Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Sep 1.
Published in final edited form as: Future Neurol. 2009 Nov 1;4(6):775–784. doi: 10.2217/fnl.09.55

Development of histone deacetylase inhibitors as therapeutics for neurological disease

Joel M Gottesfeld 1,, Massimo Pandolfo 2
PMCID: PMC2824892  NIHMSID: NIHMS171809  PMID: 20177429

Abstract

Postsynthetic modifications of histone and other chromosomal proteins by reversible acetylation and/or methylation regulate many aspects of chromatin dynamics, such as transcription, replication and DNA repair. Aberrant modification states are associated with several neurological and neuromotor diseases. Thus, small molecules that inhibit or activate the enzymes responsible for these chromatin modifications have received considerable attention as potential human therapeutics. This paper summarizes the current state of development of histone deacetylase inhibitors in a variety of neurological diseases.

Keywords: acetylation, amyotrophic lateral sclerosis, chromatin, fragile X syndrome, Friedreich's ataxia, gene regulation, histone, histone deacetylase inhibitor, Huntington's disease, myotonic dystrophy, Rett's syndrome, Rubinstein–Taybi syndrome, spinal muscular atrophy, triplet repeat disease

Chromatin structure, histone postsynthetic modifications & gene expression

The basic subunit of chromatin in eukaryotic cells is the nucleosome, consisting of an octamer of two copies each of the four core histones, H2A, H2B, H3 and H4, plus 147 base pairs (bp) of DNA [1]. A variable linker region, spanning between 20 and 100 bp, depending upon the cell type and organism, connects nucleosomes to form higher order chromatin structures. The folding of nucleosome chains into higher order domains is dependent on members of the H1 family of linker histones [2]. While the nucleosome can be regarded as the primary subunit of chromatin, different specialized histone proteins are known to be associated with particular regions of the genome, such as histone H2AZ at the promoters of active genes or the H3 variant CENP-A at centromeric heterochromatin. The precise molecular architecture of higher order chromatin structure has been debated over the years, but it is clear that such folding plays an important role in regulating access of the genome to the transcription machinery. It is also clear that the histone proteins, and postsynthetic modifications of the histones, play pivotal roles in regulating chromatin dynamics. In particular, acetylation of particular lysine residues in the amino-termini of histone H4 has been shown to regulate higher order chromatin folding [3] and lysine methylation in histone H3 and H4 has been shown to regulate the association of chromatin-associated proteins with particular genes. Of interest to various neurological diseases (discussed later), trimethylation of lysine 9 of H3 (H3K9me3) recruits heterochromatin protein 1 (HP1) through its chromodomain, resulting in gene repression [4], while H3K4me3 is associated with active genes [5]. The pattern of histone acetylation and methylation in the core histones at any genetic locus has been proposed to form a histone code that specifies gene activity [6].

Histone acetyltransferase, histone deacetylases & histone deacetylase inhibitors

Acetylation and deacetylation of histone proteins, and of other proteins involved in transcriptional regulation, have critical roles in regulating gene expression. Protein acetylation and deacetylation is modulated by the interplay between histone acetyltransferases (HATs) and histone deacetylases (HDACs). Several HATs have been identified in the human genome, including various components of transcriptional regulatory complexes and the association of the various HAT enzymes with partner proteins to form multisubunit complexes provides a mechanism to both target and regulate HAT activity at gene promoters [7]. The association of HATs with histone chaperone proteins confers acetylation specificity between N-terminal lysines (H3K9 and H3K23) and those within the histone fold domain (H3K56) [7,8]. Typically, the increases in HAT activity lead to increased gene transcription by creating a more open conformation of chromatin, whereas HDAC activity leads to repression of gene expression via condensation of the chromatin structure [9]. While HAT inhibitors and activators have been identified [10,11], far more interest has focused on the development of HDAC inhibitors (HDACIs) for various human diseases (see later).

A total of 18 HDACs (more strictly, protein deacetylases) have been identified in the human genome, including the zinc-dependent HDACs (classes I, II and IV), and the NAD+-dependent protein deacetylase enzymes (class III, or sirtuins). HDAC 1, 2, 3 and 8 belong to class I, showing homology to the yeast enzyme RPD3. Class II is further divided into class IIa (HDACs 4, 5, 7 and 9) and IIb (HDAC 6 and 10), according to their sequence homology and domain organization. HDAC11 is the lone member of class IV. The sirtuins (class III and Sirt1–7) are related to the yeast Sir2 protein and are involved in the regulation of mitochondrial activity, metabolism and aging, and have received considerable attention as potential therapeutic targets in diabetes and other diseases.

A diverse class of compounds that inhibit HDAC enzymes has been developed and several studies have focused on the use of these HDACIs as therapy for modifying histone acetylation and transcriptional activation in cell culture studies and in vivo mouse models of neurodegeneration [12]. Chemically, the HDACIs can be classified into six structural groups, including the small carboxylates, such as sodium butyrate, valproic acid and sodium phenylbutyrate; the hydroxamic acids, such as trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), and their derivatives (the benzamides CI-994 and MS-275); the epoxyketones such as the trapoxins; cyclic peptides such as apicidin and depsipeptide; and hybrid molecules containing cyclic peptide motifs and hydroxamic acid moieties [13]. As previously mentioned, the dynamic interplay between histone/protein acetylation and deacetylation serves as a central regulatory mechanism governing gene expression, which in turn guides cellular differentiation and development. It is thus quite surprising that small molecule HDACI therapeutics that inhibit this basic molecular pathway are well tolerated in humans [14] and show therapeutic promise in a wide range of human diseases including cancer, metabolic and neurological diseases.

Triplet-repeat sequences affecting chromatin structure & gene expression: fragile X & Friedreich's ataxia

In fragile X syndrome and Friedreich's ataxia, the pathological hyperexpansion of repeated sequences triggers epigenetic changes, leading to gene silencing and consequent loss of function. In both cases, the repeat expansions are in noncoding regions of the affected gene; thus, reactivation of silenced genes is an attractive therapeutic approach. In Fragile X syndrome, expansion of a CGG•CCG-repeat in the 5′-UTR of the FMR1 gene to more than 200 repeats results in gene silencing, and a concomitant loss of FMR1 protein. This expansion triggers methylation of a CpG island upstream of the FMR1 gene, hypoacetylation of associated histones and chromatin condensation. In cell lines from fragile X patients, there is a decrease in H3K4me3 with a large increase in H3K9me3 – changes that are consistent with a switch from euchromatin to heterochromatin at the FMR1 locus in the disease state [15,16]. Several reports have documented increases in FMR1 gene expression by decreasing DNA methylation with 5-azadeoxycidine, and a synergistic reactivation of the silenced gene has been reported by combination treatment with 5-azadC and the HDACIs 4-phenylbutyrate, sodium butyrate and TSA [17]. Recently, it was reported that a class III histone deacetylase, SIRT1, plays an important role in FMR1 gene silencing, and the inhibition of this enzyme with splitomicin produces significant gene reactivation [16]. The authors of this study argue that SIRT1 inhibitors may be more useful for FMR1 gene reactivation in postmitotic cells, such as neurons, since DNA methylation inhibitors require DNA replication in order to be effective [16].

