM670/671NL Swedish mutation

ATP

adenosine triphosphate

ATP13A2

ATPase type 13A2

BACE

β-secretase

BACE1-AS

BACE1-antisense

BAF

Brg1/hBrm associated factor

BAG2

B-cell chronic lymphocytic leukemia (CLL)/lymphoma 2 (BCL2)-associated athanogene 2

BBB

blood–brain barrier

BC200

brain cytoplasmic RNA 200

BDNF

brain-derived neurotrophic factor

BLCAP

bladder cancer associated protein

C

cytosine

C57BL/6 mice

C57 black 6 inbred mouse strain

CA

cornu ammonis

CAG

cytosine-adenine-guanine

cAMP

cyclic adenosine monophosphate

caspase

cysteine-dependent aspartate-directed protease

CBP

cAMP response element-binding protein binding protein

CD2AP

cluster of differentiation 2-associated protein

CD33

cluster of differentiation 33

CDK2AP

cyclin-dependent kinase 2 associated protein

C. elegans

Caenorhabditis elegans

CHD

chromodomain, helicase, DNA binding

CHRM1

muscarinic acetylcholine receptor 1

CLU

clusterin

COG3

golgi complex subunit 3

COX-2

cyclooxygenase-2

CpG

cytosine-phosphate-guanine

CR1

complement component receptor 1

CREB

cAMP response element-binding protein

CREBBP

CREB binding protein

CYFIP2

cytoplasmic FMR1-interacting protein 2

DAC

5-aza-2-deoxycytidine (decitabine)

DAF-16/FOXO

dauer 16/forkhead box O

DG

dentate gyrus

DNMT

DNA methyltransferase

DNMT3L

DNMT3-like

DP

differentiation regulated transcription factor protein

dsRNA

small double-stranded RNA

E1A

adenovirus early region 1A

E2F1

E2F transcription factor 1

EC

entorhinal cortex

EDARADD

ectodysplasin-A receptor-associated death domain

EID1

EP300 interacting inhibitor of differentiation 1

EP300

E1A-binding protein P300

ERK1

extracellular signal-regulated kinase 1

eRNA

enhancer RNA

EZH

enhancer of zeste homolog (Drosophila)

f6A

N6-formyladenosine

fAD

familial AD

FANCC

Fanconi anemia, complementation group C

FGF20

fibroblast growth factor 20

FLNA

filamin1

fPD

familial PD

FRM-0334

Forum Pharmaceuticals compound 0334

FTO

fat mass and obesity-associated protein

GABRA2

gamma-aminobutyric acid receptor subunit alpha 2

GAD-3

gastrulation defective 3

GADD45

growth arrest and DNA damage 45

GCF

granulocyte chemotactic factor

GCN

general control of amino acid synthesis

GDNF

glial cell line-derived neurotrophic factor

GFAP

glial fibrillary acidic protein

GluR

glutamate receptor

GPNMB

glycoprotein (transmembrane) nmb

GR1

glucocorticoid receptor 1

GSK-3β

glycogen synthase kinase 3β

His

histidine

H

histone protein, always followed by a number (for example, H3 in H3K4)

H2AX

histone protein H2A member X

HAT

histone acetyltransferase

Hcy

homocysteine

HD

Huntington’s disease

HDAC

histone deacetylase

HDACI

HDAC inhibitor

HdhQ150/Q7 mice

transgenic mouse model expressing one mutant HTT copy with 150 CAG repeats and a normal HTT copy with 7 CAG repeats

HEK293

human embryonic kidney 293

HKDM

histone lysine demethylase

HKMT

histone lysine methyltransferase

hm6A

N6-hydroxymethyladenosine

HRDM

histone arginine demethylase

HSP90

heat shock protein 90

HTT

Huntingtin

HuR

human antigen R

I

inosine

IBA1

ionized calcium-binding adapter molecule 1

IGF1

insulin-like growth factor 1

IKK

inhibitor of kappaB kinase

INO

inositol requiring 80

iNPC

induced neural progenitor-like cell

iPSC

induced pluripotent stem cell

IRAK1

interleukin-1 receptor-associated kinase 1

ISWI

imitation SWI

JMJD

jumonji domain containing

K

lysine (as in H3K9ac)

KAT

lysine acetyltransferase

Kcnal

potassium channel gene

L-DOPA

L-3,4-dihydroxy-phenylalanine

lincRNA

large intergenic non-coding RNA

LINE-1

long interspersed element 1

lncRNA

long ncRNA

LRRK2

leucine-rich repeat kinase 2

m1A

N1-methyladenosin

m1G

N1-methylguanine

m6A

N6-methyladenosine

MALAT

metastasis associated lung adenocarcinoma transcript 1

MAP2

microtubule-associated protein 2

MAPK

mitogen-activated protein kinase

MAPT

microtubule-associated protein tau

MAT

methionine adenosyltransferase

MBD

methyl-CpG-binding domain protein

me

methylation (as in H3K4me3)

MeCP

methyl CpG-binding protein

MetH

methionine synthase

METTL

methyltransferase-like protein

miR

microRNA

miRNA

microRNA

MPP+

1-methyl-4-phenyl-pyridinium ion

MPTP

1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine

mRNA

messenger RNA

MS4A4E

membrane-spanning 4-domains, subfamily A, member 4E

MS4A6A

membrane-spanning 4-domains, subfamily A, member 6A

MSK

mitogen- and stress-activated protein kinase

mtDNA

mitochondrial DNA

mtDNMT1

mitochondrial DNMT1

MTHF

methylenetetrahydrofolate

MTHFR

MTHF reductase

N171–82Q mice

transgenic mouse model expressing the first 171 amino acids of HTT with 82 CAG repeats at a relatively low steady-state level

N2a

Neuro-2a

NAD

nicotine adenine dinucleotide

NAT

natural antisense transcript

ncRNA

non-coding RNA

NDM29

neuroblastoma differentiation marker 29

NEAT

nuclear paraspleckle assembly transcript

NeuN

Neuronal nuclei

NF-κB

nuclear transcription factor kappa B

NFT

neurofibrillary tangle

NMDA

N-methyl-D-aspartic acid

NPTX

neuronal pentraxin

NuRD

nucleosome remodeling and histone deacetylase

p

phosphorylation (as in H3S10p)

P300

E1A-binding protein P300

PAR

promoter-associated RNA

PARK16

Parkinson disease 16

PASR

promoter-associated short RNAs

P-bodies

processing bodies

PCAF

P300/CBP-associated factor

PcG

Polycomb-group

PD

Parkinson’s disease

PDAPP mice

transgenic mice overexpressing APPind

PGC1-α

peroxisome proliferator receptor gamma coactivator-1 alpha

PICALM

phosphatidylinositol binding clathrin assembly protein

piRNA

piwi-interacting RNA

PKCδ

protein kinase Cδ

PP2A

protein phosphatase 2A

PPT

protein phosphatase

PRC1

polycomb repressive complex member Bmi1

PRC2

polycomb repressive complex member EZH2

pre-miRNA

precursor miRNA

PRMT

protein arginine methyltransferase

PROMPTS

promoter upstream transcripts

PS

presenilin

PS1ΔE9

human PS1 deleted in exon 9 mutation

PSD95

postsynaptic density protein 95

PTBP2

polypyrimidine tract binding protein 2

Psid-ps1

phosphatidylserine decarboxylase- pseudogene 1

R

arginine (as in H3R2me)

R6/2 mice

transgenic mice overexpressing exon 1 of human HTT with an expanded CAG repeat length

RAN

rat sarcoma (Ras)-related nuclear protein

rasiRNA

repeat-associated small interfering RNA

RISC

RNA-induced silencing complex

RNAi

RNA interference

RNAPII

RNA polymerase II

RNAPIII

RNA polymerase III

ROS

reactive oxygen species

rRNA

ribosomal RNA

S

serine (as inH3S10p)

S100A2

S100 calcium-binding protein A2

sAD

sporadic AD

SAH

S-adenosylhomocysteine

SAHA

suberoylanilide hydroxamic acid

SAHF

senescence-associated heterochromatin foci

SAHH

SAH hydrolase

SAM

S-adenosylmethionine

SAMP8

senescence-accelerated prone mouse 8

SAT-α

satellite-α

SB

sodium butyrate

scaRNA

small Cajal body-specific RNA

sdRNA

snoRNA-derived small RNA

SETDB1

Drosophila Su(var)3–9 and enhancer of zeste proteins (SET) domain, bifurcated 1

SHMT

serine hydroxymethyltransferase

shRNA

short hairpin RNA

siRNA

small interfering RNA

smRNA

small modulatory RNA

SIRT

sirtuin

SNCA

synuclein alpha

sncRNA

small ncRNAs

snRNA

small nuclear RNA

snoRNA

small nucleolar RNA

SNP

single nucleotide polymorphism

SORBS3

sorbin and v-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog homology 3 domain containing 3

SP

specificity factor

sPD

sporadic PD

spliRNA

splice junction-associated RNA

SPT

serine palmitoyltransferase

ss-siRNA

single-stranded siRNA

STX1B

syntaxin 1B

SUV39H

suppressor of variegation 3–9 homologue

SWI/SNF

switching defective/sucrose nonfermenting

T

threonine

TACE

TNF-α converting enzyme

TE

transposable element

TET

ten-eleven translocation

Tg19959 mice

transgenic mice overexpressing a combination of APPswe and APPind

Tg2576 mice

transgenic mice overexpressing APPswe

THF

tetrahydrofolate

TIP60

human immunodeficiency virus type 1 transactivating protein interactive protein

tiRNA

transcription initiation RNA

TNF-α

tumor necrosis factor alpha

TNKS2

tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase 2

TOM1L

target of myb1 (chicken)-like

TREM2

triggering receptor expressed on myeloid cells 2

TRF1

telomeric repeat binding factor 1

tRNA

transfer RNA

TSA

trichostatin A

TSSa-RNA

transcription start site-associated RNA

U

uracil

UCHL1

ubiquitin carboxy-terminal hydrolase L1

UTR

untranslated region

VPA

valproate/valproic acid

WTAP

Wilm’s tumor-associated protein

Y

tyrosine

YTHDF

YTH domain family

Footnotes

The authors declare no conflict of interest.

