Skip to main content

Targeting Huntington’s disease through histone deacetylases

Abstract

Huntington’s disease (HD) is a debilitating neurodegenerative condition with significant burdens on both patient and healthcare costs. Despite extensive research, treatment options for patients with this condition remain limited. Aberrant post-translational modification (PTM) of proteins is emerging as an important element in the pathogenesis of HD. These PTMs include acetylation, phosphorylation, methylation, sumoylation and ubiquitination. Several families of proteins are involved with the regulation of these PTMs. In this review, I discuss the current evidence linking aberrant PTMs and/or aberrant regulation of the cellular machinery regulating these PTMs to HD pathogenesis. Finally, I discuss the evidence suggesting that pharmacologically targeting one of these protein families the histone deacetylases may be of potential therapeutic benefit in the treatment of HD.

Introduction

Huntington’s disease (HD) is an autosomal dominant, late-onset neurodegenerative disease characterized by cognitive dysfunction, psychiatric symptoms and movement disorders (Walker 2007). The disease is caused by an expansion of a polyglutamine repeat within the amino terminus of the predominantly cytosolic protein huntingtin (Htt; DiFiglia et al. 1995; Group 1993). The expanded repeat region results in nuclear translocation and aggregation of huntingtin and has been implicated as the causative event in the pathogenesis of this disease (Klement et al. 1998; Sieradzan et al. 1999; Saudou et al. 1998).

Epigenetic regulation of gene expression involves stable and heritable changes in gene expression which are not due to changes in the primary DNA sequence. Current known epigenetic mechanisms involve the following: DNA CpG methylation, histone post-translational modifications (PTMs), gene imprinting and non-coding RNA.

The ‘histone code’ is a well-established hypothesis describing the idea that specific patterns of post-translational modifications to histones act like a molecular ‘code’ recognised and used by non-histone proteins to regulate specific chromatin functions. These modifications include acetylation, methylation, phosphorylation, sumoylation and ubiquitination, and various families of proteins have been identified which function to place or remove these PTMs. The best studied of these families are the K-acetyltransferases (KATs), histone deacetylases (HDACs), K-methyltransferases (KMTs) and K-demethylases (KDMs; Allis et al. 2007).

KATs (formerly known as either histone acetyltransferases or lysine acetyltransferases; Allis et al. 2007) function to covalently add acetyl groups to lysine residues on proteins. Likewise, KMTs add methyl groups to lysine residues either as mono-, di- or tri-methylation (Albert and Helin 2010), whilst HDACs and KDMs remove these respective modifications (Allis et al. 2007). Whilst many of these PTMs were originally identified on histones, it is becoming increasingly evident that these PTMs play important roles on many proteins leading to the notion of a ‘protein code’ exemplified by the proteins p53 (Sims and Reinberg 2008) and nuclear factor kappa-B (NFkB; Calao et al. 2008). Indeed, the Htt protein has been found to be extensively modified by several PTMs including acetylation, sumoylation, ubiquitination and phosphorylation (Table 1). Focusing on Huntington’s disease (HD), I discuss the current knowledge supporting the notion that the proteins involved with histone/protein PTMs play important regulatory roles in the pathogenesis of this disease and discuss whether inhibitors which target HDACs may have potential for use in targeting this disease.

Table 1 Sites in the Htt protein which undergo PTMs

Linking histone PTM machinery with HD

Lysine acetylation/deacetylation in HD

Much evidence has emerged directly linking KATs and HDACs with HD (Table 2). For instance, mutant Htt protein was quickly shown to form a complex with the lysine acetyltransferases KAT3A (CBP) and KAT2B (P/CAF; Nucifora et al. 2001; Steffan et al. 2001, 2000; Kazantsev et al. 1999). Once this complex is established, it in turn can then interact with p53 and repress transcription of p21WAF1/CIP1 and MDR-1 (Steffan et al. 2000). Evidence has also accumulated linking functional associations between huntingtin and HDACs. Huntingtin associates with the co-repressors mSin3a and N-CoR which recruit HDACs to repress transcription (Boutell et al. 1999; Steffan et al. 2000). Indeed in the absence or presence of ligand, mutant huntingtin has been shown to both increase nuclear co-repressor function and enhance ligand-dependent nuclear hormone receptor activation (Yohrling et al. 2003).

Table 2 Histone modifying proteins linked to HD discussed within the text

The potential importance of histone deacetylases in HD was highlighted in a Caenorhabditis elegans model of HD, where specifically targeting HDACs reduced neurodegeneration (Bates et al. 2006). Defects in microtubule-based transport have been shown to contribute to neuronal toxicity in Huntington’s disease (Muchowski et al. 2002; Hoffner et al. 2002; Trushina et al. 2003). HDAC6 has been shown to be a microtubule-associated deacetylase (Hubbert et al. 2002). Levels of acetylated tubulin have been found to be reduced in the brains of Huntington’s patients, and in vitro cell studies targeting HDAC6 resulted in the alleviation of transport- and release-defect phenotypes in the cell models via increased acetylation at lysine 40 of alpha-tubulin (Dompierre et al. 2007).

Critically, acetylation of Htt has been shown to target the mutant protein to autophagosomes for degradation. Increased acetylation at a single lysine (K444) was recently shown to facilitate the trafficking of mutant Htt into autophagosomes and significantly improved the clearance of the mutant protein by macroautophagy. In experimental models, this reversed the toxic effects of mutant huntingtin in primary striatal and cortical neurons and in a transgenic C. elegans model of HD. If the protein was altered to be resistant to acetylation, this resulted in dramatic aggregation leading to neurodegeneration in cultured neurons and in mouse brain (Jeong et al. 2009). Overexpression of I-kappa-B kinase (IKK)β, an important regulator of the NFkB transcription factor (Mankan et al. 2009), results in acetylation of Htt at K9 and phosphorylation at S13, leading to elevated nuclear translocation of Htt protein (Thompson et al. 2009).

Lysine ubiquitination/sumoylation in HD

The mutant huntingtin protein has been shown to alter levels of histone monoubiquitination, through an altered ability to interact with Bmi-1, a component of the hPRC1L E3 ubiquitin ligase complex, resulting in increased levels of monoubiquitinated histone H2A (uH2A) and aberrant gene expression (Kim et al. 2008). When gene expression patterns were examined in the brains of transgenic R6/2 mice, promoters of genes which were repressed were found to have increased levels of ubiquitinated H2A (uH2A) and decreased levels of ubiquitinated H2B (uH2B), whilst active promoters had the opposite (increased u2H2B and decreased uH2A). Furthermore, targeting histone ubiquitin in cell line models demonstrated that reducing uH2A led to the reactivation of repressed genes associated with a reduction in levels of histone lysine 9 methylation (H3K9me)2 and trimethylated histone H3 lysine 9 (H3K9me3) at the reactivated promoters. Conversely reductions of uH2B induced transcriptional repression through inhibition of monomethylation at histone H3 lysine 4 (Fig. 1; Kim et al. 2008).

Fig. 1
figure 1

Overview of epigenetic regulators affecting gene expression in Huntington’s disease. p phosphorylation, u ubiquitination, s sumoylation, ac acetylation, me methylation, ups ubiquitin/proteasome system, KAT K-acetyltransferase, KMT K-methyltransferase

Modification of proteins with polyubiquitin chains regulates many essential cellular processes including protein degradation, cell cycle, transcription, DNA repair and membrane trafficking. As discussed in previous sections, in Huntington’s disease, monoubiquitination of histones results in aberrant gene expression patterns. Evidence now links Htt ubiquitination to HD pathogenesis. Aggregation-prone proteins such as Htt have been suggested to overwhelm and impair the ubiquitin/proteasome system (UPS) in polyglutamine (polyQ) disorders. One study found that accumulation of ubiquitin conjugates in a HD disease model occurred without impairment of the ubiquitin/proteasome system (Maynard et al. 2009). However, two other studies have shown that UPS dysfunction is a consistent feature of HD pathology with impaired UPS in the synapses of HD mice (Wang et al. 2008) and with polyubiquitination chains occurring on Htt lysines 11, 48 and 63 (Bennett et al. 2007).

Htt has also been shown to be either ubiquitinated or sumoylated at the same lysine residues (K6, K9 and K15). Sumoylation stabilizes Htt and reduces its ability to form aggregates, whilst enhancing transcriptional repression, and exacerbated neurodegeneration in a drosophila model of HD, whereas ubiquitination of these residues abrogated neurodegeneration in the same model (Steffan et al. 2004). More recently, a novel striatal protein, Rhes, has been shown to associate with Htt. This protein has now been shown to induce Htt sumoylation leading to neuronal cytotoxicity (Subramaniam et al. 2009).

Lysine methylation in HD

H3K9me3 levels are elevated in both HD patients and in R6/2 transgenic mice and are associated with increased amounts of KMT1E (also known as known as ESET/SETDB1; Table 2; Ryu et al. 2006). This enzyme methylates H3K9me3, and whilst this is generally considered to be a mark of transcriptional repression and heterochromatinization (Hublitz et al. 2009), it has also been associated with active transcription genes particularly in disease states such as cancer (Wiencke et al. 2008). Within the neuronal setting, monoallelic deletion of the lysine acetyltransferase KAT3A (CBP) results in the induction of KMT1E (ESET) with concomitant increased H3K9me3 in neurons (Lee et al. 2008), suggesting that KAT3A (CBP) may be a regulator of KMT1E (ESET) gene expression and raising the notion that increased KAT activity may be a way of targeting the aberrant KMT1E (ESET)-mediated H3K9me3 levels in patients with HD (Table 2).

Deubiquitination of H2B in yeast initiates the recruitment of a complex containing the kinase Ctk1. This kinase subsequently phosphorylates the RNA polymerase II (Pol II) C-terminal domain, an event essential for the subsequent recruitment of KMT3A (Set2/HYBP) lysine methyltransferase (Wyce et al. 2007). One of the functions of this KMT is to regulate RNA Pol II elongation (Krogan et al. 2003; Wyce et al. 2007; Li et al. 2003; Xiao et al. 2003; Schaft et al. 2003; Morris et al. 2005). However, KMT3A (Set2/HYPB)-mediated H3K36 methylation also functions to recruit histone deacetylase complexes to restore normal chromatin structure in the wake of elongating Pol II (Keogh et al. 2005; Lee and Shilatifard 2007). Indeed KMT3A would appear to be essential for all H3K36me3 during gene induction both in vitro (Edmunds et al. 2008) and in vivo (Yuan et al. 2009). The human homologue of this KMT was originally described as huntingtin interacting protein B (HYPB; Sun et al. 2005a), suggesting that KMT3A (Set2/HYPB) may coordinate histone methylation and transcriptional regulation and play critical roles in the pathogenesis of HD (Sun et al. 2005a).

The Htt protein has now been shown to associate with the Polycomb repressive complex 2 (Seong et al. 2010). EZH2 (also known as KMT6) is the catalytic subunit of Polycomb repressive complex 2 (PRC2) and is a highly conserved histone methyltransferase that targets lysine-27 of histone H3 (Cao et al. 2002; Kuzmichev et al. 2002). Mutant Htt was found to be associated with reduced histone H3K27 tri-methylation, whilst recombinant wild-type Htt was found to significantly enhance the histone H3K27 tri-methylase activity of reconstituted PRC2 in vitro (Seong et al. 2010). Whilst there is currently little evidence demonstrating aberrant KDM activity in the pathogenesis of neurodegeneration, it has recently been shown that a neurospecific splice variant of KDM1 (LSD1) exists which is dynamically regulated throughout cortical development and functions to modulate neurite morphogenesis (Zibetti et al. 2010).

Lysine phosphorylation

In the R6/2 transgenic mouse model of HD, nuclear activation of the mitogen-activated protein kinase/extracellular regulated kinase (ERK) failed to induce histone H3 phosphorylation, an expected response of nuclear ERK activation. It was subsequently found that this was due to a decrease in expression of mitogen- and stress-activated kinase-1 (MSK-1), a kinase downstream ERK, critically involved in H3 phosphorylation, a finding subsequently confirmed in the striatal neurons and postmortem brains of HD patients and suggesting that aberrant MSK-1 expression is involved with the transcriptional dysregulation and striatal degeneration in patients with HD (Roze et al. 2008).

Recently, it has been shown that phosphorylation of mutant Htt by the inflammatory kinase complex (IKK) regulates additional post-translational modifications, including Htt ubiquitination, sumoylation and acetylation, resulting in increased Htt nuclear localization, cleavage and clearance mediated by lysosomal-associated membrane protein 2A and Hsc70 (Thompson et al. 2009). In particular, phosphorylation of serines (S6/S13) promotes the degradation of Htt by both the proteasome and lysosomes. These modifications, however, also result in enhanced translocation of Htt to the nucleus, which may result in both aberrant gene expression and enhanced neurodegeneration over time (Thompson et al. 2009). However, phosphorylation of Htt on serine 421 (S421) can reduce nuclear accumulation of huntingtin fragments by (a) reducing huntingtin cleavage by caspase-6, (b) affecting the levels of full-length huntingtin and (c) altering its nuclear localization (Warby et al. 2009). Phosphorylation of serine 434 (S434) by CDK5 has also been found to reduce caspase-mediated Htt cleavage at alanine residue 513 (Luo et al. 2005). Additionally, phosphorylation of Htt may also directly regulate Htt function, as phosphorylation at S431 restores its axonal transport (Zala et al. 2008), whilst phosphorylation of Htt at S431 by Akt is critical to controlling the direction of vesicle movement in neurons (Colin et al. 2008). Furthermore, phosphorylation at Htt serines S1181 and S1201 are critical to HD pathogenesis. If phosphorylation is absent, this confers toxic properties to wild-type huntingtin in a p53-dependent manner in striatal neurons and accelerates neuronal death induced by DNA damage, whilst if present they prevent this toxicity (Anne et al. 2007).

Consequences of mutant Htt and aberrant PTMs in HD

Clear links between aberrant histone post-translational modifications and disease pathogenesis have been demonstrated, and one of the most extensively studied of these modifications is histone acetylation. Indeed, acetylation of histones in brain neuronal fractions was observed as early as 1975 (Sarkander et al. 1975), and it has now been demonstrated that HDACs play important roles in both neuron differentiation (Liu et al. 2008), expression of neuron-specific genes (Lakowski et al. 2006) and the regulation of diverse cues such as maternal grooming (Weaver et al. 2004) and addiction (Renthal et al. 2007). Class II and class III histone deacetylases play pivotal roles in both neuronal proliferation and differentiation (Horio et al. 2003; Kyrylenko et al. 2003; Ajamian et al. 2003; Hoshino et al. 2003). SIRT1 has also been shown to protect primary cultures of cerebral granule neurons from FOXO-induced cell death (Brunet et al. 2004).

Because Htt has been shown to associate with the cellular machinery responsible for regulating the histone code, it has been postulated that this may lead to aberrant gene expression in patients suffering from HD. Three potential mechanisms as to how this might occur have been identified: (1) mislocalization of KATs. The association of mutant Htt and KATs affects the nuclear localization of KATs in neuronal cells into intranuclear inclusions (Steffan et al. 2000; Nucifora et al. 2001). As such depletion of this KAT from its normal localization by this aggregation may result in aberrant transcriptional control; (2) direct inhibition of KATs. Cell-free assays have shown that mutant huntingtin protein can directly inhibit KAT activity (Steffan et al. 2001), indicating a direct interference mechanism; and (3) enhanced degradation of KATs. A cell line of mutant Htt protein found that this was associated with enhanced degradation of KAT3A (CBP) indicating that the mutant Htt may actively eliminate this KAT from the affected neurons (Jiang et al. 2003, 2006; Cong et al. 2005).

As a consequence, gene expression is altered in patients with HD. Microarray gene profiling studies on HD patients have identified a subset of upregulated mRNAs that can distinguish between controls, presymptomatic HD mutation carriers and symptomatic HD patients (Borovecki et al. 2005). Furthermore, in a microarray analysis of a yeast model of expanded polyglutamine tracts, the changes in gene expression profiles closely matched those of yeast strains deleted for components of the Spt/Ada/Gcn5 acetyltransferase lysine acetyltransferase complex (Hughes et al. 2001). Additional evidence for aberrant gene expression comes from transgenic mice models of HD. For example, histone H3 is hypo-acetylated at the promoters of downregulated genes in R6/2 transgenic mice (Sadri-Vakili et al. 2007; Stack et al. 2007), whilst in transgenic N171-82Q (82Q) and R6/2 HD mice, the reduced histone 3 lysine 9 acetylation is concomitant with increased H3K9me and associated gene expression changes (Stack et al. 2007). In the sections below, I shall describe in more detail some of the mechanisms involving histone modifying complexes by which aberrant gene expression occurs in HD.