The related neurodegenerative disease Friedreich's ataxia (FRDA) is caused by expansion of the simple triplet repeat DNA sequence GAA•TTC within intron 1 of the FXN gene, encoding the essential mitochondrial protein frataxin. The GAA•TTC repeat expansion was originally believed to alter the DNA structure of the FXN gene [18] and thereby block transcription elongation by RNA polymerase II. Indeed, the loss of FXN mRNA and frataxin protein is well established as the cause of FRDA [19]. However, more recent studies indicate GAA•TTC repeats in the FXN gene caused gene silencing via an altered chromatin structure [2023]. Histone H3/H4 hypoacetylation and H3K9 methylation have been observed on FRDA FXN genes, both of which are hallmarks of heterochromatin [22]. These results have now been extended to FRDA mouse models [20,24] and to human FRDA autopsy material [20], and the consensus in the field is that FRDA is a heterochromatin gene-silencing disease. Based on these observations, we speculated that HDACIs might reverse FXN silencing by directly increasing histone acetylation on the FXN gene, leading to chromatin decondensation and active transcription. We found that a commercially available HDACI (BML-210), and derivatives we have synthesized (pimelic diphenylamides), relieve repression of the FXN gene in lymphoid cell lines derived from FRDA patients, in primary lymphocytes from donor FRDA patient blood and in the brain and heart of a mouse model for FRDA [22,24]. The HDACIs directly affect the histones associated with the expanded FXN gene in patient cells and in the mouse model, increasing acetylation at particular lysine residues on histones H3 and H4, providing evidence for the chromatin model. Unexpectedly, we found that only members of the pimelic diphenylamide family of HDACIs increase FXN gene expression, and many common and highly active HDACI, including the hydroxamates TSA and SAHA are inactive. The pimelic diphenylamides are currently under development by Repligen Corporation (MA, USA) and clinical trials in FRDA are expected in the near future.

Master epigenetic regulators: MECP2 & Rett syndrome, CBP & Rubinstein–Taybi syndrome

These diseases are caused by mutations affecting factors that mediate general epigenetic mechanisms, leading to widespread gene-expression dysregulation. Methyl-CpG-binding protein (MECP2) couples DNA methylation, an important epigenetic mark, to chromatin remodeling and changes in gene expression. Cyclic AMP response element-binding protein (CREB)-binding protein (CBP) is a major histone acetyltransferase. Rett syndrome (RTT) is caused by loss-of-function mutations in the MECP2 gene on the X chromosome (Xq28) [25]. The gene encodes the two isoforms of the methyl-CpG-binding protein (MECP2 and MECP2B). Almost all mutations occur de novo and act as X-linked dominant, meaning most affected individuals are girls. RTT is a neurodevelopmental disorder that becomes apparent after 6–18 months of apparently normal development. In its typical form, it is characterized by loss of communication skills and of the purposeful use of hands, which become engaged in stereotypic movements, progressive slowing of head growth, motor disturbances, irritability, seizures and breathing abnormalities [26]. However, MECP2 mutations cause a wide spectrum of phenotypes in addition to classical RTT, in girls as well as in boys. Girls may have a milder disease or even be asymptomatic [26]. Skewed X chromosome inactivation may explain some, but not all phenotypic variability in girls [26]. Male germline MECP2 mutations may cause a severe encephalopathy with death at birth, as seen in brothers of RTT girls, or X-linked recessive mental retardation (XLMR) [27]. MECP2 is thought to act primarily by recruiting a protein complex composed by Sin3A, RbAp46, RbAp48, and HDAC1 and HDAC2 to methylated CpG nucleotides [28]. This protein complex then carries out histone modifications and chromatin remodeling leading to silencing of the target genes. MECP2 carries out this function in the brain, while the protein MBD is active in the periphery, explaining the specific vulnerability of the brain to MECP2 mutations. Recent studies on MECP2 function revealed that it has additional roles in chromatin remodeling, RNA splicing and control of gene expression [28]. MECP2 appears to be a multifunctional protein that regulates gene expression in the brain, both repressing and activating sets of target genes [29]. Therapeutics that can affect chromatin structure and modulate gene expression may eventually be treatments for this devastating disorder, but further studies are necessary to identify potentially useful compounds or classes of compounds. HDACIs may not appear as obvious therapeutic candidates for a disease where gene de-repression is thought to play a major role. In some experimental paradigms, HDACIs have even been shown to mimic some RTT features, such as behavioral abnormalities linked to basolateral amygdala dysfunction [30]. However, the complexity of MECP2 function and in particular the discovery that some genes are activated by this protein [29] suggest that a role of these compounds in future treatments cannot be excluded.

Approximately 50% of the cases of Rubinstein–Tayby syndrome (RTS) result from heterozygous mutations in the transcriptional coactivator CBP [31] or its homolog p300 [32,33]. RTS has a complex phenotype including short stature, microcephaly, abnormal facial features (beaked nose, slightly malformed ears, highly arched palate, antimongoloid slant of eyes, and heavy or highly arched eyebrows), broad thumbs and/or great toes that may be angulated. Mental retardation is a feature of RTS, it is usually moderate, but may vary from mild to severe [34]. CBP and p300 are involved in transcriptional activation of a large number of target genes via HAT activity [35] and chromatin remodeling [36]. Several mouse models with CPB and p300 mutations have been generated. These mice have cognitive abnormalities, particularly in long-term memory [37]. Treatment with HDACIs, such as SAHA and TSA, can correct these cognitive deficits [37], suggesting that loss of HAT activity has a primary role in their pathogenesis, and by extension in the pathogenesis of mental retardation in RTS. Therefore, HDACIs may be a promising approach for the human condition, at least for its neurocognitive aspects. Interestingly, loss of CBP and p300 activity is observed in polyglutamine disorders, where these proteins interact with polyglutamine protein aggregates and are sequestered in inclusions [38]. The consequent loss of HAT activity may underlie the sensitivity of polyglutamine diseases to HDACI treatment (see later sections).