References

  1. Aarsland D, Larsen JP, Cummins JL, Laake K, 1999. Prevalence and clinical correlates of psychotic symptoms in Parkinson disease: a community-based study. Arch. Neurol. 56, 595–601. [DOI] [PubMed] [Google Scholar]
  2. Abeti R, Abramov AY, Duchen MR, 2011. Beta-amyloid activates PARP causing astrocytic metabolic failure and neuronal death. Brain 134, 1658–1672. [DOI] [PubMed] [Google Scholar]
  3. Adwan L, Zawia NH, 2013. Epigenetics: a novel therapeutic approach for the treatment of Alzheimer’s disease. Pharmacol. Ther. 139, 41–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G, Christensen J, Helin K, 2009. The H3K27me3 demethylase JMJD3 contributes to the activation of the lNK4A-ARF locus in response to oncogene- and stress-induced senescence. Genes Dev. 23, 1171–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Agirre X, Roman-Gomez J, Vazquez l., Jimenez-Velasco A, Garate L, Montiel- Duarte C, Artieda P, Cordeu L, Lahortiga l., Calasanz MJ, Heiniger A, Torres A, Minna JD, Prosper F, 2006. Abnormal methylation of the common PARK2 and PACRG promoter is associated with downregulation of gene expression in acute lymphoblastic leukemia and chronic myeloid leukemia. lnt. J. Cancer 118, 1945–1953. [DOI] [PubMed] [Google Scholar]
  6. Agis-Balboa RC, Pavelka Z, Kerimoglu C, Fischer A, 2013. Loss ofHDAC5 impairs memory function: implications for Alzheimer’s disease. J. Alzheimers Dis. 33, 35–44. [DOI] [PubMed] [Google Scholar]
  7. Aguiar RS, Peterlin BM, 2008. APOBEC3 proteins and reverse transcription. Virus Res. 134, 74–85. [DOI] [PubMed] [Google Scholar]
  8. Akbarian S, Smith MA, Jones EG, 1995. Editing for an AMPA receptor subunit RNA in prefrontal cortex and striatum in Alzheimer’s disease, Huntington’s disease and schizophrenia. Brain Res. 699, 297–304. [DOI] [PubMed] [Google Scholar]
  9. Akram A, Schmeidler J, Katsel P, Hof PR, Haroutunian V, 2010. lncreased expression of cholesterol transporter ABCA1 is highlycorrelated with severity of dementia in AD hippocampus. Brain Res. 1318, 167–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Alarcon JM, Malleret G, Touzani K, Vronskaya S, lshii S, Kandel ER, Barco A, 2004. Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron 42, 947–959. [DOI] [PubMed] [Google Scholar]
  11. Alarcón JM, Malleret G, Touzani K, Vronskaya S, lshii S, Kandel ER, Barco A, 2004. Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron 42, 947–959. [DOI] [PubMed] [Google Scholar]
  12. Allis CD, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, Pillus L, Reinberg D, Shi Y, Shiekhattar R, Shilatifard A, Workman J, Zhang Y, 2007. New nomenclature for chromatin-modifying enzymes. Cell 131, 633–636. [DOI] [PubMed] [Google Scholar]
  13. Alonso AD, Grundke-lqbal l., Barra HS, lqbal K, 1997. Abnormal phosphorylation of tau and the mechanism of Alzheimer neurofibrillary degeneration: sequestration of microtubule-associated proteins 1 and 2 and the disassembly of microtubules by the abnormal tau. Proc. Natl. Acad. Sci. U.S.A. 94, 298–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ, 2011. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341–345. [DOI] [PubMed] [Google Scholar]
  15. Alzheimer’s Association, 2013. 2013 Alzheimer’s disease facts and figures. Alzheimers Dement. 9, 208–245. [DOI] [PubMed] [Google Scholar]
  16. Amaral PP, Dinger ME, Mercer TR, Mattick JS, 2008. The eukaryotic genome as an RNA machine. Science 319, 1787–1789. [DOI] [PubMed] [Google Scholar]
  17. Ambros V, 2004. The functions of animal microRNAs. Nature 431, 350–355. [DOI] [PubMed] [Google Scholar]
  18. Ammal Kaidery N, Tarannum S, Thomas B, 2013. Epigenetic landscape of Parkinson’s disease: emerging role in disease mechanisms and therapeutic modalities. Neurother.: J. Am. Soc. Exp. Neurother. 10, 698–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Anderson J, Phan L, Hinnebusch AG, 2000. The Gcd10p/Gcd14p complex is the essential two-subunit tRNA(1-methyladenosine) methyltransferase of Saccha-romyces cerevisiae. Proc. Natl. Acad. Sci. U.S.A. 97, 5173–5178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Araki T, Sasaki Y, Milbrandt J, 2004. lncreased nuclear NAD biosynthesis and SlRT1 activation prevent axonal degeneration. Science 305, 1010–1013. [DOI] [PubMed] [Google Scholar]
  21. Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M, 2012. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11, 384–400. [DOI] [PubMed] [Google Scholar]
  22. Ashe HL, Monks J, Wijgerde M, Fraser P, Proudfoot NJ, 1997. Intergenic transcription and transinduction of the human beta-globin locus. Genes Dev. 11, 2494–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Asikainen S, Rudgalvyte M, Heikkinen L, Louhiranta K, Lakso M, Wong G, Nass R, 2010. Global microRNA expression profiling of Caenorhabditis elegans Parkinson’s disease models. J. Mol. Neurosci. 41, 210–218. [DOI] [PubMed] [Google Scholar]
  24. Athanasiadis A, Rich A, Maas S, 2004. Widespread A-to-I RNA editing of Alu-containing mRNAs in the human transcriptome. PLoS Biol. 2, e391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Babenko O, Kovalchuk I, Metz GA, 2012. Epigenetic programming of neurode-generative diseases by an adverse environment. Brain Res. 1444, 96–101. [DOI] [PubMed] [Google Scholar]
  26. Bachellerie JP, Cavaille J, Huttenhofer A, 2002. The expanding snoRNA world. Biochimie 84, 775–790. [DOI] [PubMed] [Google Scholar]
  27. Bae BI, Xu H, Igarashi S, Fujimuro M, Agrawal N, Taya Y, Hayward SD, Moran TH, Montell C, Ross CA, Snyder SH, Sawa A, 2005. p53 mediates cellular dysfunction and behavioral abnormalities in Huntington’s disease. Neuron 47, 29–41. [DOI] [PubMed] [Google Scholar]
  28. Barrachina M, Ferrer I, 2009. DNA methylation of Alzheimer disease and tauo-pathy-related genes in postmortem brain. J. Neuropathol. Exp. Neurol. 68, 880–891. [DOI] [PubMed] [Google Scholar]
  29. Barreto G, Schafer A, Marhold J, Stach D, Swaminathan SK, Handa V, Doder- lein G, Maltry N, Wu W, Lyko F, Niehrs C, 2007. Gadd45a promotes epigenetic gene activation by repair-mediated DNA demethylation. Nature 445, 671–675. [DOI] [PubMed] [Google Scholar]
  30. Barrey E, Saint-Auret G, Bonnamy B, Damas D, Boyer O, Gidrol X, 2011. Pre- microRNA and mature microRNA in human mitochondria. PLoS ONE 6, e20220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K, 2007. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837. [DOI] [PubMed] [Google Scholar]
  32. Barton AJ, Pearson RC, Najlerahim A, Harrison PJ, 1993. Pre- and postmortem influences on brain RNA. J. Neurochem. 61, 1–11. [DOI] [PubMed] [Google Scholar]
  33. Bass BL, 2002. RNA editing by adenosine deaminases that act on RNA. Annu. Rev. Biochem. 71, 817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bates EA, Victor M, Jones AK, Shi Y, Hart AC, 2006. Differential contributions of Caenorhabditis elegans histone deacetylases to huntingtin polyglutamine toxicity. J. Neurosci. 26, 2830–2838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA, 2006. Resveratrol improves health and survival of mice on a high- calorie diet. Nature 444, 337–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Beal MF, Ferrante RJ, 2004. Experimental therapeutics in transgenic mouse models of Huntington’s disease. Nat. Rev. Neurosci. 5, 373–384. [DOI] [PubMed] [Google Scholar]
  37. Beckman KB, Ames BN, 1998. The free radical theory of aging matures. Physiol. Rev. 78, 547–581. [DOI] [PubMed] [Google Scholar]
  38. Bediou B, Ryff I, Mercier B, Milliery M, Henaff MA, D’Amato T, Bonnefoy M, Vighetto A, Krolak-Salmon P, 2009. Impaired social cognition in mild Alzheimer disease. J. Geriatr. Psychiatry Neurol. 22, 130–140. [DOI] [PubMed] [Google Scholar]
  39. Behm-Ansmant I, Helm M, Motorin Y, 2011. Use of specific chemical reagents for detection of modified nucleotides in RNA. J. Nucleic Acids 2011, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Bellizzi D, D’Aquila P, Scafone T, Giordano M, Riso V, Riccio A, Passarino G, 2013. The control region of mitochondrial DNA shows an unusual CpG and non- CpG methylation pattern. DNA Res. 20, 537–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Bergman Y, Cedar H, 2013. DNA methylation dynamics in health and disease. Nat. Struct. Mol. Biol. 20, 274–281. [DOI] [PubMed] [Google Scholar]
  42. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES, 2006. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326. [DOI] [PubMed] [Google Scholar]
  43. Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M, 1999. A mammalian protein with specific demethylase activity for mCpG DNA. Nature 397, 579–583. [DOI] [PubMed] [Google Scholar]
  44. Bicchi I, Morena F, Montesano S, Polidoro M, Martino S, 2013. MicroRNAs and molecular mechanisms of neurodegeneration. Genes 4, 244–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bilen J, Liu N, Burnett BG, Pittman RN, Bonini NM, 2006. MicroRNA pathways modulate polyglutamine-induced neurodegeneration. Mol. Cell 24, 157–163. [DOI] [PubMed] [Google Scholar]
  46. Bjork GR, Wikstrom PM, Bystrom AS, 1989. Prevention of translational frame- shifting by the modified nucleoside 1-methylguanosine. Science 244, 986–989. [DOI] [PubMed] [Google Scholar]
  47. Blalock EM, Buechel HM, Popovic J, Geddes JW, Landfield PW, 2011. Microarray analyses of laser-captured hippocampus reveal distinct gray and white matter signatures associated with incipient Alzheimer’s disease. J. Chem. Neuroanat. 42, 118–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Blanco G, Fu H, Mendez C, Khosla C, Salas JA, 1996. Deciphering the biosynthetic origin of the aglycone of the aureolic acid group of anti-tumor agents. Chem. Biol. 3, 193–196. [DOI] [PubMed] [Google Scholar]
  49. Blow MJ, Grocock RJ, van Dongen S, Enright AJ, Dicks E, Futreal PA, Wooster R, Stratton MR, 2006. RNA editing of human microRNAs. Genome Biol. 7, R27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Bocklandt S, Lin W, Sehl ME, Sanchez FJ, Sinsheimer JS, Horvath S, Vilain E, 2011. Epigenetic predictor of age. PLoS ONE 6, e14821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Boissonneault V, Plante I, Rivest S, Provost P, 2009. MicroRNA-298 and micro-RNA-328 regulate expression of mouse beta-amyloid precursor protein-converting enzyme 1. J. Biol. Chem. 284, 1971–1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Boks MP, de Jong NM, Kas MJ, Vinkers CH, Fernandes C, Kahn RS, Mill J, Ophoff RA, 2012. Current status and future prospects for epigenetic psychopharmacology. Epigenetics 7, 20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Bollati V, Galimberti D, Pergoli L, Dalla Valle E, Barretta F, Cortini F, Scarpini E, Bertazzi PA, Baccarelli A, 2011. DNA methylation in repetitive elements and Alzheimer disease. Brain Behav. Immun. 25, 1078–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Bollati V, Schwartz J, Wright R, Litonjua A, Tarantini L, Suh H, Sparrow D, Vokonas P, Baccarelli A, 2009. Decline in genomic DNA methylation through aging in a cohort of elderly subjects. Mech. Ageing Dev. 130, 234–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Boon RA, Iekushi K, Lechner S, Seeger T, Fischer A, Heydt S, Kaluza D, Treguer K, Carmona G, Bonauer A, Horrevoets AJ, Didier N, Girmatsion Z, Biliczki P, Ehrlich JR, Katus HA, Muller OJ, Potente M, Zeiher AM, Hermeking H, Dimmeler S, 2013. MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–110. [DOI] [PubMed] [Google Scholar]
  56. Boon RA, Seeger T, Heydt S, Fischer A, Hergenreider E, Horrevoets AJ, Vinciguerra M, Rosenthal N, Sciacca S, Pilato M, van Heijningen P, Essers J, Brandes RP, Zeiher AM, Dimmeler S, 2011. MicroRNA-29 in aortic dilation: implications for aneurysm formation. Circ. Res. 109, 1115–1119. [DOI] [PubMed] [Google Scholar]
  57. Bottiglieri T, Godfrey P, Flynn T, Carney MW, Toone BK, Reynolds EH, 1990. Cerebrospinal fluid S-adenosylmethionine in depression and dementia: effects of treatment with parenteral and oral S-adenosylmethionine. J. Neurol. Neu-rosurg. Psychiatry 53, 1096–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Boudreau RL, McBride JL, Martins I, Shen S, Xing Y, Carter BJ, Davidson BL, 2009. Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington’s disease mice. Mol. Ther.: J. Am. Soc. Gene Ther. 17, 1053–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Boulias K, Horvitz HR, 2012. The C. elegans microRNA mir-71 acts in neurons to promote germline-mediated longevity through regulation of DAF-16/FOXO. Cell Metab. 15, 439–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Bradley-Whitman MA, Lovell MA, 2013. Epigenetic changes in the progression of Alzheimer’s disease. Mech. Ageing Dev. 134, 486–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Brahe C, Vitali T, Tiziano FD, Angelozzi C, Pinto AM, Borgo F, Moscato U, Bertini E, Mercuri E, Neri G, 2005. Phenylbutyrate increases SMN gene expression in spinal muscular atrophy patients. Eur. J. Hum. Genet.: EJHG 13, 256–259. [DOI] [PubMed] [Google Scholar]
  62. Brami-Cherrier K, Roze E, Girault JA, Betuing S, Caboche J, 2009. Role of the ERK/MSK1 signalling pathway in chromatin remodelling and brain responses to drugs of abuse. J. Neurochem. 108, 1323–1335. [DOI] [PubMed] [Google Scholar]
  63. Brasnjevic I, Lardenoije R, Schmitz C, Van Der Kolk N, Dickstein DL, Takahashi H, Hof PR, Steinbusch HWM, Rutten BPF, 2013. Region-specific neuron and synapse loss in the hippocampus of APP(SL)/PS1 knock-in mice. Transl. Neurosci. 4, 8–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Brett JO, Renault VM, Rafalski VA, Webb AE, Brunet A, 2011. The microRNA cluster miR-106b~25 regulates adult neural stem/progenitor cell proliferation and neuronal differentiation. Aging 3, 108–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Bringmann P, Luhrmann R, 1987. Antibodies specific for N6-methyladenosine react with intact snRNPs U2 and U4/U6. FEBS Lett. 213, 309–315. [DOI] [PubMed] [Google Scholar]
  66. Brohede J, Rinde M, Winblad B, Graff C, 2010. A DNA methylation study of the amyloid precursor protein gene in several brain regions from patients with familial Alzheimer disease. J. Neurogenet. 24, 179–181. [DOI] [PubMed] [Google Scholar]
  67. Brown SE, Weaver IC, Meaney MJ, Szyf M, 2008. Regional-specific global cytosine methylation and DNA methyltransferase expression in the adult rat hippocampus. Neurosci. Lett. 440, 49–53. [DOI] [PubMed] [Google Scholar]
  68. Budson AE, Solomon PR, 2012. New criteria for Alzheimer disease and mild cognitive impairment: implications for the practicing clinician. Neurologist 18, 356–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Bujnicki JM, Feder M, Radlinska M, Blumenthal RM, 2002. Structure prediction and phylogenetic analysis of a functionally diverse family of proteins homologous to the MT-A70 subunit of the human mRNA:m(6)A methyltransferase. J. Mol. Evol. 55, 431–444. [DOI] [PubMed] [Google Scholar]
  70. Byun HM, Baccarelli AA, 2014. Environmental exposure and mitochondrial epigenetics: study design and analytical challenges. Hum. Genet. 133,247–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL, 2011. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25, 1915–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Cacabelos R, 2005. Pharmacogenomics and therapeutic prospects in Alzheimer’s disease. Expert Opin. Pharmacother. 6, 1967–1987. [DOI] [PubMed] [Google Scholar]
  73. Cacabelos R, 2007. Pharmacogenetic basis for therapeutic optimization in Alzheimer’s disease. Mol. Diagn. Ther. 11, 385–405. [DOI] [PubMed] [Google Scholar]
  74. Caccamo A, Maldonado MA, Bokov AF, Majumder S, Oddo S, 2010. CBP gene transfer increases BDNF levels and ameliorates learning and memory deficits in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 107, 22687–22692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Cacciapuoti F, 2013. Lowering homocysteine levels with folic acid and B-vitamins do not reduce early atherosclerosis, but could interfere with cognitive decline and Alzheimer’s disease. J. Thromb. Thrombolysis 36, 258–262. [DOI] [PubMed] [Google Scholar]
  76. Caesar I, Gandy S, 2012. Evidence that an APOE e4’double whammy’ increases risk for Alzheimer’s disease. BMC Med. 10, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Caffrey TM, Joachim C, Paracchini S, Esiri MM, Wade-Martins R, 2006. Haplotype-specific expression of exon 10 at the human MAPT locus. Hum. Mol. Genet. 15, 3529–3537. [DOI] [PubMed] [Google Scholar]
  78. Cai M, Tian J, Zhao GH, Luo W, Zhang BR, 2011. Study of methylation levels of parkin gene promoter in Parkinson’s disease patients. Int. J. Neurosci. 121, 497–502. [DOI] [PubMed] [Google Scholar]
  79. Calabresi P, Centonze D, Gubellini P, Pisani A, Bernardi G, 2000. Acetylcholine- mediated modulation of striatal function. Trends Neurosci. 23, 120–126. [DOI] [PubMed] [Google Scholar]
  80. Campbell NRC, 1996. How safe are folic acid supplements? Arch. Intern. Med. 156, 1638–1644. [PubMed] [Google Scholar]
  81. Carrettiero DC, Hernandez I, Neveu P, Papagiannakopoulos T, Kosik KS, 2009. The cochaperone BAG2 sweeps paired helical filament-insoluble tau from the microtubule. J. Neurosci. 29, 2151–2161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, Fagan AM, Morris JC, Mawuenyega KG, Cruchaga C, Goate AM, Bales KR, Paul SM, Bateman RJ, Holtzman DM, 2011. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci. Transl. Med. 3, 89ra57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Cattaneo E, 2003. Dysfunction of wild-type huntingtin in Huntington disease. News Physiol. Sci.: Int. J. Physiol. Int. Union Physiol. Sci. Am. Physiol. Soc. 18, 34–37. [DOI] [PubMed] [Google Scholar]
  84. Cencioni C, Spallotta F, Martelli F, Valente S, Mai A, Zeiher AM, Gaetano C, 2013. Oxidative stress and epigenetic regulation in ageing and age-related diseases. Int. J. Mol. Sci. 14, 17643–17663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Cha JH, Kosinski CM, Kerner JA, Alsdorf SA, Mangiarini L, Davies SW, Penney JB, Bates GP, Young AB, 1998. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc. Natl. Acad. Sci. U.S.A. 95, 6480–6485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Chakrabarti S, Bhattacharyya D, Dasgupta D, 2000. Structural basis of DNA recognition by anticancer antibiotics, chromomycin A3, and mithramy-cin: Roles of minor groove width and ligand flexibility. Biopolymers 56, 85–95. [DOI] [PubMed] [Google Scholar]
  87. Chan A, Paskavitz J, Remington R, Rasmussen S, Shea TB, 2008. Efficacy of a vitamin/nutriceutical formulation for early-stage Alzheimer’s disease: a 1-year, open-label pilot study with an 16-month caregiver extension. Am. J. Alzheimer’s Dis. Other Dement. 23, 571–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Chan MF, van Amerongen R, Nijjar T, Cuppen E, Jones PA, Laird PW, 2001. Reduced rates of gene loss, gene silencing, and gene mutation in Dnmt1- deficient embryonic stem cells. Mol. Cell. Biol. 21, 7587–7600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Chandola U, Das R, Panda B, 2014. Role of the N6-methyladenosine RNA mark in gene regulation and its implications on development and disease. Brief. Funct. Genomics 14, 169–179. [DOI] [PubMed] [Google Scholar]
  90. Chang BS, Chen Y, Zhao YM, Bruick RK, 2007. JMJD6 is a histone arginine demethylase. Science 318, 444–447. [DOI] [PubMed] [Google Scholar]
  91. Chang S, Young BD, Li S, Qi X, Richardson JA, Olson EN, 2006. Histone deacetylase 7 maintains vascular integrity by repressing matrix metallopro-teinase 10. Cell 126, 321–334. [DOI] [PubMed] [Google Scholar]
  92. Chatterjee S, Mizar P, Cassel R, Neidl R, Selvi BR, Mohankrishna DV, Veda-murthy BM, Schneider A, Bousiges O, Mathis C, Cassel JC, Eswaramoorthy M, Kundu TK, Boutillier AL, 2013. A novel activator of CBP/p300 acetyl-transferases promotes neurogenesis and extends memory duration in adult mice. J. Neurosci. 33, 10698–10712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Chatterjee S, Zaman K, Ryu H, Conforto A, Ratan RR, 2001. Sequence-selective DNA binding drugs mithramycin A and chromomycin A3 are potent inhibitors of neuronal apoptosis induced by oxidative stress and DNA damage in cortical neurons. Ann. Neurol. 49, 345–354. [PubMed] [Google Scholar]
  94. Chen H, Dzitoyeva S, Manev H, 2012. Effect of valproic acid on mitochondrial epigenetics. Eur. J. Pharmacol. 690, 51–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Chen J, Zhou YG, Mueller-Steiner S, Chen LF, Kwon H, Yi SL, Mucke L, Li G, 2005. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappa B signaling. J. Biol. Chem. 280, 40364–40374. [DOI] [PubMed] [Google Scholar]
  96. Chen KL, Wang SS, Yang YY, Yuan RY, Chen RM, Hu CJ, 2009. The epigenetic effects of amyloid-beta(1–40) on global DNA and neprilysin genes in murine cerebral endothelial cells. Biochem. Biophys. Res. Commun. 378, 57–61. [DOI] [PubMed] [Google Scholar]
  97. Chen PS, Peng GS, Li G, Yang S, Wu X, Wang CC, Wilson B, Lu RB, Gean PW, Chuang DM, Hong JS, 2006. Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes. Mol. Psychiatry 11, 1116–1125. [DOI] [PubMed] [Google Scholar]
  98. Chen PS, Wang CC, Bortner CD, Peng GS, Wu X, Pang H, Lu RB, Gean PW, Chuang DM, Hong JS, 2007. Valproic acid and other histone deacetylase inhibitors induce microglial apoptosis and attenuate lipopolysaccharide-in-duced dopaminergic neurotoxicity. Neuroscience 149, 203–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Chen PY, Weinmann L, Gaidatzis D, Pei Y, Zavolan M, Tuschl T, Meister G, 2008. Strand-specific 5-O-methylation of siRNA duplexes controls guide strand selection and targeting specificity. RNA 14, 263–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Cheng LC, Pastrana E, Tavazoie M, Doetsch F, 2009. miR-124 regulates adult neurogenesis in the subventricularzone stem cell niche. Nat. Neurosci. 12,399–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Chestnut BA, Chang Q, Price A, Lesuisse C, Wong M, Martin LJ, 2011. Epigenetic regulation of motor neuron cell death through DNA methylation. J. Neurosci. 31, 16619–16636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Cho HJ, Liu G, Jin SM, Parisiadou L, Xie C, Yu J, Sun L, Ma B, Ding J, Vancraenenbroeck R, Lobbestael E, Baekelandt V, Taymans JM, He P, Troncoso JC, Shen Y, Cai H, 2013. MicroRNA-205 regulates the expression of Parkinson’s disease-related leucine-rich repeat kinase 2 protein. Hum. Mol. Genet. 22, 608–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Choi YC, Busch H, 1978. Modified nucleotides in T1 RNase oligonucleotides of 18S ribosomal RNA of the Novikoff hepatoma. Biochemistry 17, 2551–2560. [DOI] [PubMed] [Google Scholar]
  104. Choi YS, Hoon Jeong J, Min HK, Jung HJ, Hwang D, Lee SW, Kim Pak Y., 2011. Shot-gun proteomic analysis of mitochondrial D-loop DNA binding proteins: identification of mitochondrial histones. Mol. Biosyst. 7, 1523–1536. [DOI] [PubMed] [Google Scholar]
  105. Chooniedass-Kothari S, Emberley E, Hamedani M, Troup S, Wang X, Czosnek A, Hube F, Mutawe M, Watson P, Leygue E, 2004. The steroid receptor RNA activator is the first functional RNA encoding a protein. FEBS Lett. 566, 43–47. [DOI] [PubMed] [Google Scholar]
  106. Choudhuri S, 2011. From Waddington’s epigenetic landscape to small noncoding RNA: some important milestones in the history of epigenetics research. Toxicol. Mech. Methods 21, 252–274. [DOI] [PubMed] [Google Scholar]
  107. Chouliaras LLA, van den Hove D, Kenis G, Keitel SR, Hof P, van Os JWM, Steinbusch H, Schmitz CPF, Rutten B, 2012. Age-related increase in levels of 5-hydroxymethylcytosine in mouse hippocampus is prevented by caloric restriction. Curr. Alzheimer Res. 9, 536–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Chouliaras L, Mastroeni D, Delvaux E, Grover A, Kenis G, Hof PR, Steinbusch HWM, Coleman PD, Rutten PF, Van den Hove DLA, 2013a. Consistent decrease in global DNA methylation and hydroxymethylation in the hippocampus of Alzheimer’s disease patients. Neurobiol. Aging 34, 2091–2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Chouliaras L, Rutten BP, Kenis G, Peerbooms O, Visser PJ, Verhey F, van Os J, Steinbusch HW, van den Hove DL, 2010a. Epigenetic regulation in the pathophysiology of Alzheimer’s disease. Prog. Neurobiol. 90, 498–510. [DOI] [PubMed] [Google Scholar]
  110. Chouliaras L, Sierksma AS, Kenis G, Prickaerts J, Lemmens MA, Brasnjevic I, van Donkelaar EL, Martinez-Martinez P, Losen M, De Baets MH, Kholod N, van Leeuwen F, Hof PR, van Os J, Steinbusch HW, van den Hove DL, Rutten BP, 2010b. Gene-environment interaction research and transgenic mouse models of Alzheimer’s disease. Int. J. Alzheimers Dis. 2010, 1–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Chouliaras L, van den Hove DL, Kenis G, Dela Cruz J, Lemmens MA, van Os J, Steinbusch HW, Schmitz C, Rutten BP, 2011a. Caloric restriction attenuates age-related changes ofDNA methyltransferase 3a in mouse hippocampus. Brain Behav. Immun. 25, 616–623. [DOI] [PubMed] [Google Scholar]