PGC-1 alpha

It is well established that PPAR gamma co-activator 1 alpha (PGC-1alpha) functions as a transcriptional co-activator/co-repressor through associations with both KATs and HDACs to regulate expression of genes involved with mitochondrial energy and metabolism (Lawless et al. 2009c). PGC-1alpha has also been linked to the development and pathogenesis of HD. Small nucleotide polymorphisms (SNPs) studies have identified particular SNPs within intron-2 of the PGC-1alpha gene that are associated with a delay in age at onset of motor symptoms in patients with Huntington’s disease (Taherzadeh-Fard et al. 2009; Weydt et al. 2009). Another SNP study has identified a SNP (rs2970870) within the promoter associated with earlier onset of HD in the homozygous state and a SNP within the transcribed region (rs7665116) associated with a delayed onset (Che et al. 2011). Significantly reduced levels of PGC-1alpha occur in the muscle of HD patients and HD transgenic mice, whilst ‘knockdown’ of mutant Htt in myoblasts leads to increased PGC-1alpha (Chaturvedi et al. 2009). Reduced expression of PGC-1alpha target genes also is found in the striatum of HD patients and mouse models (Weydt et al. 2006). When PGC-1alpha knockout mice were crossbred with HD knockin mice, increased neurodegeneration of striatal neurons and motor abnormalities was found to occur. Ectopic expression of PGC-1alpha was able to partially ablate the toxic effects of mutant huntingtin in cultured striatal neurons, whilst lentiviral-mediated PGC-1alpha provided neuroprotection in the striatum of transgenic HD mice (Cui et al. 2006). When adipose stem cells were transplanted into the striatum of R6/2 HD mice, increased survival, attenuation of striatal neuron loss and reductions in levels of huntingtin aggregates were observed which correlated with elevated levels of PGC-1alpha (Lee et al. 2009).

HDAC/REST complexes

One of the best established mechanisms in the regulation of neuronal development relates to genes which are regulated by the repressor element-1 (RE1)/neuronal restrictive silencer element. A specific protein called repressor element silencing transcription factor (REST), also known as neuron-restrictive silencing factor (NRSF), binds to this element and prevents the expression of these genes in non-neuronal cells (Seth and Majzoub 2001; Belyaev et al. 2004). REST is a critical protein in development (Chen et al. 1998) and is essential for proper neuronal differentiation and maturation (Paquette et al. 2000; Abrajano et al. 2009a, b). A functional link to aberrant REST activity and selective loss of neurons has been shown in a study of Down’s syndrome (Bahn et al. 2002). In addition, a Nurr1/CoREST pathway in microglia and astrocytes has been shown to protect neurons from inflammation-induced death in Parkinson’s disease (Saijo et al. 2009).

REST contains two distinct repressor domains, one located at the N terminus and the other at the C terminus of the protein (Thiel et al. 1998; Tapia-Ramirez et al. 1997). Two distinct neuronal repressor complexes have now been isolated containing both REST and HDACs. The first of these complexes involves direct interactions with mSin3A/B at the N terminus which then recruits HDACs to repress gene expression (Grimes et al. 2000; Huang et al. 1999; Naruse et al. 1999; Roopra et al. 2000). The C-terminal repression domain associates with a novel protein called CoREST (Andres et al. 1999). This protein also interacts with HDACs through its SANT domain and has been shown to be essential for repression (You et al. 2001) and in particular actively represses genes essential for neuronal phenotype (Ballas et al. 2005). The ATP-dependent remodelling complex SWI/SNF also plays a role in REST-mediated neuronal gene regulation as CoREST recruits several SWI/SNF members, indicating that active chromatin remodelling is an element in REST-mediated repression (Battaglioli et al. 2002; Watanabe et al. 2006). CoREST complexes have also been shown to contain lysine demethylases, and recently, a KDM1–CoREST–CtBP co-repressor complex was shown to be required for late cell-lineage determination and differentiation during pituitary organogenesis (Wang et al. 2007a).

In HD, levels of a microRNA miR-9/miR-9A are downregulated. This microRNA has been identified as a critical regulator of REST/NRSF (Packer et al. 2008), and loss of this miRNA results in increased levels of nuclear REST/NRSF activity (Bithell et al. 2009). As REST/NRSF has been shown to associate with Htt (Zuccato et al. 2003), KMTs and HDACs (Gray 2009), this may result in aberrant regulation of gene expression. Indeed, in this context, REST/NRSF has been shown to regulate several miRNAs (Conaco et al. 2006; Wu and Xie 2006). Because nuclear REST/NRSF activity is increased in HD patients as a consequence of loss of miR-9/miR-9A, this results in the further loss of expression of a series of miRNAs regulated by REST/NRSF in HD patients (Bithell et al. 2009).

For instance, a novel non-coding RNA human accelerated region 1, specifically transcribed in the nervous system, is regulated by REST/NRSF and has been shown to be downregulated in HD patients (Johnson et al. 2010). Expression of another miRNA, mIR-22, has been shown to be downregulated in two transgenic mouse models of HD (YAC128 and R6/2; Lee et al. 2011). This miRNA has recently been shown to be involved with the regulation of HDAC4 in hepatocellular carcinoma, in that cancers with low levels of this miRNA had elevated levels of HDAC4 (Zhang et al. 2010). It must be noted that in the R6/2 HD mouse model, decreased levels of HDAC4 protein were observed in the cortex (Quinti et al. 2010).

Motor neuron genes modulated by HDACs

In addition to RE1/NSRE-directed repression of specific genes, motor neurons also express unique combinations of LIM-type homeodomain factors. These function to define motor column identity by regulating particular gene expression profiles (Sharma et al. 1998). LIM homeodomain transcription factors have been shown to be regulated through a complex containing HDACs (Bach et al. 1999), and as such motor neuron pathfinding may be guided through an HDAC-directed process.

HDACs within the neuronal setting of HD

E2F, HDACs and neuronal survival mechanisms

An essential feature for neuronal survival has also been linked to constitutive repression of E2F1 transcriptional activity through HDAC proteins (Boutillier et al. 2002). Elevated levels of E2F1 lead to neuronal apoptosis (Konishi and Bonni 2003; Hou et al. 2000) and enhanced immune cell proliferation (Wu et al. 2001), factors that could be deleterious in HD. Indeed, elevated levels of E2F have been found in the brains of patients suffering from HD (Pelegri et al. 2008). Subsequently, we demonstrated that HDAC inhibitors reduce levels of E2F class I proteins in vivo (Camelo et al. 2005), data which have been recapitulated by others in vitro (Suzuki et al. 2000; Said et al. 2001). These observations may therefore help to explain why HDAC inhibitors block immune cell proliferation (Dangond et al. 1998) and enhance neuronal survival (Ryu et al. 2003).

HDACs play important roles in stem cell neuronal differentiation

Increased neurogenesis has been observed in primary HD patient material (Curtis et al. 2005), and mutant Htt has been demonstrated to stimulate neuronal differentiation of embryonic and neural stem cells (Lorincz and Zawistowski 2009). HDACs play important roles in neuronal stem cell (NSC) differentiation. When Flag-tagged HDACs with reduced enzymatic activity were used within a ‘dominant negative’ setting in stem cell lines, Howard and colleagues found that mutant HDAC1 reduced neuron differentiation by 50% (Humphrey et al. 2007). During murine development, HDAC1 is expressed in neural stem cells/progenitors and glia, whereas HDAC2 is found in neural progenitors and is upregulated in post-mitotic neuroblasts and neurons, but not in fully differentiated glia (MacDonald and Roskams 2008). Using mice lacking HDAC1 or HDAC2, Olson and colleagues demonstrate that these deacetylases do not have an overt phenotype in neuronal progenitors. However, once neuron development is initiated, these mice develop severe hippocampal abnormalities, absence of cerebellar foliation, disorganization of cortical neurons and lethality by postnatal day 7, attributed to a failure of neuronal precursors to differentiate into mature neurons and excessive cell death (Montgomery et al. 2009). Prozorovski et al. (2008) have also linked the SIRT1 HDAC to differentiate neural progenitor cells. A direct role for histone deacetylases in the regulation of neural stem cell proliferation has been shown where the orphan nuclear receptor TLX, a critical regulator of stem cell proliferation, was found to associate with HDAC3 and HDAC5. Inhibition of HDAC activity or knockdown of HDAC expression led to marked induction of TLX target gene expression and dramatically reduced neural stem cell proliferation (Sun et al. 2007).

In vitro studies have also been shown to differentiate neuronal precursor cells. Trichostatin A (TSA) treatment of embryonic mouse NSCs increased neuronal differentiation of the NSCs and decreased astrocyte differentiation (Balasubramaniyan et al. 2006), whilst embryonic stem cells can also be induced to differentiate into neurons following inhibition of histone deacetylases (Yao et al. 2010). Furthermore, valproic acid has also been shown to promote neuronal differentiation by inducing proneural factors such as Ngn1, Math1 and p15 in association with H4 acetylation at their promoters indicating that the mechanism involved concerns inhibition of HDACs (Yu et al. 2009).

Inhibition of histone deacetylases in lineage-committed oligodendrocyte precursor cells activity acted as a priming event in the induction of developmental plasticity (Lyssiotis et al. 2007). A similar study examining the ability of oligodendrocyte progenitors to acquire the identity of myelin-expressing cells or choose alternative fates found that the activity of histone deacetylases was critical to these processes (Liu et al. 2007), and the transcription factor Yin Yang 1 was shown to be a critical regulator of oligodendrocyte progenitor differentiation, acting as a lineage-specific repressor of transcriptional inhibitors of myelin gene expression (Tcf4 and Id4), through the recruitment of histone deacetylase-1 to their promoters during oligodendrocyte differentiation (He et al. 2007).

REST is also critically involved with neural stem cell differentiation. Activation of REST is sufficient to cause neuronal differentiation (Su et al. 2004). REST complexes are able to both silence and repress neuronal genes in embryonic neural stem cells through the creation of chromatin environments that contain both repressive and active local epigenetic signatures (Greenway et al. 2007; Sun et al. 2005b; Ballas et al. 2005). Neurons treated with a combination of HDAC and transcription inhibitors display an acetylation and transcription-dependent increase in neural outgrowth, associated with KAT3A/KAt3B and KAT2B-dependent p53 acetylation (Gaub et al. 2010).

HDACs, ER stress and pro-inflammatory pathways in HD

Inflammation is a critical element associated with cancer (Grivennikov et al. 2010), diabetes (Lawless et al. 2009b), but also is increasingly linked to neurodegenerative disease (Esiri 2007). Endoplasmic reticulum (ER) stress is another cellular event which has also been implicated as a critical component in all these conditions (Lawless et al. 2009a). HDACs have been shown to be critical regulators of many important cellular processes, and evidence is emerging linking these enzymes in the regulation of inflammation and ER stress responses. Whilst HD is not primarily considered to be a pro-inflammatory disease as mouse models of HD show little evidence for the involvement of inflammation (Schwab et al. 2010), nevertheless it is well established that one central mediator of inflammation, NFkB, plays some role in HD pathogenesis. Over the sections below, I shall link HDACs to both of these important pathways within the context of HD.

HATs/HDACs, NFkB and HD

NFkB is a key mediator of inflammatory cascades, playing important roles in regulating NFkB activation (Lawless et al. 2009a; Mankan et al. 2009). NFkB typically consists of a heterodimeric protein comprised of a p50 and a p65(RelA) subunit. Initial studies identified lysine acetyltransferases KAT3A and KAT3B as key coactivators in regulating NFkB-driven gene expression (Gerritsen et al. 1997; Perkins et al. 1997; Wadgaonkar et al. 1999). A further lysine acetyltransferase KAT13A was also found to potentiate NFkB transactivation through interactions with p50 (Sirianni et al. 1998). It was then shown that the RelA/p65 subunit can associate with HDACs 1–3 to repress expression of NFkB-regulated genes as well as to control the induced level of expression of these genes (Ashburner et al. 2001; Hoberg et al. 2004, 2006). The histone deacetylase SIRT1 also regulates NFkB transactivation by deacetylating a critical lysine at position 310 of the RelA/p65 subunit thereby inhibiting transcription (Yeung et al. 2004). A critical regulator of NFkB activation is IKKα. NFkB transcription requires IKKα to phosphorylate silencing mediator for retinoic acid and thyroid hormone receptor which in turn stimulates the exchange of co-repressor for co-activator complexes. In the initial stages of NFkB activation, the phosphorylation event causes HDAC3 to be displaced from RelA/p65, allowing KAT3B to acetylate RelA/p65 (Hoberg et al. 2004, 2006). IL-6 and IL-8 are pro-inflammatory cytokines associated with responses to NFkB activation (Esposito et al. 2003; Dandona et al. 2004a, b), and NFkB utilises the lysine acetyltransferase activity of KAT3A/KAT3B to stimulate the transcription of these genes (Vanden Berghe et al. 1999).

The protein Daxx regulates NFkB activation by binding to a region which includes the major sites of acetylation mediated by KAT3A/KAT3B (Park et al. 2007). However, Daxx also directly associates with HDAC2 (Hollenbach et al. 2002) and so may represent a mechanism by which KATs and HDACs compete for critical lysines on NFkB subunits. In this regard, small ubiquitin-like modifier modification of KAT3A negatively modulates its transcriptional activity by recruiting a Daxx complex which contains HDAC2 (Kuo et al. 2005)

It is well established that inflammation is also a central element in HD. Elevated NFkB activity has been found in cultured cells expressing mutant Htt and striatal cells from HD transgenic mice (Khoshnan et al. 2004). Furthermore, several family members of the IKK proteins have been shown to be functionally involved in HD. The IKK proteins are critical regulators of NFkB. Indeed in the study demonstrating activation of NFkB, it was found that mutant Htt associated with and activated IKKγ (Khoshnan et al. 2004). Additional studies have shown that both IKKα and IKKβ are involved in the proteolysis of Htt in response to external cues such as DNA damage (Khoshnan et al. 2009). IKK has also been shown to phosphorylate huntingtin which targets it for degradation by the proteasome and lysosome (Thompson et al. 2009). Critically once phosphorylated, IKK regulates additional post-translational modifications, of Htt including ubiquitination, sumoylation and acetylation. Furthermore, this subsequently enhances Htt nuclear localization, cleavage and clearance mediated by lysosomal-associated membrane protein 2A and Hsc70. The authors propose that IKK activates mutant Htt clearance until an age-related loss of proteasome/lysosome function promotes accumulation of toxic post-translationally modified mutant Htt (Thompson et al. 2009).

HATs/HDACs, ER stress and HD

The ability of a cell to sense, response to and circumvent stress is essential for maintaining homeostasis. There are many ways in which stress, either endogenous or exogenous, can be manifested in a cell; these include pathogenic infection, chemical insult, genetic mutation, nutrient deprivation and even normal differentiation. The process of mutant protein folding is particularly sensitive to such insults. As such for the cellular compartments in which mutant proteins are processed and folded, there are adaptive programmes that enable both their detection and correction for more efficient processing (Rutkowski and Kaufman 2004).