Modulation of gene expression & potential therapies for spinal muscular atrophy & myotonic dystrophy

In spinal muscular atrophy (SMA) and myotonic dystrophy, increasing the expression of the SMN2 and muscleblind genes, respectively, may compensate for the primary genetic defects. Such induction may be obtained through drugs acting upon epigenetic mechanisms. SMA is an autosomal recessive disease of motor neurons that causes muscle weakness and wasting in children [39]. In the most severe form (SMA type I), weakness and hypotonia are evident at birth, affected children never sit or walk and they die within 2 years. Children with the intermediate form (SMA type II) become able to sit, but never walk and also have a severely shortened life expectancy. SMA type III is the less severe form, affected children start to walk, but eventually develop proximal muscle weakness and lose deambulation. Individuals with SMA type III survive into adult age. In some SMA type III cases (sometimes known as SMA IV), the disease onset occurs after childhood, in the third decade of life. SMA is caused by severe deficiency of a protein involved in RNA processing termed SMN, encoded by a duplicated gene on chromosome 5 [40]. The two copies, termed SMN1 and SMN2, only differ by a single nucleotide in exon 7. The CAGACAA sequence in SMN1 exon 7 is a binding site for the splicing factor SF2/ASF and acts as an exonic splicing enhancer (ESE) promoting the inclusion of exon 7 in the mature mRNA. In SNM2, this sequence has changed to TAGACAA (UAGACAA in the corresponding RNA) and does not bind SF2/ASF anymore, with loss of ESE function and frequent (>80%) skipping of exon 7 in the mature SMN2 mRNA [41]. SMA is caused by mutations that inactivate SMN1, most frequently deletions, so that SMN synthesis entirely depends on SMN2. Motor neurons are exquisitely sensitive to the consequently reduced SMN levels and undergo degeneration. Complete absence of SMN is lethal for all cell types, as shown in knockout animals [42]. The different severities of SMA type I, II and III is partly explained by a variation in SMN2 copy number and therefore in the residual amount of SMN [43]. A recent study demonstrated that the SMN2 gene is subject to silencing by DNA methylation involving MeCP2, while DNA methylation levels inversely correlate with disease severity [44]. Even though silencing of the SMN2 gene involves DNA methylation, HDACIs have been identified that boost SMN2 expression and thus increase SMN protein levels. Early studies showed that valproic acid increases SMN protein in patient-derived cell lines [45,46] and has been beneficial in mouse models of the disease [47], leading to several small, open-label trials in SMA patients. So far, improvement has been modest and limited to a subset of patients [4851]. However, other HDACIs are active in patient-derived cells, like sodium butyrate [52], sodium phenylbutirate [53] and in particular, SAHA. The latter not only appears to be much more potent than valproic acid in increasing SMN2 expression, it also significantly increases the proportion of full-length (with exon 7 included) SMN2-derived mRNA [54]. Other benzamide HDACIs may also be effective in raising SMN levels [55]. Very recently, LBH589 (panobinostat), a pan-HDACI, which is approved by the US FDA for the treatment of cutaneous T-cell lymphoma, was shown to increase full-length SMN2-derived SMN protein levels two- to three-fold. Such an increase resulted from the combined effect of the drug at both RNA and protein levels. In particular, LBH589 increased H3K9 acetylation at the SMN2 promoter, facilitated exon 7 splicing through an effect on the splicing factor hTRA2-b1, stabilized SMN by reducing its ubiquitinylation and facilitated its incorporation into the SMN complex [56]. Therefore, despite some initial disappointment with valproic acid, HDACIs remain a promising therapeutic approach for SMA.

The myotonic dystrophies (DM1 and DM2) are autosomal dominant systemic diseases resulting from expansion of noncoding, but transcribed repeat sequences (a CTG repeat in the 3′-UTR of the DMPK gene in DM1 [57] and a CCTG repeat in an intron of the ZNF9 gene in DM2 [58]). RNAs containing these sequences adopt a hairpin secondary structure and accumulate in nuclear foci [58,59]. The Drosophila muscleblind proteins (MBNL1–3) are the human homologs of the Drosophila muscleblind RNA-binding protein that regulates the splicing and maturation of a set of pre-mRNAs [60]. MBNLs bind the CUG/CCUG-containing hairpin RNA structures that accumulate in DM1 and DM2 and are sequestered in nuclear RNA foci [61]. The resulting MBNL depletion leads to misregulated maturation of multiple pre-mRNAs, which is a major contributor to DM1 and DM2 physiopathology [62]. The important role of the loss of MBNL activity in DM pathogenesis is confirmed by the development of a DM-like phenotype by mbnl-knockout mice [63] and by the ability of Mbnl overexpression to correct the phenotype of CAG repeat expansion-based DM animal models [64]. Therefore, enhancing MBNL expression may be an interesting approach to treat the myotonic dystrophies. Accordingly, the possibility that some HDACIs may induce MBNLs and be potential therapeutics for DM1 and DM2, although as yet untested, is worth exploring.

Amyotrophic lateral sclerosis

Histone deacetylase inhibition has also been proposed as a therapeutic approach for amyotrophic lateral sclerosis (ALS) on the basis that disequilibrium between HAT and HDAC activities, possibly related to CBP loss-of-function, may contribute to the pathogenesis of this lethal degenerative disease of motor neurons. Phenylbutyrate increased survival in the SOD1 G93A transgenic mouse model of ALS when used alone or in combination with an antioxidant [65]. In the same mouse model, valproic acid was also effective in extending survival and in delaying onset, particularly when used in association with the glycogen synthase kinase-3 inhibitor lithium [66]. These promising preclinical findings led to a recently published Phase IIA open-label trial of sodium phenylbutyrate in ALS patients. A total of 26 patients completed the 20-week treatment with 9–21 g/day of the medication reporting good tolerability, with no mortality and no clinically relevant laboratory changes. Histone acetylation in peripheral blood cells increased even with the lowest dose of 9 g/day [67]. Placebo-controlled Phase IIB trials with this molecule in ALS will now be needed to show whether this HDACI promises to have any clinical efficacy.