  112. Chouliaras L, van den Hove DL, Kenis G, Draanen M, Hof PR, van Os J, Steinbusch HW, Schmitz C, Rutten BP, 2013b. Histone deacetylase 2 in the mouse hippocampus: attenuation of age-related increase by caloric restriction. Curr. Alzheimer Res. 10, 868–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Chouliaras L, van den Hove DL, Kenis G, Keitel S, Hof PR, van Os J, Steinbusch HW, Schmitz C, Rutten BP, 2011b. Prevention of age-related changes in hippocampal levels of 5-methylcytidine by caloric restriction. Neurobiol. Aging 33, 1672–1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Chung K, Wallace J, Kim SY, Kalyanasundaram S, Andalman AS, Davidson TJ, Mirzabekov JJ, Zalocusky KA, Mattis J, Denisin AK, Pak S, Bernstein H, Ramakrishnan C, Grosenick L, Gradinaru V, Deisseroth K, 2013. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Chwang WB, O’Riordan KJ, Levenson JM, Sweatt JD, 2006. ERK/MAPK regulates hippocampal histone phosphorylation following contextual fear conditioning. Learn. Mem. 13, 322–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Citron M, Teplow DB, Selkoe DJ, 1995. Generation of amyloid beta protein from its precursor is sequence specific. Neuron 14, 661–670. [DOI] [PubMed] [Google Scholar]
  117. Clapier CR, Cairns BR, 2009. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 78, 273–304. [DOI] [PubMed] [Google Scholar]
  118. Cogswell JP, Ward J, Taylor IA, Waters M, Shi Y, Cannon B, Kelnar K, Kemppainen J, Brown D, Chen C, 2008. Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. J. Alzheimers Dis. 14, 27–41. [DOI] [PubMed] [Google Scholar]
  119. Condliffe D, Wong A, Troakes C, Proitsi P, Patel Y, Chouliaras L, Fernandes C, Cooper J, Lovestone S, Schalkwyk L, Mill J, Lunnon K, 2014. Cross-region reduction in 5-hydroxymethylcytosine in Alzheimer’s disease brain. Neurobiol. Aging 35, 1850–1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Conticello SG, 2008. The AID/APOBEC family of nucleic acid mutators. Genome Biol. 9, 229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Conticello SG, Thomas CJ, Petersen-Mahrt SK, Neuberger MS, 2005. Evolution of the AID/APOBEC family of polynucleotide (deoxy)cytidine deaminases. Mol. Biol. Evol. 22, 367–377. [DOI] [PubMed] [Google Scholar]
  122. Cook C, Carlomagno Y, Gendron TF, Dunmore J, Scheffel K, Stetler C, Davis M, Dickson D, Jarpe M, DeTure M, Petrucelli L, 2014. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum. Mol. Genet. 23, 104–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Cook C, Gendron TF, Scheffel K, Carlomagno Y, Dunmore J, DeTure M, Petrucelli L, 2012. Loss of HDAC6, a novel CHIP substrate, alleviates abnormal tau accumulation. Hum. Mol. Genet. 21, 2936–2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Coppede F, 2012. Genetics and epigenetics of Parkinson’s disease. Sci. World J. 2012, 489830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Coppede F, 2014. The potential of epigenetic therapies in neurodegenerative diseases. Front. Genet. 5, 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Coppede F, Mancuso M, Siciliano G, Migliore L, Murri L, 2006. Genes and the environment in neurodegeneration. Biosci. Rep. 26, 341–367. [DOI] [PubMed] [Google Scholar]
  127. Coppede F, Tannorella P, Pezzini I, Migheli F, Ricci G, Caldarazzo lenco E, Piaceri I, Polini A, Nacmias B, Monzani F, Sorbi S, Siciliano G, Migliore L, 2012. Folate, homocysteine, vitamin B12, and polymorphisms of genes participating in one-carbon metabolism in late-onset Alzheimer’s disease patients and healthy controls. Antioxid. Redox Signal. 17, 195–204. [DOI] [PubMed] [Google Scholar]
  128. Coppieters N, Dieriks BV, Lill C, Faull RL, Curtis MA, Dragunow M, 2014. Global changes in DNA methylation and hydroxymethylation in Alzheimer’s disease human brain. Neurobiol. Aging 35, 1334–1344. [DOI] [PubMed] [Google Scholar]
  129. Cortellino S, Xu J, Sannai M, Moore R, Caretti E, Cigliano A, Le Coz M, Devarajan K, Wessels A, Soprano D, 2011. Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell 146, 67–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Coufal NG, Garcia-Perez JL, Peng GE, Yeo GW, Mu Y, Lovci MT, Morell M, O’Shea KS, Moran JV, Gage FH, 2009. L1 retrotransposition in human neural progenitor cells. Nature 460, 1127–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Cui JG, Li YY, Zhao Y, Bhattacharjee S, Lukiw WJ, 2010. Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease. J. Biol. Chem. 285, 38951–38960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Czech C, Tremp G, Pradier L, 2000. Presenilins and Alzheimer’s disease: biological functions and pathogenic mechanisms. Prog. Neurobiol. 60, 363–384. [DOI] [PubMed] [Google Scholar]
  133. Daniel P, Brazier M, Cerutti I, Pieri F, Tardivel I, Desmet G, Baillet J, Chany C, 1989. Pharmacokinetic study of butyric acid administered in vivo as sodium and arginine butyrate salts. Clin. Chim. Acta 181, 255–263. [DOI] [PubMed] [Google Scholar]
  134. Darmopil S, Martin AB, De Diego IR, Ares S, Moratalla R, 2009. Genetic inactivation of dopamine D1 but not D2 receptors inhibits L-DOPA-induced dyskinesia and histone activation. Biol. Psychiatr. 66, 603–613. [DOI] [PubMed] [Google Scholar]
  135. Dasgupta S, Zhou Y, Jana M, Banik NL, Pahan K, 2003. Sodium phenylacetate inhibits adoptive transfer of experimental allergic encephalomyelitis in SJL/J mice at multiple steps. J. Immunol. 170, 3874–3882. [DOI] [PubMed] [Google Scholar]
  136. Davies MN, Volta M, Pidsley R, Lunnon K, Dixit A, Lovestone S, Coarfa C, Harris RA, Milosavljevic A, Troakes C, Al-Sarraj S, Dobson R, Schalkwyk LC, Mill J, 2012. Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood. Genome Biol. 13, R43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Day JJ, Sweatt JD, 2011. Epigenetic mechanisms in cognition. Neuron 70, 813–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. de Boni L, Tierling S, Roeber S, Walter J, Giese A, Kretzschmar HA, 2011. Next-generation sequencing reveals regional differences of the alpha-synuclein methylation state independent of Lewy body disease. Neuromol. Med. 13, 310–320. [DOI] [PubMed] [Google Scholar]
  139. de Groote ML, Verschure PJ, Rots MG, 2012. Epigenetic editing: targeted rewriting of epigenetic marks to modulate expression of selected target genes. Nucleic Acids Res. 40, 10596–10613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. De Jager PL, Srivastava G, Lunnon K, Burgess J, Schalkwyk LC, Yu L, Eaton ML, Keenan BT, Ernst J, McCabe C, Tang A, Raj T, Replogle J, Brodeur W, Gabriel S, Chai HS, Younkin C, Younkin SG, Zou F, Szyf M, Epstein CB, Schneider JA, Bernstein BE, Meissner A, Ertekin-Taner N, Chibnik LB, Kellis M, Mill J, Bennett DA, 2014. Alzheimer’s disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat. Neurosci. 17, 1156–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. de Lau LM, Breteler MM, 2006. Epidemiology of Parkinson’s disease. Lancet Neurol. 5, 525–535. [DOI] [PubMed] [Google Scholar]
  142. De Strooper B, Saftig P, Craessaerts K, Vanderstichele H, Guhde G, Annaert W, Von Figura K, Van Leuven F, 1998. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 391, 387–390. [DOI] [PubMed] [Google Scholar]
  143. Delaloy C, Liu L, Lee JA, Su H, Shen F, Yang GY, Young WL, Ivey KN, Gao FB, 2010. MicroRNA-9 coordinates proliferation and migration of human embryonic stem cell-derived neural progenitors. Cell Stem Cell 6, 323–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Delatte B, Fuks F, 2013. TET proteins: on the frenetic hunt for new cytosine modifications. Brief. Funct. Genomics 12, 191–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Delay C, Calon F, Mathews P, Hebert SS, 2011. Alzheimer-specific variants in the 3UTR of Amyloid precursor protein affect microRNA function. Mol. Neurode-gener. 6, 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Desplats P, Spencer B, Coffee E, Patel P, Michael S, Patrick C, Adame A, Rockenstein E, Masliah E, 2011. Alpha-synuclein sequesters Dnmt1 from the nucleus: a novel mechanism for epigenetic alterations in Lewy body diseases. J. Biol. Chem. 286, 9031–9037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Desrosiers R, Friderici K, Rottman F, 1974. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc. Natl. Acad. Sci. U.S.A. 71, 3971–3975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Detich N, Bovenzi V, Szyf M, 2003. Valproate induces replication-independent active DNA demethylation. J. Biol. Chem. 278, 27586–27592. [DOI] [PubMed] [Google Scholar]
  149. Devall M, Mill J, Lunnon K, 2014. The mitochondrial epigenome: a role in Alzheimer’s disease? Epigenomics-UK 6, 665–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Di Ruscio A, Ebralidze AK, Benoukraf T, Amabile G, Goff LA, Terragni J, Figueroa ME, Pontes LLDF, Alberich-Jorda M, Zhang P, 2013. DNMT1- interacting RNAs block gene-specific DNA methylation. Nature 503, 371–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. DiFiglia M, Sena-Esteves M, Chase K, Sapp E, Pfister E, Sass M, Yoder J, Reeves P, Pandey RK, Rajeev KG, Manoharan M, Sah DW, Zamore PD, Aronin N, 2007. Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and cortical neuropathology and behavioral deficits. Proc. Natl. Acad. Sci. U.S.A. 104, 17204–17209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ding H, Dolan PJ, Johnson GV, 2008. Histone deacetylase 6 interacts with the microtubule-associated protein tau. J. Neurochem. 106, 2119–2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Dinger ME, Gascoigne DK, Mattick JS, 2011. The evolution of RNAs with multiple functions. Biochimie 93, 2013–2018. [DOI] [PubMed] [Google Scholar]
  154. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigo R, Gingeras TR, 2012. Landscape of transcription in human cells. Nature 489, 101–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Dokmanovic M, Clarke C, Marks PA, 2007. Histone deacetylase inhibitors: overview and perspectives. Mol. Cancer Res.: MCR 5, 981–989. [DOI] [PubMed] [Google Scholar]
  156. Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, Sorek R, Rechavi G, 2012. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206. [DOI] [PubMed] [Google Scholar]
  157. Donmez G, Wang D, Cohen DE, Guarente L, 2010. SIRT1 suppresses betaamyloid production by activating the alpha-secretase gene ADAM10. Cell 142, 320–332. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  158. Doxakis E, 2010. Post-transcriptional regulation of alpha-synuclein expression by mir-7 and mir-153. J. Biol. Chem. 285, 12726–12734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Drouet V, Perrin V, Hassig R, Dufour N, Auregan G, Alves S, Bonvento G, Brouillet E, Luthi-Carter R, Hantraye P, Deglon N, 2009. Sustained effects of nonallele-specific Huntingtin silencing. Ann. Neurol. 65, 276–285. [DOI] [PubMed] [Google Scholar]
  160. Duclot F, Meffre J, Jacquet C, Gongora C, Maurice T, 2010. Mice knock out for the histone acetyltransferase p300/CREB binding protein-associated factor develop a resistance to amyloid toxicity. Neuroscience 167, 850–863. [DOI] [PubMed] [Google Scholar]
  161. Dunlap KA, Palmarini M, Varela M, Burghardt RC, Hayashi K, Farmer JL, Spencer TE, 2006. Endogenous retroviruses regulate periimplantation placental growth and differentiation. Proc. Natl. Acad. Sci. U.S.A. 103, 14390–14395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Durga J, van Boxtel MP, Schouten EG, Kok FJ, Jolles J, Katan MB, Verhoef P, 2007. Effect of 3-year folic acid supplementation on cognitive function in older adults in the FACIT trial: a randomised, double blind, controlled trial. Lancet 369, 208–216. [DOI] [PubMed] [Google Scholar]
  163. Dzitoyeva S, Chen H, Manev H, 2012. Effect of aging on 5-hydroxymethylcytosine in brain mitochondria. Neurobiol. Aging 33, 2881–2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Ebbel EN, Leymarie N, Schiavo S, Sharma S, Gevorkian S, Hersch S, Matson WR, Costello CE, 2010. Identification of phenylbutyrate-generated metabolites in Huntington disease patients using parallel liquid chromatography/ electrochemical array/mass spectrometry and off-line tandem mass spectrometry. Anal. Biochem. 399, 152–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Edwards TL, Scott WK, Almonte C, Burt A, Powell EH, Beecham GW, Wang LY, Zuchner S, Konidari I, Wang GF, Singer C, Nahab F, Scott B, Stajich JM, Pericak-Vance M, Haines J, Vance JM, Martin ER, 2010. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann. Hum. Genet. 74, 97–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Egorin MJ, Yuan ZM, Sentz DL, Plaisance K, Eiseman JL, 1999. Plasma pharmacokinetics of butyrate after intravenous administration of sodium butyrate or oral administration of tributyrin or sodium butyrate to mice and rats. Cancer Chemother. Pharmacol. 43, 445–453. [DOI] [PubMed] [Google Scholar]
  167. Ender C, Krek A, Friedlander MR, Beitzinger M, Weinmann L, Chen W, Pfeffer S, Rajewsky N, Meister G, 2008. A human snoRNA with microRNA-like functions. Mol. Cell 32, 519–528. [DOI] [PubMed] [Google Scholar]
  168. Epstein P, Reddy R, Henning D, Busch H, 1980. The nucleotide sequence of nuclear U6 (4.7 S) RNA. J. Biol. Chem. 255, 8901–8906. [PubMed] [Google Scholar]
  169. Espada J, Ballestar E, Santoro R, Fraga MF, Villar-Garea A, Nemeth A, Lopez- Serra L, Ropero S, Aranda A, Orozco H, 2007. Epigenetic disruption of ribosomal RNA genes and nucleolar architecture in DNA methyltransferase 1 (Dnmt1) deficient cells. Nucleic Acids Res. 35, 2191–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Eulalio A, Huntzinger E, Izaurralde E, 2008. Getting to the root of miRNA-mediated gene silencing. Cell 132, 9–14. [DOI] [PubMed] [Google Scholar]
  171. Fabian MR, Sonenberg N, Filipowicz W, 2010. Regulation of mRNA translation and stability by microRNAs. Annu. Rev. Biochem. 79, 351–379. [DOI] [PubMed] [Google Scholar]
  172. Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, Finch CE, Laurent III GS, Kenny PJ, Wahlestedt C, 2008. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of β-secretase. Nat. Med. 14, 723–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Faghihi MA, Zhang M, Huang J, Modarresi F, Van der Brug MP, Nalls MA, Cookson MR, St-Laurent G, Wahlestedt C, 2010. Evidence for natural antisense transcript-mediated inhibition of microRNA function. Genome Biol. 11, R56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Fan G, Beard C, Chen RZ, Csankovszki G, Sun Y, Siniaia M, Biniszkiewicz D, Bates B, Lee PP, Kuhn R, Trumpp A, Poon C, Wilson CB, Jaenisch R, 2001. DNA hypomethylation perturbs the function and survival of CNS neurons in postnatal animals. J. Neurosci. 21, 788–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Fan X, Liu Y, Jiang J, Ma Z, Wu H, Liu T, Liu M, Li X, Tang H, 2010. miR-20a promotes proliferation and invasion by targeting APP in human ovarian cancer cells. Acta Biochim. Biophys. Sin. 42, 318–324. [DOI] [PubMed] [Google Scholar]
  176. Fang MR, Wang J, Zhang XB, Geng Y, Hu ZY, Rudd JA, Ling SC, Chen W, Han S, 2012. The miR-124 regulates the expression of BACE1/beta-secretase correlated with cell death in Alzheimer’s disease. Toxicol. Lett. 209, 94–105. [DOI] [PubMed] [Google Scholar]
  177. Faulkner GJ, Kimura Y, Daub CO, Wani S, Plessy C, Irvine KM, Schroder K, Cloonan N, Steptoe AL, Lassmann T, Waki K, Hornig N, Arakawa T, Takahashi H, Kawai J, Forrest AR, Suzuki H, Hayashizaki Y, Hume DA, Orlando V, Grimmond SM, Carninci P, 2009. The regulated retrotransposon transcriptome of mammalian cells. Nat. Genet. 41, 563–571. [DOI] [PubMed] [Google Scholar]
  178. Feinberg AP, 2008. Epigenetics at the epicenter of modern medicine. JAMA - J. Am. Med. Assoc. 299, 1345–1350. [DOI] [PubMed] [Google Scholar]
  179. Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, Silva AJ, Fan G, 2010. Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat. Neurosci. 13, 423–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Feng S, Xiong L, Ji Z, Cheng W, Yang H, 2012. Correlation between increased ND2 expression and demethylated displacement loop of mtDNA in colorectal cancer. Mol. Med. Rep. 6, 125–130. [DOI] [PubMed] [Google Scholar]
  181. Ferrante RJ, Kowall NW, Beal MF, Martin JB, Bird ED, Richardson EP Jr., 1987. Morphologic and histochemical characteristics of a spared subset of striatal neurons in Huntington’s disease. J. Neuropathol. Exp. Neurol. 46, 12–27. [DOI] [PubMed] [Google Scholar]
  182. Ferrante RJ, Kowall NW, Beal MF, Richardson EP Jr., Bird ED, Martin JB, 1985. Selective sparing of a class of striatal neurons in Huntington’s disease. Science 230, 561–563. [DOI] [PubMed] [Google Scholar]
  183. Ferrante RJ, Kowall NW, Richardson EP, Bird ED, Martin JB, 1986. Topography of enkephalin, substance P and acetylcholinesterase staining in Huntington’s disease striatum. Neurosci. Lett. 71, 283–288. [DOI] [PubMed] [Google Scholar]
  184. Ferrante RJ, Kowall NW, Richardson EP Jr., 1991. Proliferative and degenerative changes in striatal spiny neurons in Huntington’s disease: a combined study using the section-Golgi method and calbindin D28k immunocytochemistry. J. Neurosci. 11, 3877–3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B, Smith K, Kowall NW, Ratan RR, Luthi-Carter R, Hersch SM, 2003. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington’s disease mice. J. Neurosci. 23, 9418–9427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Ferrante RJ, Ryu H, Kubilus JK, D’Mello S, Sugars KL, Lee J, Lu P, Smith K, Browne S, Beal MF, Kristal BS, Stavrovskaya l.G., Hewett S, Rubinsztein DC, Langley B, Ratan RR, 2004. Chemotherapy for the brain: the antitumor antibiotic mithramycin prolongs survival in a mouse model of Huntington’s disease. J. Neurosci. 24, 10335–10342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Fischer A, 2014. Targeting histone-modifications in Alzheimer’s disease. What is the evidence that this is a promising therapeutic avenue? Neuropharmacology 80, 95–102. [DOI] [PubMed] [Google Scholar]
  188. Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai L-H, 2007. Recovery of learning and memory is associated with chromatin remodelling. Nature 447,178–182. [DOI] [PubMed] [Google Scholar]
  189. Flores K, Wolschin F, Corneveaux JJ, Allen AN, Huentelman MJ, Amdam GV, 2012. Genome-wide association between DNA methylation and alternative splicing in an invertebrate. BMC Genomics 13, 480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Fontan-Lozano A, Suarez-Pereira l., Horrillo A, del-Pozo-Martin Y, Hmadcha A, Carrion AM, 2010. Histone H1 poly[ADP]-ribosylation regulates the chromatin alterations required for learning consolidation. J. Neurosci. 30, 13305–13313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suner D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M, 2005. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. U.S.A. 102, 10604–10609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Francis PT, Perry EK, 2007. Cholinergic and other neurotransmitter mechanisms in Parkinson’s disease, Parkinson’s disease dementia, and dementia with Lewy bodies. Mov. Disord. 22 (Suppl. 17), S351–S357. [DOI] [PubMed] [Google Scholar]
  193. Francis Y.l., Fa M, Ashraf H, Zhang H, Staniszewski A, Latchman DS, Arancio O, 2009. Dysregulation of histone acetylation in the APP/PS1 mouse model of Alzheimer’s disease. J. Alzheimers Dis. 18, 131–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Franco R, Li S, Rodriguez-Rocha H, Burns M, Panayiotidis M.l., 2010. Molecular mechanisms of pesticide-induced neurotoxicity: relevance to Parkinson’s disease. Chem.-Biol. lnteract. 188, 289–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Franich NR, Fitzsimons HL, Fong DM, Klugmann M, During MJ, Young D, 2008. AAV vector-mediated RNAi of mutant huntingtin expression is neuro- protective in a novel genetic rat model of Huntington’s disease. Mol. Ther. 16, 947–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Fu L, Guerrero CR, Zhong N, Amato NJ, Liu Y, Liu S, Cai Q, Ji D, Jin SG, Niedernhofer LJ, Pfeifer GP, Xu GL, Wang Y, 2014. Tet-mediated formation of 5-hydroxymethylcytosine in RNA. J. Am. Chem. Soc. 136, 11582–11585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Fu Y, Jia G, Pang X, Wang RN, Wang X, Li CJ, Smemo S, Dai Q, Bailey KA, Nobrega MA, 2013. FTO-mediated formation of N6-hydroxymethyladenosine and N6-formyladenosine in mammalian RNA. Nat. Commun. 4, 1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Fukuda T, 2001. Neurotoxicity of MPTP. Neuropathol. 21, 323–332. [DOI] [PubMed] [Google Scholar]
  199. Fukutani Y, Cairns NJ, Rossor MN, Lantos PL, 1996. Purkinje cell loss and astrocytosis in the cerebellum in familial and sporadic Alzheimer’s disease. Neurosci. Lett. 214, 33–36. [DOI] [PubMed] [Google Scholar]
  200. Fuso A, Nicolia V, Cavallaro RA, Ricceri L, D’Anselmi F, Coluccia P, Calamandrei G, Scarpa S, 2008. B-vitamin deprivation induces hyperhomocysteinemia and brain S-adenosylhomocysteine, depletes brain S-adenosylmethionine, and enhances PS1 and BACE expression and amyloid-beta deposition in mice. Mol. Cell. Neurosci. 37, 731–746. [DOI] [PubMed] [Google Scholar]
  201. Fuso A, Nicolia V, Pasqualato A, Fiorenza MT, Cavallaro RA, Scarpa S, 2011. Changes in Presenilin 1 gene methylation pattern in diet-induced B vitamin deficiency. Neurobiol. Aging 32, 187–199. [DOI] [PubMed] [Google Scholar]
  202. Fuso A, Nicolia V, Ricceri L, Cavallaro RA, lsopi E, Mangia F, Fiorenza MT, Scarpa S, 2012. S-adenosylmethionine reduces the progress of the Alzheimer- like features induced by B-vitamin deficiency in mice. Neurobiol. Aging 33,, 1482.e1–1482.e16. [DOI] [PubMed] [Google Scholar]
  203. Fuso A, Seminara L, Cavallaro RA, D’Anselmi F, Scarpa S, 2005. S-adenosyl- methionine/homocysteine cycle alterations modify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloid production. Mol. Cell. Neurosci. 28, 195–204. [DOI] [PubMed] [Google Scholar]
  204. Gaisler-Salomon l., Kravitz E, Feiler Y, Safran M, Biegon A, Amariglio N, Rechavi G, 2014. Hippocampus-specific deficiency in RNA editing of GluA2 in Alzheimer’s disease. Neurobiol. Aging 35, 1785–1791. [DOI] [PubMed] [Google Scholar]
  205. Gardian G, Browne SE, Choi DK, Klivenyi P, Gregorio J, Kubilus JK, Ryu H, Langley B, Ratan RR, Ferrante RJ, Beal MF, 2005. Neuroprotective effects of phenylbutyrate in the N171–82Q transgenic mouse model of Huntington’s disease. J. Biol. Chem. 280, 556–563. [DOI] [PubMed] [Google Scholar]
  206. Gavin DP, Chase KA, Sharma RP, 2013. Active DNA demethylation in postmitotic neurons: a reason for optimism. Neuropharmacology 75, 233–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Geekiyanage H, Chan C, 2011. MicroRNA-137/181c regulates serine palmitoyltransferase and in turn amyloid beta, novel targets in sporadic Alzheimer’s disease. J. Neurosci. 31, 14820–14830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Gehrke S, Imai Y, Sokol N, Lu B, 2010. Pathogenic LRRK2 negatively regulates microRNA-mediated translational repression. Nature 466, 637–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. German DC, Eisch AJ, 2004. Mouse models of Alzheimer’s disease: insight into treatment. Rev. Neurosci. 15, 353–370. [DOI] [PubMed] [Google Scholar]
  210. Gershey EL, Vidali G, Allfrey VG, 1968. Chemical studies of histone acetylation. The occurrence of epsilon-N-acetyllysine in the f2a1 histone. J. Biol. Chem. 243, 5018–5022. [PubMed] [Google Scholar]
  211. Ghose J, Sinha M, Das E, Jana NR, Bhattacharyya NP, 2011. Regulation of miR- 146a by RelA/NFkB and p53 in STHdhQ111/HdhQ111 cells, a cell model of Huntington’s disease. PLoS ONE 6, e23837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Gillardon F, Mack M, Rist W, Schnack C, Lenter M, Hildebrandt T, Hengerer B, 2008. MicroRNA and proteome expression profiling in early-symptomatic alpha-synuclein(A30P)-transgenic mice. Proteomics Clin. Appl. 2, 697–705. [DOI] [PubMed] [Google Scholar]
  213. Giordano M, Cristiani C, Crocco P, D’Aquila P, Rango FD, Pisani F, Scafone T, Tallaro F, Rose G, Passarino G, Bellizzi D, 2012. Methylation of the human mitochondrial 12S rRNA gene is correlated with aging. In: 12th International FISV Congress, Rome, Italy. [Google Scholar]
  214. Globisch D, Munzel M, Muller M, Michalakis S, Wagner M, Koch S, Bruckl T, Biel M, Carell T, 2010. Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PLoS ONE 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, James L, et al. , 1991. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349, 704–706. [DOI] [PubMed] [Google Scholar]
  216. Gokey NG, Srinivasan R, Lopez-Anido C, Krueger C, Svaren J, 2012. Developmental regulation of microRNA expression in Schwann cells. Mol. Cell. Biol. 32, 558–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Gordon CA, Hartono SR, Chedin F, 2013. Inactive DNMT3B splice variants modulate de novo DNA methylation. PLOS ONE 8, e69486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Govindarajan N, Rao P, Burkhardt S, Sananbenesi F, Schluter OM, Bradke F, Lu J, Fischer A, 2013. Reducing HDAC6 ameliorates cognitive deficits in a mouse model for Alzheimer’s disease. EMBO Mol. Med. 5, 52–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Graff J, Kim D, Dobbin MM, Tsai LH, 2011. Epigenetic regulation of gene expression in physiological and pathological brain processes. Physiol. Rev. 91, 603–649. [DOI] [PubMed] [Google Scholar]