The ER is a large cellular organelle comprising a network of interconnected, closed membrane-bound vesicles. It is the site of synthesis, folding and modification of secretory and cell-surface proteins and serves many essential functions, including the production of the components of cellular membranes, proteins, lipids and sterols (Hebert and Molinari 2007). Only correctly folded proteins are transported out of the ER whilst incompletely folded proteins are retained in the organelle to complete the folding process or to be targeted for destruction (Rapoport 2007). Due to the important roles of the ER, its proper functioning is therefore essential to cellular homeostasis. Various conditions can, however, interfere with the ER function leading to situation known as ER stress. Stress is the response of any system to perturbations of its normal state. Thus, ER stress can arise from a disturbance in protein folding which results in an accumulation of unfolded or misfolded proteins within the organelle (Lai et al. 2007). During such disturbances, in order to carry out the correct folding of proteins, the ER has evolved as a specialized protein folding machine with cellular mechanisms that promote proper folding of aberrant protein, thus preventing its aggregation. When ER homeostasis is altered by misfolded proteins, the ER responds by inducing specific genes and attempting to restore normal ER function and maintain stability (Caruso and Chevet 2007). The four elements of ER stress are (1) protein degradation, (2) endoplasmic overload response (EOR), (3) unfolded protein response (UPR) and (4) cellular death pathway. This four-stage model of ER stress toxicity helps to explain this organelles role in the onset of clinical manifestations. Two ER stress-induced signal transduction pathways have been described: the UPR (Ron and Walter 2007) and the EOR (Pahl and Baeuerle 1997). These pathways once activated attempt to adapt to the disturbance and re-establish normal ER function (Xu et al. 2005). In the case of misfolded proteins, this is known as ER-associated degradation (ERAD), the process by which these misfolded proteins are exported to the cytosol for degradation by proteasomes (Vembar and Brodsky 2008). Excessive or prolonged ER stress can overwhelm the cells ability to cope and elicits the cell death programme or apoptosis (Rao et al. 2004). Evidence linking KATs/HDACs to ER stress is not as well established as that for inflammation, with most studies utilising histone deacetylase inhibitors. However, in a recent study in hepatocytes on Mallory body (cytokeratin aggresomes) formation, decreased histone acetyltransferase and increased histone deacetylase activity were observed (Bardag-Gorce et al. 2007). In a similar model of oxidative stress-induced inclusion formation, treatment of cells with 4-phenylbutyrate (PB) was found to alleviate formation of these inclusions (Hanada et al. 2007).

In terms of neurodegeneration, evidence is clearly emerging linking ER stress to the pathogenesis of HD (Vidal et al. 2011). The first indication that ER stress may be involved with Huntington’s disease came from a study of aggresome-like perinuclear inclusions generated using a huntingtin exon 1 truncated protein model in human 293 Tet-Off cells. In depth analysis of these inclusion bodies demonstrated the presence of markers of ER stress including the molecular chaperones BiP/GRP78, Hsp70 and Hsp40 (Waelter et al. 2001).

Mutant huntingtin fragment proteins have now been shown to elevate Bip (an ER chaperone), increase levels of C/EBP homologous protein (CHOP; an ER stress-induced regulator of cell death), increase the phosphorylation of c-Jun-N-terminal kinase (a regulator of cell death) and cleavage of caspase-3 and caspase-12 (mediators of cell death; Reijonen et al. 2008). Inhibition of ER stress using salubrinal counteracted both neuronal cell death and protein aggregation by mutant Htt (Reijonen et al. 2008). Functional analysis of Htt identified an amphipathic alpha helical membrane-binding domain that can reversibly target it to vesicles and the ER (Atwal and Truant 2008; Atwal et al. 2007). Normal Htt is released from these membranes in response to ER stress and rapidly translocates into the nucleus. Once ER stress has been alleviated, the Htt is capable of nuclear export and re-association with the ER. However, this release is inhibited when huntingtin contains the polyglutamine expansion seen in Huntington’s disease. As a result, mutant huntingtin expressing cells have a perturbed ER and an increase in autophagic vesicles, indicating that one of Htt’s functions is to act as ER sentinel, potentially regulating autophagy in response to ER stress (Atwal and Truant 2008; Atwal et al. 2007). Mutant Htt has also been shown to interact with and abrogate the function of grp78, a critical ER membrane-anchored ubiquitin ligase (E3) involved in ERAD resulting in the induction of ER stress (Yang et al. 2010). Furthermore, an additional protein, SCAMP5, which functions to regulate the formation of expanded polyglutamine repeat protein aggregates has been shown to be induced in cultured striatal neurons by either ER stress or mutant HTT (Noh et al. 2009). The heat shock protein HspB8, when overexpressed, has been shown to prevent mutant Htt aggregation through an autophagic activity (Carra et al. 2005, 2008a, b, 2009).

In a presymptomatic knockin mouse model of HD, early markers of disease regulator of ribosome synthesis Rrs1 and its interacting protein 3D3/lyric (also known as metadherin and astrocyte elevated gene-1) were found to be localised in the ER, are induced by ER stress and are involved in the ER stress response in neuronal cells (Carnemolla et al. 2009). Indeed, levels of Rrs1 mRNA have been shown to be elevated in the brains of HD patients (Fossale et al. 2002). Furthermore, in the HD knockin mice, ER stress was found to occur prior to the formation of amyloid intranuclear or cytoplasmic inclusions indicating that it plays a critical role in the pathogenesis of HD (Carnemolla et al. 2009). Postmortem analysis of patient brains has found increased mRNA expression of the ER stress-related genes BiP, CHOP and Herpud1 in HD postmortem brain (Carnemolla et al. 2009).

Direct physical evidence for the association of KATs and HDACs with critical regulatory elements within the ER stress pathway is emerging. CHOP is an ER stress-inducible protein which plays a critical role in regulating programmed cell death. CHOP has recently been shown to directly associate with the histone acetyltransferase p300, and inhibition of HDACs prevents its degradation (Ohoka et al. 2007). Chromatin immunoprecipitation analyses have shown that ER stress regulators are regulated at the transcriptional level via histone post-translational modifications (Donati et al. 2006). KAT3B has been shown to bind to the promoter for the GRP78/BiP (Baumeister et al. 2005), whilst regulation of the CHOP promoter involves a complex containing JDP2 and HDAC3 (Cherasse et al. 2008).

B lymphocyte-induced maturation protein-1 (BLIMP-1) has been shown to be associated with cellular stress and is rapidly upregulated during the UPR in some models (Doody et al. 2006). This protein directly associates with HDACs to repress transcription (Yu et al. 2000) and may therefore indicate that BLIMP-1 may utilise HDACs to downregulate important genes during ER stress.

Aggregation-prone proteins have been suggested to overwhelm and impair the UPS in polyQ disorders, such as HD. Two separate studies have found that UPS dysfunction is a consistent feature of HD pathology with impaired UPS in the synapses of HD mice (Bennett et al. 2007; Wang et al. 2008). ER stress has been shown to have a general inhibitory effect on the UPS (Menendez-Benito et al. 2005), which may explain the long-term gradual accumulation of misfolded proteins in patients with HD. Intriguingly in a neurodegenerative model for spinobulbar muscular atrophy, autophagy was induced to compensate for impaired UPS function in an HDAC6-dependent manner (Pandey et al. 2007), whilst HDAC6 overexpression was sufficient to rescue the degeneration associated with UPS dysfunction in vivo in an autophagy-dependent manner (Pandey et al. 2007). Therefore, antagonists of HDAC6 could potentially be used as an intervention to induce compensatory autophagy in HD. Indeed inhibitors targeting HDAC6 have been shown to alleviate the microtubule transport- and release-defect phenotypes associated with HD (Dompierre et al. 2007). HDAC6 would therefore appear to be the critical regulator controlling cellular response pathways to cytotoxic accumulation of ubiquitinated protein aggregates and interacts with both dynenin motors and misfolded proteins to form an active transport system to transport misfolded proteins to the aggresome (Kawaguchi et al. 2003). HDAC6 also plays a role in regulating chaperone-mediated responses to cellular stress. Ubiquitinated cellular aggregates impairing proteasome activity triggers a HDAC6-dependent cell response involving the induction of major cellular chaperones via dissociation of a repressive HDAC6/heat shock factor 1/heat shock protein 90 complex (Boyault et al. 2007). These and other data showing that HDAC6 is a critical component of environmentally induced stress granules (Kwon et al. 2007) point to an essential role for HDAC6 as a cellular stress surveillance factor, where it can be both a sensor of stressful stimuli and an effector mediating and coordinating appropriate cellular responses (Matthias et al. 2008).

Targeting HD using HDACi

From the previous sections, it could be argued that targeting HDACs may be a way to target the neurodegenerative effects of HD. In the sections below, I shall summarize the current literature on the effects of HDACi in HD.

Neuroprotective effects of HDACi in models of HD

Sodium butyrate

In a cell culture model of HD, treatment of cells with sodium butyrate caused the redistribution of huntingtin-positive nuclear bodies (Kegel et al. 2002). The first indications that HDAC inhibitors may prove useful in the treatment of Huntington’s came from yeast and fly models where treatments of Huntington’s disease models were found arrest neurodegeneration or mitigated the effects on affected gene promoters (Hughes et al. 2001; Steffan et al. 2001). Sodium butyrate has also been shown to ameliorate motor deficits in mouse HD model (Ferrante et al. 2003)

Phenylbutyrate

Following on from these studies, it has since been shown that the HDAC inhibitors PB can also ameliorate Huntington motor deficits in mouse models (Gardian et al. 2005). In vitro and in vivo models of Huntington’s using ST14a/STHdh cells and R2/6 mice demonstrated a marked histone H3 hypoacetylation in downregulated genes, and treatment with HDAC inhibitors was found to both reverse the mRNA abnormalities and led to histone acetylation at the promoters of these genes (Sadri-Vakili et al. 2007).

Currently, clinical trials are undergoing in relation to the use of PB to treat HD. A phase II safety and tolerability trial (NCT00212316) has been completed, and a serum analysis has identified individual-specific patterns of ca. 20 metabolites which could provide a means for the selection of subjects for extended trials using this drug (Ebbel et al. 2010).

Valproic acid (Depakene® Depakote®)

High-dose valproate treatment of the transgenic mouse N171-82Q model of HD significantly prolonged survival and ameliorated diminished spontaneous locomotor activity, without exerting any noteworthy side effect on their behaviour or the striatal dopamine content at the dose administered (daily i.p., 100 mg/kg; Zadori et al. 2009). It must be noted that there has also been one report linking valproic acid treatment with the development of Pisa syndrome in a patient with HD (Salazar et al. 2008).

Suberoylanilide hydroxamic acid (Vorinostat, Zolinza®)

Several studies have now shown that the first FDA approved histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA; Zolinza®) has efficacy in modulating the severity of HD models. The earliest such study demonstrated that SAHA was able to ameliorate motor deficits in a mouse model of Huntington’s disease (Hockly et al. 2003).

SAHA is a pan-specific HDACi broadly inhibiting the classes I, II and IV HDACs (Khan et al. 2008), but it has also been shown to specifically downregulate HDAC7 (Dokmanovic et al. 2007). SAHA can functionally decrease the levels of HDAC7 in wild-type and the R6/2 transgenic mouse model of HD (Benn et al. 2009). Despite this, complete genetic loss of HDAC7 did not alleviate the symptoms in HD models (Benn et al. 2009). As such, HDAC7 would not appear to be the main target for the therapeutic benefits of HDACi in HD models. SAHA has also been shown to inhibit HDAC6. This deacetylase has a special function in that it deacetylates microtubules in vitro and in vivo (Hubbert et al. 2002). HDAC6 may therefore be an important target in HD as levels of tubulin acetylation were also found to be reduced in HD brains, whilst treatment with the HDACi TSA ameliorated the phenotypic effects of HD (Dompierre et al. 2007). Indeed, microtubule dynamics associated with HDAC6 have also been shown to be of broad potential interest in conditions of neurodegeneration. In Alzheimer’s disease, the protein tau has been shown to inhibit HDAC6 resulting in increased microtubule acetylation and prevents the induction of autophagy by inhibiting proteasome function (Perez et al. 2009). Similarly, HDAC6 has also been shown to regulate the cellular location of Parkin, a protein-ubiquitin E3 ligase linked to Parkinson’s disease (Jiang et al. 2008).

Other HDACi

In the R6/2 transgenic mouse model of HD, a novel pimelic diphenylamide HDAC inhibitor, HDACi 4b, significantly improved motor performance, overall appearance and body weight of symptomatic mice, with associated significant attenuation of gross brain-size decline and striatal atrophy (Thomas et al. 2008).

Potential use of HDACi to alleviate ER stress in HD

Data from in vitro studies are emerging demonstrating the efficacy of the histone deacetylase inhibitor 4-phenylbutyrate in relieving ER stress in cell line models of cystic fibrosis (Rubenstein and Zeitlin 2000) and mutant alpha1-ATZ liver disease (Burrows et al. 2000). Since these initial observations, several studies have shown that 4-phenylbutyrate may act as a chemical chaperone to relieve ER stress induced in models of ischemia (Qi et al. 2004; Vilatoba et al. 2005). Similar data have emerged for models of cataract formation (Mulhern et al. 2007), Parkinson’s disease (Kubota et al. 2006), retinitis pigmentosa (Bonapace et al. 2004), glaucoma (Yam et al. 2007a) and reconfirmation of its ability to alleviate stress in cystic fibrosis (Vij et al. 2006).

In diabetes, 4-phenylbutyrate has also been shown to both relieve ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. This occurs by the restoration of systemic insulin sensitivity, resolution of fatty liver disease and enhancement of insulin action in liver, muscle and adipose tissues (Ozcan et al. 2006).

ER stress in liver-induced ischemia has been shown to be ameliorated by treatments with 4-phenylbutyrate (Vilatoba et al. 2005). This compound has also been shown to alleviate oxidative stress-induced ER tress in cultured hepatocytes and hepatoma cells (Hanada et al. 2007) and prevents ER stress-mediated aggregate formation in a model of hereditary haemochromatosis (de Almeida et al. 2007).

Valproate/valproic acid, another histone deacetylase inhibitor, has also been shown to have potential in the treatment of ER stress. Initial studies demonstrated that treatment of cells with valproate caused the upregulation of GRP78/BiP, a key ER-mediated chaperone (Wang et al. 1999). Subsequent studies confirmed that valproate could increase the expression of additional important endoplasmic reticulum stress proteins, GRP94 and calreticulin (Bown et al. 2000; Chen et al. 2000; Shao et al. 2006). Valproate has also been shown to protect against oxidative stress induced protein damage and had provided neuroprotection in the same model system (Wang et al. 2003; Shao et al. 2005; Cui et al. 2007).

Using HDACi to potentially modulate chaperone activity in HD

A plethora of studies have clearly shown that histone deacetylase inhibitors can reactivate or alter gene expression. However, histone deacetylases and histone deacetylase inhibitors may also play important roles in regulating chaperone expression and function. Obviously, this has important implications in conditions such as HD where aberrant misfolding of proteins can result in ER stress (Vidal et al. 2011) and can be highlighted by the fact that HDAC6 is recognised as a major regulator of cellular responses aimed at counteracting the effects of misfolded protein accumulation (Kovacs et al. 2005; Boyault et al. 2006, 2007; Gao et al. 2007). Inhibition or depletion of HDAC6 leads to induction of Hsp90 acetylation resulting in inhibition of its chaperone activity (Fiskus et al. 2007; Wang et al. 2007b; Scroggins et al. 2007; Edwards et al. 2007; Bali et al. 2005; Kong et al. 2006). The class I HDACs (HDACs 1–3) have also been shown to associate with the ATP-dependent chaperone Hsp70, and this association enhances deacetylase catalytic activity (Johnson et al. 2002).

Histone deacetylase inhibitors have emerged as potential therapeutic agents in the relief of ER stress induced in cell line models. Many of these studies have utilised the chemical PB. For example, in a hepatocyte cell line model of oxidative stress induced ER stress, treatment of cells with PB was found to alleviate the ER stress in these cells (Hanada et al. 2007). Other examples of the benefits of PB as a chemical chaperone relieving apoptosis and/or ER stress have been reported in models of eye disease (Bonapace et al. 2004; Yam et al. 2007a), defective trafficking of nephrin in kidney (Liu et al. 2004), rescue of protein trafficking in the lysosomal storage disorder Fabry disease (Yam et al. 2007b), correction of autodominant hypoparathyroidism-induced apoptosis (Datta et al. 2007) and relief of ER stress-mediated programmed cell death in Arabidopsis (Watanabe and Lam 2007).

Other evidence for the potential of HDACi influencing chaperone-like activities to relieve ER stress are emerging. In cystic fibrosis models, phenylbutyrate can restore CFTR trafficking and function (Zeitlin et al. 2002; Singh et al. 2006; Rubenstein and Zeitlin 1998, 2000; Rubenstein and Lyons 2001; Rubenstein et al. 1997; Lim et al. 2004; Kontopoulos et al. 2006; Choo-Kang and Zeitlin 2001), through the induction of Hsp90 (Choo-Kang and Zeitlin 2001), and additional lung conditions for which beneficial responses have been observed using HDACi as chemical chaperones include respiratory distress syndrome (Cheong et al. 2006) and emphysema (Burrows et al. 2000). HDACi can also act as chemical chaperones in the liver. For example, PB was used to protect liver cells from ER stress-mediated apoptosis induced by liver ischemia (Vilatoba et al. 2005), can enhance the cell-surface expression and transport capacity of mutated bile salt export pumps (Hayashi and Sugiyama 2007) and also reduce ER stress-induced formation of Mallory bodies in hepatocytes (Hanada et al. 2007).