HDACIs in polyglutamine repeat disorders

The CAG repeat/polyglutamine (polyQ) expansion disorders include Huntington's disease (HD), the spinocerebellar ataxias (SCAs) 1, 2, 3, 6, 7, 8 and 17, spinobulbar muscular atrophy (SBMA) and dentatorubral pallidoluysian atrophy (DRPLA). All of these disorders are progressive in nature and result in severe neurological disturbances. A general mechanism of gene expression dysregulation may be common to these diseases, owing to the effect of nuclear protein microaggregates or inclusions on transcription factors or other components of the transcriptional machinery. Furthermore, some of the mutated proteins are transcriptional regulators themselves (Ataxin 7 in SCA7 and TBP in SCA17), whose activity is affected by the presence of the expanded polyglutamine tract. While the nature of these intranuclear inclusions remains controversial (pathogenic or protective), they represent a pathological hallmark for HD, as well as for many other polyQ diseases [68]. Studies in cell culture, yeast, Drosophila and, more recently, mouse models of polyQ disease, have indicated that HDACIs might provide a useful class of agents to ameliorate the transcriptional deficits in HD and some of the SCAs [6972]. Four studies have examined the potential therapeutic effects of the HDACIs SAHA [70], sodium butyrate [71], phenylbutyrate [73] and a pimelic diphenylamide [74] in HD mouse models. Two studies using R6/2 transgenic mice found that sodium butyrate and SAHA treatment improved motor performance and demonstrated neuroprotective effects. No effects on Htt aggregation or HD transgene expression were detected by either compound [70,71]. In both studies, treatment with these HDACIs increased global acetylation of histones. In a third study, HD-N171-82Q mice were treated with phenylbutyrate after the onset of symptoms, in order to determine whether HDACIs could reverse deficits already in progress [73]. While no effects on motor performance were noted, phenylbutyrate treatment did increase survival, which was associated with increased acetylation of histones H3 and H4. In addition, phenylbutyrate displayed neuroprotective effects of decreasing striatal atrophy and reducing ventricular enlargement [73]. A recent study indicated that one of the pimelic diphenylamides developed for FRDA (HDACI 4b) also shows clinical efficacy in the R6/2 transgenic mouse model for HD [74]. This HDACI ameliorated the motor and behavioral symptoms associated with disease progression in these mice and corrected transcriptional abnormalities associated with polyQ-expanded Htt protein in the mouse brain. Importantly, unlike the other HDACIs tested in HD mouse models, this compound partially prevented the weight loss that is associated with disease progression in the mouse model, a feature that is also present in human HD [74].

Future perspective

The HDACI SAHA (and vorinostat) has been approved by the FDA for the treatment of cutaneous T-cell lymphoma [14], the first HDACI specifically approved as a human therapeutic. Earlier drugs, such as 4-phenylbutyrate and valproic acid, are indeed HDACIs, but were approved based on efficacy as anticonvulsant and mood-stabilizing drugs (valproic acid) and for patients with urea cycle disorders (4-PBA). Some of the recent clinical trials with these compounds have been mentioned previously. A search using an online database [101] reveals that 70 trials are currently underway or planned to test the efficacy of various HDACI as monotherapies or combination therapies, with most of these trials testing HDACI as cancer therapeutics. The majority of current HDACI relatively nonspecifically, targeting each of the members of a particular class of HDACs with comparable IC50 values [75]. Although current HDACI are relatively well tolerated in humans, they have been associated with numerous side effects, such as cardiac arrhythmia, bone marrow depression, clotting disorders, diarrhea, fatigue and electrolyte disturbances in Phase I and II studies [76]. We expect that compounds that specifically inhibit a subset of the HDACs, targeted to obtain a desired clinical outcome, will be less toxic and will substantially broaden the therapeutic uses of HDACI in neurological diseases. In this regard, we recently reported that the HDACI 06, which is active in a mouse model of FRDA [24], specifically targeted HDAC3, with at least a tenfold specificity toward this enzyme over other class I HDACs, and essentially no activity against class II HDACs [77]. Derivatives of this compound are under development as therapeutics for FRDA and HD. Similarly, other isoform-selective HDACIs have been shown to be neuroprotective in oxidative stress-induced cellular models for neurodegeneration, without the cytotoxic effects of hyroxamate-based inhibitors. These include cysteine-based moieties for zinc chelation [78] and HDAC6-selective mercaptoacet-amines [79]. In contrast to the HDAC3-selective compounds that show efficacy in the FRDA and HD models previously mentioned, these latter studies demonstrated that similar benzamides, as well as hydroxamates and other thiol-containing compounds, were ineffective in protecting primary cortical neurons from homocyseate-induced stress [79]. Thus, inhibition of class II HDACs might be more beneficial than inhibition of class I HDACs in neurological diseases such as ALS, while inhibition of specific class I HDACs, such as HDAC3, may well be the best therapeutic approach in diseases where transcription defects are the core pathology of the disease. To avoid the potential toxic effects of HDACI treatment, a recent study demonstrated that pulsed inhibition of HDACs in cortical neurons induce neuroprotection without apparent toxicity [80]. Thus, pulsed treatment regimens might be effective in humans without the side effects found in cancer clinical trials with hydoxamate-based HDACIs [76]. The combination of new inhibitors that show improved selectivity for different members of the various HDAC classes along with innovative treatment regimens will likely yield therapeutic benefit for the diseases described in this paper. Researchers in the field hope that within the next few years, human clinical trials will be initiated for these diseases, and there is reasonable hope that approved therapeutics should appear by the middle of the next decade.

Executive summary.

Chromatin structure, histone postsynthetic modifications & gene expression

  • The higher order structure of chromatin is controlled by postsynthetic modifications of the histone proteins, such as acetylation and methylation of specific lysine residues. These modification states of the histones dictate whether or not a region of the genome is accessible to the transcription machinery of the cell.

Histone acetyltransferases, histone deacetylases & histone deacetylation inhibitors

  • The human genome encodes numerous enzymes responsible for both histone acetylation (histone acetyltransferases [HATs]) and histone deacetylation (histone deacetylases [HDACs]). Modulation of the activities of these enzymes with small molecules has been shown to regulate gene expression.

  • Various small molecule HDAC inhibitors have been identified and considerable effort has been expended to derive molecules that are selective for various members of the four classes of HDAC enzymes.

Triplet-repeat sequences affecting chromatin structure & gene expression: fragile X & Friedreich's ataxia

  • These diseases are caused by chromatin-mediated silencing of particular genes: FMR1 in the case of fragile X syndrome and frataxin in the case of Friedreich's ataxia.

  • Small molecules, such as DNA methyltransferase inhibitors and HDAC inhibitors have been shown to reactivate these genes and offer potential therapeutic approaches.

Master epigenetic regulators: methyl-CpG-binding protein & Rett syndrome, CBP & Rubinstein–Taybi syndrome

  • These diseases arise from mutations in two central epigenetic regulators, the methyl-CpG-binding protein 2 in Rett syndrome and the histone acetyltransferase, cyclic AMP response element-binding protein (CREB)-binding protein, in Rubinstein–Taybi syndrome.