  220. Graff J, Rei D, Guan JS, Wang WY, Seo J, Hennig KM, Nieland TJ, Fass DM, Kao PF, Kahn M, Su SC, Samiei A, Joseph N, Haggarty SJ, Delalle I, Tsai LH, 2012. An epigenetic blockade of cognitive functions in the neurodegener- ating brain. Nature 483, 222–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Graveland GA, Williams RS, DiFiglia M, 1985. Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington’s disease. Science 227, 770–773. [DOI] [PubMed] [Google Scholar]
  222. Green KN, Steffan JS, Martinez-Coria H, Sun X, Schreiber SS, Thompson LM, LaFerla FM, 2008. Nicotinamide restores cognition in Alzheimer’s disease transgenic mice via a mechanism involving sirtuin inhibition and selective reduction of Thr231-phosphotau. J. Neurosci. 28, 11500–11510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Greer EL, Shi Y, 2012. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13, 343–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Gregoretti IV, Lee YM, Goodson HV, 2004. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J. Mol. Biol. 338, 17–31. [DOI] [PubMed] [Google Scholar]
  225. Grewal SI, Jia S, 2007. Heterochromatin revisited. Nat. Rev. Genet. 8, 35–46. [DOI] [PubMed] [Google Scholar]
  226. Groen T, 2010. DNA methylation and Alzheimer’s disease. Epigenet. Aging 315–326. [Google Scholar]
  227. Guan J-S, Haggarty SJ, Giacometti E, Dannenberg J-H, Joseph N, Gao J, Nieland TJ, Zhou Y, Wang X, Mazitschek R, 2009. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 459, 55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Guarente L, Picard F, 2005. Calorie restriction - the SIR2 connection. Cell 120, 473–482. [DOI] [PubMed] [Google Scholar]
  229. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cru- chaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J, Alzheimer Genetic Analysis G, 2013. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Guerrero-Bosagna C, Skinner MK, 2012. Environmentally induced epigenetic transgenerational inheritance of phenotype and disease. Mol. Cell. Endocrinol. 354, 3–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Guffanti A, Simchovitz A, Soreq H, 2014. Emerging bioinformatics approaches for analysis of NGS-derived coding and non-coding RNAs in neurodegenerative diseases. Front. Cell. Neurosci. 8, 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Guintivano J, Aryee MJ, Kaminsky ZA, 2013. A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics 8, 290–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Guo JU, Su Y, Shin JH, Shin J, Li H, Xie B, Zhong C, Hu S, Le T, Fan G, Zhu H, Chang Q, Gao Y, Ming GL, Song H, 2013. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat. Neurosci. 17, 215–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Guo JU, Su Y, Zhong C, Ming GL, Song H, 2011. Hydroxylation of 5-methylcy-tosine by TET1 promotes active DNA demethylation in the adult brain. Cell 145, 423–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD, 2010. Histone methylation regulates memory formation. J. Neurosci. 30, 3589–3599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Haan MN, Miller JW, Aiello AE, Whitmer RA, Jagust WJ, Mungas DM, Allen LH, Green R, 2007. Homocysteine, B vitamins, and the incidence of dementia and cognitive impairment: results from the Sacramento Area Latino Study on Aging. Am. J. Clin. Nutr. 85, 511–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Habibi E, Masoudi-Nejad A, Abdolmaleky HM, Haggarty SJ, 2011. Emerging roles of epigenetic mechanisms in Parkinson’s disease. Funct. Integr. Genomics 11, 523–537. [DOI] [PubMed] [Google Scholar]
  238. Hackl M, Brunner S, Fortschegger K, Schreiner C, Micutkova L, Muck C, Laschober GT, Lepperdinger G, Sampson N, Berger P, Herndler-Brandstet- ter D, Wieser M, Kuhnel H, Strasser A, Rinnerthaler M, Breitenbach M, Mildner M, Eckhart L, Tschachler E, Trost A, Bauer JW, Papak C, Traja- noski Z, Scheideler M, Grillari-Voglauer R, Grubeck-Loebenstein B, Jansen- Durr P, Grillari J, 2010. miR-17, miR-19b, miR-20a, and miR-106a are down- regulated in human aging. Aging Cell 9, 291–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Hagen G, Dennig J, Preiss A, Beato M, Suske G, 1995. Functional analyses of the transcription factor Sp4 reveal properties distinct from Sp1 and Sp3. J. Biol. Chem. 270, 24989–24994. [DOI] [PubMed] [Google Scholar]
  240. Haggarty SJ, Koeller KM, Wong JC, Grozinger CM, Schreiber SL, 2003. Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc. Natl. Acad. Sci. U.S.A. 100, 4389–4394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Haigis MC, Sinclair DA, 2010. Mammalian sirtuins: biological insights and disease relevance. Annu. Rev. Pathol. 5, 253–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Han J, Lee Y, Yeom KH, Kim YK, Jin H, Kim VN, 2004. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 18, 3016–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Han J, Pedersen JS, Kwon SC, Belair CD, Kim Y-K, Yeom K-H, Yang W-Y, Haussler D, Blelloch R, Kim VN, 2009. Posttranscriptional crossregulation between Drosha and DGCR8. Cell 136, 75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Handel AE, Ebers GC, Ramagopalan SV, 2010. Epigenetics: molecular mechanisms and implications for disease. Trends Mol. Med. 16, 7–16. [DOI] [PubMed] [Google Scholar]
  245. Hansen KD, Timp W, Bravo HC, Sabunciyan S, Langmead B, McDonald OG, Wen B, Wu H, Liu Y, Diep D, Briem E, Zhang K, Irizarry RA, Feinberg AP, 2011. Increased methylation variation in epigenetic domains across cancer types. Nat. Genet. 43, 768–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Harada F, Kato N, Nishimura S, 1980. The nucleotide sequence of nuclear 4.8S RNA of mouse cells. Biochem. Biophys. Res. Commun. 95, 1332–1340. [DOI] [PubMed] [Google Scholar]
  247. Hardy J, 2006. A hundred years of Alzheimer’s disease research. Neuron 52, 3–13. [DOI] [PubMed] [Google Scholar]
  248. Hardy J, Selkoe DJ, 2002. The amyloid hypothesis ofAlzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356. [DOI] [PubMed] [Google Scholar]
  249. Hardy JA, Higgins GA, 1992. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185. [DOI] [PubMed] [Google Scholar]
  250. Hargreaves DC, Crabtree GR, 2011. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 21, 396–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Harms KL, Chen XB, 2007. Histone deacetylase 2 modulates p53 transcriptional activities through regulation of p53-DNA binding activity. Cancer Res. 67, 3145–3152. [DOI] [PubMed] [Google Scholar]
  252. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A, 2009. Genome-wide association study identifies variants at CLU and P1CALM associated with Alzheimer’s disease. Nat. Genet. 41, 1088–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Harper SQ, Staber PD, He X, Eliason SL, Martins IH, Mao Q, Yang L, Kotin RM, Paulson HL, Davidson BL, 2005. RNA interference improves motor and neuropathological abnormalities in a Huntington’s disease mouse model. Proc. Natl. Acad. Sci. U.S.A. 102, 5820–5825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. He L, Hannon GJ, 2004. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 5, 522. [DOI] [PubMed] [Google Scholar]
  255. He YF, Li BZ, Li Z, Liu P, Wang Y, Tang Q, Ding J, Jia Y, Chen Z, Li L, Sun Y, Li X, Dai Q, Song CX, Zhang K, He C, Xu GL, 2011. Tet-mediated formation of 5-carboxylcytosine and its excision byTDG in mammalian DNA. Science 333, 1303–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Hebert SS, De Strooper B, 2009. Alterations of the microRNA network cause neurodegenerative disease. Trends Neurosci. 32, 199–206. [DOI] [PubMed] [Google Scholar]
  257. Hebert SS, Horre K, Nicolai L, Bergmans B, Papadopoulou AS, Delacourte A, De Strooper B, 2009. MicroRNA regulation of Alzheimer’s amyloid precursor protein expression. Neurobiol. Dis. 33, 422–428. [DOI] [PubMed] [Google Scholar]
  258. Hebert SS, Horre K, Nicolai L, Papadopoulou AS, Mandemakers W, Silahtar-oglu AN, Kauppinen S, Delacourte A, De Strooper B, 2008. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/beta-secretase expression. Proc. Natl. Acad. Sci. U.S.A. 105, 6415–6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Hebert SS, Papadopoulou AS, Smith P, Galas MC, Planel E, Silahtaroglu AN, Sergeant N, Buee L, De Strooper B, 2010. Genetic ablation of Dicer in adult forebrain neurons results in abnormal tau hyperphosphorylation and neurodegeneration. Hum. Mol. Genet. 19, 3959–3969. [DOI] [PubMed] [Google Scholar]
  260. Hebert SS, Sergeant N, Buee L, 2012. MicroRNAs and the regulation of tau metabolism. Int. J. Alzheimers Dis. 2012, 406561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Hebert SS, Wang W-X, Zhu Q, Nelson PT, 2013. A study of small RNAs from cerebral neocortex of pathology-verified Alzheimer’s disease, dementia with lewy bodies, hippocampal sclerosis, frontotemporal lobar dementia, and nondemented human controls. J. Alzheimer’s Dis. 35, 335–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Henikoff S,Matzke MA, 1997. Exploringand explaining epigeneticeffects. Trends Genet. 13, 293–295. [DOI] [PubMed] [Google Scholar]
  263. Hernandez DG, Nalls MA, Gibbs JR, Arepalli S, van der Brug M, Chong S, Moore M, Longo DL, Cookson MR, Traynor BJ, Singleton AB, 2011. Distinct DNA methylation changes highly correlated with chronological age in the human brain. Hum. Mol. Genet. 20, 1164–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Hirschey MD, 2011. Old enzymes, new tricks: sirtuins are NAD(+)-dependent de- acylases. Cell Metab. 14, 718–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Ho L, Crabtree GR, 2010. Chromatin remodelling during development. Nature 463, 474–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Hockly E, Richon VM, Woodman B, Smith DL, Zhou X, Rosa E, Sathasivam K, Ghazi-Noori S, Mahal A, Lowden PA, Steffan JS, Marsh JL, Thompson LM, Lewis CM, Marks PA, Bates GP, 2003. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc. Natl. Acad. Sci. U.S.A. 100, 2041–2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Hogarth P, Lovrecic L, Krainc D, 2007. Sodium phenylbutyrate in Huntington’s disease: a dose-finding study. Mov. Disord. 22, 1962–1964. [DOI] [PubMed] [Google Scholar]
  268. Hollingworth P, Harold D, Sims R, Gerrish A, Lambert J-C, Carrasquillo MM, Abraham R, Hamshere ML, Pahwa JS, Moskvina V, 2011. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 43, 429–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Holmes AP, Wood SH, Merry BJ, de Magalhaes JP, 2013. A-to-1 RNA editing does not change with age in the healthy male rat brain. Biogerontology 14, 395–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Hong EE, Okitsu CY, Smith AD, Hsieh CL, 2013. Regionally specific and genome-wide analyses conclusively demonstrate the absence of CpG methyl- ation in human mitochondrial DNA. Mol. Cell. Biol. 33, 2683–2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Horvath S, 2013. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Houlden H, Singleton AB, 2012. The genetics and neuropathology ofParkinson’s disease. Acta Neuropathol. 124, 325–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Houston I, Peter CJ, Mitchell A, Straubhaar J, Rogaev E, Akbarian S, 2013. Epigenetics in the human brain. Neuropsychopharmacol. 38, 183–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. How Kit A, Nielsen HM, Tost J, 2012. DNA methylation based biomarkers: practical considerations and applications. Biochimie 94, 2314–2337. [DOI] [PubMed] [Google Scholar]
  275. Hoyaux D, Decaestecker C, Heizmann CW, Vogl T, Schafer BW, Salmon I, Kiss R, Pochet R, 2000. S100 proteins in Corpora amylacea from normal human brain. Brain Res. 867, 280–288. [DOI] [PubMed] [Google Scholar]
  276. Hoyer S, Lee SK, Loffler T, Schliebs R, 2000. Inhibition of the neuronal insulin receptor. An in vivo model for sporadic Alzheimer disease? Ann. N. Y. Acad. Sci. 920, 256–258. [DOI] [PubMed] [Google Scholar]
  277. Hsieh J, Eisch AJ, 2010. Epigenetics, hippocampal neurogenesis, and neuropsychiatric disorders: unraveling the genome to understand the mind. Neurobiol. Dis. 39, 73–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Hu HY, Guo S, Xi J, Yan Z, Fu N, Zhang X, Menzel C, Liang H, Yang H, Zhao M, Zeng R, Chen W, Paabo S, Khaitovich P, 2011a. MicroRNA expression and regulation in human, chimpanzee, and macaque brains. PLoS Genet. 7, e1002327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Hu J, Matsui M, Gagnon KT, Schwartz JC, Gabillet S, Arar K, Wu J, Bezproz- vanny I, Corey DR, 2009. Allele-specific silencing of mutant huntingtin and ataxin-3 genes by targeting expanded CAG repeats in mRNAs. Nat. Biotechnol. 27, 478–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Hu XL, Wang Y, Shen Q, 2012. Epigenetic control on cell fate choice in neural stem cells. Protein Cell 3, 278–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Hu Y, Chopra V, Chopra R, Locascio JJ, Liao Z, Ding H, Zheng B, Matson WR, Ferrante RJ, Rosas HD, Hersch SM, Scherzer CR, 2011b. Transcriptional modulator H2A histone family, member Y (H2AFY) marks Huntington disease activity in man and mouse. Proc. Natl. Acad. Sci. U.S.A. 108, 17141–17146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Huang B, Schiefer J, Sass C, Landwehrmeyer GB, Kosinski CM, Kochanek S, 2007. High-capacity adenoviral vector-mediated reduction of huntingtin aggregate load in vitro and in vivo. Hum. Gene Ther. 18, 303–311. [DOI] [PubMed] [Google Scholar]
  283. Huang HY, Lin SZ, Chen WF, Li KW, Kuo JS, Wang MJ, 2011. Urocortin modulates dopaminergic neuronal survival via inhibition of glycogen synthase kinase-3beta and histone deacetylase. Neurobiol. Aging 32, 1662–1677. [DOI] [PubMed] [Google Scholar]
  284. Huda A, Jordan IK, 2009. Epigenetic regulation of mammalian genomes by transposable elements. Ann. N. Y. Acad. Sci. 1178, 276–284. [DOI] [PubMed] [Google Scholar]
  285. Hunt LT, Dayhoff MO, 1977. Amino-terminal sequence identity of ubiquitin and the nonhistone component of nuclear protein A24. Biochem. Biophys. Res. Commun. 74, 650–655. [DOI] [PubMed] [Google Scholar]
  286. Hutnick LK, Golshani P, Namihira M, Xue Z, Matynia A, Yang XW, Silva AJ, Schweizer FE, Fan G, 2009. DNA hypomethylation restricted to the murine forebrain induces cortical degeneration and impairs postnatal neuronal maturation. Hum. Mol. Genet. 18, 2875–2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Huynh JL, Casaccia P, 2013. Epigenetic mechanisms in multiple sclerosis: implications for pathogenesis and treatment. Lancet Neurol. 12, 195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Hwang JY, Aromolaran KA, Zukin RS, 2013. Epigenetic mechanisms in stroke and epilepsy. Neuropsychopharmacol. 38, 167–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Hyman BT, Elvhage TE, Reiter J, 1994. Extracellular signal regulated kinases. Localization of protein and mRNA in the human hippocampal formation in Alzheimer’s disease. Am. J. Pathol. 144, 565–572. [PMC free article] [PubMed] [Google Scholar]
  290. Iacobazzi V, Castegna A, Infantino V, Andria G, 2013. Mitochondrial DNA methylation as a next-generation biomarker and diagnostic tool. Mol. Genet. Metab. 110, 25–34. [DOI] [PubMed] [Google Scholar]
  291. Igarashi S, Morita H, Bennett KM, Tanaka Y, Engelender S, Peters MF, Cooper JK, Wood JD, Sawa A, Ross CA, 2003. Inducible PC12 cell model of Huntington’s disease shows toxicity and decreased histone acetylation. Neuroreport 14, 565–568. [DOI] [PubMed] [Google Scholar]
  292. Imai Y, Gehrke S, Wang HQ, Takahashi R, Hasegawa K, Oota E, Lu B, 2008. Phosphorylation of 4E-BP by LRRK2 affects the maintenance of dopaminergic neurons in Drosophila. EMBO J. 27, 2432–2443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Iqbal K, Liu F, Gong CX, Alonso Adel C., Grundke-Iqbal I, 2009. Mechanisms of tau-induced neurodegeneration. Acta Neuropathol. 118, 53–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Iqbal K, Tellez-Nagel I, Grundke-Iqbal I, 1974. Protein abnormalities in Huntington’s chorea. Brain Res. 76, 178–184. [DOI] [PubMed] [Google Scholar]
  295. Ishimaru N, Fukuchi M, Hirai A, Chiba Y, Tamura T, Takahashi N, Tabuchi A, Tsuda M, Shiraishi M, 2010. Differential epigenetic regulation of BDNF and NT-3 genes by trichostatin A and 5-aza-2-deoxycytidine in Neuro-2a cells. Biochem. Biophys. Res. Commun. 394, 173–177. [DOI] [PubMed] [Google Scholar]
  296. Iskandar BJ, Rizk E, Meier B, Hariharan N, Bottiglieri T, Finnell RH, Jarrard DF, Banerjee RV, Skene JP, Nelson A, 2010. Folate regulation of axonal regeneration in the rodent central nervous system through DNA methylation. J. Clin. Invest. 120, 1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van Gorp S, Nazor KL, Boscolo FS, 2012. Probing sporadic and familial Alzheimer/’s disease using induced pluripotent stem cells. Nature 482, 216–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Ito S, Shen L, Dai Q, Wu SC, Collins LB, Swenberg JA, He C, Zhang Y, 2011. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carbox-ylcytosine. Science 333, 1300–1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Jackman JE, Montange RK, Malik HS, Phizicky EM, 2003. Identification of the yeast gene encoding the tRNA m1G methyltransferase responsible for modification at position 9. RNA 9, 574–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Jackson M, Krassowska A, Gilbert N, Chevassut T, Forrester L, Ansell J, Ramsahoye B, 2004. Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells. Mol. Cell. Biol. 24, 8862–8871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Jady BE, Bertrand E, Kiss T, 2004. Human telomerase RNA and box H/ACA scaRNAs share a common Cajal body-specific localization signal. J. Cell. Biol. 164, 647–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Jaenisch R, Bird A, 2003. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet. 33, 245–254. [DOI] [PubMed] [Google Scholar]
  303. Jankovic J, 2008. Parkinson’s disease: clinical features and diagnosis. J. Neurol. Neurosurg. Psychiatry 79, 368–376. [DOI] [PubMed] [Google Scholar]
  304. Jee CD, Lee HS, Bae SI, Yang HK, Lee YM, Rho MS, Kim WH, 2005. Loss of caspase-1 gene expression in human gastric carcinomas and cell lines. Int. J. Oncol. 26, 1265–1271. [PubMed] [Google Scholar]
  305. Jellinger KA, 2009. Formation and development of Lewy pathology: a critical update. J. Neurol. 256 (Suppl. 3), 270–279. [DOI] [PubMed] [Google Scholar]
  306. Jenuwein T, Allis CD, 2001. Translating the histone code. Science 293,1074–1080. [DOI] [PubMed] [Google Scholar]
  307. Jeon Y, Sarma K, Lee JT, 2012. New and Xisting regulatory mechanisms of X chromosome inactivation. Curr. Opin. Genet. Dev. 22, 62–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang Y-G, 2011. N6-methyladenosine in nuclear RNA is a major substrate of the obesity- associated FTO. Nat. Chem. Biol. 7, 885–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Jia H, Kast RJ, Steffan JS, Thomas EA, 2012a. Selective histone deacetylase (HDAC) inhibition imparts beneficial effects in Huntington’s disease mice: implications for the ubiquitin-proteasomal and autophagy systems. Hum. Mol. Genet. 21, 5280–5293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  310. Jia H, Pallos J, Jacques V, Lau A, Tang B, Cooper A, Syed A, Purcell J, Chen Y, Sharma S, Sangrey GR, Darnell SB, Plasterer H, Sadri-Vakili G, Gottesfeld JM, Thompson LM, Rusche JR, Marsh JL, Thomas EA, 2012b. Histone deacetylase (HDAC) inhibitors targeting HDAC3 and HDAC1 ameliorate polyglutamine-elicited phenotypes in model systems of Huntington’s disease. Neurobiol. Dis. 46, 351–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Jiang N, Yan X, Zhou W, Zhang Q, Chen H, Zhang Y, Zhang X, 2008. NMR- based metabonomic investigations into the metabolic profile ofthe senescence- accelerated mouse. J. Proteome Res. 7, 3678–3686. [DOI] [PubMed] [Google Scholar]
  312. Jin H, Kanthasamy A, Ghosh A, Yang Y, Anantharam V, Kanthasamy AG, 2011a. alpha-Synuclein negatively regulates protein kinase Cdelta expression to suppress apoptosis in dopaminergic neurons by reducing p300 histone acetyltransferase activity. J. Neurosci. 31, 2035–2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Jin SG, Wu X, Li AX, Pfeifer GP, 2011b. Genomic mapping of5-hydroxymethyl-cytosine in the human brain. Nucleic Acids Res. 39, 5015–5024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Johnson AA, Sarthi J, Pirooznia SK, Reube W, Elefant F, 2013. Increasing Tip60 HAT levels rescues axonal transport defects and associated behavioral phenotypes in a Drosophila Alzheimer’s disease model. J. Neurosci. 33, 7535–7547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Johnson R, Guigo R, 2014. The RIDL hypothesis: transposable elements as functional domains of long noncoding RNAs. RNA 20, 959–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Johnson R, Zuccato C, Belyaev ND, Guest DJ, Cattaneo E, Buckley NJ, 2008. A microRNA-based gene dysregulation pathway in Huntington’s disease. Neuro-biol. Dis. 29, 438–445. [DOI] [PubMed] [Google Scholar]
  317. Johnsson P, Ackley A, Vidarsdottir L, Lui WO, Corcoran M, Grander D, Morris KV, 2013. A pseudogene long-noncoding-RNA network regulates PTEN transcription and translation in human cells. Nat. Struct. Mol. Biol. 20, 440–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, Rujescu D, Hampel H, Giegling I, Andreassen OA, Engedal K, Ulstein I, Djurovic S, Ibrahim-Verbaas C, Hofman A, Ikram MA, van Duijn CM, Thorsteinsdottir U, Kong A, Stefansson K, 2013. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 368, 107–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Jowaed A, Schmitt I, Kaut O, Wüllner U, 2010. Methylation regulates alpha-synuclein expression and is decreased in Parkinson’s disease patients’ brains. J. Neurosci. 30, 6355–6359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Julien C, Tremblay C, Emond V, Lebbadi M, Salem N Jr., Bennett DA, Calon F, 2009. SIRT1 decrease parallels the accumulation of tau in Alzheimer disease. J. Neuropathol. Exp. Neurol. 68, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Jung JW, Lee S, Seo MS, Park SB, Kurtz A, Kang SK, Kang KS, 2010. Histone deacetylase controls adult stem cell aging by balancing the expression of polycomb genes and jumonji domain containing 3. Cell. Mol. Life Sci. 67, 1165–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Junn E, Lee KW, Jeong BS, Chan TW, Im JY, Mouradian MM, 2009. Repression of alpha-synuclein expression and toxicity by microRNA-7. Proc. Natl. Acad. Sci. U.S.A. 106, 13052–13057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Junn E, Mouradian MM, 2012. MicroRNAs in neurodegenerative diseases and their therapeutic potential. Pharmacol. Ther. 133, 142–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Jurkowski TP, Meusburger M, Phalke S, Helm M, Nellen W, Reuter G, Jeltsch A, 2008. Human DNMT2 methylates tRNA(Asp) molecules using a DNA methyl- transferase-like catalytic mechanism. RNA 14, 1663–1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Kaidi A, Jackson SP, 2013. KAT5 tyrosine phosphorylation couples chromatin sensing to ATM signalling. Nature 498, 70–74. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  326. Kanthasamy A, Jin H, Anantharam V, Sondarva G, Rangasamy V, Rana A, Kanthasamy A, 2012. Emerging neurotoxic mechanisms in environmental factors-induced neurodegeneration. Neurotoxicology 33, 833–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermuller J, Hofacker IL, Bell I, Cheung E, Drenkow J, Dumais E, Patel S, Helt G, Ganesh M, Ghosh S, Piccolboni A, Sementchenko V, Tammana H, Gingeras TR, 2007. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 316, 1484–1488. [DOI] [PubMed] [Google Scholar]
  328. Kawahara Y, Megraw M, Kreider E, Iizasa H, Valente L, Hatzigeorgiou AG, Nishikura K, 2008. Frequency and fate of microRNA editing in human brain. Nucleic Acids Res. 36, 5270–5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Kawakami K, Nakamura A, Ishigami A, Goto S, Takahashi R, 2009. Age-related difference of site-specific histone modifications in rat liver. Biogerontology 10, 415–421. [DOI] [PubMed] [Google Scholar]
  330. Kellner S, Burhenne J, Helm M, 2010. Detection of RNA modifications. RNA Biol. 7, 237–247. [DOI] [PubMed] [Google Scholar]
  331. Kelly-Sell MJ, Kim YH, Straus S, Benoit B, Harrison C, Sutherland K, Armstrong R, Weng WK, Showe LC, Wysocka M, Rook AH, 2012. The histone deacetylase inhibitor, romidepsin, suppresses cellular immune functions of cutaneous T-cell lymphoma patients. Am. J. Hematol. 87, 354–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Kennedy BJ, 1972. Mithramycin therapy in testicular cancer. J. Urol. 107,429–432. [DOI] [PubMed] [Google Scholar]
  333. Kennedy BP, Bottiglieri T, Arning E, Ziegler MG, Hansen LA, Masliah E, 2004. Elevated S-adenosylhomocysteine in Alzheimer brain: influence on methyl- transferases and cognitive function. J. Neural Transm. 111, 547–567. [DOI] [PubMed] [Google Scholar]