However, it must be noted that within the neuronal setting, initial studies on mood stabilizing drugs such as valproic acid demonstrated increased expression of ER stress proteins in cerebral cortex, hippocampus, neuronal and glial cells (Wang et al. 1999; Bown et al. 2000; Chen et al. 2000; Corson et al. 2004; Wang et al. 2001; Shao et al. 2006).

Use of HDACi in patients with HD

Initial clinical trials testing the use of valproate in the treatment of HD proved disappointing and did not show any clinical benefit (Bachman et al. 1977; Pearce et al. 1977; Symington et al. 1978). More recent studies have, however, demonstrated some benefit in HD patients either as specific monotherapy (Saft et al. 2006) or as a combination therapy with olanzipine (Grove et al. 2000).

Intensive pharmaceutical efforts have generated many agents targeting histone deacetylases, and of these, currently two have received FDA approval, SAHA/Vorinostat (Zolinza) and FK228/Romidepsin (Istodax), and both are currently licensed for the treatment of cutaneous T cell lymphoma (Mann et al. 2007a; b; Hymes 2010). Several histone deacetylase inhibitors which are undergoing clinical trials have, or currently include, patients with neurodegenerative diseases including HD, and as these trials proceed, a better understanding of the potential side effects and dose-limiting toxicities of these drugs will emerge for patients with these conditions. For the most part, the first generation inhibitors have shown well-tolerated safety profiles (Lawless et al. 2009a).

A randomized, double-blind and placebo-controlled phase I clinical trial by Elixir Pharmaceuticals of an orally active, selective Sirt1 inhibitor SEN0014196 (6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide), has been initiated as a disease-modifying therapy in Huntington’s disease, but results from this are not yet known.

Likewise, a phase I study examining the safety and tolerability of phenylbutyrate in HD (PHEND-HD) has recently been completed (Clinical Trials identifier—NCT00212316). In this study, phenylbutyrate was found to be both safe and well tolerated in early symptomatic HD subjects taking 15 g daily, and levels leukocyte histone acetylation were measurable, indicating that HDAC inhibition was occurring in response to treatment (Moser et al. 2008) A similar dose escalation/de-escalation study using sodium phenylbutyrate in HD patients agreed with the MTD of 15 g daily (Hogarth et al. 2007), and at 12 g/daily was found to correct gene expression of a 12 gene biomarker set previously identified by this group (Borovecki et al. 2005).

Future directions

Clearly, the potential utility of HDACi for the treatment of HD exists. Several HDAC inhibitors have good safety/tolerability profiles and are undergoing clinical trials in neurodegenerative disease. It therefore seems imperative that these drugs should continue to be tested for their potential therapeutic role in patients afflicted with HD. Further clinical trials are, however, required to demonstrate their efficacy in alleviating the symptoms of this disease, and indeed, it may be that HDACi may be more effective in a combinatorial setting with other agents, particularly for diseases such as HD. These and other questions remain to be addressed, but it is clear that these inhibitors may play important roles in both targeting neurodegenerative conditions through their activity on gene transcription and potentially through additionally alleviating ER stress or modulating chaperone activity. Further experiments will, however, be required to functionally address these possibilities.

References

  • Abrajano JJ, Qureshi IA, Gokhan S, Zheng D, Bergman A, Mehler MF (2009a) Differential deployment of REST and CoREST promotes glial subtype specification and oligodendrocyte lineage maturation. PLoS ONE 4(11):e7665

    Google Scholar 

  • Abrajano JJ, Qureshi IA, Gokhan S, Zheng D, Bergman A, Mehler MF (2009b) REST and CoREST modulate neuronal subtype specification, maturation and maintenance. PLoS ONE 4(12):e7936

    Google Scholar 

  • Aiken CT, Steffan JS, Guerrero CM, Khashwji H, Lukacsovich T, Simmons D, Purcell JM, Menhaji K, Zhu YZ, Green K, Laferla F, Huang L, Thompson LM, Marsh JL (2009) Phosphorylation of threonine 3: implications for Huntingtin aggregation and neurotoxicity. J Biol Chem 284(43):29427–29436

    PubMed  CAS  PubMed Central  Google Scholar 

  • Ajamian F, Suuronen T, Salminen A, Reeben M (2003) Upregulation of class II histone deacetylases mRNA during neural differentiation of cultured rat hippocampal progenitor cells. Neurosci Lett 346(1–2):57–60

    PubMed  CAS  Google Scholar 

  • Albert M, Helin K (2010) Histone methyltransferases in cancer. Semin Cell Dev Biol 21(2):209–220

    PubMed  CAS  Google Scholar 

  • 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(4):633–636

    PubMed  CAS  Google Scholar 

  • Andres ME, Burger C, Peral-Rubio MJ, Battaglioli E, Anderson ME, Grimes J, Dallman J, Ballas N, Mandel G (1999) CoREST: a functional corepressor required for regulation of neural- specific gene expression. Proc Natl Acad Sci USA 96(17):9873–9878

    PubMed  CAS  PubMed Central  Google Scholar 

  • Anne SL, Saudou F, Humbert S (2007) Phosphorylation of huntingtin by cyclin-dependent kinase 5 is induced by DNA damage and regulates wild-type and mutant huntingtin toxicity in neurons. J Neurosci 27(27):7318–7328

    PubMed  CAS  Google Scholar 

  • Ashburner BP, Westerheide SD, Baldwin AS Jr (2001) The p65 (RelA) subunit of NF-kappaB interacts with the histone deacetylase (HDAC) corepressors HDAC1 and HDAC2 to negatively regulate gene expression. Mol Cell Biol 21(20):7065–7077

    PubMed  CAS  PubMed Central  Google Scholar 

  • Atwal RS, Truant R (2008) A stress sensitive ER membrane-association domain in Huntingtin protein defines a potential role for Huntingtin in the regulation of autophagy. Autophagy 4(1):91–93

    PubMed  Google Scholar 

  • Atwal RS, Xia J, Pinchev D, Taylor J, Epand RM, Truant R (2007) Huntingtin has a membrane association signal that can modulate huntingtin aggregation, nuclear entry and toxicity. Hum Mol Genet 16(21):2600–2615

    PubMed  CAS  Google Scholar 

  • Bach I, Rodriguez-Esteban C, Carriere C, Bhushan A, Krones A, Rose DW, Glass CK, Andersen B, Izpisua Belmonte JC, Rosenfeld MG (1999) RLIM inhibits functional activity of LIM homeodomain transcription factors via recruitment of the histone deacetylase complex. Nat Genet 22(4):394–399

    PubMed  CAS  Google Scholar 

  • Bachman DS, Butler IJ, McKhann GM (1977) Long-term treatment of juvenile Huntington’s chorea with dipropylacetic acid. Neurology 27(2):193–197

    PubMed  CAS  Google Scholar 

  • Bahn S, Mimmack M, Ryan M, Caldwell MA, Jauniaux E, Starkey M, Svendsen CN, Emson P (2002) Neuronal target genes of the neuron-restrictive silencer factor in neurospheres derived from fetuses with Down’s syndrome: a gene expression study. Lancet 359(9303):310–315

    PubMed  CAS  Google Scholar 

  • Balasubramaniyan V, Boddeke E, Bakels R, Kust B, Kooistra S, Veneman A, Copray S (2006) Effects of histone deacetylation inhibition on neuronal differentiation of embryonic mouse neural stem cells. Neuroscience 143(4):939–951

    PubMed  CAS  Google Scholar 

  • Bali P, Pranpat M, Bradner J, Balasis M, Fiskus W, Guo F, Rocha K, Kumaraswamy S, Boyapalle S, Atadja P, Seto E, Bhalla K (2005) Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 280(29):26729–26734

    PubMed  CAS  Google Scholar 

  • Ballas N, Grunseich C, Lu DD, Speh JC, Mandel G (2005) REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell 121(4):645–657

    PubMed  CAS  Google Scholar 

  • Bardag-Gorce F, Dedes J, French BA, Oliva JV, Li J, French SW (2007) Mallory body formation is associated with epigenetic phenotypic change in hepatocytes in vivo. Exp Mol Pathol 83(2):160–168

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(10):2830–2838

    PubMed  CAS  Google Scholar 

  • Battaglioli E, Andres ME, Rose DW, Chenoweth JG, Rosenfeld MG, Anderson ME, Mandel G (2002) REST repression of neuronal genes requires components of the hSWI.SNF complex. J Biol Chem 277(43):41038–41045

    PubMed  CAS  Google Scholar 

  • Baumeister P, Luo S, Skarnes WC, Sui G, Seto E, Shi Y, Lee AS (2005) Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers. Mol Cell Biol 25(11):4529–4540

    PubMed  CAS  PubMed Central  Google Scholar 

  • Belyaev ND, Wood IC, Bruce AW, Street M, Trinh JB, Buckley NJ (2004) Distinct RE-1 silencing transcription factor-containing complexes interact with different target genes. J Biol Chem 279(1):556–561

    PubMed  CAS  Google Scholar 

  • Benn CL, Butler R, Mariner L, Nixon J, Moffitt H, Mielcarek M, Woodman B, Bates GP (2009) Genetic knock-down of HDAC7 does not ameliorate disease pathogenesis in the R6/2 mouse model of Huntington’s disease. PLoS ONE 4(6):e5747

    PubMed  PubMed Central  Google Scholar 

  • Bennett EJ, Shaler TA, Woodman B, Ryu KY, Zaitseva TS, Becker CH, Bates GP, Schulman H, Kopito RR (2007) Global changes to the ubiquitin system in Huntington’s disease. Nature 448(7154):704–708

    PubMed  CAS  Google Scholar 

  • Bithell A, Johnson R, Buckley NJ (2009) Transcriptional dysregulation of coding and non-coding genes in cellular models of Huntington’s disease. Biochem Soc Trans 37(Pt 6):1270–1275

    PubMed  CAS  Google Scholar 

  • Bonapace G, Waheed A, Shah GN, Sly WS (2004) Chemical chaperones protect from effects of apoptosis-inducing mutation in carbonic anhydrase IV identified in retinitis pigmentosa 17. Proc Natl Acad Sci USA 101(33):12300–12305

    PubMed  CAS  PubMed Central  Google Scholar 

  • Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, Rosas HD, Hersch SM, Hogarth P, Bouzou B, Jensen RV, Krainc D (2005) Genome-wide expression profiling of human blood reveals biomarkers for Huntington’s disease. Proc Natl Acad Sci USA 102(31):11023–11028

    PubMed  CAS  PubMed Central  Google Scholar 

  • Boutell JM, Thomas P, Neal JW, Weston VJ, Duce J, Harper PS, Jones AL (1999) Aberrant interactions of transcriptional repressor proteins with the Huntington’s disease gene product, huntingtin. Hum Mol Genet 8(9):1647–1655

    PubMed  CAS  Google Scholar 

  • Boutillier AL, Trinh E, Loeffler JP (2002) Constitutive repression of E2F1 transcriptional activity through HDAC proteins is essential for neuronal survival. Ann NY Acad Sci 973:438–442

    PubMed  CAS  Google Scholar 

  • Bown CD, Wang JF, Young LT (2000) Increased expression of endoplasmic reticulum stress proteins following chronic valproate treatment of rat C6 glioma cells. Neuropharmacology 39(11):2162–2169

    PubMed  CAS  Google Scholar 

  • Boyault C, Gilquin B, Zhang Y, Rybin V, Garman E, Meyer-Klaucke W, Matthias P, Muller CW, Khochbin S (2006) HDAC6-p97/VCP controlled polyubiquitin chain turnover. EMBO J 25(14):3357–3366

    PubMed  CAS  PubMed Central  Google Scholar 

  • Boyault C, Zhang Y, Fritah S, Caron C, Gilquin B, Kwon SH, Garrido C, Yao TP, Vourc’h C, Matthias P, Khochbin S (2007) HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates. Genes Dev 21(17):2172–2181

    PubMed  CAS  PubMed Central  Google Scholar 

  • Brill LM, Xiong W, Lee KB, Ficarro SB, Crain A, Xu Y, Terskikh A, Snyder EY, Ding S (2009) Phosphoproteomic analysis of human embryonic stem cells. Cell Stem Cell 5(2):204–213

    PubMed  CAS  PubMed Central  Google Scholar 

  • Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA, Alt FW, Greenberg ME (2004) Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303(5666):2011–2015

    PubMed  CAS  Google Scholar 

  • Burrows JA, Willis LK, Perlmutter DH (2000) Chemical chaperones mediate increased secretion of mutant alpha 1-antitrypsin (alpha 1-AT) Z: a potential pharmacological strategy for prevention of liver injury and emphysema in alpha 1-AT deficiency. Proc Natl Acad Sci USA 97(4):1796–1801

    PubMed  CAS  PubMed Central  Google Scholar 

  • Calao M, Burny A, Quivy V, Dekoninck A, Van Lint C (2008) A pervasive role of histone acetyltransferases and deacetylases in an NF-kappaB-signaling code. Trends Biochem Sci 33(7):339–349

    PubMed  CAS  Google Scholar 

  • Camelo S, Iglesias AH, Hwang D, Due B, Ryu H, Smith K, Gray SG, Imitola J, Duran G, Assaf B, Langley B, Khoury SJ, Stephanopoulos G, De Girolami U, Ratan RR, Ferrante RJ, Dangond F (2005) Transcriptional therapy with the histone deacetylase inhibitor trichostatin A ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol 164(1–2):10–21

    PubMed  CAS  Google Scholar 

  • Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y (2002) Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298(5595):1039–1043

    PubMed  CAS  Google Scholar 

  • Carnemolla A, Fossale E, Agostoni E, Michelazzi S, Calligaris R, De Maso L, Del Sal G, MacDonald ME, Persichetti F (2009) Rrs1 is involved in endoplasmic reticulum stress response in Huntington disease. J Biol Chem 284(27):18167–18173

    PubMed  CAS  PubMed Central  Google Scholar 

  • Carra S, Sivilotti M, Chavez Zobel AT, Lambert H, Landry J (2005) HspB8, a small heat shock protein mutated in human neuromuscular disorders, has in vivo chaperone activity in cultured cells. Hum Mol Genet 14(12):1659–1669

    PubMed  CAS  Google Scholar 

  • Carra S, Seguin SJ, Lambert H, Landry J (2008a) HspB8 chaperone activity toward poly(Q)-containing proteins depends on its association with Bag3, a stimulator of macroautophagy. J Biol Chem 283(3):1437–1444

    CAS  Google Scholar 

  • Carra S, Seguin SJ, Landry J (2008b) HspB8 and Bag3: a new chaperone complex targeting misfolded proteins to macroautophagy. Autophagy 4(2):237–239

    CAS  Google Scholar 

  • Carra S, Brunsting JF, Lambert H, Landry J, Kampinga HH (2009) HspB8 participates in protein quality control by a non-chaperone-like mechanism that requires eIF2{alpha} phosphorylation. J Biol Chem 284(9):5523–5532

    PubMed  CAS  Google Scholar 

  • Caruso ME, Chevet E (2007) Systems biology of the endoplasmic reticulum stress response. Subcell Biochem 43:277–298

    PubMed  Google Scholar 

  • Chaturvedi RK, Adhihetty P, Shukla S, Hennessy T, Calingasan N, Yang L, Starkov A, Kiaei M, Cannella M, Sassone J, Ciammola A, Squitieri F, Beal MF (2009) Impaired PGC-1alpha function in muscle in Huntington’s disease. Hum Mol Genet 18(16):3048–3065

    PubMed  CAS  PubMed Central  Google Scholar 

  • Che HV, Metzger S, Portal E, Deyle C, Riess O, Nguyen HP (2011) Localization of sequence variations in PGC-1alpha influence their modifying effect in Huntington disease. Mol Neurodegener 6(1):1

    PubMed  CAS  PubMed Central  Google Scholar 

  • Chen ZF, Paquette AJ, Anderson DJ (1998) NRSF/REST is required in vivo for repression of multiple neuronal target genes during embryogenesis. Nat Genet 20(2):136–142

    PubMed  CAS  Google Scholar 

  • Chen B, Wang JF, Young LT (2000) Chronic valproate treatment increases expression of endoplasmic reticulum stress proteins in the rat cerebral cortex and hippocampus. Biol Psychiatry 48(7):658–664