  • Thus, HDAC inhibitors offer promise for therapy in Rett syndrome and Rubinstein–Taybi syndrome.

Modulation of gene expression & potential therapies for spinal muscular atrophy & myotonic dystrophy

  • Spinal muscular atrophy is caused by a mutation in the survival of motor neuron (SMN1) gene, and activation of the related SMN2 gene with HDAC inhibitors has been shown to increase SMN protein levels in both cellular and animal models of this disease.

  • Myotonic dystrophy type 1 is caused by a toxic RNA gain-of-function, where long CUG repeat RNA sequesters essential proteins involved in RNA splicing, such as the human homolog of the Drosophila protein muscleblind (MBNL1). Thus, activation of MBNL1 gene expression with small molecules has been proposed as a potential therapy.

Amyotrophic lateral sclerosis

  • Histone deacetylase inhibition has also been proposed as a therapeutic approach for amyotrophic lateral sclerosis (ALS), based on the hypothesis that a disequilibrium between HAT and HDAC activities may contribute to the pathogenesis of this lethal degenerative disease of motor neurons.

  • Clinical trials of sodium phenylbutyrate in ALS patients have begun.

Histone deacetylase inhibitors in polyglutamine repeat disorders

  • Dysregulation of gene expression may be a common element among the polyQ repeat disorders such as Huntington's disease and the spinocerebellar ataxias. Polyglutamine repeats cause protein aggregation or inclusions in the nucleus, and result in sequestration of transcription factors or other components of the transcriptional machinery.

  • Histone deacetylase inhibitors have proved beneficial in mouse models for HD.

Conclusion & future perspective

  • Clinical trials of several nonselective first-generation HDAC inhibitors, such as valproic acid and 4-phenylbutyrate, have been performed in neurodegenerative and neuromuscular diseases; however, these molecules have met with limited success.

  • Histone deacetylase inhibitors that are selective for specific members of the various classes of HDAC enzymes are expected to be less toxic and would show a better therapeutic outcome.

  • Pulsed treatment regimens might also be more beneficial and would likely show lower levels of unwanted side effects, such as those observed with the US FDA approved HDAC inhibitor, suberoylanilide hydroxamic acid (and vorinostat).

Footnotes

Financial & competing interests disclosure: Studies in the authors' laboratories are funded by the National Institutes of Neurological Disorders and Stroke, NIH (to Joel M Gottesfeld); the Friedreich's Ataxia Research Alliance, Ataxia UK, GoFAR, Ataxia Ireland (to both authors); Fondazione CRT (Italy), the Belgian National Scientific Research Funds (FNRS), the Belgian Ministry of Scientific Policy (Program IAP6; to Massimo Pandolfo). Joel M Gottesfeld serves as a consultant to Repligen Corporation (Waltham, MA, USA) and declares a competing financial interest. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Contributor Information

Joel M Gottesfeld, Department of Molecular Biology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA 92037, USA, Tel.: +1 858 784 8913, Fax: +1 858 784 8965.

Massimo Pandolfo, Email: massimo.pandolfo@ulb.ac.be, Service de Neurologie, Laboratoire de, Neurologie Expérimentale, Hôpital, Erasme, ULB, 808 Route de Lennik, B-1070 Brussels, Belgium, Tel.: +32 2555 3429, Fax: +32 2555 3942.

Bibliography

Papers of special note have been highlighted as:

▪ of interest

▪▪ of considerable interest

  • 1.Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997;389:251–260. doi: 10.1038/38444. [DOI] [PubMed] [Google Scholar]; ▪▪ Describes the structure of the nucleosome at atomic resolution and provides details for how nucleosomes might contribute to gene regulation.
  • 2.Hayes JJ, Hansen JC. Nucleosomes and the chromatin fiber. Curr Opin Genet Dev. 2001;11:124–129. doi: 10.1016/s0959-437x(00)00168-4. [DOI] [PubMed] [Google Scholar]
  • 3.Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL. Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science. 2006;311:844–847. doi: 10.1126/science.1124000. [DOI] [PubMed] [Google Scholar]; ▪ Demonstrates that a particular histone modification can have a profound influence on chromatin structure and possibly gene expression.
  • 4.Jacobs SA, Khorasanizadeh S. Structure of HP1 chromodomain bound to a lysine 9-methylated histone H3 tail. Science. 2002;295:2080–2083. doi: 10.1126/science.1069473. [DOI] [PubMed] [Google Scholar]
  • 5.Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ. How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat Rev Struct Mol Biol. 2007;14:1025–1040. doi: 10.1038/nsmb1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41–45. doi: 10.1038/47412. [DOI] [PubMed] [Google Scholar]; ▪▪ A model is proposed for how histone postsynthetic modifications control gene activity.
  • 7.Berndsen CE, Denu JM. Catalysis and substrate selection by histone/protein lysine acetyltransferases. Curr Opin Struct Biol. 2008;18:682–689. doi: 10.1016/j.sbi.2008.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Williams SK, Truong D, Tyler JK. Acetylation in the globular core of histone H3 on lysine-56 promotes chromatin disassembly during transcriptional activation. Proc Natl Acad Sci USA. 2008;105:9000–9005. doi: 10.1073/pnas.0800057105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Elgin SC, Grewal SL. Heterochromatin: silence is golden. Curr Biol. 2003;13:R895–R898. doi: 10.1016/j.cub.2003.11.006. [DOI] [PubMed] [Google Scholar]
  • 10.Balasubramanyam K, Swaminathan V, Ranganathan A, Kundu TK. Small molecule modulators of histone acetyltransferase p300. J Biol Chem. 2003;278:19134–19140. doi: 10.1074/jbc.M301580200. [DOI] [PubMed] [Google Scholar]
  • 11.Arif M, Pradhan SK, Thanuja GR, et al. Mechanism of p300 specific histone acetyltransferase inhibition by small molecules. J Med Chem. 2009;22:267–277. doi: 10.1021/jm800657z. [DOI] [PubMed] [Google Scholar]
  • 12.Langley B, Gensert JM, Beal MF, Ratan RR. Remodeling chromatin and stress resistance in the central nervous system: histone deacetylase inhibitors as novel and broadly effective neuroprotective agents. Curr Drug Targets CNS Neurol Disord. 2005;4:41–50. doi: 10.2174/1568007053005091. [DOI] [PubMed] [Google Scholar]
  • 13.Drummond DC, Noble CO, Kirpotin DB, Guo Z, Scott GK, Benz CC. Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol. 2005;45:495–528. doi: 10.1146/annurev.pharmtox.45.120403.095825. [DOI] [PubMed] [Google Scholar]
  • 14.Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007;12:1247–1252. doi: 10.1634/theoncologist.12-10-1247. [DOI] [PubMed] [Google Scholar]; ▪ Landmark approval of a histone deacetylase inhibitor as a human therapeutic.
  • 15.Coffee B, Zhang F, Ceman S, Warren ST, Reines D. Histone modifications depict an aberrantly heterochromatinized FMR1 gene in fragile x syndrome. Am J Hum Genet. 2002;71:923–932. doi: 10.1086/342931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Biacsi R, Kumari D, Usdin K. Sirt1 inhibition alleviates gene silencing in fragile X mental retardation syndrome. PLoS Genet. 2008;4:E1000017. doi: 10.1371/journal.pgen.1000017. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Role of a sirtuin in FMR1 gene silencing is revealed in this paper.
  • 17.Chiurazzi P, Pomponi MG, Pietrobono R, Bakker CE, Neri G, Oostra BA. Synergistic effect of histone hyperacetylation and DNA demethylation in the reactivation of the FMR1 gene. Hum Mol Genet. 1999;8:2317–2323. doi: 10.1093/hmg/8.12.2317. [DOI] [PubMed] [Google Scholar]
  • 18.Sakamoto N, Chastain PD, Parniewski P, et al. Sticky DNA: self-association properties of long GAA.TTC repeats in R.R.Y triplex structures from Friedreich's ataxia. Mol Cell. 1999;3:465–475. doi: 10.1016/s1097-2765(00)80474-8. [DOI] [PubMed] [Google Scholar]
  • 19.Campuzano V, Montermini L, Molto MD, et al. Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996;271:1423–1427. doi: 10.1126/science.271.5254.1423. [DOI] [PubMed] [Google Scholar]
  • 20.Al-Mahdawi S, Pinto RM, Ismail O, et al. The Friedreich ataxia GAA repeat expansion mutation induces comparable epigenetic changes in human and transgenic mouse brain and heart tissues. Hum Mol Genet. 2008;17:735–746. doi: 10.1093/hmg/ddm346. [DOI] [PubMed] [Google Scholar]
  • 21.Greene E, Mahishi L, Entezam A, Kumari D, Usdin K. Repeat-induced epigenetic changes in intron 1 of the frataxin gene and its consequences in Friedreich ataxia. Nucl Acids Res. 2007;35:3383–3390. doi: 10.1093/nar/gkm271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Herman D, Jenssen K, Burnett R, Soragni E, Perlman SL, Gottesfeld JM. Histone deacetylase inhibitors reverse gene silencing in Friedreich's ataxia. Nature Chem Biol. 2006;2:551–558. doi: 10.1038/nchembio815. [DOI] [PubMed] [Google Scholar]
  • 23.Saveliev A, Everett C, Sharpe T, Webster Z, Festenstein R. DNA triplet repeats mediate heterochromatin-protein-1-sensitive variegated gene silencing. Nature. 2003;422:909–913. doi: 10.1038/nature01596. [DOI] [PubMed] [Google Scholar]; ▪▪ Evidence is presented demonstrating that triplet repeat sequences influence heterochromatin formation, which may be central to the pathology of many neurodegenerative diseases.
  • 24.Rai M, Soragni E, Jenssen K, et al. HDAC inhibitors correct frataxin deficiency in a Friedreich ataxia mouse model. PLoS ONE. 2008;3(4):E1958. doi: 10.1371/journal.pone.0001958. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Identification of an histone deacetylase (HDAC) inhibitor that reverses heterochromatin-mediated gene silencing in a Friedreich's ataxia mouse model.
  • 25.Amir RE, Van Den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Gen. 1999;23:185–188. doi: 10.1038/13810. [DOI] [PubMed] [Google Scholar]; ▪ The molecular basis for Rett syndrome is elucidated.
  • 26.Weaving LS, Ellaway CJ, Gecz J, Christodoulou J. Rett syndrome: clinical review and genetic update. J Med Gen. 2005;42:1–7. doi: 10.1136/jmg.2004.027730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Villard R. MECP2 mutations in males. J Med Gen. 2007;44:417–423. doi: 10.1136/jmg.2007.049452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hite KC, Adams VH, Hansen JC. Recent advances in MECP2 structure and function. Biochem Cell Biol. 2009;87:219–227. doi: 10.1139/o08-115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chahrour M, Jung SY, Shaw C, et al. MECP2, a key contributor to neurological disease, activates and represses transcription. Science. 2008;320:1224–1229. doi: 10.1126/science.1153252. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪▪ The surprising result that methyl-CpG-binding protein 2 may be involved in both the positive and negative regulation of gene expression is reported.
  • 30.Adachi M, Autry AE, Covington HER, Monteggia LM. MECP2-mediated transcription repression in the basolateral amygdala may underlie heightened anxiety in a mouse model of Rett syndrome. J Neurosci. 2009;29:4218–4227. doi: 10.1523/JNEUROSCI.4225-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Petrij F, Giles RH, Dauwerse HG, et al. Rubinstein–Taybi syndrome is caused by mutations in the transcriptional co-activator CBP. Nature. 1995;376:348–351. doi: 10.1038/376348a0. [DOI] [PubMed] [Google Scholar]
  • 32.Lundblad JR, Kwok RP, Laurance ME, Harter MI, Goodman RH. Adenoviral E1A-associated protein p300 as a functional homologue of the transcriptional co-activator CBP. Nature. 1995;374:85–88. doi: 10.1038/374085a0. [DOI] [PubMed] [Google Scholar]
  • 33.Roelfsema JH, White SJ, Ariyurek Y, et al. Genetic heterogeneity in Rubinstein–Taybi syndrome: mutations in both the CBP and p300 genes can cause disease. Am J Hum Gen. 2005;76:572–580. doi: 10.1086/429130. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Mutations in histone acetyltransferases are the cause of Rubinstein–Tayby syndrome.
  • 34.Roelfsema JH, Peters DJ. Rubinstein–Taybi syndrome: clinical and molecular overview. Expert Rev Mol Med. 2007;9:1–16. doi: 10.1017/S1462399407000415. [DOI] [PubMed] [Google Scholar]
  • 35.Bannister AJ, Kouzarides T. The CBP coactivator is a histone acetyltransferase. Nature. 1996;384:641–643. doi: 10.1038/384641a0. [DOI] [PubMed] [Google Scholar]
  • 36.Shikama N, Chan HM, Krstic-Demonacos M, et al. Functional interaction between nucleosome assembly proteins and p300/CREB-binding protein family coactivators. Mol Cell Biol. 2000;20:8933–8943. doi: 10.1128/mcb.20.23.8933-8943.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Alarcón JM, Malleret G, Touzani K, et al. Chromatin acetylation, memory, and LTP are impaired in CBP+/- mice: a model for the cognitive deficit in Rubinstein–Taybi syndrome and its amelioration. Neuron. 2004;42:947–959. doi: 10.1016/j.neuron.2004.05.021. [DOI] [PubMed] [Google Scholar]
  • 38.McCampbell A, Taylor JP, Taye AA, et al. CREB-binding protein sequestration by expanded polyglutamine. Hum Mol Genet. 2000;9:2197–2202. doi: 10.1093/hmg/9.14.2197. [DOI] [PubMed] [Google Scholar]
  • 39.Lunn MR, Wang CH. Spinal muscular atrophy. Lancet. 2008;371:2120–2133. doi: 10.1016/S0140-6736(08)60921-6. [DOI] [PubMed] [Google Scholar]
  • 40.Lefebvre S, Burglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell. 1995;80:155–165. doi: 10.1016/0092-8674(95)90460-3. [DOI] [PubMed] [Google Scholar]
  • 41.Lorson CL, Strasswimmer J, Yao JM, et al. SMN oligomerization defect correlates with spinal muscular atrophy severity. Nat Genet. 1998;19:63–66. doi: 10.1038/ng0598-63. [DOI] [PubMed] [Google Scholar]
  • 42.Schmid A, Didonato CJ. Animal models of spinal muscular atrophy. J Child Neurol. 2007;22:1004–1012. doi: 10.1177/0883073807305667. [DOI] [PubMed] [Google Scholar]
  • 43.Swoboda KJ, Prior TW, Scott CB, et al. Natural history of denervation in SMA: relation to age, SMN2 copy number, and function. Ann Neurol. 2005;57:704–712. doi: 10.1002/ana.20473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hauke J, Riessland M, Lunke S, et al. Survival motor neuron gene 2 silencing by DNA methylation correlates with spinal muscular atrophy disease severity and can be bypassed by histone deacetylase inhibition. Hum Mol Genet. 2009;18:304–317. doi: 10.1093/hmg/ddn357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sumner CJ, Huynh TN, Markowitz JA, et al. Valproic acid increases SMN levels in spinal muscular atrophy patient cells. Ann Neurol. 2003;54:647–654. doi: 10.1002/ana.10743. [DOI] [PubMed] [Google Scholar]; ▪ Molecular mechanisms and potential therapeutics for spinal muscular atrophy are reported in these two papers.
  • 46.Brichta L, Hofmann Y, Hahnen E, et al. Valproic acid increases the SMN2 protein level: a well-known drug as a potential therapy for spinal muscular atrophy. Hum Mol Genet. 2003;12:2481–2489. doi: 10.1093/hmg/ddg256. [DOI] [PubMed] [Google Scholar]
  • 47.Tsai LK, Tsai MS, Ting CH, Li H. Multiple therapeutic effects of valproic acid in spinal muscular atrophy model mice. J Mol Med. 2008;86:1243–1254. doi: 10.1007/s00109-008-0388-1. [DOI] [PubMed] [Google Scholar]
  • 48.Brichta L, Holker L, Haug K, Klockgether T, Wirth B. In vivo activation of SMN in spinal muscular atrophy carriers and patients treated with valproate. Ann Neurol. 2006;59:970–975. doi: 10.1002/ana.20836. [DOI] [PubMed] [Google Scholar]
  • 49.Weihl CC, Connolly AM, Pestronk A. Valproate may improve strength and function in patients with type III/IV spinal muscle atrophy. Neurology. 2006;67:500–501. doi: 10.1212/01.wnl.0000231139.26253.d0. [DOI] [PubMed] [Google Scholar]
  • 50.Tsai LK, Yang CC, Hwu WL, Li H. Valproic acid treatment in six patients with spinal muscular atrophy. Eur J Neurol. 2007;14:E8–E9. doi: 10.1111/j.1468-1331.2007.01992.x. [DOI] [PubMed] [Google Scholar]
  • 51.Swoboda KJ, Scott CB, Reyna SP, et al. Phase II open label study of valproic acid in spinal muscular atrophy. PLoS One. 2009;4:E5268. doi: 10.1371/journal.pone.0005268. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Clinical studies of valproic acid in spinal muscular atrophy reveal potential benefits of HDAC inhibitor therapy.
  • 52.Chang JG, Hsieh-Li HM, Jong YJ, Wang NM, Tsai CH, Li H. Treatment of spinal muscular atrophy by sodium butyrate. Proc Natl Acad Sci USA. 2001;98:9808–9813. doi: 10.1073/pnas.171105098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Andreassi C, Angelozzi C, Tiziano FD, et al. Phenylbutyrate increases SMN expression in vitro: relevance for treatment of spinal muscular atrophy. Eur J Hum Genet. 2004;12:59–65. doi: 10.1038/sj.ejhg.5201102. [DOI] [PubMed] [Google Scholar]
  • 54.Hahnen E, Eyupoglu IY, Brichta L, et al. In vitro and ex vivo evaluation of second-generation histone deacetylase inhibitors for the treatment of spinal muscular atrophy. J Neurochem. 2006;98:193–202. doi: 10.1111/j.1471-4159.2006.03868.x. [DOI] [PubMed] [Google Scholar]
  • 55.Riessland M, Brichta L, Hahnen E, Wirth B. The benzamide M344, a novel histone deacetylase inhibitor, significantly increases SMN2 RNA/protein levels in spinal muscular atrophy cells. Hum Genet. 2006;120:101–110. doi: 10.1007/s00439-006-0186-1. [DOI] [PubMed] [Google Scholar]
  • 56.Garbes L, Riessland M, Hölker I, et al. LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate. Hum Mol Genet. 2009 doi: 10.1093/hmg/ddp313. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 57.Fu YH, Pizzuti A, Fenwick RGJ, et al. An unstable triplet repeat in a gene related to myotonic muscular dystrophy. Science. 1995;255:1256–1258. doi: 10.1126/science.1546326. [DOI] [PubMed] [Google Scholar]
  • 58.Liquori CL, Ricker K, Moseley ML, et al. Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science. 2001;293:864–867. doi: 10.1126/science.1062125. [DOI] [PubMed] [Google Scholar]
  • 59.Taneja KL, Mccurrach M, Schalling M, Housman D, Singer RH. Foci of trinucleotide repeat transcripts in nuclei of myotonic dystrophy cells and tissues. J Cell Biol. 1995;128:995–1002. doi: 10.1083/jcb.128.6.995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pascual M, Vicente M, Monferrer L, Artero R. The muscleblind family of proteins: an emerging class of regulators of developmentally programmed alternative splicing. Differentiation. 2006;74:65–80. doi: 10.1111/j.1432-0436.2006.00060.x. [DOI] [PubMed] [Google Scholar]
  • 61.Mankodi A, Urbinati CR, Yuan QP, et al. Muscleblind localizes to nuclear foci of aberrant RNA in myotonic dystrophy types 1 and 2. Hum Mol Genet. 2001;10:2165–2170. doi: 10.1093/hmg/10.19.2165. [DOI] [PubMed] [Google Scholar]
  • 62.Ranum LP, Day JW. Myotonic dystrophy: RNA pathogenesis comes into focus. Am J Hum Genet. 2004;7:793–804. doi: 10.1086/383590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kanadia RN, Johnstone KA, Mankodi A, et al. A muscleblind knockout model for myotonic dystrophy. Science. 2003;302:1978–1980. doi: 10.1126/science.1088583. [DOI] [PubMed] [Google Scholar]
  • 64.Kanadia RN, Shin J, Yuan Y, et al. Reversal of RNA missplicing and myotonia after muscleblind overexpression in a mouse poly(CUG) model for myotonic dystrophy. Proc Natl Acad Sci USA. 2006;103:11748–11753. doi: 10.1073/pnas.0604970103. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪▪ Increased expression of muscleblind as a therapeutic approach for myotonic dystrophy.
  • 65.Petri S, Kiaei M, Kipiani K, et al. Additive neuroprotective effects of a histone deacetylase inhibitor and a catalytic antioxidant in a transgenic mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2006;22:40–49. doi: 10.1016/j.nbd.2005.09.013. [DOI] [PubMed] [Google Scholar]; ▪ Describes the potential therapeutics for amyotrophic lateral sclerosis.
  • 66.Feng HL, Leng Y, Ma CH, Zhang J, Ren M, Chuang DM. Combined lithium and valproate treatment delays disease onset, reduces neurological deficits and prolongs survival in an amyotrophic lateral sclerosis mouse model. Neuroscience. 2008;155:567–572. doi: 10.1016/j.neuroscience.2008.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cudkowicz ME, Andres PL, Macdonald SA, et al. Phase 2 study of sodium phenylbutyrate in ALS. Amyotroph Lateral Scler. 2009;10:99–106. doi: 10.1080/17482960802320487. [DOI] [PubMed] [Google Scholar]
  • 68.Zoghbi HY, Orr HT. Glutamine repeats and neurodegeneration. Ann Rev Neurosci. 2000;23:217–247. doi: 10.1146/annurev.neuro.23.1.217. [DOI] [PubMed] [Google Scholar]
  • 69.Steffan JS, Bodai L, Pallos J, et al. Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature. 2001;413:739–743. doi: 10.1038/35099568. [DOI] [PubMed] [Google Scholar]
  • 70.Hockly E, Richon VM, Woodman B, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington's disease. Proc Natl Acad Sci USA. 2003;100:2041–2046. doi: 10.1073/pnas.0437870100. [DOI] [PMC free article] [PubMed] [Google Scholar]; References [69] and [70] point to HDAC inhibitors as therapeutics for Huntington's disease.
  • 71.Ferrante RJ, Kubilus JK, Lee J, et al. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J Neurosci. 2003;23:9418–9427. doi: 10.1523/JNEUROSCI.23-28-09418.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jung J, Bonini N. CREB-binding protein modulates repeat instability in a Drosophila model for polyQ disease. Science. 2007;315:1857–1859. doi: 10.1126/science.1139517. [DOI] [PubMed] [Google Scholar]
  • 73.Gardian G, Browne SE, Choi DK, et al. Neuroprotective effects of phenylbutyrate in the N171–82Q transgenic mouse model of Huntington's disease. J Biol Chem. 2005;280:556–563. doi: 10.1074/jbc.M410210200. [DOI] [PubMed] [Google Scholar]
  • 74.Thomas EA, Coppola G, Desplats PA, et al. The HDAC inhibitor 4b ameliorates the disease phenotype and transcriptional abnormalities in Huntington's disease transgenic mice. Proc Natl Acad Sci USA. 2008;105:15564–15569. doi: 10.1073/pnas.0804249105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Beckers T, Burkhardt C, Wieland H, et al. Distinct pharmacological properties of second generation HDAC inhibitors with the benzamide or hydroxamate head group. Int J Cancer. 2007;121:1138. doi: 10.1002/ijc.22751. [DOI] [PubMed] [Google Scholar]
  • 76.Bruserud Ø, Stapnes C, Ersvaer E, Gjertsen BT, Ryningen A. Histone deacetylase inhibitors in cancer treatment: a review of the clinical toxicity and the modulation of gene expression in cancer cell. Curr Pharm Biotechnol. 2007;8:388–400. doi: 10.2174/138920107783018417. [DOI] [PubMed] [Google Scholar]
  • 77.Chou CJ, Herman D, Gottesfeld JM. Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class I histone deacetylases. J Biol Chem. 2008;283:35402–35409. doi: 10.1074/jbc.M807045200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Butler KV, He R, Mclaughlin K, Vistoli G, Langley B, Kozikowski AP. Stereoselective HDAC inhibition from cysteine-derived zinc-binding groups. ChemMedChem. 2009;4:1292–1301. doi: 10.1002/cmdc.200900088. [DOI] [PubMed] [Google Scholar]
  • 79.Chen Y, He R, Chen Y, D'annibale MA, Langley B, Kozikowski AP. Studies of benzamide- and thiol-based histone deacetylase inhibitors in models of oxidative-stress-induced neuronal death: identification of some HDAC3-selective inhibitors. ChemMedChem. 2009;4:842–852. doi: 10.1002/cmdc.200800461. [DOI] [PubMed] [Google Scholar]
  • 80.Langley B, D'Annibale MA, Suh K, et al. Pulse inhibition of histone deacetylases induces complete resistance to oxidative death in cortical neurons without toxicity and reveals a role for cytoplasmic p21(waf1/cip1) in cell cycle-independent neuroprotection. J Neurosci. 2008;28:163–176. doi: 10.1523/JNEUROSCI.3200-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪▪ Potential treatment regimens for neurodegenerative diseases are discussed.

Website

RESOURCES