  334. Khairallah MI, Kassem LA, 2011. Alzheimer’s disease: current status of etio- pathogenesis and therapeutic strategies. Pak. J. Biol. Sci. 14, 257–272. [DOI] [PubMed] [Google Scholar]
  335. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, Regev A, Lander ES, Rinn JL, 2009. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. U.S.A. 106, 11667–11672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  336. Kidd SK, Schneider JS, 2010. Protection of dopaminergic cells from MPP+-medi- ated toxicity by histone deacetylase inhibition. Brain Res. 1354, 172–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Kidd SK, Schneider JS, 2011. Protective effects of valproic acid on the nigrostriatal dopamine system in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience 194, 189–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Kilgore M, Miller CA, Fass DM, Hennig KM, Haggarty SJ, Sweatt JD, Rum-baugh G, 2009. Inhibitors of class 1 histone deacetylases reverse contextual memory deficits in a mouse model of Alzheimer’s disease. Neuropsychophar-macol.: Off. Publ. Am. Coll. Neuropsychopharmacol. 35, 870–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, Puigserver P, Sinclair DA, Tsai LH, 2007a. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. Embo J. 26, 3169–3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Kim DD, Kim TT, Walsh T, Kobayashi Y, Matise TC, Buyske S, Gabriel A, 2004. Widespread RNA editing of embedded alu elements in the human transcriptome. Genome Res. 14, 1719–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LI, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, McKinney-Free-man S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, Hanna J, Murakami P, Jaenisch R, Weissleder R, Orkin SH, Weissman IL, Feinberg AP, Daley GQ, 2010a. Epigenetic memory in induced pluripotent stem cells. Nature 467, 285–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Kim MS, Akhtar MW, Adachi M, Mahgoub M, Bassel-Duby R, Kavalali ET, Olson EN, Monteggia LM, 2012. An essential role for histone deacetylase 4 in synaptic plasticity and memory formation. J. Neurosci. 32, 10879–10886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Kim SH, Jeong JW, Park JA, Lee JW, Seo JH, Jung BK, Bae MK, Kim KW, 2007b. Regulation of the HIF-1alpha stability by histone deacetylases. Oncol. Rep. 17, 647–651. [PubMed] [Google Scholar]
  344. Kim T-K, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S, 2010b. Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Kirilyuk A, Shimoji M, Catania J, Sahu G, Pattabiraman N, Giordano A, Albanese C, Mocchetti I, Toretsky JA, Uversky VN, Avantaggiati ML, 2012. An intrinsically disordered region of the acetyltransferase p300 with similarity to prion-like domains plays a role in aggregation. PLoS ONE 7, e48243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Klafki HW, Staufenbiel M, Kornhuber J, Wiltfang J, 2006. Therapeutic approaches to Alzheimer’s disease. Brain 129, 2840–2855. [DOI] [PubMed] [Google Scholar]
  347. Kleinsmith LJ, Allfrey VG, Mirsky AE, 1966. Phosphorylation of nuclear protein early in the course of gene activation in lymphocytes. Science 154, 780–781. [DOI] [PubMed] [Google Scholar]
  348. Klose RJ, Bird AP, 2006. Genomic DNA methylation: the mark and its mediators. Trends Biochem. Sci. 31, 89–97. [DOI] [PubMed] [Google Scholar]
  349. Kondo Y, Issa J-PJ, 2003. Enrichment for histone H3 lysine 9 methylation at Alu repeats in human cells. J. Biol. Chem. 278, 27658–27662. [DOI] [PubMed] [Google Scholar]
  350. Kontopoulos E, Parvin JD, Feany MB, 2006. Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum. Mol. Genet. 15, 3012–3023. [DOI] [PubMed] [Google Scholar]
  351. Kopin IJ, 1987. Toxins and Parkinson’s disease: MPTP parkinsonism in humans and animals. Adv. Neurol. 45, 137–144. [PubMed] [Google Scholar]
  352. Korzus E, Rosenfeld MG, Mayford M, 2004. CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron 42, 961–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Koshibu K, Graff J, Beullens M, Heitz FD, Berchtold D, Russig H, Farinelli M, Bollen M, Mansuy IM, 2009. Protein phosphatase 1 regulates the histone code for long-term memory. J. Neurosci. 29, 13079–13089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Kouzarides T, 2007. Chromatin modifications and their function. Cell 128, 693–705. [DOI] [PubMed] [Google Scholar]
  355. Kowall NW, Ferrante RJ, Martin JB, 1987. Patterns of cell loss in Huntington’s disease. Trends Neurosci. 10, 24–29. [Google Scholar]
  356. Koziol MJ, Rinn JL, 2010. RNA traffic control of chromatin complexes. Curr. Opin. Genet. Dev. 20, 142–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Kremer B, Goldberg P, Andrew SE, Theilmann J, Telenius H, Zeisler J, Squitieri F, Lin B, Bassett A, Almqvist E, et al. , 1994. A worldwide study of the Huntington’s disease mutation. The sensitivity and specificity of measuring CAG repeats. N.Engl. J. Med. 330, 1401–1406. [DOI] [PubMed] [Google Scholar]
  358. Kriaucionis S, Heintz N, 2009. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324, 929–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Krützfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M, 2005. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438, 685–689. [DOI] [PubMed] [Google Scholar]
  360. Kudriashova IB, Kirnos MD, Vaniushin BF, 1976. DNA-methylase activities from animal mitochondria and nuclei: different specificity of DNA methylation. Biokhimiia 41, 1968–1977. [PubMed] [Google Scholar]
  361. Kuhn DE, Nuovo GJ, Terry AV Jr., Martin MM, Malana GE, Sansom SE, Pleister AP, Beck WD, Head E, Feldman DS, Elton TS, 2010. Chromosome 21-derived microRNAs provide an etiological basis for aberrant protein expression in human Down syndrome brains. J. Biol. Chem. 285, 1529–1543. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  362. Kumar A, Seghal N, Naidu PS, Padi SS, Goyal R, 2007. Colchicines-induced neurotoxicity as an animal model of sporadic dementia of Alzheimer’s type. Pharmacol. Rep. 59, 274. [PubMed] [Google Scholar]
  363. Ladd-Acosta C, Pevsner J, Sabunciyan S, Yolken RH, Webster MJ, Dinkins T, Callinan PA, Fan J-B, Potash JB, Feinberg AP, 2007. DNA methylation signatures within the human brain. Am. J. Hum. Genet. 81, 1304–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Lagger G, O’Carroll D, Rembold M, Khier H, Tischler J, Weitzer G, Schuetten- gruber B, Hauser C, Brunmeir R, Jenuwein T, Seiser C, 2002. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J. 21, 2672–2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Lahiri DK, Maloney B, Zawia NH, 2009. The LEARn model: an epigenetic explanation for idiopathic neurobiological diseases. Mol. Psychiatry 14, 992–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Lakhal S, El Andaloussi S, O’Loughlin AJ, Li J, Wood MM, 2013. RNAi therapeutic delivery by exosomes In: RNA Interference from Biology to Therapeu-tics Springer, pp. 185–205. [Google Scholar]
  367. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, Funke R, Gage D, Harris K, Heaford A, Howland J, Kann L, Lehoczky J, LeVine R, McEwan P, McKernan K, Meldrim J, Mesirov JP, Miranda C, Morris W, Naylor J, Raymond C, Rosetti M, Santos R, Sheridan A, Sougnez C, Stange-Thomann N, Stojanovic N, Subramanian A, Wyman D, Rogers J, Sulston J, Ainscough R, Beck S, Bentley D, Burton J, Clee C, Carter N, Coulson A, Deadman R, Deloukas P, Dunham A, Dunham I, Durbin R, French L, Grafham D, Gregory S, Hubbard T, Humphray S, Hunt A, Jones M, Lloyd C, McMurray A, Matthews L, Mercer S, Milne S, Mullikin JC, Mungall A, Plumb R, Ross M, Shownkeen R, Sims S, Waterston RH, Wilson RK, Hillier LW, McPherson JD, Marra MA, Mardis ER, Fulton LA, Chinwalla AT, Pepin KH, Gish WR, Chissoe SL, Wendl MC, Delehaunty KD, Miner TL, Delehaunty A, Kramer JB, Cook LL, Fulton RS, Johnson DL, Minx PJ, Clifton SW, Hawkins T, Branscomb E, Predki P, Richardson P, Wenning S, Slezak T, Doggett N, Cheng JF, Olsen A, Lucas S, Elkin C, Uberbacher E, Frazier M, Gibbs RA, Muzny DM, Scherer SE, Bouck JB, Sodergren EJ, Worley KC, Rives CM, Gorrell JH, Metzker ML, Naylor SL, Kucherlapati RS, Nelson DL, Weinstock GM, Sakaki Y, Fujiyama A, Hattori M, Yada T, Toyoda A, Itoh T, Kawagoe C, Watanabe H, Totoki Y, Taylor T, Weissenbach J, Heilig R, Saurin W, Artiguenave F, Brottier P, Bruls T, Pelletier E, Robert C, Wincker P, Smith DR, Doucette-Stamm L, Rubenfield M, Weinstock K, Lee HM, Dubois J, Rosenthal A, Platzer M, Nyakatura G, Taudien S, Rump A, Yang H, Yu J, Wang J, Huang G, Gu J, Hood L, Rowen L, Madan A, Qin S, Davis RW, Federspiel NA, Abola AP, Proctor MJ, Myers RM, Schmutz J, Dickson M, Grimwood J, Cox DR, Olson MV, Kaul R, Raymond C, Shimizu N, Kawasaki K, Minoshima S, Evans GA, Athanasiou M, Schultz R, Roe BA, Chen F, Pan H, Ramser J, Lehrach H, Reinhardt R, McCombie WR, de la Bastide M, Dedhia N, Blocker H, Hornischer K, Nordsiek G, Agarwala R, Aravind L, Bailey JA, Bateman A, Batzoglou S, Birney E, Bork P, Brown DG, Burge CB, Cerutti L, Chen HC, Church D, Clamp M, Copley RR, Doerks T, Eddy SR, Eichler EE, Furey TS, Galagan J, Gilbert JG, Harmon C, Hayashizaki Y, Haussler D, Hermjakob H, Hokamp K, Jang W, Johnson LS, Jones TA, Kasif S, Kaspryzk A, Kennedy S, Kent WJ, Kitts P, Koonin EV, Korf I, Kulp D, Lancet D, Lowe TM, McLysaght A, Mikkelsen T, Moran JV, Mulder N, Pollara VJ, Ponting CP, Schuler G, Schultz J, Slater G, Smit AF, Stupka E, Szustakowski J, Thierry-Mieg D, Thierry-Mieg J, Wagner L, Wallis J, Wheeler R, Williams A, Wolf YI, Wolfe KH, Yang SP, Yeh RF, Collins F, Guyer MS, Peterson J, Felsenfeld A, Wetterstrand KA, Patrinos A, Morgan MJ, de Jong P, Catanese JJ, Osoegawa K, Shizuya H, Choi S, Chen YJ, International Human Genome Sequencing, C., 2001. Initial sequencing and analysis of the human genome. Nature 409, 860–921. [DOI] [PubMed] [Google Scholar]
  368. Lee J, Hagerty S, Cormier KA, Kim J, Kung AL, Ferrante RJ, Ryu H, 2008. Monoallele deletion of CBP leads to pericentromeric heterochromatin condensation through ESET expression and histone H3 (K9) methylation. Hum. Mol. Genet. 17, 1774–1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Lee J, Hwang YJ, Kim KY, Kowall NW, Ryu H, 2013a. Epigenetic mechanisms of neurodegeneration in Huntington’s disease. Neurother. 10, 664–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Lee J, Hwang YJ, Shin JY, Lee WC, Wie J, Kim KY, Lee MY, Hwang D, Ratan RR, Pae AN, Kowall NW, So I, Kim JI, Ryu H, 2013b. Epigenetic regulation of cholinergic receptor M1 (CHRM1) by histone H3K9me3 impairs Ca(2+) signaling in Huntington’s disease. Acta Neuropathol. 125, 727–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  371. Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, Ferrante TC, Terry R, Jeanty SS, Li C, Amamoto R, Peters DT, Turczyk BM, Marblestone AH, Inverso SA, Bernard A, Mali P, Rios X, Aach J, Church GM, 2014. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Lee RC, Feinbaum RL, Ambros V, 1993. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843–854. [DOI] [PubMed] [Google Scholar]
  373. Lee S, Lemere CA, Frost JL, Shea TB, 2012. Dietary supplementation with S-adenosyl methionine delayed amyloid-beta and tau pathology in 3xTg-AD mice. J. Alzheimers Dis. 28, 423–431. [DOI] [PubMed] [Google Scholar]
  374. Lee ST, Chu K, Im WS, Yoon HJ, Im JY, Park JE, Park KH, Jung KH, Lee SK, Kim M, Roh JK, 2011. Altered microRNA regulation in Huntington’s disease models. Exp. Neurol. 227, 172–179. [DOI] [PubMed] [Google Scholar]
  375. Lee VM, Goedert M, Trojanowski JQ, 2001. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 24, 1121–1159. [DOI] [PubMed] [Google Scholar]
  376. Lemere CA, Lopera F, Kosik KS, Lendon CL, Ossa J, Saido TC, Yamaguchi H, Ruiz A, Martinez A, Madrigal L, Hincapie L, Arango JC, Anthony DC, Koo EH, Goate AM, Selkoe DJ, Arango JC, 1996. The E280A presenilin 1 Alzheimer mutation produces increased A beta 42 deposition and severe cerebellar pathology. Nat. Med. 2, 1146–1150. [DOI] [PubMed] [Google Scholar]
  377. Leng Y, Marinova Z, Reis-Fernandes MA, Nau H, Chuang DM, 2010. Potent neuroprotective effects of novel structural derivatives of valproic acid: potential roles of HDAC inhibition and HSP70 induction. Neurosci. Lett. 476, 127–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Levanon EY, Eisenberg E, Yelin R, Nemzer S, Hallegger M, Shemesh R, Fligel- man ZY, Shoshan A, Pollock SR, Sztybel D, Olshansky M, Rechavi G, Jantsch MF, 2004. Systematic identification of abundant A-to-I editing sites in the human transcriptome. Nat. Biotechnol. 22, 1001–1005. [DOI] [PubMed] [Google Scholar]
  379. Levenson JM, Roth TL, Lubin FD, Miller CA, Huang I-C, Desai P, Malone LM, Sweatt JD, 2006. Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus. J. Biol. Chem. 281, 15763–15773. [DOI] [PubMed] [Google Scholar]
  380. Li N, Bates DJ, An J, Terry DA, Wang E, 2011. Up-regulation of key microRNAs, and inverse down-regulation of their predicted oxidative phosphorylation target genes, during aging in mouse brain. Neurobiol. Aging 32, 944–955. [DOI] [PubMed] [Google Scholar]
  381. Li SH, Cheng AL, Zhou H, Lam S, Rao M, Li H, Li XJ, 2002. Interaction of Huntington disease protein with transcriptional activator Sp1. Mol. Cell. Biol. 22, 1277–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Liang C, Zhu H, Xu Y, Huang L, Ma C, Deng W, Liu Y, Qin C, 2012. MicroRNA- 153 negatively regulates the expression of amyloid precursor protein and amyloid precursor-like protein 2. Brain Res. 1455, 103–113. [DOI] [PubMed] [Google Scholar]
  383. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM, 2005. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769–773. [DOI] [PubMed] [Google Scholar]
  384. Lisch D, 2012. Regulation of transposable elements in maize. Curr. Opin. Plant Biol. 15, 511–516. [DOI] [PubMed] [Google Scholar]
  385. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, Nery JR, Lee L, Ye Z, Ngo Q-M, 2009. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Lithner CU, Hernandez CM, Nordberg A, Sweatt JD, 2009. Epigenetic changes related to beta-amyloid-implications for Alzheimer’s disease. Alzheimer’s Dement. 5, P304. [Google Scholar]
  387. Liu C, Teng ZQ, Santistevan NJ, Szulwach KE, Guo W, Jin P, Zhao X, 2010a. Epigenetic regulation of miR-184 by MBD1 governs neural stem cell proliferation and differentiation. Cell Stem Cell 6, 433–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Liu F, Gong CX, 2008. Tau exon 10 alternative splicing and tauopathies. Mol. Neurodegener. 3, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  389. Liu HP, Lin WY, Wu BT, Liu SH, Wang WF, Tsai CH, Lee CC, Tsai FJ, 2010b. Evaluation of the poly(ADP-ribose) polymerase-1 gene variants in Alzheimer’s disease. J. Clin. Lab. Anal. 24, 182–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Liu L, Li Y, Tollefsbol TO, 2008. Gene-environment interactions and epigenetic basis of human diseases. Curr. Issues Mol. Biol. 10, 25–36. [PMC free article] [PubMed] [Google Scholar]
  391. Liu R, Lei JX, Luo C, Lan X, Chi L, Deng P, Lei S, Ghribi O, Liu QY, 2012a. Increased EID1 nuclear translocation impairs synaptic plasticity and memory function associated with pathogenesis of Alzheimer’s disease. Neurobiol. Dis. 45, 902–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Liu W, Liu C, Zhu J, Shu P, Yin B, Gong Y, Qiang B, Yuan J, Peng X, 2012b. MicroRNA-16 targets amyloid precursor protein to potentially modulate Alzheimer’s-associated pathogenesis in SAMP8 mice. Neurobiol. Aging 33, 522–534. [DOI] [PubMed] [Google Scholar]
  393. Lombardi MS, Jaspers L, Spronkmans C, Gellera C, Taroni F, Di Maria E, Donato SD, Kaemmerer WF, 2009. A majority of Huntington’s disease patients may be treatable by individualized allele-specific RNA interference. Exp. Neurol. 217, 312–319. [DOI] [PubMed] [Google Scholar]
  394. Long JM, Lahiri DK, 2011. MicroRNA-101 downregulates Alzheimer’s amyloid-β precursor protein levels in human cell cultures and is differentially expressed. Biochem. Biophys. Res. Commun. 404, 889–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Long JM, Ray B, Lahiri DK, 2012. MicroRNA-153 physiologically inhibits expression of amyloid-beta precursor protein in cultured human fetal brain cells and is dysregulated in a subset of alzheimer disease patients. J. Biol. Chem. 287, 31298–31310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Lopatina N, Haskell JF, Andrews LG, Poole JC, Saldanha S, Tollefsbol T, 2002. Differential maintenance and de novo methylating activity by three DNA methyltransferases in aging and immortalized fibroblasts. J. Cell Biochem. 84, 324–334. [DOI] [PubMed] [Google Scholar]
  397. Lovestone S, Reynolds CH, 1997. The phosphorylation of tau: a critical stage in neurodevelopment and neurodegenerative processes. Neuroscience 78, 309–324. [DOI] [PubMed] [Google Scholar]
  398. Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ, 1997. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389, 251–260. [DOI] [PubMed] [Google Scholar]
  399. Lukiw WJ, 2007. Micro-RNA speciation in fetal, adult and Alzheimer’s disease hippocampus. Neuroreport 18, 297–300. [DOI] [PubMed] [Google Scholar]
  400. Lukiw WJ, Handley P, Wong L, Crapper McLachlan DR, 1992. BC200 RNA in normal human neocortex, non-Alzheimer dementia (NAD), and senile dementia of the Alzheimer type (AD). Neurochem. Res. 17, 591–597. [DOI] [PubMed] [Google Scholar]
  401. Lukiw WJ, Zhao Y, Cui JG, 2008. An NF-kappaB-sensitive micro RNA-146a- mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J. Biol. Chem. 283, 31315–31322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Lunnon K, Ibrahim Z, Proitsi P, Lourdusamy A, Newhouse S, Sattlecker M, Furney S, Saleem M, Soininen H, Kloszewska I, Mecocci P, Tsolaki M, Vellas B, Coppola G, Geschwind D, Simmons A, Lovestone S, Dobson R, Hodges A, AddNeuroMed C, 2012. Mitochondrial dysfunction and immune activation are detectable in early Alzheimer’s disease blood. J. Alzheimers Dis. 30, 685–710. [DOI] [PubMed] [Google Scholar]
  403. Lunnon K, Mill J, 2013. Epigenetic studies in Alzheimer’s disease: current findings, caveats, and considerations for future studies. Am. J. Med. Genet. Part B: Neuropsychiatr. Genet. 162b, 789–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Lunnon K, Smith R, Hannon E, De Jager PL, Srivastava G, Volta M, Troakes C, Al-Sarraj S, Burrage J, Macdonald R, Condliffe D, Harries LW, Katsel P, Haroutunian V, Kaminsky Z, Joachim C, Powell J, Lovestone S, Bennett DA, Schalkwyk LC, Mill J, 2014. Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease. Nat. Neurosci. 17, 1164–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Lyko F, Foret S, Kucharski R, Wolf S, Falckenhayn C, Maleszka R, 2010. The honey bee epigenomes: differential methylation of brain DNA in queens and workers. PLoS Biol. 8, e1000506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  406. Ma DK, Jang MH, Guo JU, Kitabatake Y, Chang ML, Pow-Anpongkul N, Flavell RA, Lu B, Ming GL, Song H, 2009. Neuronal activity-induced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science 323, 1074–1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H, 2010. Epigenetic choreographers of neurogenesis in the adult mammalian brain. Nat. Neurosci. 13, 1338–1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. MacDonald JL, Roskams AJ, 2008. Histone deacetylases 1 and 2 are expressed at distinct stages of neuro-glial development. Dev. Dyn. 237, 2256–2267. [DOI] [PubMed] [Google Scholar]
  409. MacDonald ME, Ambrose CM, Duyao MP, Myers RH, Lin C, Srinidhi L, Barnes G, Taylor SA, James M, Groot N, 1993. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 72, 971–983. [DOI] [PubMed] [Google Scholar]
  410. Machida Y, Okada T, Kurosawa M, Oyama F, Ozawa K, Nukina N, 2006. rAAV- mediated shRNA ameliorated neuropathology in Huntington disease model mouse. Biochem. Biophys. Res. Commun. 343, 190–197. [DOI] [PubMed] [Google Scholar]
  411. Madrigano J, Baccarelli A, Mittleman MA, Sparrow D, Vokonas PS, Tarantini L, Schwartz J, 2012. Aging and epigenetics: longitudinal changes in gene-specific DNA methylation. Epigenetics 7, 63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Maes OC, An J, Sarojini H, Wang E, 2008. Murine microRNAs implicated in liver functions and aging process. Mech. Ageing Dev. 129, 534–541. [DOI] [PubMed] [Google Scholar]
  413. Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, Antonini A, Martelli F, Capogrossi MC, 2011. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 18, 1628–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  414. Majello B, De Luca P, Suske G, Lania L, 1995. Differential transcriptional regulation of c-myc promoter through the same DNA binding sites targeted by Sp1-like proteins. Oncogene 10, 1841–1848. [PubMed] [Google Scholar]
  415. Malouf R, Grimley Evans J, Areosa Sastre A, 2005. Folic acid with or without vitamin B12 for cognition and dementia. Cochrane Libr. 4, CD004514. [DOI] [PubMed] [Google Scholar]
  416. Manev H, Dzitoyeva S, Chen H, 2012. Mitochondrial DNA: a blind spot in neuroepigenetics. Biomol. Concepts 3, 107–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, Lawton M, Trottier Y, Lehrach H, Davies SW, 1996. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87, 493–506. [DOI] [PubMed] [Google Scholar]
  418. Margis R, Margis R, Rieder CR, 2011. Identification of blood microRNAs associated to Parkinsonis disease. J. Biotechnol. 152, 96–101. [DOI] [PubMed] [Google Scholar]
  419. Marinova Z, Leng Y, Leeds P, Chuang DM, 2011. Histone deacetylase inhibition alters histone methylation associated with heat shock protein 70 promoter modifications in astrocytes and neurons. Neuropharmacology 60, 1109–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Marinova Z, Ren M, Wendland JR, Leng Y, Liang MH, Yasuda S, Leeds P, Chuang DM, 2009. Valproic acid induces functional heat-shock protein 70 via class I histone deacetylase inhibition in cortical neurons: a potential role of Sp1 acetylation. J. Neurochem. 111, 976–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Marks PA, 2010. The clinical development of histone deacetylase inhibitors as targeted anticancer drugs. Expert Opin. Investig. Drugs 19, 1049–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Marks PA, Breslow R, 2007. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat. Biotechnol. 25,84–90. [DOI] [PubMed] [Google Scholar]
  423. Marks PA, Xu WS, 2009. Histone deacetylase inhibitors: potential in cancer therapy. J. Cell Biochem. 107, 600–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Marmorstein R, Roth SY, 2001. Histone acetyltransferases: function, structure, and catalysis. Curr. Opin. Genet. Dev. 11, 155–161. [DOI] [PubMed] [Google Scholar]
  425. Marques S, Lemos R, Ferreiro E, Martins M, de Mendonca A, Santana I, Outeiro T, Pereira C, 2012. Epigenetic regulation of BACE1 in Alzheimer’s disease patients and in transgenic mice. Neuroscience 220, 256–266. [DOI] [PubMed] [Google Scholar]
  426. Martens JH, O’Sullivan RJ, Braunschweig U, Opravil S, Radolf M, Steinlein P, Jenuwein T, 2005. The profile of repeat-associated histone lysine methylation states in the mouse epigenome. EMBO J. 24, 800–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  427. Massone S, Ciarlo E, Vella S, Nizzari M, Florio T, Russo C, Cancedda R, Pagano A, 2012. NDM29, a RNA polymerase III-dependent non coding RNA, promotes amyloidogenic processing of APP and amyloid ß secretion. Biochim. Biophys. Acta: Mol. Cell Res. 1823, 1170–1177. [DOI] [PubMed] [Google Scholar]
  428. Massone S, Vassallo I, Fiorino G, Castelnuovo M, Barbieri F, Borghi R, Tabaton M, Robello M, Gatta E, Russo C, Florio T, Dieci G, Cancedda R, Pagano A, 2011. 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease. Neu-robiol. Dis. 41, 308–317. [DOI] [PubMed] [Google Scholar]
  429. Mastroeni D, Chouliaras L, Grover A, Liang WS, Hauns K, Rogers J, Coleman PD, 2013. Reduced RAN expression and disrupted transport between cytoplasm and nucleus; a key event in Alzheimer’s disease pathophysiology. PLOS ONE 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J, 2010. Epigenetic changes in Alzheimer’s disease: decrements in DNA methylation. Neurobiol. Aging 31, 2025–2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  431. Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J, 2011. Epigenetic mechanisms in Alzheimer’s disease. Neurobiol. Aging 32, 1161–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Matsubara M, Tanaka T, Terato H, Ohmae E, Izumi S, Katayanagi K, Ide H, 2004. Mutational analysis of the damage-recognition and catalytic mechanism of human SMUG1 DNA glycosylase. Nucleic Acids Res. 32, 5291–5302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Matsumoto L, Takuma H, Tamaoka A, Kurisaki H, Date H, Tsuji S, Iwata A, 2010CpG demethylation enhances alpha-synuclein expression and affects the pathogenesis of Parkinson’s disease. PLoS ONE 5, e15522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Mattick JS, 2010. RNA as the substrate for epigenome-environment interactions. Bioessays 32, 548–552. [DOI] [PubMed] [Google Scholar]