    PubMed  CAS  Google Scholar 

  • Cheong N, Madesh M, Gonzales LW, Zhao M, Yu K, Ballard PL, Shuman H (2006) Functional and trafficking defects in ATP binding cassette A3 mutants associated with respiratory distress syndrome. J Biol Chem 281(14):9791–9800

    PubMed  CAS  Google Scholar 

  • Cherasse Y, Chaveroux C, Jousse C, Maurin AC, Carraro V, Parry L, Fafournoux P, Bruhat A (2008) Role of the repressor JDP2 in the amino acid-regulated transcription of CHOP. FEBS Lett 582(10):1537–1541

    PubMed  CAS  Google Scholar 

  • Choo-Kang LR, Zeitlin PL (2001) Induction of HSP70 promotes DeltaF508 CFTR trafficking. Am J Physiol Lung Cell Mol Physiol 281(1):L58–L68

    PubMed  CAS  Google Scholar 

  • Colin E, Zala D, Liot G, Rangone H, Borrell-Pages M, Li XJ, Saudou F, Humbert S (2008) Huntingtin phosphorylation acts as a molecular switch for anterograde/retrograde transport in neurons. EMBO J 27(15):2124–2134

    PubMed  CAS  PubMed Central  Google Scholar 

  • Conaco C, Otto S, Han JJ, Mandel G (2006) Reciprocal actions of REST and a microRNA promote neuronal identity. Proc Natl Acad Sci USA 103(7):2422–2427

    PubMed  CAS  PubMed Central  Google Scholar 

  • Cong SY, Pepers BA, Evert BO, Rubinsztein DC, Roos RA, van Ommen GJ, Dorsman JC (2005) Mutant huntingtin represses CBP, but not p300, by binding and protein degradation. Mol Cell Neurosci 30(4):560–571

    PubMed  CAS  Google Scholar 

  • Corson TW, Woo KK, Li PP, Warsh JJ (2004) Cell-type specific regulation of calreticulin and Bcl-2 expression by mood stabilizer drugs. Eur Neuropsychopharmacol 14(2):143–150

    PubMed  CAS  Google Scholar 

  • Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D (2006) Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 127(1):59–69

    PubMed  CAS  Google Scholar 

  • Cui J, Shao L, Young LT, Wang JF (2007) Role of glutathione in neuroprotective effects of mood stabilizing drugs lithium and valproate. Neuroscience 144(4):1447–1453

    PubMed  CAS  Google Scholar 

  • Curtis MA, Penney EB, Pearson J, Dragunow M, Connor B, Faull RL (2005) The distribution of progenitor cells in the subependymal layer of the lateral ventricle in the normal and Huntington’s disease human brain. Neuroscience 132(3):777–788

    PubMed  CAS  Google Scholar 

  • Dandona P, Aljada A, Bandyopadhyay A (2004a) Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol 25(1):4–7

    CAS  Google Scholar 

  • Dandona P, Aljada A, Chaudhuri A, Mohanty P (2004b) Endothelial dysfunction, inflammation and diabetes. Rev Endocr Metab Disord 5(3):189–197

    CAS  Google Scholar 

  • Dangond F, Hafler DA, Tong JK, Randall J, Kojima R, Utku N, Gullans SR (1998) Differential display cloning of a novel human histone deacetylase (HDAC3) cDNA from PHA-activated immune cells. Biochem Biophys Res Commun 242(3):648–652

    PubMed  CAS  Google Scholar 

  • Datta R, Waheed A, Shah GN, Sly WS (2007) Signal sequence mutation in autosomal dominant form of hypoparathyroidism induces apoptosis that is corrected by a chemical chaperone. Proc Natl Acad Sci USA 104(50):19989–19994

    PubMed  CAS  PubMed Central  Google Scholar 

  • Daub H, Olsen JV, Bairlein M, Gnad F, Oppermann FS, Korner R, Greff Z, Keri G, Stemmann O, Mann M (2008) Kinase-selective enrichment enables quantitative phosphoproteomics of the kinome across the cell cycle. Mol Cell 31(3):438–448

    PubMed  CAS  Google Scholar 

  • de Almeida SF, Picarote G, Fleming JV, Carmo-Fonseca M, Azevedo JE, de Sousa M (2007) Chemical chaperones reduce endoplasmic reticulum stress and prevent mutant HFE aggregate formation. J Biol Chem 282(38):27905–27912

    PubMed  Google Scholar 

  • Dephoure N, Zhou C, Villen J, Beausoleil SA, Bakalarski CE, Elledge SJ, Gygi SP (2008) A quantitative atlas of mitotic phosphorylation. Proc Natl Acad Sci USA 105(31):10762–10767

    PubMed  CAS  PubMed Central  Google Scholar 

  • DiFiglia M, Sapp E, Chase K, Schwarz C, Meloni A, Young C, Martin E, Vonsattel JP, Carraway R, Reeves SA et al (1995) Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons. Neuron 14(5):1075–1081

    PubMed  CAS  Google Scholar 

  • Dokmanovic M, Perez G, Xu W, Ngo L, Clarke C, Parmigiani RB, Marks PA (2007) Histone deacetylase inhibitors selectively suppress expression of HDAC7. Mol Cancer Ther 6(9):2525–2534

    PubMed  CAS  Google Scholar 

  • Dompierre JP, Godin JD, Charrin BC, Cordelieres FP, King SJ, Humbert S, Saudou F (2007) Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci 27(13):3571–3583

    PubMed  CAS  Google Scholar 

  • Donati G, Imbriano C, Mantovani R (2006) Dynamic recruitment of transcription factors and epigenetic changes on the ER stress response gene promoters. Nucleic Acids Res 34(10):3116–3127

    PubMed  CAS  PubMed Central  Google Scholar 

  • Doody GM, Stephenson S, Tooze RM (2006) BLIMP-1 is a target of cellular stress and downstream of the unfolded protein response. Eur J Immunol 36(6):1572–1582

    PubMed  CAS  Google Scholar 

  • 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 LC/EC-array/MS and off-line tandem MS. Anal Biochem 399:152–161

    PubMed  CAS  PubMed Central  Google Scholar 

  • Edmunds JW, Mahadevan LC, Clayton AL (2008) Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation. EMBO J 27(2):406–420

    PubMed  CAS  PubMed Central  Google Scholar 

  • Edwards A, Li J, Atadja P, Bhalla K, Haura EB (2007) Effect of the histone deacetylase inhibitor LBH589 against epidermal growth factor receptor-dependent human lung cancer cells. Mol Cancer Ther 6(9):2515–2524

    PubMed  CAS  Google Scholar 

  • Esiri MM (2007) The interplay between inflammation and neurodegeneration in CNS disease. J Neuroimmunol 184(1–2):4–16

    PubMed  CAS  Google Scholar 

  • Esposito K, Nappo F, Giugliano F, Di Palo C, Ciotola M, Barbieri M, Paolisso G, Giugliano D (2003) Cytokine milieu tends toward inflammation in type 2 diabetes. Diab Care 26(5):1647

    Google Scholar 

  • 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(28):9418–9427

    PubMed  CAS  Google Scholar 

  • Fiskus W, Ren Y, Mohapatra A, Bali P, Mandawat A, Rao R, Herger B, Yang Y, Atadja P, Wu J, Bhalla K (2007) Hydroxamic acid analogue histone deacetylase inhibitors attenuate estrogen receptor-alpha levels and transcriptional activity: a result of hyperacetylation and inhibition of chaperone function of heat shock protein 90. Clin Cancer Res 13(16):4882–4890

    PubMed  CAS  Google Scholar 

  • Fossale E, Wheeler VC, Vrbanac V, Lebel LA, Teed A, Mysore JS, Gusella JF, MacDonald ME, Persichetti F (2002) Identification of a presymptomatic molecular phenotype in Hdh CAG knock-in mice. Hum Mol Genet 11(19):2233–2241

    PubMed  CAS  Google Scholar 

  • Gao YS, Hubbert CC, Lu J, Lee YS, Lee JY, Yao TP (2007) Histone deacetylase 6 regulates growth factor-induced actin remodeling and endocytosis. Mol Cell Biol 27(24):8637–8647

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(1):556–563

    PubMed  CAS  Google Scholar 

  • Gaub P, Tedeschi A, Puttagunta R, Nguyen T, Schmandke A, Di Giovanni S (2010) HDAC inhibition promotes neuronal outgrowth and counteracts growth cone collapse through CBP/p300 and P/CAF-dependent p53 acetylation. Cell Death Differ 17:1392–1408

    PubMed  CAS  Google Scholar 

  • Gerritsen ME, Williams AJ, Neish AS, Moore S, Shi Y, Collins T (1997) CREB-binding protein/p300 are transcriptional coactivators of p65. ProcNatlAcad Sci USA 94(7):2927–2932

    CAS  Google Scholar 

  • Gray SG (2009) Histone deacetylase inhibitors as a therapeutic modality in multiple sclerosis. In: Zouali M (ed) The epigenetics of autoimmune diseases. Wiley-Blackwell, Chichester, pp 403–432

    Google Scholar 

  • Greenway DJ, Street M, Jeffries A, Buckley NJ (2007) RE1 silencing transcription factor maintains a repressive chromatin environment in embryonic hippocampal neural stem cells. Stem Cells 25(2):354–363

    PubMed  CAS  Google Scholar 

  • Grimes JA, Nielsen SJ, Battaglioli E, Miska EA, Speh JC, Berry DL, Atouf F, Holdener BC, Mandel G, Kouzarides T (2000) The co-repressor mSin3A is a functional component of the REST-CoREST repressor complex. J Biol Chem 275(13):9461–9467

    PubMed  CAS  Google Scholar 

  • Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140(6):883–899

    PubMed  CAS  PubMed Central  Google Scholar 

  • Group THsDCR (1993) A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 72(6):971–983

    Google Scholar 

  • Grove VE Jr, Quintanilla J, DeVaney GT (2000) Improvement of Huntington’s disease with olanzapine and valproate. N Engl J Med 343(13):973–974

    PubMed  Google Scholar 

  • Gu X, Greiner ER, Mishra R, Kodali R, Osmand A, Finkbeiner S, Steffan JS, Thompson LM, Wetzel R, Yang XW (2009) Serines 13 and 16 are critical determinants of full-length human mutant huntingtin induced disease pathogenesis in HD mice. Neuron 64(6):828–840

    PubMed  CAS  PubMed Central  Google Scholar 

  • Hanada S, Harada M, Kumemura H, Bishr Omary M, Koga H, Kawaguchi T, Taniguchi E, Yoshida T, Hisamoto T, Yanagimoto C, Maeyama M, Ueno T, Sata M (2007) Oxidative stress induces the endoplasmic reticulum stress and facilitates inclusion formation in cultured cells. J Hepatol 47(1):93–102

    PubMed  CAS  Google Scholar 

  • Hayashi H, Sugiyama Y (2007) 4-Phenylbutyrate enhances the cell surface expression and the transport capacity of wild-type and mutated bile salt export pumps. Hepatology 45(6):1506–1516

    PubMed  CAS  Google Scholar 

  • He Y, Dupree J, Wang J, Sandoval J, Li J, Liu H, Shi Y, Nave KA, Casaccia-Bonnefil P (2007) The transcription factor Yin Yang 1 is essential for oligodendrocyte progenitor differentiation. Neuron 55(2):217–230

    PubMed  CAS  PubMed Central  Google Scholar 

  • Hebert DN, Molinari M (2007) In and out of the ER: protein folding, quality control, degradation, and related human diseases. Physiol Rev 87(4):1377–1408

    PubMed  CAS  Google Scholar 

  • Hoberg JE, Yeung F, Mayo MW (2004) SMRT derepression by the IkappaB kinase alpha: a prerequisite to NF-kappaB transcription and survival. Mol Cell 16(2):245–255

    PubMed  CAS  Google Scholar 

  • Hoberg JE, Popko AE, Ramsey CS, Mayo MW (2006) IkappaB kinase alpha-mediated derepression of SMRT potentiates acetylation of RelA/p65 by p300. Mol Cell Biol 26(2):457–471

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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 USA 100(4):2041–2046

    PubMed  CAS  PubMed Central  Google Scholar 

  • Hoffner G, Kahlem P, Djian P (2002) Perinuclear localization of huntingtin as a consequence of its binding to microtubules through an interaction with beta-tubulin: relevance to Huntington’s disease. J Cell Sci 115(Pt 5):941–948

    PubMed  CAS  Google Scholar 

  • Hogarth P, Lovrecic L, Krainc D (2007) Sodium phenylbutyrate in Huntington’s disease: a dose-finding study. Mov Disord 22(13):1962–1964

    PubMed  Google Scholar 

  • Hollenbach AD, McPherson CJ, Mientjes EJ, Iyengar R, Grosveld G (2002) Daxx and histone deacetylase II associate with chromatin through an interaction with core histones and the chromatin-associated protein Dek. J Cell Sci 115(Pt 16):3319–3330

    PubMed  CAS  Google Scholar 

  • Horio Y, Hisahara S, Sakamoto J (2003) Functional analysis of SIR2. Nippon Yakurigaku Zasshi 122(Suppl):30P–32P

    PubMed  Google Scholar 

  • Hoshino M, Tagawa K, Okuda T, Murata M, Oyanagi K, Arai N, Mizutani T, Kanazawa I, Wanker EE, Okazawa H (2003) Histone deacetylase activity is retained in primary neurons expressing mutant huntingtin protein. J Neurochem 87(1):257–267

    PubMed  CAS  Google Scholar 

  • Hou ST, Callaghan D, Fournier MC, Hill I, Kang L, Massie B, Morley P, Murray C, Rasquinha I, Slack R, MacManus JP (2000) The transcription factor E2F1 modulates apoptosis of neurons. J Neurochem 75(1):91–100

    PubMed  CAS  Google Scholar 

  • Huang Y, Myers SJ, Dingledine R (1999) Transcriptional repression by REST: recruitment of Sin3A and histone deacetylase to neuronal genes. Nat Neurosci 2(10):867–872

    PubMed  CAS  Google Scholar 

  • Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang XF, Yao TP (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417(6887):455–458

    PubMed  CAS  Google Scholar 

  • Hublitz P, Albert M, Peters AH (2009) Mechanisms of transcriptional repression by histone lysine methylation. Int J Dev Biol 53(2–3):335–354

    PubMed  CAS  Google Scholar 

  • Hughes RE, Lo RS, Davis C, Strand AD, Neal CL, Olson JM, Fields S (2001) Altered transcription in yeast expressing expanded polyglutamine. Proc Natl Acad Sci USA 98(23):13201–13206

    PubMed  CAS  PubMed Central  Google Scholar 

  • Humbert S, Bryson EA, Cordelieres FP, Connors NC, Datta SR, Finkbeiner S, Greenberg ME, Saudou F (2002) The IGF-1/Akt pathway is neuroprotective in Huntington’s disease and involves Huntingtin phosphorylation by Akt. Dev Cell 2(6):831–837

    PubMed  CAS  Google Scholar 

  • Humphrey GW, Wang YH, Hirai T, Padmanabhan R, Panchision DM, Newell LF, McKay RD, Howard BH (2007) Complementary roles for histone deacetylases 1, 2, and 3 in differentiation of pluripotent stem cells. Differentiation 76:348–256

    PubMed  PubMed Central  Google Scholar 

  • Hymes KB (2010) The role of histone deacetylase inhibitors in the treatment of patients with cutaneous T-cell lymphoma. Clin Lymphoma Myeloma Leuk 10(2):98–109

    PubMed  CAS  Google Scholar 

  • Jeong H, Then F, Melia TJ Jr, Mazzulli JR, Cui L, Savas JN, Voisine C, Paganetti P, Tanese N, Hart AC, Yamamoto A, Krainc D (2009) Acetylation targets mutant huntingtin to autophagosomes for degradation. Cell 137(1):60–72

    PubMed  CAS  PubMed Central  Google Scholar 

  • Jiang H, Nucifora FC Jr, Ross CA, DeFranco DB (2003) Cell death triggered by polyglutamine-expanded huntingtin in a neuronal cell line is associated with degradation of CREB-binding protein. Hum Mol Genet 12(1):1–12

    PubMed  Google Scholar 

  • Jiang H, Poirier MA, Liang Y, Pei Z, Weiskittel CE, Smith WW, DeFranco DB, Ross CA (2006) Depletion of CBP is directly linked with cellular toxicity caused by mutant huntingtin. Neurobiol Dis 23(3):543–551

    PubMed  CAS  Google Scholar 

  • Jiang Q, Ren Y, Feng J (2008) Direct binding with histone deacetylase 6 mediates the reversible recruitment of parkin to the centrosome. J Neurosci 28(48):12993–13002

    PubMed  CAS  PubMed Central  Google Scholar 

  • Johnson CA, White DA, Lavender JS, O’Neill LP, Turner BM (2002) Human class I histone deacetylase complexes show enhanced catalytic activity in the presence of ATP and co-immunoprecipitate with the ATP-dependent chaperone protein Hsp70. J Biol Chem 277(11):9590–9597

    PubMed  CAS  Google Scholar 

  • Johnson R, Richter N, Jauch R, Gaughwin PM, Zuccato C, Cattaneo E, Stanton LW (2010) The human accelerated region 1 noncoding RNA is repressed by REST in Huntington’s disease. Physiol Genomics 41:269

    PubMed  CAS  Google Scholar 

  • Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115(6):727–738

    PubMed  CAS  Google Scholar 

  • Kazantsev A, Preisinger E, Dranovsky A, Goldgaber D, Housman D (1999) Insoluble detergent-resistant aggregates form between pathological and nonpathological lengths of polyglutamine in mammalian cells. Proc Natl Acad Sci USA 96(20):11404–11409

    PubMed  CAS  PubMed Central  Google Scholar 

  • Kegel KB, Meloni AR, Yi Y, Kim YJ, Doyle E, Cuiffo BG, Sapp E, Wang Y, Qin ZH, Chen JD, Nevins JR, Aronin N, DiFiglia M (2002) Huntingtin is present in the nucleus, interacts with the transcriptional corepressor C-terminal binding protein, and represses transcription. J Biol Chem 277(9):7466–7476

    PubMed  CAS  Google Scholar 

  • Keogh MC, Kurdistani SK, Morris SA, Ahn SH, Podolny V, Collins SR, Schuldiner M, Chin K, Punna T, Thompson NJ, Boone C, Emili A, Weissman JS, Hughes TR, Strahl BD, Grunstein M, Greenblatt JF, Buratowski S, Krogan NJ (2005) Cotranscriptional set2 methylation of histone H3 lysine 36 recruits a repressive Rpd3 complex. Cell 123(4):593–605

    PubMed  CAS  Google Scholar 

  • Khan N, Jeffers M, Kumar S, Hackett C, Boldog F, Khramtsov N, Qian X, Mills E, Berghs SC, Carey N, Finn PW, Collins LS, Tumber A, Ritchie JW, Jensen PB, Lichenstein HS, Sehested M (2008) Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem J 409(2):581–589

    PubMed  CAS  Google Scholar 

  • Khoshnan A, Ko J, Watkin EE, Paige LA, Reinhart PH, Patterson PH (2004) Activation of the IkappaB kinase complex and nuclear factor-kappaB contributes to mutant huntingtin neurotoxicity. J Neurosci 24(37):7999–8008

    PubMed  CAS  Google Scholar 

  • Khoshnan A, Ko J, Tescu S, Brundin P, Patterson PH (2009) IKKalpha and IKKbeta regulation of DNA damage-induced cleavage of huntingtin. PLoS ONE 4(6):e5768

    PubMed  PubMed Central  Google Scholar 

  • Kim MO, Chawla P, Overland RP, Xia E, Sadri-Vakili G, Cha JH (2008) Altered histone monoubiquitylation mediated by mutant huntingtin induces transcriptional dysregulation. J Neurosci 28(15):3947–3957

    PubMed  CAS  PubMed Central  Google Scholar 

  • Klement IA, Skinner PJ, Kaytor MD, Yi H, Hersch SM, Clark HB, Zoghbi HY, Orr HT (1998) Ataxin-1 nuclear localization and aggregation: role in polyglutamine- induced disease in SCA1 transgenic mice. Cell 95(1):41–53

    PubMed  CAS  Google Scholar 

  • Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J (2006) Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol 26(6):2019–2028

    PubMed  CAS  PubMed Central  Google Scholar 

  • Konishi Y, Bonni A (2003) The E2F-Cdc2 cell-cycle pathway specifically mediates activity deprivation-induced apoptosis of postmitotic neurons. J Neurosci 23(5):1649–1658

    PubMed  CAS  Google Scholar 

  • Kontopoulos E, Parvin JD, Feany MB (2006) Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum Mol Genet 15(20):3012–3023

    PubMed  CAS  Google Scholar 

  • Kovacs JJ, Murphy PJ, Gaillard S, Zhao X, Wu JT, Nicchitta CV, Yoshida M, Toft DO, Pratt WB, Yao TP (2005) HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor. Mol Cell 18(5):601–607

    PubMed  CAS  Google Scholar 

  • Krogan NJ, Kim M, Tong A, Golshani A, Cagney G, Canadien V, Richards DP, Beattie BK, Emili A, Boone C, Shilatifard A, Buratowski S, Greenblatt J (2003) Methylation of histone H3 by Set2 in Saccharomyces cerevisiae is linked to transcriptional elongation by RNA polymerase II. Mol Cell Biol 23(12):4207–4218

    PubMed  CAS  PubMed Central  Google Scholar 

  • Kubota K, Niinuma Y, Kaneko M, Okuma Y, Sugai M, Omura T, Uesugi M, Uehara T, Hosoi T, Nomura Y (2006) Suppressive effects of 4-phenylbutyrate on the aggregation of Pael receptors and endoplasmic reticulum stress. J Neurochem 97(5):1259–1268

    PubMed  CAS  Google Scholar 

  • Kuo HY, Chang CC, Jeng JC, Hu HM, Lin DY, Maul GG, Kwok RP, Shih HM (2005) SUMO modification negatively modulates the transcriptional activity of CREB-binding protein via the recruitment of Daxx. Proc Natl Acad Sci USA 102(47):16973–16978

    PubMed  CAS  PubMed Central  Google Scholar 

  • Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D (2002) Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes Dev 16(22):2893–2905

    PubMed  CAS  PubMed Central  Google Scholar 

  • Kwon S, Zhang Y, Matthias P (2007) The deacetylase HDAC6 is a novel critical component of stress granules involved in the stress response. Genes Dev 21(24):3381–3394

    PubMed  CAS  PubMed Central  Google Scholar 

  • Kyrylenko S, Kyrylenko O, Suuronen T, Salminen A (2003) Differential regulation of the Sir2 histone deacetylase gene family by inhibitors of class I and II histone deacetylases. Cell Mol Life Sci 60(9):1990–1997

    PubMed  CAS  Google Scholar 

  • Lai E, Teodoro T, Volchuk A (2007) Endoplasmic reticulum stress: signaling the unfolded protein response. Physiol Bethesda 22:193–201

    CAS  Google Scholar 

  • Lakowski B, Roelens I, Jacob S (2006) CoREST-like complexes regulate chromatin modification and neuronal gene expression. J Mol Neurosci 29(3):227–239

    PubMed  CAS  Google Scholar 

  • Lawless MW, Norris S, O’Byrne KJ, Gray SG (2009a) Targeting histone deacetylases for the treatment of disease. J Cell Mol Med 13(5):826–852

    CAS  Google Scholar 

  • Lawless MW, Norris S, O’Byrne KJ, Gray SG (2009b) Targeting histone deacetylases for the treatment of immune, endocrine & metabolic disorders. Endocr Metab Immune Disord Drug Targets 9(1):84–107

    CAS  Google Scholar 

  • Lawless MW, O’Byrne KJ, Gray SG (2009c) Oxidative stress induced lung cancer and COPD: opportunities for epigenetic therapy. J Cell Mol Med 13(9a):2800–2821

    CAS  Google Scholar 

  • Lee JS, Shilatifard A (2007) A site to remember: H3K36 methylation a mark for histone deacetylation. Mutat Res 618(1–2):130–134

    PubMed  CAS  Google Scholar 

  • 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(12):1774–1782

    PubMed  CAS  PubMed Central  Google Scholar 

  • Lee ST, Chu K, Jung KH, Im WS, Park JE, Lim HC, Won CH, Shin SH, Lee SK, Kim M, Roh JK (2009) Slowed progression in models of Huntington disease by adipose stem cell transplantation. Ann Neurol 66(5):671–681

    PubMed  CAS  Google Scholar 

  • 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(1):172–179

    PubMed  CAS  Google Scholar 

  • Li B, Howe L, Anderson S, Yates JR 3rd, Workman JL (2003) The Set2 histone methyltransferase functions through the phosphorylated carboxyl-terminal domain of RNA polymerase II. J Biol Chem 278(11):8897–8903

    PubMed  CAS  Google Scholar 

  • Li H, Xing X, Ding G, Li Q, Wang C, Xie L, Zeng R, Li Y (2009) SysPTM: a systematic resource for proteomic research on post-translational modifications. Mol Cell Proteomics 8(8):1839–1849

    PubMed  CAS  PubMed Central  Google Scholar 

  • Lim M, McKenzie K, Floyd AD, Kwon E, Zeitlin PL (2004) Modulation of deltaF508 cystic fibrosis transmembrane regulator trafficking and function with 4-phenylbutyrate and flavonoids. Am J Respir Cell Mol Biol 31(3):351–357

    PubMed  CAS  Google Scholar 

  • Liu XL, Done SC, Yan K, Kilpelainen P, Pikkarainen T, Tryggvason K (2004) Defective trafficking of nephrin missense mutants rescued by a chemical chaperone. J Am Soc Nephrol 15(7):1731–1738

    PubMed  CAS  Google Scholar 

  • Liu A, Han YR, Li J, Sun D, Ouyang M, Plummer MR, Casaccia-Bonnefil P (2007) The glial or neuronal fate choice of oligodendrocyte progenitors is modulated by their ability to acquire an epigenetic memory. J Neurosci 27(27):7339–7343

    PubMed  CAS  PubMed Central  Google Scholar 

  • Liu H, Hu Q, Kaufman A, D’Ercole AJ, Ye P (2008) Developmental expression of histone deacetylase 11 in the murine brain. J Neurosci Res 86(3):537–543

    PubMed  CAS  PubMed Central  Google Scholar 

  • Lorincz MT, Zawistowski VA (2009) Expanded CAG repeats in the murine Huntington’s disease gene increases neuronal differentiation of embryonic and neural stem cells. Mol Cell Neurosci 40(1):1–13

    PubMed  CAS  PubMed Central  Google Scholar 

  • Luo S, Vacher C, Davies JE, Rubinsztein DC (2005) Cdk5 phosphorylation of huntingtin reduces its cleavage by caspases: implications for mutant huntingtin toxicity. J Cell Biol 169(4):647–656

    PubMed  CAS  PubMed Central  Google Scholar 

  • Lyssiotis CA, Walker J, Wu C, Kondo T, Schultz PG, Wu X (2007) Inhibition of histone deacetylase activity induces developmental plasticity in oligodendrocyte precursor cells. Proc Natl Acad Sci USA 104(38):14982–14987

    PubMed  CAS  PubMed Central  Google Scholar 

  • MacDonald JL, Roskams AJ (2008) Histone deacetylases 1 and 2 are expressed at distinct stages of neuro-glial development. Dev Dyn 237(8):2256–2267

    PubMed  Google Scholar 

  • Mankan AK, Lawless MW, Gray SG, Kelleher D, McManus R (2009) NF-kappaB regulation: the nuclear response. J Cell Mol Med 13(4):631–643

    PubMed  CAS  PubMed Central  Google Scholar 

  • Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R (2007a) FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12(10):1247–1252

    CAS  Google Scholar 

  • Mann BS, Johnson JR, He K, Sridhara R, Abraham S, Booth BP, Verbois L, Morse DE, Jee JM, Pope S, Harapanhalli RS, Dagher R, Farrell A, Justice R, Pazdur R (2007b) Vorinostat for treatment of cutaneous manifestations of advanced primary cutaneous T-cell lymphoma. Clin Cancer Res 13(8):2318–2322

    CAS  Google Scholar 

  • Matthias P, Yoshida M, Khochbin S (2008) HDAC6 a new cellular stress surveillance factor. Cell Cycle 7(1):7–10

    PubMed  CAS  Google Scholar 

  • Maynard CJ, Bottcher C, Ortega Z, Smith R, Florea BI, Diaz-Hernandez M, Brundin P, Overkleeft HS, Li JY, Lucas JJ, Dantuma NP (2009) Accumulation of ubiquitin conjugates in a polyglutamine disease model occurs without global ubiquitin/proteasome system impairment. Proc Natl Acad Sci USA 106(33):13986–13991

    PubMed  CAS  PubMed Central  Google Scholar 

  • Menendez-Benito V, Verhoef LG, Masucci MG, Dantuma NP (2005) Endoplasmic reticulum stress compromises the ubiquitin–proteasome system. Hum Mol Genet 14(19):2787–2799

    PubMed  CAS  Google Scholar 

  • Montgomery RL, Hsieh J, Barbosa AC, Richardson JA, Olson EN (2009) Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci USA 106(19):7876–7881

    PubMed  CAS  PubMed Central  Google Scholar 

  • Morris SA, Shibata Y, Noma K, Tsukamoto Y, Warren E, Temple B, Grewal SI, Strahl BD (2005) Histone H3 K36 methylation is associated with transcription elongation in Schizosaccharomyces pombe. Eukaryot Cell 4(8):1446–1454

    PubMed  CAS  PubMed Central  Google Scholar 

  • Moser K, Biglan K, Ross C, Langbehn D, Aylward E, Stout J, Queller S, Carlozzi N, Duff K, Beglinger L, Paulsen J, Tomusk A, Lifer S, Hastings S, Dawson J, Walker B, Whitlock K, Johnson S, Pacifici R, Hersch S, Dorsey E, Katz R, Tempkin T, Wheelock V, Schwartz G, Corey-Bloom J, Mattis P, Feigin A, Young P, McArthur D, Perlman S, Higginson C, Carr L, Sigvardt K, Chirieac M, Shinaman A, Shoulson I, Kane A, Peavy G, Goldstein J, Jacobson M, Lessig S, Wasserman L, Kayson E, Tang C, Zgaljardic D, Ma Y, Dhawan V, Guttman M, Eidelberg D, Peng S, Kingsley P, Rosas H, Gevorkian S, Oakes D, Matson W, Massood T, Latourelle J, Mysore J, Fossale E, Gillis T, Gusella J, MacDonald M, Myers R, Yastrubetskaya O, Preston J, Chiu E, Goh A, Oster E, Bausch J, Kayson E, Quaid K, Sims S, Swenson M, Harrison J, Moskowitz C, Stepanov N, Suter G, Westphal B, Johnson S, Langbehn D, Paulsen J, Nopoulos P, Beglinger L, Johnson H, Magnotta V, Pierson R, Lipe H, Bird T, McCusker E, Lownie A, Lechich A, Montas S, Duckett A, Klager J, Sandler S, Pae A, Apostol B, Simmons D, Zuccato C, Illes K, Pallos J, Casale M, Kathuria S, Cattaneo E, Marsh J, Thompson L, Patzke H, Chesworth R, Li Z, Rahil G, Wang J, Smith J, Huet F, Shapiro G, Leit S, Beaulieu P, Raeppel F, Fournel M, Sainte-Croix H, Nolan S, Albayya F, Barbier A, Besterman J, Ahlijanian M, Deziel R, Aubeeluck A, Buchanan H, Ross C, Biglan K, Landwehrmeyer B, Whitlock K, Carlozzi N, Mickes L, Lee J, Kim R, Di Toro B, Fine E, Cahill T, Johnson D, Goldstein J, Peavy G, Jacobson M, Goodman L, Como P, Cha J, Beck C, Adams M, Chadwick G, de Blieck E, McCallum C, Deuel L, Clarke A, Stewart R, Adams W, Paulson H, Fiedorowicz J, Hanson J, Ramza N, Priller J, Ecker D (2008) Inaugural Huntington disease clinical research symposium organized by the Huntington Study Group. Neurotherapeutics 5(2):362–375

    Google Scholar 

  • Muchowski PJ, Ning K, D’Souza-Schorey C, Fields S (2002) Requirement of an intact microtubule cytoskeleton for aggregation and inclusion body formation by a mutant huntingtin fragment. Proc Natl Acad Sci USA 99(2):727–732

    PubMed  CAS  PubMed Central  Google Scholar 

  • Mulhern ML, Madson CJ, Kador PF, Randazzo J, Shinohara T (2007) Cellular osmolytes reduce lens epithelial cell death and alleviate cataract formation in galactosemic rats. Mol Vis 13:1397–1405

    PubMed  CAS  Google Scholar 

  • Naruse Y, Aoki T, Kojima T, Mori N (1999) Neural restrictive silencer factor recruits mSin3 and histone deacetylase complex to repress neuron-specific target genes. Proc Natl Acad Sci USA 96(24):13691–13696

    PubMed  CAS  PubMed Central  Google Scholar 

  • Noh JY, Lee H, Song S, Kim NS, Im W, Kim M, Seo H, Chung CW, Chang JW, Ferrante RJ, Yoo YJ, Ryu H, Jung YK (2009) SCAMP5 links endoplasmic reticulum stress to the accumulation of expanded polyglutamine protein aggregates via endocytosis inhibition. J Biol Chem 284(17):11318–11325

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(5512):2423–2428

    PubMed  CAS  Google Scholar 

  • Ohoka N, Hattori T, Kitagawa M, Onozaki K, Hayashi H (2007) Critical and functional regulation of CHOP (C/EBP homologous protein) through the N-terminal portion. J Biol Chem 282(49):35687–35694

    PubMed  CAS  Google Scholar 

  • Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, Gorgun CZ, Hotamisligil GS (2006) Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 313(5790):1137–1140

    PubMed  Google Scholar 

  • Packer AN, Xing Y, Harper SQ, Jones L, Davidson BL (2008) The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington’s disease. J Neurosci 28(53):14341–14346

    PubMed  CAS  PubMed Central  Google Scholar 

  • Pahl HL, Baeuerle PA (1997) The ER-overload response: activation of NF-kappa B. Trends Biochem Sci 22(2):63–67

    PubMed  CAS  Google Scholar 

  • Pandey UB, Nie Z, Batlevi Y, McCray BA, Ritson GP, Nedelsky NB, Schwartz SL, DiProspero NA, Knight MA, Schuldiner O, Padmanabhan R, Hild M, Berry DL, Garza D, Hubbert CC, Yao TP, Baehrecke EH, Taylor JP (2007) HDAC6 rescues neurodegeneration and provides an essential link between autophagy and the UPS. Nature 447(7146):859–863

    PubMed  CAS  Google Scholar 

  • Paquette AJ, Perez SE, Anderson DJ (2000) Constitutive expression of the neuron-restrictive silencer factor (NRSF)/REST in differentiating neurons disrupts neuronal gene expression and causes axon pathfinding errors in vivo. Proc Natl Acad Sci USA 97(22):12318–12323

    PubMed  CAS  PubMed Central  Google Scholar 

  • Pardo R, Colin E, Regulier E, Aebischer P, Deglon N, Humbert S, Saudou F (2006) Inhibition of calcineurin by FK506 protects against polyglutamine-huntingtin toxicity through an increase of huntingtin phosphorylation at S421. J Neurosci 26(5):1635–1645

    PubMed  CAS  Google Scholar 

  • Park J, Lee JH, La M, Jang MJ, Chae GW, Kim SB, Tak H, Jung Y, Byun B, Ahn JK, Joe CO (2007) Inhibition of NF-kappaB acetylation and its transcriptional activity by Daxx. J Mol Biol 368(2):388–397

    PubMed  CAS  Google Scholar 

  • Pearce I, Heathfield KW, Pearce MJ (1977) Valproate sodium in Huntington chorea. Arch Neurol 34(5):308–309

    PubMed  CAS  Google Scholar 

  • Pelegri C, Duran-Vilaregut J, del Valle J, Crespo-Biel N, Ferrer I, Pallas M, Camins A, Vilaplana J (2008) Cell cycle activation in striatal neurons from Huntington’s disease patients and rats treated with 3-nitropropionic acid. Int J Dev Neurosci 26(7):665–671

    PubMed  CAS  Google Scholar 

  • Perez M, Santa-Maria I, Gomez de Barreda E, Zhu X, Cuadros R, Cabrero JR, Sanchez-Madrid F, Dawson HN, Vitek MP, Perry G, Smith MA, Avila J (2009) Tau—an inhibitor of deacetylase HDAC6 function. J Neurochem 109(6):1756–1766

    PubMed  CAS  Google Scholar 

  • Perkins ND, Felzien LK, Betts JC, Leung K, Beach DH, Nabel GJ (1997) Regulation of NF-kappaB by cyclin-dependent kinases associated with the p300 coactivator. Science 275(5299):523–527

    PubMed  CAS  Google Scholar 

  • Prozorovski T, Schulze-Topphoff U, Glumm R, Baumgart J, Schroter F, Ninnemann O, Siegert E, Bendix I, Brustle O, Nitsch R, Zipp F, Aktas O (2008) Sirt1 contributes critically to the redox-dependent fate of neural progenitors. Nat Cell Biol 10(4):385–394

    PubMed  CAS  Google Scholar 

  • Qi X, Hosoi T, Okuma Y, Kaneko M, Nomura Y (2004) Sodium 4-phenylbutyrate protects against cerebral ischemic injury. Mol Pharmacol 66(4):899–908

    PubMed  CAS  Google Scholar 

  • Quinti L, Chopra V, Rotili D, Valente S, Amore A, Franci G, Meade S, Valenza M, Altucci L, Maxwell MM, Cattaneo E, Hersch S, Mai A, Kazantsev A (2010) Evaluation of histone deacetylases as drug targets in Huntington’s disease models. Study of HDACs in brain tissues from R6/2 and CAG140 knock-in HD mouse models and human patients and in a neuronal HD cell model. PLoS Curr 2: RRN1172. doi:10.1371/currents.RRN1172

  • Rangone H, Poizat G, Troncoso J, Ross CA, MacDonald ME, Saudou F, Humbert S (2004) The serum- and glucocorticoid-induced kinase SGK inhibits mutant huntingtin-induced toxicity by phosphorylating serine 421 of huntingtin. Eur J Neurosci 19(2):273–279

    PubMed  Google Scholar 

  • Rao RV, Ellerby HM, Bredesen DE (2004) Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ 11(4):372–380

    PubMed  CAS  Google Scholar 

  • Rapoport TA (2007) Protein translocation across the eukaryotic endoplasmic reticulum and bacterial plasma membranes. Nature 450(7170):663–669

    PubMed  CAS  Google Scholar 

  • Reijonen S, Putkonen N, Norremolle A, Lindholm D, Korhonen L (2008) Inhibition of endoplasmic reticulum stress counteracts neuronal cell death and protein aggregation caused by N-terminal mutant huntingtin proteins. Exp Cell Res 314(5):950–960

    PubMed  CAS  Google Scholar 

  • Renthal W, Maze I, Krishnan V, Covington HE 3rd, Xiao G, Kumar A, Russo SJ, Graham A, Tsankova N, Kippin TE, Kerstetter KA, Neve RL, Haggarty SJ, McKinsey TA, Bassel-Duby R, Olson EN, Nestler EJ (2007) Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli. Neuron 56(3):517–529

    PubMed  CAS  Google Scholar 

  • Ron D, Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8(7):519–529

    PubMed  CAS  Google Scholar 

  • Roopra A, Sharling L, Wood IC, Briggs T, Bachfischer U, Paquette AJ, Buckley NJ (2000) Transcriptional repression by neuron-restrictive silencer factor is mediated via the SIN3-histone deacetylase complex. Mol Cell Biol 20(6):2147–2157

    PubMed  CAS  PubMed Central  Google Scholar 

  • Roze E, Betuing S, Deyts C, Marcon E, Brami-Cherrier K, Pages C, Humbert S, Merienne K, Caboche J (2008) Mitogen- and stress-activated protein kinase-1 deficiency is involved in expanded-huntingtin-induced transcriptional dysregulation and striatal death. FASEB J 22(4):1083–1093

    PubMed  CAS  Google Scholar 

  • Rubenstein RC, Zeitlin PL (1998) A pilot clinical trial of oral sodium 4-phenylbutyrate (Buphenyl) in deltaF508-homozygous cystic fibrosis patients: partial restoration of nasal epithelial CFTR function. Am J Respir Crit Care Med 157(2):484–490

    PubMed  CAS  Google Scholar 

  • Rubenstein RC, Zeitlin PL (2000) Sodium 4-phenylbutyrate downregulates Hsc70: implications for intracellular trafficking of DeltaF508-CFTR. Am J Physiol Cell Physiol 278(2):C259–C267

    PubMed  CAS  Google Scholar 

  • Rubenstein RC, Lyons BM (2001) Sodium 4-phenylbutyrate downregulates HSC70 expression by facilitating mRNA degradation. Am J Physiol Lung Cell Mol Physiol 281(1):L43–L51

    PubMed  CAS  Google Scholar 

  • Rubenstein RC, Egan ME, Zeitlin PL (1997) In vitro pharmacologic restoration of CFTR-mediated chloride transport with sodium 4-phenylbutyrate in cystic fibrosis epithelial cells containing delta F508-CFTR. J Clin Invest 100(10):2457–2465

    PubMed  CAS  PubMed Central  Google Scholar 

  • Rutkowski DT, Kaufman RJ (2004) A trip to the ER: coping with stress. Trends Cell Biol 14(1):20–28

    PubMed  CAS  Google Scholar 

  • Ryu H, Lee J, Olofsson BA, Mwidau A, Deodeoglu A, Escudero M, Flemington E, Azizkhan-Clifford J, Ferrante RJ, Ratan RR (2003) Histone deacetylase inhibitors prevent oxidative neuronal death independent of expanded polyglutamine repeats via an Sp1-dependent pathway. Proc Natl Acad Sci USA 100(7):4281–4286

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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) trimethylation in Huntington’s disease. Proc Natl Acad Sci USA 103(50):19176–19181

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(11):1293–1306

    PubMed  CAS  Google Scholar 

  • Saft C, Lauter T, Kraus PH, Przuntek H, Andrich JE (2006) Dose-dependent improvement of myoclonic hyperkinesia due to Valproic acid in eight Huntington’s disease patients: a case series. BMC Neurol 6:11

    PubMed  PubMed Central  Google Scholar 

  • Said TK, Moraes RC, Sinha R, Medina D (2001) Mechanisms of suberoylanilide hydroxamic acid inhibition of mammary cell growth. Breast Cancer Res 3(2):122–133

    PubMed  CAS  PubMed Central  Google Scholar 

  • Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, Gage FH, Glass CK (2009) A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell 137(1):47–59

    PubMed  CAS  PubMed Central  Google Scholar 

  • Salazar Z, Tschopp L, Calandra C, Micheli F (2008) Pisa syndrome and parkinsonism secondary to valproic acid in Huntington’s disease. Mov Disord 23(16):2430–2431

    PubMed  Google Scholar 

  • Sarkander HI, Fleischer-Lambropoulos H, Brade WP (1975) A comparative study of histone acetylation in neuronal and glial nuclei enriched rat brain fractions. FEBS Lett 52(1):40–43

    PubMed  CAS  Google Scholar 

  • Saudou F, Finkbeiner S, Devys D, Greenberg ME (1998) Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 95(1):55–66

    PubMed  CAS  Google Scholar 

  • Schaft D, Roguev A, Kotovic KM, Shevchenko A, Sarov M, Shevchenko A, Neugebauer KM, Stewart AF (2003) The histone 3 lysine 36 methyltransferase, SET2, is involved in transcriptional elongation. Nucleic Acids Res 31(10):2475–2482

    PubMed  CAS  PubMed Central  Google Scholar 

  • Schilling B, Gafni J, Torcassi C, Cong X, Row RH, LaFevre-Bernt MA, Cusack MP, Ratovitski T, Hirschhorn R, Ross CA, Gibson BW, Ellerby LM (2006) Huntingtin phosphorylation sites mapped by mass spectrometry. Modulation of cleavage and toxicity. J Biol Chem 281(33):23686–23697

    PubMed  CAS  Google Scholar 

  • Schwab C, Klegeris A, McGeer PL (2010) Inflammation in transgenic mouse models of neurodegenerative disorders. Biochim Biophys Acta 1802(10):889–902

    PubMed  CAS  Google Scholar 

  • Scroggins BT, Robzyk K, Wang D, Marcu MG, Tsutsumi S, Beebe K, Cotter RJ, Felts S, Toft D, Karnitz L, Rosen N, Neckers L (2007) An acetylation site in the middle domain of Hsp90 regulates chaperone function. Mol Cell 25(1):151–159

    PubMed  CAS  PubMed Central  Google Scholar 

  • Seong IS, Woda JM, Song JJ, Lloret A, Abeyrathne PD, Woo CJ, Gregory G, Lee JM, Wheeler VC, Walz T, Kingston RE, Gusella JF, Conlon RA, Macdonald ME (2010) Huntingtin facilitates Polycomb repressive complex 2. Hum Mol Genet 19(4):573–583

    PubMed  CAS  PubMed Central  Google Scholar 

  • Seth KA, Majzoub JA (2001) Repressor element silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) can act as an enhancer as well as a repressor of corticotropin-releasing hormone gene transcription. J Biol Chem 276(17):13917–13923

    PubMed  CAS  Google Scholar 

  • Shao L, Young LT, Wang JF (2005) Chronic treatment with mood stabilizers lithium and valproate prevents excitotoxicity by inhibiting oxidative stress in rat cerebral cortical cells. Biol Psychiatry 58(11):879–884

    PubMed  CAS  Google Scholar 

  • Shao L, Sun X, Xu L, Young LT, Wang JF (2006) Mood stabilizing drug lithium increases expression of endoplasmic reticulum stress proteins in primary cultured rat cerebral cortical cells. Life Sci 78(12):1317–1323

    PubMed  CAS  Google Scholar 

  • Sharma K, Sheng HZ, Lettieri K, Li H, Karavanov A, Potter S, Westphal H, Pfaff SL (1998) LIM homeodomain factors Lhx3 and Lhx4 assign subtype identities for motor neurons. Cell 95(6):817–828

    PubMed  CAS  Google Scholar 

  • Sieradzan KA, Mechan AO, Jones L, Wanker EE, Nukina N, Mann DM (1999) Huntington’s disease intranuclear inclusions contain truncated, ubiquitinated huntingtin protein. Exp Neurol 156(1):92–99

    PubMed  CAS  Google Scholar 

  • Sims RJ 3rd, Reinberg D (2008) Is there a code embedded in proteins that is based on post-translational modifications? Nat Rev Mol Cell Biol 9(10):815–820

    PubMed  CAS  Google Scholar 

  • Singh OV, Vij N, Mogayzel PJ Jr, Jozwik C, Pollard HB, Zeitlin PL (2006) Pharmacoproteomics of 4-phenylbutyrate-treated IB3-1 cystic fibrosis bronchial epithelial cells. J Proteome Res 5(3):562–571

    PubMed  CAS  Google Scholar 

  • Sirianni N, Naidu S, Pereira J, Pillotto RF, Hoffman EP (1998) Rett syndrome: confirmation of X-linked dominant inheritance, and localization of the gene to Xq28. Am J Hum Genet 63(5):1552–1558

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(10):1164–1175

    PubMed  CAS  Google Scholar 

  • Steffan JS, Kazantsev A, Spasic-Boskovic O, Greenwald M, Zhu YZ, Gohler H, Wanker EE, Bates GP, Housman DE, Thompson LM (2000) The Huntington’s disease protein interacts with p53 and CREB-binding protein and represses transcription. Proc Natl Acad Sci USA 97(12):6763–6768

    PubMed  CAS  PubMed Central  Google Scholar 

  • 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(6857):739–743

    PubMed  CAS  Google Scholar 

  • Steffan JS, Agrawal N, Pallos J, Rockabrand E, Trotman LC, Slepko N, Illes K, Lukacsovich T, Zhu YZ, Cattaneo E, Pandolfi PP, Thompson LM, Marsh JL (2004) SUMO modification of Huntingtin and Huntington’s disease pathology. Science 304(5667):100–104

    PubMed  CAS  Google Scholar 

  • Su X, Kameoka S, Lentz S, Majumder S (2004) Activation of REST/NRSF target genes in neural stem cells is sufficient to cause neuronal differentiation. Mol Cell Biol 24(18):8018–8025

    PubMed  CAS  PubMed Central  Google Scholar 

  • Subramaniam S, Sixt KM, Barrow R, Snyder SH (2009) Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity. Science 324(5932):1327–1330