  435. Mattick JS, 2011. The central role of RNA in human development and cognition. FEBS Lett. 585, 1600–1616. [DOI] [PubMed] [Google Scholar]
  436. Mattick JS, Mehler MF, 2008. RNA editing, DNA recoding and the evolution of human cognition. Trends Neurosci. 31, 227–233. [DOI] [PubMed] [Google Scholar]
  437. Mattson MP, 2003. Methylation and acetylation in nervous system development and neurodegenerative disorders. Ageing Res. Rev. 2, 329–342. [DOI] [PubMed] [Google Scholar]
  438. Mattson MP, Cheng B, Davis D, Bryant K, Lieberburg I, Rydel RE, 1992. Beta-Amyloid peptides destabilize calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity. J. Neurosci. 12, 376–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  439. Maurice T, Duclot F, Meunier J, Naert G, Givalois L, Meffre J, Celerier A, Jacquet C, Copois V, Mechti N, Ozato K, Gongora C, 2008. Altered memory capacities and response to stress in p300/CBP-associated factor (PCAF) histone acetylase knockout mice. Neuropsychopharmacol. 33, 1584–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  440. Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ, 2010. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 330, 1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Maxwell ES, Fournier MJ, 1995. The small nucleolar RNAs. Annu. Rev. Biochem. 64, 897–934. [DOI] [PubMed] [Google Scholar]
  442. Mazin AL, 2009. Suicidal function of DNA methylation in age-related genome disintegration. Ageing Res. Rev. 8, 314–327. [DOI] [PubMed] [Google Scholar]
  443. McBride JL, Boudreau RL, Harper SQ, Staber PD, Monteys AM, Martins I, Gilmore BL, Burstein H, Peluso RW, Polisky B, Carter BJ, Davidson BL, 2008. Artificial miRNAs mitigate shRNA-mediated toxicity in the brain: implications for the therapeutic development of RNAi. Proc. Natl. Acad. Sci. U.S.A. 105, 5868–5873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. McCampbell A, Taye AA, Whitty L, Penney E, Steffan JS, Fischbeck KH, 2001. Histone deacetylase inhibitors reduce polyglutamine toxicity. Proc. Natl. Acad. Sci. U.S.A. 98, 15179–15184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  445. McFarland KN, Das S, Sun TT, Leyfer D, Xia E, Sangrey GR, Kuhn A, Luthi- Carter R, Clark TW, Sadri-Vakili G, 2012. Genome-wide histone acetylation is altered in a transgenic mouse model of Huntington’s disease. PLoS ONE 7, e41423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  446. McMahon JA, Green TJ, Skeaff CM, Knight RG, Mann JI, Williams SM, 2006. A controlled trial of homocysteine lowering and cognitive performance. N. Engl. J. Med. 354, 2764–2772. [DOI] [PubMed] [Google Scholar]
  447. McQuown SC, Barrett RM, Matheos DP, Post RJ, Rogge GA, Alenghat T, Mullican SE, Jones S, Rusche JR, Lazar MA, Wood MA, 2011. HDAC3 is a critical negative regulator of long-term memory formation. J. Neurosci. 31, 764–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Meaburn EL, Schalkwyk LC, Mill J, 2010. Allele-specific methylation in the human genome: implications for genetic studies of complex disease. Epigenetics 5, 578–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Menghini R, Casagrande V, Cardellini M, Martelli E, Terrinoni A, Amati F, Vasa-Nicotera M, Ippoliti A, Novelli G, Melino G, Lauro R, Federici M, 2009. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation 120, 1524–1532. [DOI] [PubMed] [Google Scholar]
  450. Mercer TR, Mattick JS, 2013. Structure and function of long noncoding RNAs in epigenetic regulation. Nat. Struct. Mol. Biol. 20, 300–307. [DOI] [PubMed] [Google Scholar]
  451. Mercer TR, Wilhelm D, Dinger ME, Solda G, Korbie DJ, Glazov EA, Truong V, Schwenke M, Simons C, Matthaei KI, Saint R, Koopman P, Mattick JS, 2011. Expression of distinct RNAs from 3’ untranslated regions. Nucleic Acids Res. 39, 2393–2403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  452. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR, 2012. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell 149, 1635–1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Michan S, Sinclair D, 2007. Sirtuins in mammals: insights into their biological function. Biochem. J. 404, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  454. Mielcarek M, Benn CL, Franklin SA, Smith DL, Woodman B, Marks PA, Bates GP, 2011. SAHA decreases HDAC 2 and 4 levels in vivo and improves molecular phenotypes in the R6/2 mouse model of Huntington’s disease. PLoS ONE 6, e27746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  455. Migliore L, Coppede F, 2009. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat. Res. 667, 82–97. [DOI] [PubMed] [Google Scholar]
  456. Mill J, 2011. Toward an integrated genetic and epigenetic approach to Alzheimer’s disease. Neurobiol. Aging 32, 1188–1191. [DOI] [PubMed] [Google Scholar]
  457. Mill J, Heijmans BT, 2013. From promises to practical strategies in epigenetic epidemiology. Nat. Rev. Genet. 14, 585–594. [DOI] [PubMed] [Google Scholar]
  458. Miller AA, Kurschel E, Osieka R, Schmidt CG, 1987. Clinical pharmacology of sodium butyrate in patients with acute leukemia. Eur. J. Cancer Clin. Oncol. 23, 1283–1287. [DOI] [PubMed] [Google Scholar]
  459. Miller CA, Sweatt JD, 2007. Covalent modification of DNA regulates memory formation. Neuron 53, 857–869. [DOI] [PubMed] [Google Scholar]
  460. Milutinovic S, Brown SE, Zhuang Q, Szyf M, 2004. DNA methyltransferase 1 knock down induces gene expression by a mechanism independent of DNA methylation and histone deacetylation. J. Biol. Chem. 279, 27915–27927. [DOI] [PubMed] [Google Scholar]
  461. Min SW, Cho SH, Zhou Y, Schroeder S, Haroutunian V, Seeley WW, Huang EJ, Shen Y, Masliah E, Mukherjee C, Meyers D, Cole PA, Ott M, Gan L, 2010. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67, 953–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  462. Minamide LS, Striegl AM, Boyle JA, Meberg PJ, Bamburg JR, 2000. Neurodegenerative stimuli induce persistent ADF/cofilin-actin rods that disrupt distal neurite function. Nat. Cell Biol. 2, 628–636. [DOI] [PubMed] [Google Scholar]
  463. Minones-Moyano E, Porta S, Escaramis G, Rabionet R, Iraola S, Kagerbauer B, Espinosa-Parrilla Y, Ferrer I, Estivill X, Marti E, 2011. MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function. Hum. Mol. Genet. 20, 3067–3078. [DOI] [PubMed] [Google Scholar]
  464. Minucci S, Pelicci PG, 2006. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat. Rev. Cancer 6, 38–51. [DOI] [PubMed] [Google Scholar]
  465. Mohamed JS, Lopez MA, Boriek AM, 2010. Mechanical stretch up-regulates microRNA-26a and induces human airway smooth muscle hypertrophy by suppressing glycogen synthase kinase-3ß. J. Biol. Chem. 285, 29336–29347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Moore K, McKnight AJ, Craig D, O’Neill F, 2014. Epigenome-wide association study for Parkinson’s disease. Neuromol. Med. 16, 845–855. [DOI] [PubMed] [Google Scholar]
  467. Moreira PR, Guimaraes MM, Guimaraes AL, Diniz MG, Gomes CC, Brito JA, Gomez RS, 2009. Methylation of P16, P21, P27, RB1 and P53 genes in odontogenic keratocysts. J. Oral Pathol. Med. 38, 99–103. [DOI] [PubMed] [Google Scholar]
  468. Morgan AR, Hamilton G, Turic D, Jehu L, Harold D, Abraham R, Hollingworth P, Moskvina V, Brayne C, Rubinsztein DC, Lynch A, Lawlor B, Gill M, O’Donovan M, Powell J, Lovestone S, Williams J, Owen MJ, 2008. Association analysis of528 intra-genic SNPs in a region ofchromosome 10 linked to late onset Alzheimer’s disease. Am. J. Med. Genet. Part B: Neuropsychiatr. Genet. 147B, 727–731. [DOI] [PubMed] [Google Scholar]
  469. Morris BJ, 2013. Seven sirtuins for seven deadly diseases of aging. Free Radic. Biol. Med. 56, 133–171. [DOI] [PubMed] [Google Scholar]
  470. Morrison LD, Smith DD, Kish SJ, 1996. Brain S-adenosylmethionine levels are severely decreased in Alzheimer’s disease. J. Neurochem. 67, 1328–1331. [DOI] [PubMed] [Google Scholar]
  471. Morrissy AS, Griffith M, Marra MA, 2011. Extensive relationship between antisense transcription and alternative splicing in the human genome. Genome Res. 21, 1203–1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  472. Morse DP, Aruscavage PJ, Bass BL, 2002. RNA hairpins in noncoding regions of human brain and Caenorhabditis elegans mRNA are edited by adenosine deaminases that act on RNA. Proc. Natl. Acad. Sci. U.S.A. 99, 7906–7911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  473. Muerkoster SS, Werbing V, Koch D, Sipos B, Ammerpohl O, Kalthoff H, Tsao MS, Folsch UR, Schafer H, 2008. Role of myofibroblasts in innate chemore-sistance of pancreatic carcinoma - epigenetic downregulation of caspases. Int. J. Cancer 123, 1751–1760. [DOI] [PubMed] [Google Scholar]
  474. Mukaetova-Ladinska EB, Harrington CR, Roth M, Wischik CM, 1996. Alterations in tau protein metabolism during normal aging. Dementia 7, 95–103. [DOI] [PubMed] [Google Scholar]
  475. Muller GB, Olsson L, 2003. Epigenesis and epigenetics. Keyw. Concepts Evol. Dev. Biol. 114. [Google Scholar]
  476. Muller T, Schrotter A, Loosse C, Pfeiffer K, Theiss C, Kauth M, Meyer HE, Marcus K, 2013. A ternary complex consisting ofAICD, FE65, and TIP60 down- regulates Stathmin1. Biochim. Biophys. Acta 1834, 387–394. [DOI] [PubMed] [Google Scholar]
  477. Munns TW, Sims HF, Liszewski MK, 1977. Immunospecific retention of oligonucleotides possessing N6-methyladenosine and 7-methylguanosine. J. Biol. Chem. 252, 3102–3104. [PubMed] [Google Scholar]
  478. Munzel M, Globisch D, Bruckl T, Wagner M, Welzmiller V, Michalakis S, Muller M, Biel M, Carell T, 2010. Quantification of the sixth DNA base hydroxymethylcytosine in the brain. Angew. Chem. Int. Ed. Engl. 49, 5375–5377. [DOI] [PubMed] [Google Scholar]
  479. Muramatsu M, Sankaranand VS, Anant S, Sugai M, Kinoshita K, Davidson NO, Honjo T, 1999. Specific expression of activation-induced cytidine deaminase (AID), a novel member of the RNA-editing deaminase family in germinal center B cells. J. Biol. Chem. 274, 18470–18476. [DOI] [PubMed] [Google Scholar]
  480. Murray K, 1964. The occurrence of epsilon-N-methyl lysine in histones. Biochemistry 3, 10–15. [DOI] [PubMed] [Google Scholar]
  481. Mus E, Hof PR, Tiedge H, 2007. Dendritic BC200 RNA in aging and in Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 104, 10679–10684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  482. Myers RH, MacDonald ME, Koroshetz WJ, Duyao MP, Ambrose CM, Taylor SA, Barnes G, Srinidhi J, Lin CS, Whaley WL, et al. , 1993. De novo expansion of a (CAG)n repeat in sporadic Huntington’s disease. Nat. Genet. 5, 168–173. [DOI] [PubMed] [Google Scholar]
  483. Myung NH, Zhu X, Kruman II, Castellani RJ, Petersen RB, Siedlak SL, Perry G, Smith MA, Lee HG, 2008. Evidence of DNA damage in Alzheimer disease: phosphorylation of histone H2AX in astrocytes. Age 30, 209–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Naimark D, Jackson E, Rockwell E, Jeste DV, 1996. Psychotic symptoms in Parkinson’s disease patients with dementia. J. Am. Geriatr. Soc. 44, 296–299. [DOI] [PubMed] [Google Scholar]
  485. Naj AC, Jun G, Beecham GW, Wang L-S, Vardarajan BN, Buros J, Gallins PJ, Buxbaum JD, Jarvik GP, Crane PK, 2011. Common variants at MS4A4/ MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat. Genet. 43, 436–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Nakamori M, Thornton C, 2010. Epigenetic changes and non-coding expanded repeats. Neurobiol. Dis. 39, 21–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  487. Nakamura A, Kawakami K, Kametani F, Nakamoto H, Goto S, 2010. Biological significance of protein modifications in aging and calorie restriction. Ann. N. Y. Acad. Sci. 1197, 33–39. [DOI] [PubMed] [Google Scholar]
  488. Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW, 2003. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716. [DOI] [PubMed] [Google Scholar]
  489. Nass MM, 1973. Differential methylation of mitochondrial and nuclear DNA in cultured mouse, hamster and virus-transformed hamster cells. In vivo and in vitro methylation. J. Mol. Biol. 80, 155–175. [DOI] [PubMed] [Google Scholar]
  490. Navaratnam N, Sarwar R, 2006. An overview of cytidine deaminases. Int. J. Hematol. 83, 195–200. [DOI] [PubMed] [Google Scholar]
  491. Neumann H, Daly MJ, 2013. Variant TREM2 as risk factor for Alzheimer’s disease. N. Engl. J. Med. 368, 182–184. [DOI] [PubMed] [Google Scholar]
  492. Ng CW, Yildirim F, Yap YS, Dalin S, Matthews BJ, Velez PJ, Labadorf A, Housman DE, Fraenkel E, 2013. Extensive changes in DNA methylation are associated with expression of mutant huntingtin. Proc. Natl. Acad. Sci. U.S.A. 110, 2354–2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  493. Nicholas A, de Magalhaes JP, Kraytsberg Y, Richfield EK, Levanon EY, Khrapko K, 2010. Age-related gene-specific changes of A-to-I mRNA editing in the human brain. Mech. Ageing Dev. 131, 445–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  494. Nicholas AP, Lubin FD, Hallett PJ, Vattem P, Ravenscroft P, Bezard E, Zhou S, Fox SH, Brotchie JM, Sweatt JD, Standaert DG, 2008. Striatal histone modifications in models of levodopa-induced dyskinesia. J. Neurochem. 106, 486–494. [DOI] [PubMed] [Google Scholar]
  495. Nishikura K, 2006. Editor meets silencer: crosstalk between RNA editing and RNA interference. Nat. Rev. Mol. Cell Biol. 7, 919–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  496. Nishino J, Kim I, Chada K, Morrison SJ, 2008. Hmga2 promotes neural stem cell self-renewal in young but not old mice by reducing p16< sup> Ink4a and p19< sup> Arf expression. Cell 135, 227–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  497. Niwa R, Zhou F, Li C, Slack FJ, 2008. The expression of the Alzheimer’s amyloid precursor protein-like gene is regulated by developmental timing microRNAs and their targets in Caenorhabditis elegans. Dev. Biol. 315, 418–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Noren Hooten N, Abdelmohsen K, Gorospe M, Ejiogu N, Zonderman AB, Evans MK, 2010. microRNA expression patterns reveal differential expression of target genes with age. PLoS ONE 5, e10724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  499. Nucifora FC Jr., Sasaki M, Peters MF, Huang H, Cooper JK, Yamada M, Takahashi H, Tsuji S, Troncoso J, Dawson VL, Dawson TM, Ross CA, 2001. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 291, 2423–2428. [DOI] [PubMed] [Google Scholar]
  500. Nuutinen T, Suuronen T, Kauppinen A, Salminen A, 2010. Valproic acid stimulates clusterin expression in human astrocytes: implications for Alzheimer’s disease. Neurosci. Lett. 475, 64–68. [DOI] [PubMed] [Google Scholar]
  501. Nuutinen T, Suuronen T, Kyrylenko S, Huuskonen J, Salminen A, 2005. Induction of clusterin/apoJ expression by histone deacetylase inhibitors in neural cells. Neurochem. Int. 47, 528–538. [DOI] [PubMed] [Google Scholar]
  502. Obeid R, Schadt A, Dillmann U, Kostopoulos P, Fassbender K, Herrmann W, 2009. Methylation status and neurodegenerative markers in Parkinson disease. Clin. Chem. 55, 1852–1860. [DOI] [PubMed] [Google Scholar]
  503. Ogawa O, Zhu X, Lee HG, Raina A, Obrenovich ME, Bowser R, Ghanbari HA, Castellani RJ, Perry G, Smith MA, 2003. Ectopic localization of phosphory-lated histone H3 in Alzheimer’s disease: a mitotic catastrophe? Acta Neuro- pathol. 105, 524–528. [DOI] [PubMed] [Google Scholar]
  504. Okano M, Bell DW, Haber DA, Li E, 1999. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257. [DOI] [PubMed] [Google Scholar]
  505. Okazawa H, 2003. Polyglutamine diseases: a transcription disorder? Cell. Mol. Life Sci. 60, 1427–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  506. Oliveira AM, Estevez MA, Hawk JD, Grimes S, Brindle PK, Abel T, 2011. Subregion-specific p300 conditional knock-out mice exhibit long-term memory impairments. Learn. Mem. 18, 161–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  507. Oliveira AM, Hemstedt TJ, Bading H, 2012. Rescue of aging-associated decline in Dnmt3a2 expression restores cognitive abilities. Nat. Neurosci. 15, 1111–1113. [DOI] [PubMed] [Google Scholar]
  508. Oliveira AM, Wood MA, McDonough CB, Abel T, 2007. Transgenic mice expressing an inhibitory truncated form of p300 exhibit long-term memory deficits. Learn. Mem. 14, 564–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  509. Olivieri F, Lazzarini R, Recchioni R, Marcheselli F, Rippo MR, Di Nuzzo S, Albertini MC, Graciotti L, Babini L, Mariotti S, Spada G, Abbatecola AM, Antonicelli R, Franceschi C, Procopio AD, 2013. MiR-146a as marker of senescence-associated pro-inflammatory status in cells involved in vascular remodelling. Age 35, 1157–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  510. Ooi SK, Bestor TH, 2008. The colorful history of active DNA demethylation. Cell 133, 1145–1148. [DOI] [PubMed] [Google Scholar]
  511. Ooi SK, Qiu C, Bernstein E, Li K, Jia D, Yang Z, Erdjument-Bromage H,Tempst P, Lin SP, Allis CD, Cheng X, Bestor TH, 2007. DNMT3L connects unmethy- lated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  512. Orr BA, Haffner MC, Nelson WG, Yegnasubramanian S, Eberhart CG, 2012. Decreased 5-hydroxymethylcytosine is associated with neural progenitor phenotype in normal brain and shorter survival in malignant glioma. PLoS ONE 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  513. Outeiro TF, Kontopoulos E, Altmann SM, Kufareva I, Strathearn KE, Amore AM, Volk CB, Maxwell MM, Rochet JC, McLean PJ, Young AB, Abagyan R, Feany MB, Hyman BT, Kazantsev AG, 2007. Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson’s disease. Science 317, 516–519. [DOI] [PubMed] [Google Scholar]
  514. Pan K, Chen Y, Roth M, Wang W, Wang S, Yee AS, Zhang X, 2013. HBP1-mediated transcriptional regulation of DNA methyltransferase 1 and its impact on cell senescence. Mol. Cell. Biol. 33, 887–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  515. Parra M, Mahmoudi T, Verdin E, 2007. Myosin phosphatase dephosphorylates HDAC7, controls its nucleocytoplasmic shuttling, and inhibits apoptosis in thymocytes. Genes Dev. 21, 638–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  516. Pasinetti GM, Ho L, Dooley C, Abbi B, Lange G, 2012. Select non-coding RNA in blood components provide novel clinically accessible biological surrogates for improved identification of traumatic brain injury in OEF/OIF veterans. Am. J. Neurodegener. Dis. 1, 88–98. [PMC free article] [PubMed] [Google Scholar]
  517. Patel N, Hoang D, Miller N, Ansaloni S, Huang QH, Rogers JT, Lee JC, Saunders AJ, 2008. MicroRNAs can regulate human APP levels. Mol. Neuro-degener. 3, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  518. Pauler FM, Sloane MA, Huang R, Regha K, Koerner MV, Tamir I, Sommer A, Aszodi A, Jenuwein T, Barlow DP, 2009. H3K27me3 forms BLOCs over silent genes and intergenic regions and specifies a histone banding pattern on a mouse autosomal chromosome. Genome Res. 19, 221–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  519. Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, Agis-Balboa RC, Cota P, Wittnam JL, Gogol-Doering A, Opitz L, 2010. Altered histone acetylation is associated with age-dependent memory impairment in mice. Science 328, 753–756. [DOI] [PubMed] [Google Scholar]
  520. Peng GS, Li G, Tzeng NS, Chen PS, Chuang DM, Hsu YD, Yang S, Hong JS, 2005. Valproate pretreatment protects dopaminergic neurons from LPS-in- duced neurotoxicity in rat primary midbrain cultures: role of microglia. Brain Res. Molecular Brain Res. 134, 162–169. [DOI] [PubMed] [Google Scholar]
  521. Perez M, Santa-Maria I, Barreda D, Gomez E, Zhu X, Cuadros R, Cabrero JR, Sanchez-Madrid F, Dawson HN, Vitek MP, 2009. Tau-an inhibitor of dea- cetylase HDAC6 function. J. Neurochem. 109, 1756–1766. [DOI] [PubMed] [Google Scholar]
  522. Perry G, Roder H, Nunomura A, Takeda A, Friedlich AL, Zhu X, Raina AK, Holbrook N, Siedlak SL, Harris PL, Smith MA, 1999. Activation of neuronal extracellular receptor kinase (ERK) in Alzheimer disease links oxidative stress to abnormal phosphorylation. Neuroreport 10, 2411–2415. [DOI] [PubMed] [Google Scholar]
  523. Perry M, Chalkley R, 1982. Histone acetylation increases the solubility of chromatin and occurs sequentially over most of the chromatin. A novel model for the biological role of histone acetylation. J. Biol. Chem. 257, 7336–7347. [PubMed] [Google Scholar]
  524. Perry RP, Kelley DE, Friderici K, Rottman F, 1975. The methylated constituents of L cell messenger RNA: evidence for an unusual cluster at the 5’ terminus. Cell 4, 387–394. [DOI] [PubMed] [Google Scholar]
  525. Persengiev S, Kondova I, Otting N, Koeppen AH, Bontrop RE, 2011. Genome- wide analysis of miRNA expression reveals a potential role for miR-144 in brain aging and spinocerebellar ataxia pathogenesis. Neurobiol. Aging 32,, 2316.e17–2316.e27. [DOI] [PubMed] [Google Scholar]
  526. Phalke S, Nickel O, Walluscheck D, Hortig F, Onorati MC, Reuter G, 2009. Retrotransposon silencing and telomere integrity in somatic cells of Drosophila depends on the cytosine-5 methyltransferase DNMT2. Nat. Genet. 41, 696–702. [DOI] [PubMed] [Google Scholar]
  527. Pidsley R, Mill J, 2011. Epigenetic studies of psychosis: current findings, methodological approaches, and implications for postmortem research. Biol. Psy-chiatr. 69, 146–156. [DOI] [PubMed] [Google Scholar]
  528. Pina B, Martinez P, Suau P, 1988. Differential acetylation of core histones in rat cerebral cortex neurons during development and aging. Eur. J. Biochem. 174, 311–315. [DOI] [PubMed] [Google Scholar]
  529. Ping X-L, Sun B-F, Wang L, Xiao W, Yang X, Wang W-J, Adhikari S, Shi Y, Lv Y, Chen Y-S, 2014. Mammalian WTAP is a regulatory subunit of the RNA N6- methyladenosine methyltransferase. Cell Res. 24, 177–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  530. Pirola CJ, Gianotti TF, Burgueno AL, Rey-Funes M, Loidl CF, Mallardi P, San Martino J, Castaflo GO, Sookoian S, 2012. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut 62, 1356–1363. [DOI] [PubMed] [Google Scholar]
  531. Pirooznia SK, Sarthi J, Johnson AA, Toth MS, Chiu K, Koduri S, Elefant F, 2012. Tip60 HAT activity mediates APP induced lethality and apoptotic cell death in the CNS of a Drosophila Alzheimer’s disease model. PLoS ONE 7, e41776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  532. Plagnol V, Nalls MA, Bras JM, Hernandez DG, Sharma M, Sheerin U-M, Saad M, Simon-Sanchez J, Schulte C, Lesage S, 2011. A two-stage meta-analysis identifies several new loci for Parkinson’s disease. PLoS Genet. 7, e1002142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  533. Podlisny MB, Tolan DR, Selkoe DJ, 1991. Homology ofthe amyloid beta protein precursor in monkey and human supports a primate model for beta amyloidosis in Alzheimer’s disease. Am. J. Pathol. 138, 1423–1435. [PMC free article] [PubMed] [Google Scholar]
  534. Politz JC, Hogan EM, Pederson T, 2009. MicroRNAs with a nucleolar location. RNA 15, 1705–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  535. Popp C, Dean W, Feng S, Cokus SJ, Andrews S, Pellegrini M, Jacobsen SE, Reik W, 2010. Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature 463, 1101–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  536. Portela A, Esteller M, 2010. Epigenetic modifications and human disease. Nat. Biotechnol. 28, 1057–1068. [DOI] [PubMed] [Google Scholar]
  537. Prado L, Lombo F, Brana A, Mendez C, Rohr J, Salas J, 1999. Analysis of two chromosomal regions adjacent to genes for a type II polyketide synthase involved in the biosynthesis of the antitumor polyketide mithramycin in Streptomyces argillaceus. MGG 261, 216–225. [DOI] [PubMed] [Google Scholar]
  538. Preker P, Nielsen J, Kammler S, Lykke-Andersen S, Christensen MS, Mapen-dano CK, Schierup MH, Jensen TH, 2008. RNA exosome depletion reveals transcription upstream of active human promoters. Science 322, 1851–1854. [DOI] [PubMed] [Google Scholar]
  539. Pruitt K, Zinn RL, Ohm JE, McGarvey KM, Kang SHL, Watkins DN, Herman JG, Baylin SB, 2006. Inhibition of SIRT1 reactivates silenced cancer genes without loss of promoter DNA hypermethylation. PLoS Genet. 2, 344–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  540. Przedborski S, Vila M, 2003. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model: a tool to explore the pathogenesis of Parkinson’s disease. Ann. N. Y. Acad. Sci. 991, 189–198. [PubMed] [Google Scholar]
  541. Qiang L, Fujita R, Yamashita T, Angulo S, Rhinn H, Rhee D, Doege C, Chau L, Aubry L, Vanti WB, 2011. Directed conversion ofAlzheimer’s disease patient skin fibroblasts into functional neurons. Cell 146, 359–371. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  542. Qing H, He G, Ly PT, Fox CJ, Staufenbiel M, Cai F, Zhang Z, Wei S, Sun X, Chen CH, Zhou W, Wang K, Song W, 2008. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse models. J. Exp. Med. 205, 2781–2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  543. Qu Z, Adelson DL, 2012. Evolutionary conservation and functional roles ofncRNA. Front. Genet. 3, 205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  544. Quintas A, de Solis AJ, Diez-Guerra FJ, Carrascosa JM, Bogonez E, 2012. Age-associated decrease of SIRT1 expression in rat hippocampus: prevention by late onset caloric restriction. Exp. Gerontol. 47, 198–201. [DOI] [PubMed] [Google Scholar]