    PubMed  CAS  PubMed Central  Google Scholar 

  • Sun XJ, Wei J, Wu XY, Hu M, Wang L, Wang HH, Zhang QH, Chen SJ, Huang QH, Chen Z (2005a) Identification and characterization of a novel human histone H3 lysine 36-specific methyltransferase. J Biol Chem 280(42):35261–35271

    CAS  Google Scholar 

  • Sun YM, Greenway DJ, Johnson R, Street M, Belyaev ND, Deuchars J, Bee T, Wilde S, Buckley NJ (2005b) Distinct profiles of REST interactions with its target genes at different stages of neuronal development. Mol Biol Cell 16(12):5630–5638

    CAS  Google Scholar 

  • 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 USA 104(39):15282–15287

    PubMed  CAS  PubMed Central  Google Scholar 

  • Suzuki T, Yokozaki H, Kuniyasu H, Hayashi K, Naka K, Ono S, Ishikawa T, Tahara E, Yasui W (2000) Effect of trichostatin A on cell growth and expression of cell cycle- and apoptosis-related molecules in human gastric and oral carcinoma cell lines. Int J Cancer 88(6):992–997

    PubMed  CAS  Google Scholar 

  • Symington GR, Leonard DP, Shannon PJ, Vajda FJ (1978) Sodium valproate in Huntington’s disease. Am J Psychiatry 135(3):352–354

    PubMed  CAS  Google Scholar 

  • Taherzadeh-Fard E, Saft C, Andrich J, Wieczorek S, Arning L (2009) PGC-1alpha as modifier of onset age in Huntington disease. Mol Neurodegener 4:10

    PubMed  PubMed Central  Google Scholar 

  • Tapia-Ramirez J, Eggen BJ, Peral-Rubio MJ, Toledo-Aral JJ, Mandel G (1997) A single zinc finger motif in the silencing factor REST represses the neural-specific type II sodium channel promoter. Proc Natl Acad Sci USA 94(4):1177–1182

    PubMed  CAS  PubMed Central  Google Scholar 

  • Thiel G, Lietz M, Cramer M (1998) Biological activity and modular structure of RE-1-silencing transcription factor (REST), a repressor of neuronal genes. J Biol Chem 273(41):26891–26899

    PubMed  CAS  Google Scholar 

  • 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 USA 105(40):15564–15569

    PubMed  CAS  PubMed Central  Google Scholar 

  • Thompson LM, Aiken CT, Kaltenbach LS, Agrawal N, Illes K, Khoshnan A, Martinez-Vincente M, Arrasate M, O’Rourke JG, Khashwji H, Lukacsovich T, Zhu YZ, Lau AL, Massey A, Hayden MR, Zeitlin SO, Finkbeiner S, Green KN, LaFerla FM, Bates G, Huang L, Patterson PH, Lo DC, Cuervo AM, Marsh JL, Steffan JS (2009) IKK phosphorylates Huntingtin and targets it for degradation by the proteasome and lysosome. J Cell Biol 187(7):1083–1099

    PubMed  CAS  PubMed Central  Google Scholar 

  • Trushina E, Heldebrant MP, Perez-Terzic CM, Bortolon R, Kovtun IV, Badger JD 2nd, Terzic A, Estevez A, Windebank AJ, Dyer RB, Yao J, McMurray CT (2003) Microtubule destabilization and nuclear entry are sequential steps leading to toxicity in Huntington’s disease. Proc Natl Acad Sci USA 100(21):12171–12176

    PubMed  CAS  PubMed Central  Google Scholar 

  • Vanden Berghe W, De Bosscher K, Boone E, Plaisance S, Haegeman G (1999) The nuclear factor-kappa B engages CBP/p300 and histone acetyltransferase activity for transcriptional activation of the interleukin-6 gene promoter. J Biol Chem 274(45):32091–32098

    PubMed  CAS  Google Scholar 

  • Vembar SS, Brodsky JL (2008) One step at a time: endoplasmic reticulum-associated degradation. Nat Rev Mol Cell Biol 9(12):944–957

    PubMed  CAS  PubMed Central  Google Scholar 

  • Vidal R, Caballero B, Couve A, Hetz C (2011) Converging pathways in the occurrence of endoplasmic reticulum (ER) stress in Huntington’s disease. Curr Mol Med 11(1):1–12

    PubMed  CAS  Google Scholar 

  • Vij N, Fang S, Zeitlin PL (2006) Selective inhibition of endoplasmic reticulum-associated degradation rescues DeltaF508-cystic fibrosis transmembrane regulator and suppresses interleukin-8 levels: therapeutic implications. J Biol Chem 281(25):17369–17378

    PubMed  CAS  Google Scholar 

  • Vilatoba M, Eckstein C, Bilbao G, Smyth CA, Jenkins S, Thompson JA, Eckhoff DE, Contreras JL (2005) Sodium 4-phenylbutyrate protects against liver ischemia reperfusion injury by inhibition of endoplasmic reticulum-stress mediated apoptosis. Surgery 138(2):342–351

    PubMed  Google Scholar 

  • Wadgaonkar R, Phelps KM, Haque Z, Williams AJ, Silverman ES, Collins T (1999) CREB-binding protein is a nuclear integrator of nuclear factor-kappaB and p53 signaling. J BiolChem 274(4):1879–1882

    CAS  Google Scholar 

  • Waelter S, Boeddrich A, Lurz R, Scherzinger E, Lueder G, Lehrach H, Wanker EE (2001) Accumulation of mutant huntingtin fragments in aggresome-like inclusion bodies as a result of insufficient protein degradation. Mol Biol Cell 12(5):1393–1407

    PubMed  CAS  PubMed Central  Google Scholar 

  • Walker FO (2007) Huntington’s disease. Lancet 369(9557):218–228

    PubMed  CAS  Google Scholar 

  • Wang JF, Bown C, Young LT (1999) Differential display PCR reveals novel targets for the mood-stabilizing drug valproate including the molecular chaperone GRP78. Mol Pharmacol 55(3):521–527

    PubMed  CAS  Google Scholar 

  • Wang JF, Bown CD, Chen B, Young LT (2001) Identification of mood stabilizer-regulated genes by differential-display PCR. Int J Neuropsychopharmacol 4(1):65–74

    PubMed  CAS  Google Scholar 

  • Wang JF, Azzam JE, Young LT (2003) Valproate inhibits oxidative damage to lipid and protein in primary cultured rat cerebrocortical cells. Neuroscience 116(2):485–489

    PubMed  CAS  Google Scholar 

  • Wang J, Scully K, Zhu X, Cai L, Zhang J, Prefontaine GG, Krones A, Ohgi KA, Zhu P, Garcia-Bassets I, Liu F, Taylor H, Lozach J, Jayes FL, Korach KS, Glass CK, Fu XD, Rosenfeld MG (2007a) Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 446(7138):882–887

    CAS  Google Scholar 

  • Wang Y, Wang SY, Zhang XH, Zhao M, Hou CM, Xu YJ, Du ZY, Yu XD (2007b) FK228 inhibits Hsp90 chaperone function in K562 cells via hyperacetylation of Hsp70. Biochem Biophys Res Commun 356(4):998–1003

    CAS  Google Scholar 

  • Wang J, Wang CE, Orr A, Tydlacka S, Li SH, Li XJ (2008) Impaired ubiquitin–proteasome system activity in the synapses of Huntington’s disease mice. J Cell Biol 180(6):1177–1189

    PubMed  CAS  PubMed Central  Google Scholar 

  • Warby SC, Doty CN, Graham RK, Shively J, Singaraja RR, Hayden MR (2009) Phosphorylation of huntingtin reduces the accumulation of its nuclear fragments. Mol Cell Neurosci 40(2):121–127

    PubMed  CAS  Google Scholar 

  • Watanabe H, Mizutani T, Haraguchi T, Yamamichi N, Minoguchi S, Yamamichi-Nishina M, Mori N, Kameda T, Sugiyama T, Iba H (2006) SWI/SNF complex is essential for NRSF-mediated suppression of neuronal genes in human nonsmall cell lung carcinoma cell lines. Oncogene 25(3):470–479

    PubMed  CAS  Google Scholar 

  • Watanabe N, Lam E (2007) BAX inhibitor-1 modulates endoplasmic reticulum stress-mediated programmed cell death in Arabidopsis. J Biol Chem 283(6):3200–3210

    PubMed  Google Scholar 

  • 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(8):847–854

    PubMed  CAS  Google Scholar 

  • Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler TK, Strand AD, Cui L, Beyer RP, Easley CN, Smith AC, Krainc D, Luquet S, Sweet IR, Schwartz MW, La Spada AR (2006) Thermoregulatory and metabolic defects in Huntington’s disease transgenic mice implicate PGC-1alpha in Huntington’s disease neurodegeneration. Cell Metab 4(5):349–362

    PubMed  CAS  Google Scholar 

  • Weydt P, Soyal SM, Gellera C, Didonato S, Weidinger C, Oberkofler H, Landwehrmeyer GB, Patsch W (2009) The gene coding for PGC-1alpha modifies age at onset in Huntington’s disease. Mol Neurodegener 4:3

    PubMed  PubMed Central  Google Scholar 

  • Wiencke JK, Zheng S, Morrison Z, Yeh RF (2008) Differentially expressed genes are marked by histone 3 lysine 9 trimethylation in human cancer cells. Oncogene 27(17):2412–2421

    PubMed  CAS  Google Scholar 

  • Wu J, Xie X (2006) Comparative sequence analysis reveals an intricate network among REST, CREB and miRNA in mediating neuronal gene expression. Genome Biol 7(9):R85

    PubMed  PubMed Central  Google Scholar 

  • Wu L, Timmers C, Maiti B, Saavedra HI, Sang L, Chong GT, Nuckolls F, Giangrande P, Wright FA, Field SJ, Greenberg ME, Orkin S, Nevins JR, Robinson ML, Leone G (2001) The E2F1-3 transcription factors are essential for cellular proliferation. Nature 414(6862):457–462

    PubMed  CAS  Google Scholar 

  • Wyce A, Xiao T, Whelan KA, Kosman C, Walter W, Eick D, Hughes TR, Krogan NJ, Strahl BD, Berger SL (2007) H2B ubiquitylation acts as a barrier to Ctk1 nucleosomal recruitment prior to removal by Ubp8 within a SAGA-related complex. Mol Cell 27(2):275–288

    PubMed  CAS  Google Scholar 

  • Xia Q, Cheng D, Duong DM, Gearing M, Lah JJ, Levey AI, Peng J (2008) Phosphoproteomic analysis of human brain by calcium phosphate precipitation and mass spectrometry. J Proteome Res 7(7):2845–2851

    PubMed  CAS  PubMed Central  Google Scholar 

  • Xiao T, Hall H, Kizer KO, Shibata Y, Hall MC, Borchers CH, Strahl BD (2003) Phosphorylation of RNA polymerase II CTD regulates H3 methylation in yeast. Genes Dev 17(5):654–663

    PubMed  CAS  PubMed Central  Google Scholar 

  • Xu C, Bailly-Maitre B, Reed JC (2005) Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 115(10):2656–2664

    PubMed  CAS  PubMed Central  Google Scholar 

  • Yam GH, Gaplovska-Kysela K, Zuber C, Roth J (2007a) Sodium 4-phenylbutyrate acts as a chemical chaperone on misfolded myocilin to rescue cells from endoplasmic reticulum stress and apoptosis. Invest Ophthalmol Vis Sci 48(4):1683–1690

    Google Scholar 

  • Yam GH, Roth J, Zuber C (2007b) 4-Phenylbutyrate rescues trafficking incompetent mutant alpha-galactosidase A without restoring its functionality. Biochem Biophys Res Commun 360(2):375–380

    CAS  Google Scholar 

  • Yanai A, Huang K, Kang R, Singaraja RR, Arstikaitis P, Gan L, Orban PC, Mullard A, Cowan CM, Raymond LA, Drisdel RC, Green WN, Ravikumar B, Rubinsztein DC, El-Husseini A, Hayden MR (2006) Palmitoylation of huntingtin by HIP14 is essential for its trafficking and function. Nat Neurosci 9(6):824–831

    PubMed  CAS  PubMed Central  Google Scholar 

  • Yang H, Liu C, Zhong Y, Luo S, Monteiro MJ, Fang S (2010) Huntingtin interacts with the cue domain of gp78 and inhibits gp78 binding to ubiquitin and p97/VCP. PLoS ONE 5(1):e8905

    PubMed  PubMed Central  Google Scholar 

  • Yao X, Zhang JR, Huang HR, Dai LC, Liu QJ, Zhang M (2010) Histone deacetylase inhibitor promotes differentiation of embryonic stem cells into neural cells in adherent monoculture. Chin Med J Engl 123(6):734–738

    PubMed  CAS  Google Scholar 

  • Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, Mayo MW (2004) Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 23(12):2369–2380

    PubMed  CAS  PubMed Central  Google Scholar 

  • Yohrling GJ, Farrell LA, Hollenberg AN, Cha JH (2003) Mutant huntingtin increases nuclear corepressor function and enhances ligand-dependent nuclear hormone receptor activation. Mol Cell Neurosci 23(1):28–38

    PubMed  CAS  Google Scholar 

  • You A, Tong JK, Grozinger CM, Schreiber SL (2001) CoREST is an integral component of the CoREST–human histone deacetylase complex. Proc Natl Acad Sci USA 98(4):1454–1458

    PubMed  CAS  PubMed Central  Google Scholar 

  • Yu J, Angelin-Duclos C, Greenwood J, Liao J, Calame K (2000) Transcriptional repression by blimp-1 (PRDI-BF1) involves recruitment of histone deacetylase. Mol Cell Biol 20(7):2592–2603

    PubMed  CAS  PubMed Central  Google Scholar 

  • Yu IT, Park JY, Kim SH, Lee JS, Kim YS, Son H (2009) Valproic acid promotes neuronal differentiation by induction of proneural factors in association with H4 acetylation. Neuropharmacology 56(2):473–480

    PubMed  CAS  Google Scholar 

  • Yuan W, Xie J, Long C, Erdjument-Bromage H, Ding X, Zheng Y, Tempst P, Chen S, Zhu B, Reinberg D (2009) Heterogeneous nuclear ribonucleoprotein L Is a subunit of human KMT3a/Set2 complex required for H3 Lys-36 trimethylation activity in vivo. J Biol Chem 284(23):15701–15707

    PubMed  CAS  PubMed Central  Google Scholar 

  • Zadori D, Geisz A, Vamos E, Vecsei L, Klivenyi P (2009) Valproate ameliorates the survival and the motor performance in a transgenic mouse model of Huntington’s disease. Pharmacol Biochem Behav 94(1):148–153

    PubMed  CAS  Google Scholar 

  • Zala D, Colin E, Rangone H, Liot G, Humbert S, Saudou F (2008) Phosphorylation of mutant huntingtin at S421 restores anterograde and retrograde transport in neurons. Hum Mol Genet 17(24):3837–3846

    PubMed  CAS  Google Scholar 

  • Zeitlin PL, Diener-West M, Rubenstein RC, Boyle MP, Lee CK, Brass-Ernst L (2002) Evidence of CFTR function in cystic fibrosis after systemic administration of 4-phenylbutyrate. Mol Ther 6(1):119–126

    PubMed  CAS  Google Scholar 

  • Zhang J, Yang Y, Yang T, Liu Y, Li A, Fu S, Wu M, Pan Z, Zhou W (2010) microRNA-22, downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br J Cancer 103(8):1215–1220

    PubMed  CAS  PubMed Central  Google Scholar 

  • Zibetti C, Adamo A, Binda C, Forneris F, Toffolo E, Verpelli C, Ginelli E, Mattevi A, Sala C, Battaglioli E (2010) Alternative splicing of the histone demethylase LSD1/KDM1 contributes to the modulation of neurite morphogenesis in the mammalian nervous system. J Neurosci 30(7):2521–2532

    PubMed  CAS  Google Scholar 

  • Zuccato C, Tartari M, Crotti A, Goffredo D, Valenza M, Conti L, Cataudella T, Leavitt BR, Hayden MR, Timmusk T, Rigamonti D, Cattaneo E (2003) Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nat Genet 35(1):76–83

    PubMed  CAS  Google Scholar 

Download references

Conflict of interest statement

Dr. Gray declares no financial relationship.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steven G. Gray.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Gray, S.G. Targeting Huntington’s disease through histone deacetylases. Clin Epigenet 2, 257–277 (2011). https://0-doi-org.brum.beds.ac.uk/10.1007/s13148-011-0025-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1007/s13148-011-0025-7

Keywords