  545. Raasch P, Schmitz U, Patenge N, Vera J, Kreikemeyer B, Wolkenhauer O, 2010. Non-coding RNA detection methods combined to improve usability, reproducibility and precision. BMC Bioinform. 11, 491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  546. Raber J, Wong D, Buttini M, Orth M, Bellosta S, Pitas RE, Mahley RW, Mucke L, 1998. Isoform-specific effects of human apolipoprotein E on brain function revealed in ApoE knockout mice: increased susceptibility offemales. Proc. Natl. Acad. Sci. U.S.A. 95, 10914–10919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  547. Rai K, Chidester S, Zavala CV, Manos EJ, James SR, Karpf AR, Jones DA, Cairns BR, 2007. Dnmt2 functions in the cytoplasm to promote liver, brain, and retina development in zebrafish. Genes Dev. 21, 261–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  548. Rai K, Huggins IJ, James SR, Karpf AR, Jones DA, Cairns BR, 2008. DNA demethylation in zebrafish involves the coupling ofa deaminase, a glycosylase, and gadd45. Cell 135, 1201–1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  549. Raiber EA, Beraldi D, Ficz G, Burgess HE, Branco MR, Murat P, Oxley D, Booth MJ, Reik W, Balasubramanian S, 2012. Genome-wide distribution of 5-formylcytosine in embryonic stem cells is associated with transcription and depends on thymine DNA glycosylase. Genome Biol. 13, R69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  550. Ralston SH, 1994. Pathogenesis and management of cancer associated hypercal- caemia. Cancer. Surv. 21, 179–196. [PubMed] [Google Scholar]
  551. Rao JS, Keleshian VL, Klein S, Rapoport SI, 2012. Epigenetic modifications in frontal cortex from Alzheimer’s disease and bipolar disorder patients. Transl. Psychiatry 2, e132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  552. Rauhala HE, Porkka KP, Saramaki OR, Tammela TL, Visakorpi T, 2008. Clusterin is epigenetically regulated in prostate cancer. Int. J. Cancer 123, 1601–1609. [DOI] [PubMed] [Google Scholar]
  553. Razin A, Riggs AD, 1980. DNA methylation and gene function. Science 210, 604–610. [DOI] [PubMed] [Google Scholar]
  554. Relton CL, Davey Smith G, 2012. Two-step epigenetic Mendelian randomization: a strategy for establishing the causal role of epigenetic processes in pathways to disease. Int. J. Epidemiol. 41, 161–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  555. Remington R, Chan A, Paskavitz J, Shea TB, 2009. Efficacy of a vitamin/nutri-ceutical formulation for moderate-stage to later-stage Alzheimer’s disease: a placebo-controlled pilot study. Am. J. Alzheimer’s Dis. Other Dement. 24, 27–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  556. Ricobaraza A, Cuadrado-Tejedor M, Marco S, Perez-Otano I, Garcia-Osta A, 2012. Phenylbutyrate rescues dendritic spine loss associated with memory deficits in a mouse model of Alzheimer disease. Hippocampus 22,1040–1050. [DOI] [PubMed] [Google Scholar]
  557. Ricobaraza A, Cuadrado-Tejedor M, Perez-Mediavilla A, Frechilla D, Del Rio J, Garcia-Osta A, 2009. Phenylbutyrate ameliorates cognitive deficit and reduces tau pathology in an Alzheimer’s disease mouse model. Neuropsychopharmacol. 34, 1721–1732. [DOI] [PubMed] [Google Scholar]
  558. Riedel O, Klotsche J, Spottke A, Deuschl G, Forstl H, Henn F, Heuser I, Oertel W, Reichmann H, Riederer P, Trenkwalder C, Dodel R, Wittchen HU, 2008. Cognitive impairment in 873 patients with idiopathic Parkinson’s disease. Results from the German Study on Epidemiology of Parkinson’s Disease with Dementia (GEPAD). J. Neurol. 255, 255–264. [DOI] [PubMed] [Google Scholar]
  559. Ristow M, Zarse K, 2010. How increased oxidative stress promotes longevity and metabolic health: the concept of mitochondrial hormesis (mitohormesis). Exp. Gerontol. 45, 410–418. [DOI] [PubMed] [Google Scholar]
  560. Robertson KD, Jones PA, 1998. The human ARF cell cycle regulatory gene promoter is a CpG island which can be silenced by DNA methylation and down-regulated by wild-type p53. Mol. Cell. Biol. 18, 6457–6473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  561. Rodriguez-Lebron E, Denovan-Wright EM, Nash K, Lewin AS, Mandel RJ, 2005. Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice. Mol. Ther. 12, 618–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  562. Ronan JL, Wu W, Crabtree GR, 2013. From neural development to cognition: unexpected roles for chromatin. Nat. Rev. Genet. 14, 347–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  563. Rosato RR, Grant S, 2005. Histone deacetylase inhibitors: insights into mechanisms of lethality. Expert Opin. Ther. Targets 9, 809–824. [DOI] [PubMed] [Google Scholar]
  564. Ross CA, 1997. Intranuclear neuronal inclusions: a common pathogenic mechanism for glutamine-repeat neurodegenerative diseases? Neuron 19, 1147–1150. [DOI] [PubMed] [Google Scholar]
  565. Ross CA, Poirier MA, 2004. Protein aggregation and neurodegenerative disease. Nat. Med. 10 Suppl., S10–S17. [DOI] [PubMed] [Google Scholar]
  566. Rossi LE, Avila DE, Spallanzani RG, Ziblat A, Fuertes MB, Lapyckyj L, Croci DO, Rabinovich GA, Domaica CI, Zwirner NW, 2012. Histone deacetylase inhibitors impair NK cell viability and effector functions through inhibition of activation and receptor expression. J. Leukocyte Biol. 91, 321–331. [DOI] [PubMed] [Google Scholar]
  567. Rottman F, Shatkin AJ, Perry RP, 1974. Sequences containing methylated nucleotides at the 5’ termini of messenger RNAs: possible implications for processing. Cell 3, 197–199. [DOI] [PubMed] [Google Scholar]
  568. Roy A, Ghosh A, Jana A, Liu X, Brahmachari S, Gendelman HE, Pahan K, 2012. Sodium phenylbutyrate controls neuroinflammatory and antioxidant activities and protects dopaminergic neurons in mouse models of Parkinson’s disease. PLoS ONE 7, e38113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  569. Rutten BP, Brasnjevic I, Steinbusch HW, Schmitz C, 2010. Caloric restriction and aging but not overexpression of SOD1 affect hippocampal volumes in mice. Mech. Ageing Dev. 131, 574–579. [DOI] [PubMed] [Google Scholar]
  570. Ryan WG, 1977. Treatment of Paget’s disease of bone with mithramycin. Clin. Orthop. Relat. Res. 127, 106–110. [PubMed] [Google Scholar]
  571. Ryan JM, Cristofalo VJ, 1972. Histone acetylation during aging of human cells in culture. Biochem. Biophys. Res. Commun. 48, 735–742. [DOI] [PubMed] [Google Scholar]
  572. Ryu H, Barrup M, Kowall NW, McKee AC, 2008. P3–260: epigenetic modification in a monozygotic twin with Alzheimer’s disease. Alzheimer’s Dement. 4, T598. [Google Scholar]
  573. Ryu H, Lee J, Hagerty SW, Soh BY, McAlpin SE, Cormier KA, Smith KM, Ferrante RJ, 2006. ESET/SETDB1 gene expression and histone H3 (K9) tri- methylation in Huntington’s disease. Proc. Natl. Acad. Sci. U.S.A. 103, 19176–19181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  574. Ryu H, Rosas HD, Hersch SM, Ferrante RJ, 2005. The therapeutic role of creatine in Huntington’s disease. Pharmacol. Ther. 108, 193–207. [DOI] [PubMed] [Google Scholar]
  575. Sadri-Vakili G, Bouzou B, Benn CL, Kim MO, Chawla P, Overland RP, Glajch KE, Xia E, Qiu Z, Hersch SM, Clark TW, Yohrling GJ, Cha JH, 2007. Histones associated with downregulated genes are hypo-acetylated in Huntington’s disease models. Hum. Mol. Genet. 16, 1293–1306. [DOI] [PubMed] [Google Scholar]
  576. Saikia M, Fu Y, Pavon-Eternod M, He CA, Pan T, 2010. Genome-wide analysis of N-1-methyl-adenosine modification in human tRNAs. RNA 16, 1317–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  577. Saito T, Matsuba Y, Mihira N, Takano J, Nilsson P, Itohara S, Iwata N, Saido TC, 2014. Single App knock-in mouse models of Alzheimer’s disease. Nat. Neurosci. 17, 661–663. [DOI] [PubMed] [Google Scholar]
  578. Saito Y, Jones PA, 2006. Epigenetic activation of tumor suppressor microRNAs in human cancer cells. Cell Cycle 5, 2220–2222. [DOI] [PubMed] [Google Scholar]
  579. Salminen A, Tapiola T, Korhonen P, Suuronen T, 1998. Neuronal apoptosis induced by histone deacetylase inhibitors. Brain Res. Molecular Brain Res. 61, 203–206. [DOI] [PubMed] [Google Scholar]
  580. Santosh B, Varshney A, Yadava PK, 2014. Non-coding RNAs: biological functions and applications. Cell Biochem. Funct. 33, 14–22. [DOI] [PubMed] [Google Scholar]
  581. Saraiya AA, Wang CC, 2008. snoRNA, a novel precursor of microRNA in Giardia lamblia. PLoS Pathog. 4, e1000224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  582. Sarg B, Koutzamani E, Helliger W, Rundquist I, Lindner HH, 2002. Postsynthetic trimethylation of histone H4 at lysine 20 in mammalian tissues is associated with aging. J. Biol. Chem. 277, 39195–39201. [DOI] [PubMed] [Google Scholar]
  583. Sarthi J, Elefant F, 2011. dTip60 HAT activity controls synaptic bouton expansion at the Drosophila neuromuscular junction. PLoS ONE 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  584. Sasaki T, Maier B, Bartke A, Scrable H, 2006. Progressive loss of S1RT1 with cell cycle withdrawal. Aging cell 5, 413–422. [DOI] [PubMed] [Google Scholar]
  585. Sawicka A, Seiser C, 2012. Histone H3 phosphorylation - a versatile chromatin modification for different occasions. Biochimie 94, 2193–2201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  586. Sawyer SL, Emerman M, Malik HS, 2004. Ancient adaptive evolution of the primate antiviral DNA-editing enzyme APOBEC3G. PLoS Biol. 2, E275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  587. Scarpa S, Fuso A, D’Anselmi F, Cavallaro RA, 2003. Presenilin 1 gene silencing by S-adenosylmethionine: a treatment for Alzheimer disease? FEBS Lett. 541, 145–148. [DOI] [PubMed] [Google Scholar]
  588. Schaefer M, Hagemann S, Hanna K, Lyko F, 2009. Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Cancer Res. 69, 8127–8132. [DOI] [PubMed] [Google Scholar]
  589. Schipper HM, Maes OC, Chertkow HM, Wang E, 2007. MicroRNA expression in Alzheimer blood mononuclear cells. Gene Regul. Syst. Biol. 1, 263–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  590. Schmeisser K, Mansfeld J, Kuhlow D, Weimer S, Priebe S, Heiland I, Birringer M, Groth M, Segref A, Kanfi Y, Price NL, Schmeisser S, Schuster S, Pfeiffer AFH, Guthke R, Platzer M, Hoppe T, Cohen HY, Zarse K, Sinclair DA, Ristow M, 2013. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat. Chem. Biol. 9, 693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  591. Schmitz KM, Schmitt N, Hoffmann-Rohrer U, Schafer A, Grummt I, Mayer C, 2009. TAF12 recruits Gadd45a and the nucleotide excision repair complex to the promoter of rRNA genes leading to active DNA demethylation. Mol. Cell 33, 344–353. [DOI] [PubMed] [Google Scholar]
  592. Schonrock N, Humphreys DT, Preiss T, Gotz J, 2012. Target Gene Repression Mediated by miRNAs miR-181c and miR-9 Both of Which Are Down-regulated by Amyloid-beta. J. Mol. Neurosci. 46, 324–335. [DOI] [PubMed] [Google Scholar]
  593. Schouten M, Buijink MR, Lucassen PJ, Fitzsimons CP, 2012. New neurons in agingbrains: molecular control by small non-codingRNAs. Front. Neurosci. 6, 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  594. Schrijvers EM, Koudstaal PJ, Hofman A, Breteler MM, 2011. Plasma clusterin and the risk of Alzheimer disease. JAMA 305, 1322–1326. [DOI] [PubMed] [Google Scholar]
  595. Schumann GG, 2007. APOBEC3 proteins: major players in intracellular defence against LINE-1-mediated retrotransposition. Biochem. Soc. Trans. 35, 637–642. [DOI] [PubMed] [Google Scholar]
  596. Sebastiani P, Montano M, Puca A, Solovieff N, Kojima T, Wang MC, Melista E, Meltzer M, Fischer SE, Andersen S, Hartley SH, Sedgewick A, Arai Y, Bergman A, Barzilai N, Terry DF, Riva A, Anselmi CV, Malovini A, Kitamoto A, Sawabe M, Arai T, Gondo Y, Steinberg MH, Hirose N, Atzmon G, Ruvkun G, Baldwin CT, Perls TT, 2009. RNA editing genes associated with extreme old age in humans and with lifespan in C. elegans. PLoS ONE 4, e8210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  597. Seila AC, Calabrese JM, Levine SS, Yeo GW, Rahl PB, Flynn RA, Young RA, Sharp PA, 2008. Divergent transcription from active promoters. Science 322, 1849–1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  598. Selkoe DJ, 1993. Physiological production of the beta-amyloid protein and the mechanism of Alzheimer’s disease. Trends Neurosci. 16, 403–409. [DOI] [PubMed] [Google Scholar]
  599. Selkoe DJ, 2012. Preventing Alzheimer’s disease. Science 337, 1488–1492. [DOI] [PubMed] [Google Scholar]
  600. Serot JM, Christmann D, Dubost T, Bene MC, Faure GC, 2001. CSF-folate levels are decreased in late-onset AD patients. J. Neural Transm. 108, 93–99. [DOI] [PubMed] [Google Scholar]
  601. Sethi P, Lukiw WJ, 2009. Micro-RNA abundance and stability in human brain: specific alterations in Alzheimer’s disease temporal lobe neocortex. Neurosci. Lett. 459, 100–104. [DOI] [PubMed] [Google Scholar]
  602. Sharma RP, 2012. Blood chromatin as a biosensor of the epigenetic milieu: a tool for studies in living psychiatric patients. Epigenomics-UK 4, 551–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  603. Sharma RP, Tun N, Grayson DR, 2008. Depolarization induces downregulation of DNMT1 and DNMT3a in primary cortical cultures. Epigenetics 3, 74–80. [DOI] [PubMed] [Google Scholar]
  604. Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K, Tsuda T, Mar L, Foncin JF, Bruni AC, Montesi MP, Sorbi S, Rainero I, Pinessi L, Nee L, Chumakov I, Pollen D, Brookes A, Sanseau P, Polinsky RJ, Wasco W, Da Silva HA, Haines JL, Perkicak-Vance MA, Tanzi RE, Roses AD, Fraser PE, Rommens JM, St George-Hyslop PH, 1995. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 375, 754–760. [DOI] [PubMed] [Google Scholar]
  605. Shiio Y, Eisenman RN, 2003. Histone sumoylation is associated with transcriptional repression. Proc. Natl. Acad. Sci. U.S.A. 100, 13225–13230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  606. Shimba S, Bokar JA, Rottman F, Reddy R, 1995. Accurate and efficient N-6- adenosine methylation in spliceosomal U6 small nuclear RNA by HeLa cell extract in vitro. Nucleic Acids Res. 23, 2421–2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  607. Shmookler Reis RJ, Goldstein S, 1983. Mitochondrial DNA in mortal and immortal human cells, Genome number, integrity, and methylation. J. Biol. Chem. 258, 9078–9085. [PubMed] [Google Scholar]
  608. Shock LS, Thakkar PV, Peterson EJ, Moran RG, Taylor SM, 2011. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc. Natl. Acad. Sci. U.S.A. 108, 3630–3635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  609. Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, McKay DM, Tintner R, Frangione B, et al. , 1992. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science 258, 126–129. [DOI] [PubMed] [Google Scholar]
  610. Siddiqui A, Chinta SJ, Mallajosyula JK, Rajagopolan S, Hanson I, Rane A, Melov S, Andersen JK, 2012. Selective binding of nuclear alpha-synuclein to the PGC1alpha promoter under conditions of oxidative stress may contribute to losses in mitochondrial function: implications for Parkinson’s disease. Free Radic. Biol. Med. 53, 993–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  611. Siegmund KD, Connor CM, Campan M, Long TI, Weisenberger DJ, Biniszkie- wicz D, Jaenisch R, Laird PW, Akbarian S, 2007. DNA methylation in the human cerebral cortex is dynamically regulated throughout the life span and involves differentiated neurons. PLoS ONE 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  612. Silva PN, Gigek CO, Leal MF, Bertolucci PH, de Labio RW, Payao SL, Smith Mde A, 2008. Promoter methylation analysis of S1RT3, SMARCA5, HTERT and CDH1 genes in aging and Alzheimer’s disease. J. Alzheimers Dis. 13, 173–176. [DOI] [PubMed] [Google Scholar]
  613. Simoes-Pires C, Zwick V, Nurisso A, Schenker E, Carrupt PA, Cuendet M, Cronin-Furman E, Borland M, Bergquist K, Bennett J, 2013. HDAC6 as a target for neurodegenerative diseases: what makes it different from the other HDACs? Mol. Neurodegener. 8, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  614. Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan- Ruiz C, Lichtner P, Scholz SW, Hernandez DG, Kruger R, Federoff M, Klein C, Goate A, Perlmutter J, Bonin M, Nalls MA, Illig T, Gieger C, Houlden H, Steffens M, Okun MS, Racette BA, Cookson MR, Foote KD, Fernandez HH,Traynor BJ, Schreiber S, Arepalli S, Zonozi R, Gwinn K, van der Brug M, Lopez G, Chanock SJ, Schatzkin A, Park Y, Hollenbeck A, Gao J, Huang X, Wood NW, Lorenz D, Deuschl G, Chen H, Riess O, Hardy JA, Singleton AB, Gasser T, 2009. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat. Genet. 41, 1308–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  615. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K, 2003. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 302, 841. [DOI] [PubMed] [Google Scholar]
  616. Sleegers K, Lambert JC, Bertram L, Cruts M, Amouyel P, Van Broeckhoven C, 2010. The pursuit of susceptibility genes for Alzheimer’s disease: progress and prospects. Trends Genet. 26, 84–93. [DOI] [PubMed] [Google Scholar]
  617. Slooter AJ, Cruts M, Kalmijn S, Hofman A, Breteler MM, Van Broeckhoven C, van Duijn CM, 1998. Risk estimates of dementia by apolipoprotein E genotypes from a population-based incidence study: the Rotterdam Study. Arch. Neurol. 55, 964–968. [DOI] [PubMed] [Google Scholar]
  618. Smith P, Al Hashimi A, Girard J, Delay C, Hebert SS, 2011a. In vivo regulation of amyloid precursor protein neuronal splicing by microRNAs. J. Neurochem. 116, 240–247. [DOI] [PubMed] [Google Scholar]
  619. Smith PY, Delay C, Girard J, Papon M-A, Planel E, Sergeant N, Buee L, Hebert SS, 2011b. MicroRNA-132 loss is associated with tau exon 10 inclusion in progressive supranuclear palsy. Hum. Mol. Genet. 20, 4016–4024. [DOI] [PubMed] [Google Scholar]
  620. Smith WW, Pei Z, Jiang H, Dawson VL, Dawson TM, Ross CA, 2006. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat. Neurosci. 9, 1231–1233. [DOI] [PubMed] [Google Scholar]
  621. Smrt RD, Szulwach KE, Pfeiffer RL, Li X, Guo W, Pathania M, Teng ZQ, Luo Y, Peng J, Bordey A, Jin P, Zhao X, 2010. MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase mind bomb-1. Stem Cells 28, 1060–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  622. So K, Tamura G, Honda T, Homma N, Endoh M, Togawa N, Nishizuka S, Motoyama T, 2006. Quantitative assessment of RUNX3 methylation in neoplastic and non-neoplastic gastric epithelia using a DNA microarray. Pathol. Int. 56, 571–575. [DOI] [PubMed] [Google Scholar]
  623. Somel M, Guo S, Fu N, Yan Z, Hu HY, Xu Y, Yuan Y, Ning Z, Hu Y, Menzel C, Hu H, Lachmann M, Zeng R, Chen W, Khaitovich P, 2010. MicroRNA, mRNA, and protein expression link development and aging in human and macaque brain. Genome Res. 20, 1207–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  624. Sommer G, Kralisch S, Lipfert J, Weise S, Krause K, Jessnitzer B, Lossner U, Blüher M, Stumvoll M, Fasshauer M, 2009. Amyloid precursor protein expression is induced by tumor necrosis factor a in 3T3-L1 adipocytes. J. Cell. Biochem. 108, 1418–1422. [DOI] [PubMed] [Google Scholar]
  625. Sommer M, Poliak N, Upadhyay S, Ratovitski E, Nelkin BD, Donehower LA, Sidransky D, 2006. DeltaNp63alpha overexpression induces downregulation of Sirt1 and an accelerated aging phenotype in the mouse. Cell Cycle 5, 2005–2011. [DOI] [PubMed] [Google Scholar]
  626. Song C-X, Szulwach KE, Dai Q, Fu Y, Mao S-Q, Lin L, Street C, Li Y, Poidevin M, Wu H, 2013. Genome-wide profiling of 5-formylcytosine reveals its roles in epigenetic priming. Cell 153, 678–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  627. Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG, 2010. Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration. Mol. Pharmacol. 77, 621–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  628. Song C, Kanthasamy A, Jin H, Anantharam V, Kanthasamy AG, 2011a. Paraquat induces epigenetic changes by promoting histone acetylation in cell culture models of dopaminergic degeneration. Neurotoxicology 32, 586–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  629. Song CX, Szulwach KE, Fu Y, Dai Q, Yi C, Li X, Li Y, Chen CH, Zhang W, Jian X, Wang J, Zhang L, Looney TJ, Zhang B, Godley LA, Hicks LM, Lahn BT, Jin P, He C, 2011b. Selective chemical labeling reveals the genome-wide distribution of 5-hydroxymethylcytosine. Nat. Biotechnol. 29, 68–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  630. Sonntag K-C, 2010. MicroRNAs and deregulated gene expression networks in neurodegeneration. Brain Res. 1338, 48–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  631. Sontag E, Nunbhakdi-Craig V, Sontag JM, Diaz-Arrastia R, Ogris E, Dayal S, Lentz SR, Arning E, Bottiglieri T, 2007. Protein phosphatase 2A methyl- transferase links homocysteine metabolism with tau and amyloid precursor protein regulation. J. Neurosci. 27, 2751–2759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  632. Sparks DL, Scheff SW, Hunsaker JC 3rd, Liu H, Landers T, Gross DR, 1994. Induction of Alzheimer-like beta-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol. Exp. Neurol. 126, 88–94. [DOI] [PubMed] [Google Scholar]
  633. St Laurent R, O’Brien LM, Ahmad ST, 2013. Sodium butyrate improves locomotor impairment and early mortality in a rotenone-induced Drosophila model of Parkinson’s disease. Neuroscience 246, 382–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  634. Stack EC, Del Signore SJ, Luthi-Carter R, Soh BY, Goldstein DR, Matson S, Goodrich S, Markey AL, Cormier K, Hagerty SW, Smith K, Ryu H, Ferrante RJ, 2007. Modulation of nucleosome dynamics in Huntington’s disease. Hum. Mol. Genet. 16, 1164–1175. [DOI] [PubMed] [Google Scholar]
  635. Stan AD, Ghose S, Gao XM, Roberts RC, Lewis-Amezcua K, Hatanpaa KJ, Tamminga CA, 2006. Human postmortem tissue: what quality markers matter? Brain Res. 1123, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  636. Steffan JS, Bodai L, Pallos J, Poelman M, McCampbell A, Apostol BL, Kazantsev A, Schmidt E, Zhu YZ, Greenwald M, Kurokawa R, Housman DE, Jackson GR, Marsh JL, Thompson LM, 2001. Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature 413, 739–743. [DOI] [PubMed] [Google Scholar]
  637. Stilling RM, Benito E, Gertig M, Barth J, Capece V, Burkhardt S, Bonn S, Fischer A, 2014. De-regulation of gene expression and alternative splicing affects distinct cellular pathways in the aging hippocampus. Front. Cell. Neurosci. 8, 373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  638. Strosznajder JB, Czapski GA, Adamczyk A, Strosznajder RP, 2012. Poly(ADP- ribose) polymerase-1 in amyloid beta toxicity and Alzheimer’s disease. Mol. Neurobiol. 46, 78–84. [DOI] [PubMed] [Google Scholar]
  639. Su Y, Ryder J, Li B, Wu X, Fox N, Solenberg P, Brune K, Paul S, Zhou Y, Liu F, Ni B, 2004. Lithium, a common drug for bipolar disorder treatment, regulates amyloid-beta precursor protein processing. Biochemistry 43, 6899–6908. [DOI] [PubMed] [Google Scholar]
  640. Suarez-Quian CA, Goldstein SR, Pohida T, Smith PD, Peterson JI, Wellner E, Ghany M, Bonner RF, 1999. Laser capture microdissection of single cells from complex tissues. BioTechniques 26, 328–335. [DOI] [PubMed] [Google Scholar]
  641. Sugai F, Yamamoto Y, Miyaguchi K, Zhou Z, Sumi H, Hamasaki T, Goto M, Sakoda S, 2004. Benefit of valproic acid in suppressing disease progression of ALS model mice. Eur. J. Neurosci. 20, 3179–3183. [DOI] [PubMed] [Google Scholar]
  642. Sugars KL, Rubinsztein DC, 2003. Transcriptional abnormalities in Huntington disease. Trends Genet. 19, 233–238. [DOI] [PubMed] [Google Scholar]
  643. Sun G, Yu RT, Evans RM, Shi Y, 2007. Orphan nuclear receptor TLX recruits histone deacetylases to repress transcription and regulate neural stem cell proliferation. Proc. Natl. Acad. Sci. U.S.A. 104, 15282–15287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  644. Sun Z, Terragni J, Borgaro JG, Liu Y, Yu L, Guan S, Wang H, Sun D, Cheng X, Zhu Z, Pradhan S, Zheng Y, 2013. High-resolution enzymatic mapping of genomic 5-hydroxymethylcytosine in mouse embryonic stem cells. Cell Rep. 3, 567–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  645. Suuronen T, Nuutinen T, Ryhanen T, Kaarniranta K, Salminen A, 2007. Epigenetic regulation of clusterin/apolipoprotein J expression in retinal pigment epithelial cells. Biochem. Biophys. Res. Commun. 357, 397–401. [DOI] [PubMed] [Google Scholar]
  646. Suzuki MM, Bird A, 2008. DNA methylation landscapes: provocative insights from epigenomics. Nat. Rev. Genet. 9, 465–476. [DOI] [PubMed] [Google Scholar]
  647. Szebenyi G, Morfini GA, Babcock A, Gould M, Selkoe K, Stenoien DL, Young M, Faber PW, MacDonald ME, McPhaul MJ, Brady ST, 2003. Neuropatho- genic forms of huntingtin and androgen receptor inhibit fast axonal transport. Neuron 40, 41–52. [DOI] [PubMed] [Google Scholar]
  648. Szulwach KE, Li XK, Smrt RD, Li YJ, Luo YP, Lin L, Santistevan NJ, Li WD, Zhao XY, Jin P, 2010. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J. Cell. Biol. 189, 127–U181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  649. Szyf M, 2014. Epigenetics, a key for unlocking complex CNS disorders? Therapeutic implications. Eur. Neuropsychopharmacol. 25, 682–702. [DOI] [PubMed] [Google Scholar]
  650. Taft RJ, Glazov EA, Lassmann T, Hayashizaki Y, Carninci P, Mattick JS, 2009. Small RNAs derived from snoRNAs. RNA 15, 1233–1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  651. Taganov KD, Boldin MP, Chang K-J, Baltimore D, 2006. NF-kB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. U.S.A. 103, 12481–12486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  652. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S, 2007. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872. [DOI] [PubMed] [Google Scholar]
  653. Tan MJ, Luo H, Lee S, Jin FL, Yang JS, Montellier E, Buchou T, Cheng ZY, Rousseaux S, Rajagopal N, Lu ZK, Ye Z, Zhu Q, Wysocka J, Ye Y, Khochbin S, Ren B, Zhao YM, 2011. Identification of 67 Histone Marks and Histone Lysine Crotonylation as a New Type of Histone Modification. Cell 146, 1015–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  654. Tanaka T, Weisblum B, 1975. Systematic difference in the methylation of ribo- somal ribonucleic acid from gram-positive and gram-negative bacteria. J. Bacteriol. 123, 771–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  655. Tanzi RE, 2012. The genetics of Alzheimer disease. Cold Spring Harb. Perspect. Med. 2, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  656. Tanzi RE, Kovacs DM, Kim TW, Moir RD, Guenette SY, Wasco W, 1996. The gene defects responsible for familial Alzheimer’s disease. Neurobiol. Dis. 3, 159–168. [DOI] [PubMed] [Google Scholar]
  657. Thakur MK, Kanungo MS, 1981. Methylation of chromosomal proteins and DNA of rat brain and its modulation by estradiol and calcium during aging. Exp. Gerontol. 16, 331–336. [DOI] [PubMed] [Google Scholar]
  658. Thambisetty M, Simmons A, Velayudhan L, Hye A, Campbell J, Zhang Y, Wahlund LO, Westman E, Kinsey A, Guntert A, Proitsi P, Powell J, Causevic M, Killick R, Lunnon K, Lynham S, Broadstock M, Choudhry F, Howlett DR, Williams RJ, Sharp SI, Mitchelmore C, Tunnard C, Leung R, Foy C, O’Brien D, Breen G, Furney SJ, Ward M, Kloszewska I, Mecocci P, Soininen H, Tsolaki M, Vellas B, Hodges A, Murphy DG, Parkins S, Richardson JC, Resnick SM, Ferrucci L, Wong DF, Zhou Y, Muehlboeck S, Evans A, Francis PT, Spenger C, Lovestone S, 2010. Association ofplasma clusterin concentration with severity, pathology, and progression in Alzheimer disease. Arch. Gen. Psychiatry 67, 739–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  659. The University of Manchester, 2014. miRBase, pp. 21. [Google Scholar]
  660. Thomas B, Beal MF, 2011. Molecular insights into Parkinson’s disease. F1000 Med. Rep. 3, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  661. Thomas B, Matson S, Chopra V, Sun L, Sharma S, Hersch S, Rosas HD, Scherzer C, Ferrante R, Matson W, 2013. A novel method for detecting 7- methyl guanine reveals aberrant methylation levels in Huntington disease. Anal. Biochem. 436, 112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  662. Thomas EA, 2006. Striatal specificity of gene expression dysregulation in Huntington’s disease. J. Neurosci. Res. 84, 1151–1164. [DOI] [PubMed] [Google Scholar]
  663. Thomas EA, Coppola G, Desplats PA, Tang B, Soragni E, Burnett R, Gao F, Fitzgerald KM, Borok JF, Herman D, Geschwind DH, Gottesfeld JM, 2008. The HDAC inhibitor 4b ameliorates the disease phenotype and transcriptional abnormalities in Huntington’s disease transgenic mice. Proc. Natl. Acad. Sci. U.S.A. 105, 15564–15569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  664. Thompson RF, Atzmon G, Gheorghe C, Liang HQ, Lowes C, Greally JM, Barzilai N, 2010. Tissue-specific dysregulation of DNA methylation in aging. Aging cell 9, 506–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  665. Tian C, Liu Q, Ma K, Wang Y, Chen Q, Ambroz R, Klinkebiel DL, Li Y, Huang Y, Ding J, Wu J, Zheng JC, 2013. Characterization of induced neural progenitors from skin fibroblasts by a novel combination of defined factors. Sci. Rep. 3, 1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  666. Tian F, Marini AM, Lipsky RH, 2010. Effects of histone deacetylase inhibitor Trichostatin A on epigenetic changes and transcriptional activation of Bdnf promoter 1 by rat hippocampal neurons. Ann. N. Y. Acad. Sci. 1199, 186–193. [DOI] [PubMed] [Google Scholar]
  667. Tiraboschi P, Hansen LA, Thal LJ, Corey-Bloom J, 2004. The importance of neuritic plaques and tangles to the development and evolution of AD. Neurology 62, 1984–1989. [DOI] [PubMed] [Google Scholar]
  668. Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Ukitsu M, Genda Y, 1999. The methylation status of cytosines in a tau gene promoter region alters with age to downregulate transcriptional activity in human cerebral cortex. Neurosci. Lett. 275, 89–92. [DOI] [PubMed] [Google Scholar]
  669. Trapp BD, Nave KA, 2008. Multiple sclerosis: an immune or neurodegenerative disorder? Annu. Rev. Neurosci. 31, 247–269. [DOI] [PubMed] [Google Scholar]
  670. Trapp J, Meier R, Hongwiset D, Kassack MU, Sippl W,Jung M, 2007. Structure- activity studies on suramin analogues as inhibitors of NAD + -dependent his- tone deacetylases (sirtuins). Chem. Med. Chem. 2, 1419–1431. [DOI] [PubMed] [Google Scholar]
  671. Trojanowski JQ, Goedert M, Iwatsubo T, Lee VM, 1998. Fatal attractions: abnormal protein aggregation and neuron death in Parkinson’s disease and Lewy body dementia. Cell Death Differ. 5, 832–837. [DOI] [PubMed] [Google Scholar]
  672. Trushina E, Dyer RB, Badger JD 2nd, Ure D, Eide L, Tran DD, Vrieze BT, Legendre-Guillemin V, McPherson PS, Mandavilli BS, Van Houten B, Zeitlin S,McNiven M, Aebersold R, Hayden M, Parisi JE, Seeberg E, Dragatsis I, Doyle K, Bender A, Chacko C, McMurray CT, 2004. Mutant huntingtin impairs axonal trafficking in mammalian neurons in vivo and in vitro. Mol. Cell. Biol. 24, 8195–8209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  673. Tschöp K, Engeland K, 2007. Cell cycle-dependent transcription of cyclin B2 is influenced by DNA methylation but is independent of methylation in the CDE and CHR elements. FEBS J. 274, 5235–5249. [DOI] [PubMed] [Google Scholar]
  674. Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, Tsukamoto AS, Gage FH, Weissman IL, 2000. Direct isolation of human central nervous system stem cells. Proc. Natl. Acad. Sci. U.S.A. 97, 14720–14725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  675. Ueda K, Omachi A, Kawaichi M, Hayaishi O, 1975. Natural occurrence of poly(ADP-ribosyl) histones in rat liver. Proc. Natl. Acad. Sci. U.S.A. 72, 205–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  676. Umylny B, Presting G, Efird JT, Klimovitsky BI, Ward WS, 2007. Most human Alu and murine B1 repeats are unique. J. Cell Biochem. 102, 110–121. [DOI] [PubMed] [Google Scholar]
  677. Urdinguio RG, Sanchez-Mut JV, Esteller M, 2009. Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies. Lancet Neurol. 8, 1056–1072. [DOI] [PubMed] [Google Scholar]
  678. Valente L, Nishikura K, 2005. ADAR gene family and A-to-I RNA editing: diverse roles in posttranscriptional gene regulation. Prog. Nucleic Acid Res. Mol. Biol. 79, 299–338. [DOI] [PubMed] [Google Scholar]
  679. van de Berg WD, Hepp DH, Dijkstra AA, Rozemuller JA, Berendse HW, Foncke E, 2012. Patterns of alpha-synuclein pathology in incidental cases and clinical subtypes of Parkinson’s disease. Parkinsonism Relat. Disord. 18 (Suppl. 1), S28–S30. [DOI] [PubMed] [Google Scholar]
  680. van den Hove DL, Chouliaras L, Rutten BP, 2012. The role of 5-hydroxymethyl- cytosine in aging and Alzheimer’s disease: current status and prospects for future studies. Curr. Alzheimer Res. 9, 545–549. [DOI] [PubMed] [Google Scholar]
  681. van den Hove DLA, Kompotis K, Lardenoije R, Kenis G, Mill J, Steinbusch HW, Lesch K-P, de Strooper B, Fitzsimons CP, Rutten BPF, 2014. Epigenetically regulated microRNAs in Alzheimer’s disease. Neurobiol. Aging 35, 731–745. [DOI] [PubMed] [Google Scholar]
  682. Vaquero A, Scher M, Erdjument-Bromage H, Tempst P, Serrano L, Reinberg D, 2007. SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature 450, 440–444. [DOI] [PubMed] [Google Scholar]
  683. Vasa-Nicotera M, Chen H, Tucci P, Yang AL, Saintigny G, Menghini R, Mahe C, Agostini M, Knight RA, Melino G, 2011. miR-146a is modulated in human endothelial cell with aging. Atherosclerosis 217, 326–330. [DOI] [PubMed] [Google Scholar]
  684. Vecsey CG, Hawk JD, Lattal KM, Stein JM, Fabian SA, Attner MA, Cabrera SM, McDonough CB, Brindle PK, Abel T, Wood MA, 2007. Histone dea- cetylase inhibitors enhance memory and synaptic plasticity via CREB:CBP- dependent transcriptional activation. J. Neurosci. 27, 6128–6140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  685. Vega RB, Matsuda K, Oh J, Barbosa AC, Yang XL, Meadows E, McAnally J, Pomajzl C, Shelton JM, Richardson JA, Karsenty G, Olson EN, 2004. Histone deacetylase 4 controls chondrocyte hypertrophy during skeletogenesis. Cell 119, 555–566. [DOI] [PubMed] [Google Scholar]
  686. Veldman BA, Wijn AM, Knoers N, Praamstra P, Horstink MW, 1998. Genetic and environmental risk factors in Parkinson’s disease. Clin. Neurol. Neurosurg. 100, 15–26. [DOI] [PubMed] [Google Scholar]
  687. Verma A, Verma N, 2011. Induced pluripotent stem cells and promises of neuroregenerative medicine. Neurol. India 59, 555–557. [DOI] [PubMed] [Google Scholar]
  688. Villar-Menendez I, Blanch M, Tyebji S, Pereira-Veiga T, Albasanz JL, Martin M, Ferrer I, Perez-Navarro E, Barrachina M, 2013. Increased 5-methylcytosine and decreased 5-hydroxymethylcytosine levels are associated with reduced striatal A2AR levels in Huntington’s disease. Neuromol. Med. 15, 295–309. [DOI] [PubMed] [Google Scholar]
  689. Vogel-Ciernia A, Matheos DP, Barrett RM, Kramär EA, Azzawi S, Chen Y, Magnan CN, Zeller M, Sylvain A, Haettig J, 2013. The neuron-specific chromatin regulatory subunit BAF53b is necessary for synaptic plasticity and memory. Nat. Neurosci. 16, 552–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  690. Vogel-Ciernia A, Wood MA, 2014. Neuron-specific chromatin remodeling: a missing link in epigenetic mechanisms underlying synaptic plasticity, memory, and intellectual disability disorders. Neuropharmacology 80, 18–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  691. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr., 1985. Neuropathological classification of Huntington’s disease. J. Neuropathol. Exp. Neurol. 44, 559–577. [DOI] [PubMed] [Google Scholar]
  692. Voutsinas GE, Stavrou EF, Karousos G, Dasoula A, Papachatzopoulou A, Syrrou M, Verkerk AJ, van der Spek P, Patrinos GP, Stöger R, 2010. Allelic imbalance of expression and epigenetic regulation within the alpha-synuclein wild-type and p. Ala53Thr alleles in Parkinson disease. Hum. Mutat. 31, 685–691. [DOI] [PubMed] [Google Scholar]
  693. Walker MP, LaFerla FM, Oddo SS, Brewer GJ, 2013. Reversible epigenetic histone modifications and Bdnf expression in neurons with aging and from a mouse model of Alzheimer’s disease. Age 35, 519–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  694. Wang CM, Tsai SN, Yew TW, Kwan YW, Ngai SM, 2010a. Identification of histone methylation multiplicities patterns in the brain of senescence-accelerated prone mouse 8. Biogerontology 11, 87–102. [DOI] [PubMed] [Google Scholar]
  695. Wang G, van der Walt JM, Mayhew G, Li YJ, Zuchner S, Scott WK, Martin ER, Vance JM, 2008a. Variation in the miRNA-433 binding site of FGF20 confers risk for Parkinson disease by overexpression of alpha-synuclein. Am. J. Hum. Genet. 82, 283–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  696. Wang H, Liu J, Zong Y, Xu Y, Deng W, Zhu H, Liu Y, Ma C, Huang L, Zhang L, Qin C, 2010b. miR-106b aberrantly expressed in a double transgenic mouse model for Alzheimer’s disease targets TGF-beta type II receptor. Brain Res. 1357, 166–174. [DOI] [PubMed] [Google Scholar]
  697. Wang J, Yu JT, Tan MS, Jiang T, Tan L, 2013. Epigenetic mechanisms in Alzheimer’s disease: implications for pathogenesis and therapy. Ageing Res. Rev. 12, 1024–1041. [DOI] [PubMed] [Google Scholar]
  698. Wang SC, Oelze B, Schumacher A, 2008b. Age-specific epigenetic drift in late- onset Alzheimer’s disease. PLoS ONE 3, e2698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  699. Wang W-X, Rajeev BW, Stromberg AJ, Ren N, Tang G, Huang Q, Rigoutsos I, Nelson PT, 2008c. The expression of microRNA miR-107 decreases early in Alzheimer’s disease and may accelerate disease progression through regulation of β-site amyloid precursor protein-cleaving enzyme 1. J. Neurosci. 28, 1213–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  700. Wang WX, Huang Q, Hu Y, Stromberg AJ, Nelson PT, 2011. Patterns of microRNA expression in normal and early Alzheimer’s disease human temporal cortex: white matter versus gray matter. Acta Neuropathol. 121, 193–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  701. Wang WX, Wilfred BR, Madathil SK, Tang G, Hu Y, Dimayuga J, Stromberg AJ, Huang Q, Saatman KE, Nelson PT, 2010c. miR-107 regulates granulin/ progranulin with implications for traumatic brain injury and neurodegenerative disease. Am. J. Pathol. 177, 334–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  702. Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, Fu Y, Parisien M, Dai Q, Jia G, Ren B, Pan T, He C, 2014a. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  703. Wang X, Nagl NG, Wilsker D, Van Scoy M, Pacchione S, Yaciuk P, Dallas PB, Moran E, 2004. Two related ARID family proteins are alternative subunits of human SWI/SNF complexes. Biochem. J. 383, 319–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  704. Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z, Zhao JC, 2014b. N6-methyla- denosine modification destabilizes developmental regulators in embryonic stem cells. Nat. Cell. Biol. 16, 191–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  705. Wang Z, Kai L, Day M, Ronesi J, Yin HH, Ding J, Tkatch T, Lovinger DM, Surmeier DJ, 2006. Dopaminergic control of corticostriatal long-term synaptic depression in medium spiny neurons is mediated by cholinergic interneurons. Neuron 50, 443–452. [DOI] [PubMed] [Google Scholar]
  706. Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney MJ, 2004. Epigenetic programming by maternal behavior. Nat. Neurosci. 7, 847–854. [DOI] [PubMed] [Google Scholar]
  707. Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW, 1975. A protein factor essential for microtubule assembly. Proc. Natl. Acad. Sci. U.S.A. 72, 1858–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  708. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB, 2009. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  709. Wenk GL, 2003. Neuropathologic changes in Alzheimer’s disease. J. Clin. Psychiatry 64 (Suppl. 9), 7–10. [PubMed] [Google Scholar]
  710. Werner A, 2005. Natural antisense transcripts. RNA Biol. 2, 53–62. [DOI] [PubMed] [Google Scholar]
  711. West RL, Lee JM, Maroun LE, 1995. Hypomethylation of the amyloid precursor protein gene in the brain of an Alzheimer’s disease patient. J. Mol. Neurosci. 6, 141–146. [DOI] [PubMed] [Google Scholar]
  712. Westberry JM, Trout AL, Wilson ME, 2011. Epigenetic regulation of estrogen receptor beta expression in the rat cortex during aging. Neuroreport 22, 428–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  713. Whittemore SR, Sanon HR, Wood PM, 1993. Concurrent isolation and characterization of oligodendrocytes, microglia and astrocytes from adult human spinal cord. Int. J. Dev. Neurosci.: Off. J. Int. Soc. Dev. Neurosci. 11, 755–764. [DOI] [PubMed] [Google Scholar]
  714. Wilson AG, 2008. Epigenetic regulation of gene expression in the inflammatory response and relevance to common diseases. J. Periodontol. 79, 1514–1519. [DOI] [PubMed] [Google Scholar]
  715. Wojda U, Kuznicki J, 2013. Alzheimer’s disease modeling: ups, downs, and perspectives for human induced pluripotent stem cells. J. Alzheimers Dis. 34, 563–588. [DOI] [PubMed] [Google Scholar]
  716. Wood H, 2013. Neurodegenerative disease: altered DNA methylation and RNA splicing could be key mechanisms in Huntington disease. Nat. Rev. Neurol. 9, 119. [DOI] [PubMed] [Google Scholar]
  717. Wood MA, Attner MA, Oliveira AMM, Brindle PK, Abel T, 2006. A transcription factor-binding domain of the coactivator CBP is essential for longterm memory and the expression of specific target genes. Learn. Mem. 13, 609–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  718. Wu H, Coskun V, Tao J, Xie W, Ge W, Yoshikawa K, Li E, Zhang Y, Sun YE, 2010. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 329, 444–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  719. Wu X, Chen PS, Dallas S, Wilson B, Block ML, Wang CC, Kinyamu H, Lu N, Gao X, Leng Y, Chuang DM, Zhang W, Lu RB, Hong JS, 2008. Histone deacetylase inhibitors up-regulate astrocyte GDNF and BDNF gene transcription and protect dopaminergic neurons. Int. J. Neuropsychopharmacol. 11, 1123–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  720. Xiong M, Zhang T, Zhang LM, Lu SD, Huang YL, Sun FY, 2008. Caspase inhibition attenuates accumulation of beta-amyloid by reducing beta-secretase production and activity in rat brains after stroke. Neurobiol. Dis. 32, 433–441. [DOI] [PubMed] [Google Scholar]
  721. Xiong Y, Zhao K, Wu J, Xu Z, Jin S, Zhang YQ, 2013. HDAC6 mutations rescue human tau-induced microtubule defects in Drosophila. Proc. Natl. Acad. Sci. U.S.A. 110, 4604–4609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  722. Xu T, Li L, Huang C, Li X, Peng Y, Li J, 2014. MicroRNA-323–3p with clinical potential in rheumatoid arthritis. Alzheimer’s disease and ectopic pregnancy. Expert Opin. Ther. Targets 18, 153–158. [DOI] [PubMed] [Google Scholar]
  723. Xu WS, Parmigiani RB, Marks PA, 2007. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26, 5541–5552. [DOI] [PubMed] [Google Scholar]
  724. Xu Z, Li H, Jin P, 2012. Epigenetics-based therapeutics for neurodegenerative disorders. Curr. Transl. Geriatr. Exp. Gerontol. Rep. 1, 229–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  725. Yamada K, Mizuno M, Nabeshima T, 2002. Role for brain-derived neurotrophic factor in learning and memory. Life Sci. 70, 735–744. [DOI] [PubMed] [Google Scholar]
  726. Yamagata K, Urakami K, Ikeda K, Ji Y, Adachi Y, Arai H, Sasaki H, Sato K, Nakashima K, 2001. High expression of apolipoprotein E mRNA in the brains with sporadic Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 12, 57–62. [DOI] [PubMed] [Google Scholar]
  727. Yao J, Hennessey T, Flynt A, Lai E, Beal MF, Lin MT, 2010. MicroRNA-related cofilin abnormality in Alzheimer’s disease. PLoS ONE 5, e15546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  728. Ye X, Zerlanko B, Zhang R, Somaiah N, Lipinski M, Salomoni P, Adams PD, 2007. Definition of pRB- and p53-dependent and -independent steps in HIRA/ ASF1a-mediated formation of senescence-associated heterochromatin foci. Mol. Cell. Biol. 27, 2452–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  729. Yoon SY, Choi HI, Choi JE, Sul CA, Choi JM, Kim DH, 2007. Methotrexate decreases PP2A methylation and increases tau phosphorylation in neuron. Biochem. Biophys. Res. Commun. 363, 811–816. [DOI] [PubMed] [Google Scholar]
  730. Yu D, Pendergraff H, Liu J, Kordasiewicz HB, Cleveland DW, Swayze EE, Lima WF, Crooke ST, Prakash TP, Corey DR, 2012. Single-stranded RNAs use RNAi to potently and allele-selectively inhibit mutant huntingtin expression. Cell 150, 895–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  731. Yu JT, Tan L, 2012. The role of clusterin in Alzheimer’s disease: pathways, pathogenesis, and therapy. Mol. Neurobiol. 45, 314–326. [DOI] [PubMed] [Google Scholar]
  732. Yu S, Zhang JZ, Zhao CL, Zhang HY, Xu Q, 2004. Isolation and characterization of the CD133+ precursors from the ventricular zone of human fetal brain by magnetic affinity cell sorting. Biotechnol. Lett. 26, 1131–1136. [DOI] [PubMed] [Google Scholar]
  733. Zeng Y, Yi R, Cullen BR, 2003. MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc. Natl. Acad. Sci. U.S.A. 100, 9779–9784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  734. Zhang CE, Tian Q, Wei W, Peng JH, Liu GP, Zhou XW, Wang Q, Wang DW, Wang JZ, 2008. Homocysteine induces tau phosphorylation by inactivating protein phosphatase 2A in rat hippocampus. Neurobiol. Aging 29,1654–1665. [DOI] [PubMed] [Google Scholar]
  735. Zhang CL, McKinsey TA, Chang SR, Antos CL, Hill JA, Olson EN, 2002. Class I! histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell 110, 479–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  736. Zhang K, Schrag M, Crofton A, Trivedi R, Vinters H, Kirsch W, 2012. Targeted proteomics for quantification of histone acetylation in Alzheimer’s disease. Proteomics 12, 1261–1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  737. Zhang Y, Friedlander R, 2011. Using non-coding small RNAs to develop therapies for Huntington’s disease. Gene Ther. 18, 1139–1149. [DOI] [PubMed] [Google Scholar]
  738. Zhang Y, Li M, Drozda M, Chen M, Ren S, Mejia Sanchez RO, Leavitt BR, Cattaneo E, Ferrante RJ, Hayden MR, Friedlander RM, 2003. Depletion of wild-type huntingtin in mouse models ofneurologic diseases. J. Neurochem. 87, 101–106. [DOI] [PubMed] [Google Scholar]
  739. Zhang Y, Reinberg D, 2001. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 15, 2343–2360. [DOI] [PubMed] [Google Scholar]
  740. Zhang ZY, Schluesener HJ, 2013. Oral administration of histone deacetylase inhibitor MS-275 ameliorates neuroinflammation and cerebral amyloidosis and improves behavior in a mouse model. J. Neuropathol. Exp. Neurol. 72, 178–185. [DOI] [PubMed] [Google Scholar]
  741. Zhao C, Sun G, Li S, Shi Y, 2009. A feedback regulatory loop involving microRNA- 9 and nuclear receptor TLX in neural stem cell fate determination. Nat. Struct. Mol. Biol. 16, 365–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  742. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Grunblatt E, Moran LB, Mandel SA, Riederer P, Miller RM, Federoff HJ, Wullner U, Papapetropoulos S, Youdim MB, Cantuti-Castelvetri I, Young AB, Vance JM, Davis RL, Hedreen JC, Adler CH, Beach TG, Graeber MB, Middleton FA, Rochet JC, Scherzer CR, Global PDGEC, 2010. PGC-1alpha, a potential therapeutic target for early intervention in Parkinson’s disease. Sci. Transl. Med. 2, 52ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  743. Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang C-M, Li CJ, Vagbo CB, Shi Y, Wang W-L, Song S-H, 2013. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  744. Zhou W, Bercury K, Cummiskey J, Luong N, Lebin J, Freed CR, 2011. Phenylbutyrate up-regulates the DJ-1 protein and protects neurons in cell culture and in animal models of Parkinson disease. J. Biol. Chem. 286,14941–14951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  745. Zhou XW, Gustafsson JA, Tanila H, Bjorkdahl C, Liu R, Winblad B, Pei JJ, 2008. Tau hyperphosphorylation correlates with reduced methylation of protein phosphatase 2A. Neurobiol. Dis. 31, 386–394. [DOI] [PubMed] [Google Scholar]
  746. Zhu B, Zheng Y, Angliker H, Schwarz S, Thiry S, Siegmann M, Jost J-P, 2000a. 5-Methylcytosine DNA glycosylase activity is also present in the human MBD4 (G/T mismatch glycosylase) and in a related avian sequence. Nucleic Acids Res 28, 4157–4165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  747. Zhu B, Zheng Y, Hess D, Angliker H, Schwarz S, Siegmann M, Thiry S, Jost J-P, 2000b. 5-methylcytosine-DNA glycosylase activity is present in a cloned G/T mismatch DNA glycosylase associated with the chicken embryo DNA demeth- ylation complex. Proc. Natl. Acad. Sci. U.S.A. 97, 5135–5139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  748. Zhu HC, Wang LM, Wang M, Song B, Tan S, Teng JF, Duan DX, 2012. MicroRNA-195 downregulates Alzheimer’s disease amyloid-beta production by targeting BACE1. Brain Res. Bull. 88, 596–601. [DOI] [PubMed] [Google Scholar]
  749. Zhu M, Li WW, Lu CZ, 2014. Histone decacetylase inhibitors prevent mitochondrial fragmentation and elicit early neuroprotection against MPP+. CNS Neurosci. Therapeut. 20, 308–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  750. Zhu X, Castellani RJ, Takeda A, Nunomura A, Atwood CS, Perry G, Smith MA, 2001. Differential activation of neuronal ERK, JNK/SAPK and p38 in Alzheimer disease: the ‘two hit’ hypothesis. Mech. Ageing Dev. 123, 39–46. [DOI] [PubMed] [Google Scholar]
  751. Ziller MJ, Gu H, Muller F, Donaghey J, Tsai LT, Kohlbacher O, De Jager PL, Rosen ED, Bennett DA, Bernstein BE, Gnirke A, Meissner A, 2013. Charting a dynamic DNA methylation landscape of the human genome. Nature 500, 477–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  752. Zong Y, Wang H, Dong W, Quan X, Zhu H, Xu Y, Huang L, Ma C, Qin C, 2011. miR-29c regulates BACE1 protein expression. Brain Res. 1395, 108–115. [DOI] [PubMed] [Google Scholar]
  753. Zovoilis A, Agbemenyah HY, Agis-Balboa RC, Stilling RM, Edbauer D, Rao P, Farinelli L, Delalle I, Schmitt A, Falkai P, 2011. microRNA-34c is a novel target to treat dementias. EMBO J. 30, 4299–4308. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES