www.fgks.org   »   [go: up one dir, main page]

Academia.eduAcademia.edu
Traffic 2012; 13: 771–779  2012 John Wiley & Sons A/S doi:10.1111/j.1600-0854.2012.01347.x Review HDAC6 at the Intersection of Neuroprotection and Neurodegeneration Constantin d’Ydewalle1,2 , Elke Bogaert1,2 and Ludo Van Den Bosch1,2∗ 1 Vesalius Research Center, VIB, Campus Gasthuisberg, Leuven, Belgium 2 Laboratory of Neurobiology, KU Leuven, Leuven, Belgium *Corresponding author: Ludo Van Den Bosch, ludo.vandenbosch@vib-kuleuven.be Histone deacetylase 6 (HDAC6) catalyzes multiple reactions. We summarize the current knowledge on HDAC6, its targets and functions. Among others, HDAC6 recognizes damaged proteins and assures that these proteins are destroyed by autophagy. On the other hand, HDAC6 also modifies the tracks used by the clearance mechanism so that axonal transport becomes less efficient. We hypothesize that a disturbance in the equilibrium between the different functions of HDAC6 could play an important role in neurodegeneration. Key words: autophagy, axonal transport, deacetylation, HDAC6, microtubules, neurodegeneration, post-translational modifications Received 27 September 2011, revised and accepted for publication 23 February 2012, uncorrected manuscript published online 28 February 2012, published online 26 March 2012 Histone deacetylase (HDAC) enzymes are intriguing targets involved in a broad range of biological processes, including genetic, epigenetic and oncogenic processes (1–4). HDACs ensure the reversible acetylation of histones and play a crucial role in transcriptional regulation and histone metabolism (1,5). Given their importance in these processes, a wide range of medicinal chemistry studies aimed to develop HDAC inhibitors to treat tumors and other malignancies (2,4). Recently, the attention of scientists working in the field of neurodegeneration has been drawn to HDACs, and in particular to HDAC6, as it could be a key player in many neurodegenerative disorders. In this review, we will summarize the current knowledge on HDAC6, its targets and functions in autophagy and axonal transport, and we propose a new model in which HDAC6 orchestrates its activities in neurodegenerative diseases. HDAC6 and the HDAC Family The HDAC family consists of 18 members classified into two groups: zinc-dependent enzymes (HDAC1–11) and nicotinamide adenine dinucleotide (NAD+ )-dependent enzymes (sirtuin 1–7) (Figure 1). On the basis of sequence homology of the catalytic domain to yeast, HDACs have been further classified into different classes. Class I HDACs (HDAC1, 2, 3 and 8) are homologous to the yeast Rpd3 protein (Figure 1). They are ubiquitously expressed and are generally detected in the nucleus. Class II comprises HDAC4, 5, 7 and 9 (class IIa) as well as HDAC6 and 10 (class IIb) (Figure 1). Class II HDACs share sequence homology with the yeast Hda1 protein and shuttle between the nucleus and the cytoplasm. HDAC11 is the only member of class IV HDACs (Figure 1). It shows similarity to the catalytic domains of both class I and II enzymes. Class III HDACs comprise the NAD+ dependent sirtuins that are homologous to the yeast Sir2 protein (Figure 1). The amino acid sequence outside the catalytic domain is highly variable suggesting that these enzymes might exert their deacetylating function on other substrates than histones. Moreover, HDAC6 is unique as it is the only HDAC that contains two functional N-terminally located catalytic sites in combination with an ubiquitin-binding domain at the C-terminal part of the protein (Figure 1) (6). HDAC10 contains a second leucine-rich region that is thought to represent a catalytically inactive deacetylase domain (7). HDAC6 is also the only HDAC with an exclusive cytoplasmic localization, suggesting that its activity is histone-independent and does not influence transcriptional processes (6). Target Proteins for HDAC6-Mediated Deacetylation A major breakthrough in the search of molecular targets was the discovery of acetylated tubulin in polymerized microtubules as a substrate for HDAC6 (6,8,9). Pharmacological inhibition or downregulation of HDAC6 in cellular and animal models increases acetylated tubulin abundance (6,8–13). Several proteins acting through different cellular pathways regulate HDAC6-mediated deacetylation of microtubules. The Akt-dependent glycogen synthase kinase-3β (GSK-3β) increases the deacetylase activity of HDAC6 on tubulin as shown in hippocampal neurons (Figure 2)(12). Although GSK-3β and HDAC6 do colocalize, a direct interaction between these two proteins has not been demonstrated (12). Alternatively, the tubulin polymerization-promoting protein (TPPP/p25) is an unstructured protein that aligns www.traffic.dk 771 d’Ydewalle et al. Figure 1: Classification of HDAC enzymes. Zinc (Zn)-dependent or NAD+ -dependent HDACs are shown to scale and are categorized into classes (I–IV) according to their homology to yeast. Class I HDACs (1, 2, 3 and 8) show homology to Rpd3. Class II HDACs, including 4, 5, 7 and 9 (class IIa) as well as 6 and 10 (class IIb), are homologs of yeast Hda1. Class III HDACs comprise SIRT1–7 and show homology to yeast Sir2. Class IV HDACs (HDAC11) show homology to both class I and II HDACs. HDAC6 is the only HDAC with two N-terminal active deacetylating domains and a C-terminal ubiquitin-binding domain. along the microtubule network and affects microtubule dynamics and cell motility (14). Binding of TPPP/p25 to HDAC6 directly inhibits the deacetylating function of HDAC6 in HeLa cells (Figure 2) (14). Finally, the microtubule-associated protein Tau, which ensures stabilization of the microtubular network, also binds to HDAC6 and decreases the deacetylase activity of HDAC6 in neuronal cell lines (Figure 2) (15,16). In line with these findings, Tau knockout mice demonstrate decreased acetylated tubulin levels as the inhibitory effect of Tau on HDAC6 is abolished (16). Recently, it has been found that Tau is deacetylated mainly by sirtuin-1, a class III HDAC, and to a lesser extent by sirtuin-2 and HDAC6 (17). Thus, HDAC6 plays a regulatory role in the stability of microtubule dynamics, thereby influencing neuronal extension and polarization of neurons (8,11). Acetylation is required for the recruitment and anchoring of molecular motor proteins coupled to their cargo and to microtubules (18). It has been proposed that HDAC6 might regulate axonal transport either directly by binding to motor proteins or indirectly by altering the acetylation state of microtubules. HDAC6 indeed interacts with core components of the dynactin complex such as p150(glued) and actin-related protein-1 (Arp1) in non-neuronal cell lines (Figure 2) (6,9). Moreover, inhibition or knockdown of HDAC6 in cultured neurons increases the axonal transport of mitochondria by increasing acetylated tubulin amounts and the concomitant recruitment of motor protein dynein and kinesin-1 to microtubules (10,12). Microtubules are in a dynamic equilibrium consisting of stages of depolymerization, repolymerization and stable microtubules. Microtubules containing high levels of acetylated tubulin are more resistant to drug-induced depolymerization indicating that tubulin acetylation is 772 a marker for stable microtubules in fibroblasts (8). Overexpression of HDAC6 induces depolymerization and subsequent deacetylation, indicating that tubulin dimers and monomers are deacetylated, while acetylation only occurs in polymerized microtubules (8). In cultured neurons, HDAC6 is enriched at the distal ends of axons where it interacts with end-binding protein-1 (EB-1) and Arp1, both accumulating at growing tips of microtubules (Figure 2) (9). Moreover, inhibition or downregulation of HDAC6 results in reduced axonal length and decreased growth and shrinkage velocity of microtubules (9,11). As HDAC6 affects microtubule dynamics, it has been suggested that this enzyme also regulates cell motility. However, global HDAC6-mediated deacetylation of tubulin in mouse embryonic fibroblasts is not sufficient to induce changes in cell motility (19). Upon stimulation with growth factors, HDAC6 translocates to actin-enriched membrane ruffles where it binds and deacetylates cortactin (Figure 2) (19,20). The latter protein is a cortical actin-binding protein and promotes the polymerization and rearrangement of the actin cytoskeleton, especially the actin cortex around the cellular periphery. Deacetylation of cortactin changes its binding capacity to F-actin, eventually altering actin polymerization (20). More recently, redox regulatory proteins peroxiredoxin I (PRX1) and II (PRX2) were identified as specific targets of HDAC6 in non-neuronal cell lines (21). PRX1 and PRX2 are antioxidant enzymes that reduce H2 O2 levels (Figure 2). Acetylation of PRX increases their reducing activity, their resistance to superoxidation and to transition to high-molecular mass complexes. Inhibition of HDAC6 deacetylase activity with a consequent accumulation of acetylated PRX could lead to a beneficial increase in antioxidant activity (21). Traffic 2012; 13: 771–779 Histone Deacetylase 6 in Neurodegeneration Figure 2: HDAC6 and its substrates for deacetylation. HDAC6 interacts with a wide variety of targets. HDAC6 binds to and deacetylates polymerized microtubules. It also interacts with EB-1, p150(glued) and Arp1 of the dynactin motor complex, affecting microtubule dynamics and cell motility. While the Akt-dependent GSK-3β increases HDAC6-mediated tubulin deacetylation, both Tau and TPPP/p25 inhibit this reaction. HDAC6 also interacts with and deacetylates cortactin, changing its binding affinity to filamentous actin and ultimately changing actin polymerization. HDAC6 deacetylates PRX1 and PRX2 altering their reducing capacity of water peroxide (H2 O2 ) to hydrogen (H2 O). Finally, HSP90 is deacetylated by HDAC6 affecting the downstream targets of HSP90. Finally, the molecular ATP-dependent chaperone heatshock protein 90 (HSP90) is also a substrate of HDAC6 (Figure 2) (22). The reversible acetylation of HSP90 regulates its chaperone activity (22–24). As a chaperone protein, HSP90 is a critical player in a wide variety of cellular signaling pathways as it facilitates the structural maturation and assembly of its client proteins. HSP90 binds to multiple client proteins, including heat-shock factor 1 (HSF1) under basal conditions. In HDAC6-deficient regulatory T cells, HSF1-regulated genes including HSP90 were upregulated (25,26). The proteins encoded by these genes showed an increased acetylation state compared to wild-type cells (25,26). Furthermore, as we will discuss in detail in the next chapter, HDAC6 senses the presence of ubiquitinated aggregates and consequently induces the expression of major cellular chaperones by triggering the dissociation of a repressive HDAC6/HSF1/HSP90 complex and subsequent HSF1 activation (27). Inhibition of HDAC6 also induces a reduction in binding of HSP90 to vascular endothelial growth factor receptor 1 and 2 (VEGFR1/Flt1 and VEGFR2/Flk1, respectively), followed by increased binding of HSP70 to these receptors (28). Knockdown of HDAC6 leads to a depletion of VEGFR1/Flt1 and VEGFR2/Flk1 (28). These results indicate that HDAC6 plays an important role in the HSP90-mediated Traffic 2012; 13: 771–779 regulation of VEGFRs, and possibly other targets of HSP90 (28). Interestingly, vascular endothelial growth factor (VEGF) has protective effects in motor neuron disease, as shown both in vitro and in vivo (29–32). The direct or indirect effects of HDAC6-mediated deacetylation on molecular chaperones that are associated with VEGF (and its receptors) and neurodegeneration suggest that HDAC6 might play a role in neurodegenerative diseases. Exciting information has been obtained in transgenic mice with targeted disruption of HDAC6. Young HDAC6 knockout mice do not show any overt phenotype (33). Genetic ablation of HDAC6 leads to hyperacetylation of microtubules and HSP90 without any obvious abnormal effects on development and survival of these mice (33). Another strain of HDAC6 knockout mice demonstrate ubiquitin-positive aggregates and apoptotic cell death, both pathogenic hallmarks of neurodegeneration, in the brain from 6 months of age on (34). Similarly, a neurodegenerative phenotype was observed in Drosophila with selective HDAC6 deficiency in photoreceptor neurons (34). Together, these in vivo data indicate that HDAC6 plays a crucial protective role in neurodegeneration. 773 d’Ydewalle et al. Figure 3: The role of HDAC6 in autophagy. A finely tuned balance between VCP/p97 and HDAC6 directs ubiquitinated aggregates either to the UPS or to the autophagic pathway. HDAC6 binds to ubiquitinated aggregates via its ubiquitin-binding domain as well as to dynein motor proteins. A dynein-mediated retrograde transport leads to accumulation of aggregates into an aggresome at the perinuclear region. HDAC6 also contributes to autophagosome formation by interacting with ATGs. HDAC6 also participates in the fusion of autophagosomes and lysosomes to autophagolysosomes, the final step prior to lysosomal degradation of ubiquitinated aggregates. FUS/TLS and TDP-43 bind to HDAC6 mRNA in stress granules. Role of HDAC6 in Autophagy Cells deal with protein aggregates in multiple ways. In several of these processes, HDAC6 has been shown to play a crucial role. Autophagocytosis is a catabolic process in which degradation of damaged organelles and/or ubiquitinated proteins and aggregates occurs through the lysosomal pathway. Microautophagy is a process in which the lysosomes directly engulf cytoplasm by invagination, protrusion or septation of the lysosomallimiting membrane. Macroautophagy, also referred to as autophagy, on the other hand, sequesters damaged organelles and ubiquitinated aggregates in a doublemembrane vesicle called autophagosomes that fuse with lysosomes. Neuronal survival is dependent on autophagy due to its postmitotic nature, the polarized morphology and active protein trafficking (35). The majority of studies suggest that autophagy is a compensatory mechanism that is activated when the ubiquitin proteasome system (UPS) is impaired (36,37). It is still unclear, however, whether autophagy and the UPS are independent processes (38). Nevertheless, as HDAC6 contains an ubiquitin-binding domain, it was suggested that it also plays a role in protein quality control (34,37,39). As stated above, HDAC6 inhibition triggers the dissociation of a repressive HDAC6/HSP90/HSF1 complex and leads to HSF1 activation and induces the expression of 774 HSF1-responsive genes including molecular chaperones (Figure 3) (27). These chaperones are intrinsically associated with ubiquitinated misfolded proteins that are prone to aggregation. Nevertheless, HDAC6 is not required for autophagy activation per se. It is rather recruited to poly-ubiquitinated autophagic substrates (27,34,39). A finely tuned balance between valosin-containing protein (VCP)/p97 and HDAC6 directs the fate of ubiquitinated aggregates (Figure 3). VCP/p97 promotes ubiquitin chain turnover and protein degradation through the UPS, whereas HDAC6 favors the accumulation of ubiquitinated proteins and subsequent perinuclear aggregation (27,40,41). This hypothesis is strengthened by the observation that VCP/p97 and HDAC6 interact with each other (Figure 3) (41). However, the functional consequences of this interaction are not yet clear. Aggregation can occur both in the cell body and in extended processes of neurons, while the aggresome (i.e. accumulated aggregates) is formed at the perinuclear region. This implies that ubiquitinated aggregates are transported retrogradely toward this region. An elegant study demonstrated that HDAC6 associates with the dynein motor protein complex and recruits ubiquitinated protein aggregates to dynein (Figure 3) (39). Next, autophagic vacuoles are formed that encapsulate the ubiquitinated aggregates. The formation of these Traffic 2012; 13: 771–779 Histone Deacetylase 6 in Neurodegeneration autophagosomes depends on autophagy-related (Atg) proteins that are molecular determinants during nucleation, elongation and maturation of these vacuoles. Pharmacological inhibition and ablation of HDAC6 interferes with the recruitment of Atg-8/LC3 to the aggresome (Figure 3) (37). HDAC6 also stimulates autophagosome–lysosome fusion by promoting F-actin remodeling in a cortactindependent manner (Figure 3) (34). Lysosomal recruitment also requires an intact microtubule structure as druginduced depolymerization of microtubules disrupts perinuclear accumulation of lysosomes (Figure 3) (37,42). Conversely, HDAC6 deficiency leads to failure of autophagosome maturation, protein aggregate build-up and neurodegeneration (34). Another essential part of the cellular response to protein aggregation is the reversible translational suppression. This process occurs in cytoplasmic stress granules (SGs) that are dense globules containing stress proteins and untranslated mRNAs. SGs are formed in a microtubuledependent manner during cellular stress and are protective in nature (42). Like autophagy, SGs are formed when the UPS is impaired, suggesting that both autophagy and translational suppression occur simultaneously (43). HDAC6 appears to colocalize and interact with Ras-GAP SH3 domain-binding protein (G3BP), an SG marker, indicating that HDAC6 might contribute to SG formation (Figure 3) (42). The contribution of HDAC6 to SG formation depends on its two hdac domains (42). Pharmacological inhibition or genetic disruption of HDAC6 indeed abolishes SG formation (42). The deacetylating function of HDAC6 is thus critically important for SG formation. Whether G3BP or other SG markers are activated directly or indirectly by HDAC6-mediated deacetylation is not known. Blocking the dynein motor proteins does not affect the localization of HDAC6, while it does impair SG formation (42). Like in the autophagic pathway, an intact and densely acetylated microtubular network seems to contribute to SG formation, possibly by enhancing trafficking of SG components (42). TAR DNA-binding protein-43 (TDP-43) and fused in sarcoma/translocated in liposarcoma (FUS/TLS) are RNAbinding proteins that are found in SGs. TDP-43 and FUS/TLS form a functional complex that binds to HDAC6 mRNA (Figure 3). Silencing either TDP-43 or FUS/TLS reduces HDAC6 mRNA levels, indicating a crucial link between these RNA-binding proteins and HDAC6 (44). Competition experiments indicated that TDP-43 and FUS/TLS occupy overlapping binding sites on HDAC6 mRNA (44). A Drosophila model in which TDP-43 is silenced confirmed the decreased HDAC6 expression. In cells in which TDP-43 is silenced, cellular aggregate formation and increased cytotoxicity are found (45). Finally, HDAC6 overexpression is able to rescue the phenotype of a Drosophila model of spinobulbar muscular Traffic 2012; 13: 771–779 atrophy, indicating that HDAC6 might partially counteract the pathogenesis of neurodegenerative disorders characterized by an impaired UPS (36). All together, these data strongly suggest that HDAC6 is a master regulator of the protective response to cytotoxic protein aggregation through autophagy and SG formation. HDAC6 is a Key Enzyme in Neurodegenerative Diseases Recently, it has become clear that microtubule acetylation and microtubule dynamics play a crucial role in flagellar transport of cilia. Moreover, HDAC6 has been shown to be an important tubulin deacetylase, in particular of axonemal microtubules. HDAC6 thus might be a promising therapeutic target for ciliopathies that give rise to a wide variety of symptoms such as multiorgan failure with variable central nervous system involvement (46–48). Ubiquitinated aggregates are a common feature of many neurodegenerative disorders. Given its role in autophagic degradation of ubiquitinated aggregates, HDAC6 has been associated with a wide variety of neurodegenerative diseases. Alzheimer’s disease is the most common form of dementia and is characterized by amyloid plaques and neurofibrillary Tau tangles in and around cortical neurons. In patients with Alzheimer’s disease, contradicting information has been obtained. Although HDAC6 appears to be upregulated, acetylated tubulin levels are increased in neurons containing neurofibrillary Tau pathology (15,16). Inhibition of the proteasome system in cellular models potentiates the interaction between HDAC6 and Tau, and facilitates perinuclear aggregation of Tau independently of the deacetylase activity (15). The characteristic hallmarks of Parkinson’s disease (PD) include movement-related symptoms (such as shaking, rigidity and slowness of movement) and difficulty with walking caused by selective neuronal death in the substantia nigra. Cellular models demonstrated that HDAC6 expression increases and colocalizes with mutant α-synuclein, one of the genetic causes of familial PD, in the perinuclear region forming aggresome-like structures (49). HDAC6 deficiency blocks the formation of aggregates and interferes with autophagy (49). Immunohistochemical and ultrastructural analysis of post mortem tissue from patients with PD and dementia with Lewy bodies (DLBs) demonstrated strong HDAC6 positivity on the filamentous structures within Lewy bodies (39,50). Parkin, associated with early-onset PD, promotes mitophagy by catalyzing mitochondrial ubiquitination (51). This recruits ubiquitinbinding autophagic components HDAC6 and p62, leading to mitochondrial clearance (51). In a Drosophila model for PD, depletion of HDAC6 significantly increases the loss of dopaminergic neurons, retinal degeneration and locomotor dysfunction (52). Conversely, overexpression 775 d’Ydewalle et al. of HDAC6 promotes inclusion formation and suppresses the α-synuclein-induced loss of neurons and retinal degeneration (52). Huntington’s disease (HD) is caused by an increase in CAG repeats (>28 repeats) in the Huntingtin (HTT ) gene [poly(Q) in Htt] and leads to defects in muscle coordination (‘chorea’), cognitive decline and dementia. In HD brains, acetylated tubulin levels are decreased (10). In vitro models of HD demonstrated that HDAC6 inhibition restores acetylated tubulin abundance and subsequently the axonal transport defects observed in diseased states (10). However, genetic disruption of HDAC6 in HD mice indicated that the aggregate load as well as the axonal transport defects are not restored, while tubulin acetylation is increased (53). HDAC6 deficiency in HD mice also does not influence the disease progression at the behavioral level (53). Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by the selective loss of motor neurons in the spinal cord, brainstem and motor cortex. The symptoms include spasticity and rapidly progressive weakness and muscle atrophy followed by death due to respiratory failure. Ubiquitinated aggregates have been observed in several models as well as in patients, indicating that HDAC6 might be involved. This notion is supported by the association of mutations in TDP-43 and FUS/TLS as a cause of familial ALS, which form a complex and bind to HDAC6 mRNA in SGs. In addition, mutations in VCP/p97 have been associated with ALS (54). Mutant VCP mice demonstrate progressive muscle weakness, TDP-43 pathology in muscle and brain. Although it has been suggested that an impaired nuclear factor-κB (NFκB) pathway or decreased ATP levels might underlie the phenotype in mouse and Drosophila models, respectively, the close links between VCP, TDP-43 and HDAC6 suggest that HDAC6 might contribute to disease pathogenesis (54–56). Recently, mutations in the gene encoding ubiquilin-2 (UBQLN-2 ) have been associated with hereditary ALS (57). Ubiquilin-2 physically associates with both the proteasome and ubiquitin ligases and thus functionally links the ubiquitination machinery to the UPS. These findings further indicate that the autophagic pathway might be compromised in the underlying disease mechanism of ALS (57). Treatment with an HDAC inhibitor also delays disease progression and increases survival in a mouse model of ALS, further supporting the notion that HDAC6 is a central enzyme in the disease (58). Moreover, rapid muscle denervation due to axonal loss and defects in axonal transport is observed in cellular and animal models of ALS in which HDAC6 also might play a role (18,59). All together, these observations point toward a potential role of HDAC6 in ALS pathology. However, a cross-breeding experiment with HDAC6−/− mice and mouse models of ALS will help to elucidate the exact role of HDAC6 in ALS pathology. Charcot-Marie-Tooth (CMT) disease is the most common inherited disorder of the peripheral nervous system. 776 Clinical symptoms include progressive weakness and atrophy of distal limb muscles, sensory defects, skeletal deformities and steppage gait. Approximately 40 causative genes have been associated with CMT. Although biochemical studies are sparse, some of the mutant proteins have been shown to form aggregates in cellular models of CMT (60–65). Moreover, mouse models recapitulating mutant small heat-shock protein B1 (HSPB1)-induced CMT demonstrate reduced acetylated tubulin levels in peripheral nerves, leading to severe axonal transport defects (13,62). Although the exact link between mutant HSPB1 and HDAC6 remains elusive, pharmacological inhibition of HDAC6 rescues the phenotype of mutant HSPB1 mice and restores the axonal transport defects (13). Increased expression of HDAC6 is observed after injury to neurons (66). Pharmacological and genetic approaches demonstrate that HDAC6 inhibition promotes survival and regeneration of neurons, indicating that HDAC6 is a target for both regeneration and protection in neurodegenerative diseases (66). Hypothesis: HDAC6 Combines Protective and Degenerative Functions To reconcile both protective and degenerative functions of HDAC6, we propose a two-hit model in which both functions of HDAC6 lead to disease. The combined presence of deacetylating domains and an ubiquitinbinding motif within the same enzyme implies that there is a structural and functional link between both functions. In many neurodegenerative disorders, toxic ubiquitinated aggregates accumulate during the disease process (Figure 4). HDAC6-mediated HSF1 activation increases stress-response protein levels in order to target misfolded proteins to the UPS, which represents the first line of defense of the neuron (Figure 4). This process eventually saturates and/or the UPS becomes impaired. When this occurs, HDAC6 drives the ubiquitinated aggregates to the autophagy pathway by shifting the balance from the UPS to autophagy through its interaction with VCP/p97 (Figure 4) (27,36,37). HDAC6 also recruits the aggregates through dynein motors that transport them to the perinuclear region for aggresome formation (Figure 4) (39). Simultaneously, HDAC6 contributes to SG formation in order to preserve untranslated mRNAs (Figure 4) (42,43). In general, HDAC6-mediated deacetylation of its substrates is considered to have a beneficial effect on neuronal functioning as described above. Under basal conditions, HDAC6 might have only moderate deacetylating capacity. It has been suggested that an excess of HDAC6 favors the accumulation of ubiquitinated aggregates (67). The accumulation of HDAC6 thus could lead to increased tubulin and cortactin deacetylation eventually causing a disturbance of microtubular and actin dynamics, Traffic 2012; 13: 771–779 Histone Deacetylase 6 in Neurodegeneration Figure 4: Proposed roles of HDAC6 in neuroprotection and neurodegeneration. Over time, aggregate-prone proteins form insoluble ubiquitin-positive aggregates that lead to impairment of the UPS. VCP/p97 shifts the equilibrium from the UPS to the autophagic pathway. HDAC6 is recruited to ubiquitinated aggregates. Once recruited, HDAC6 plays both neuroprotective and neurotoxic functions. HDAC6 is involved in stress granule formation in which translation is silenced in stressed cells. Dynein motor proteins interact with HDAC6 bound to ubiquitinated aggregates. This complex is then transported to the perinuclear region where the aggresome is formed. HDAC6 recruitment to aggregates also leads to a local accumulation of HDAC6 causing hyperdeacetylation of its substrates. Deacetylation of HSP90 and peroxiredoxins causes toxic insults, while deacetylation of tubulin and cortactin leads to cytoskeletal alterations. The accumulation of toxic insults and changes in microtubule and actin dynamics eventually lead to neurodegeneration. respectively (Figure 4). Both are crucial cytoskeletal structures for neurons and their axons. Moreover, deacetylation of tubulin potentially compromises axonal transport, as recruitment and anchoring of motor proteins are reduced (10,13). Increased HDAC6-mediated deacetylation of PRXs and HSP90, combined with the perinuclear load of aggresomes, could lead to higher incidence of toxic insults (including increased presence of aggregate-prone proteins and reactive oxygen species) eventually inducing neurodegenerative diseases (Figure 4). In conclusion, our model illustrates the main neuroprotective function of HDAC6 as a clearance mechanism for ubiquitinated proteins. At later stages of the disease, the equilibrium could shift to effects that contribute to neurodegeneration, including a disturbance of the axonal transport. An immediate consequence of this hypothesis is that therapeutic strategies should focus on the negative aspects of HDAC6, leaving the beneficial effects unaffected. Acknowledgments Research of the authors is supported by grants from the Fund for Scientific Research Flanders (FWO-Vlaanderen), the University of Leuven, the Belgian government (Interuniversity Attraction Poles, programme P6/43 of the Belgian Federal Science Policy Office), the Association Belge contre Traffic 2012; 13: 771–779 les Maladies neuro-Musculaires (ABMM), the Association Française contre les Myopathies (AFM), the Frick Foundation for Amyotrophic Lateral Sclerosis Research, the Muscular Dystrophy Association, the European Community’s Health Seventh Framework Programme (FP7/2007-2013 under grant agreement 259867) and the Latran Foundation. C. d. Y. is supported by the Agency for Innovation by Science and Technology in Flanders (IWT-Vlaanderen). E. B. is a postdoctoral fellow of FWOVlaanderen. References 1. Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004;429:457–463. 2. Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 2006;5:769–784. 3. Grunstein M. Histone acetylation in chromatin structure and transcription. Nature 1997;389:349–352. 4. Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov 2002;1:287–299. 5. Wade PA, Pruss D, Wolffe AP. Histone acetylation: chromatin in action. Trends Biochem Sci 1997;22:128–132. 6. Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang X-F, Yao T-P. HDAC6 is a microtubule-associated deacetylase. Nature 2002;417:455–458. 7. Tong JJ, Liu J, Bertos NR, Yang X-J. Identification of HDAC10, a novel class II human histone deacetylase containing a leucine-rich domain. Nucleic Acids Res 2002;30:1114–1123. 8. Matsuyama A, Shimazu T, Sumida Y, Saito A, Yoshimatsu Y, Seigneurin-Berny D, Osada H, Komatsu Y, Nishino N, Khochbin S, Horinouchi S, Yoshida M. In vivo destabilization of dynamic microtubules by HDAC6-mediated deacetylation. EMBO J 2002;21:6820–6831. 9. Zilberman Y, Ballestrem C, Carramusa L, Mazitschek R, Khochbin S, Bershadsky A. Regulation of microtubule dynamics by inhibition 777 d’Ydewalle et al. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. of the tubulin deacetylase HDAC6. J Cell Sci 2009;122: 3531–3541. Dompierre JP, Godin JD, Charrin BC, Cordelières FP, King SJ, Humbert S, Saudou F. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci 2007;27:3571–3583. Tapia M, Wandosell F, Garrido JJ. Impaired function of HDAC6 slows down axonal growth and interferes with axon initial segment development. PLoS ONE 2010;5:e12908. Chen S, Owens GC, Makarenkova H, Edelman DB. HDAC6 regulates mitochondrial transport in hippocampal neurons. PLoS ONE 2010;5:e10848. d’Ydewalle C, Krishnan J, Chiheb DM, Van Damme P, Irobi J, Kozikowski AP, Vanden Berghe P, Timmerman V, Robberecht W, Van Den Bosch L. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1–induced Charcot-Marie-Tooth disease. Nat Med 2011;17:968–974. Tokesi N, Lehotzky A, Horvath I, Szabo B, Olah J, Lau P, Ovadi J. TPPP/p25 promotes tubulin acetylation by inhibiting histone deacetylase 6. J Biol Chem 2010;285:17896–17906. Ding H, Dolan PJ, Johnson GVW. Histone deacetylase 6 interacts with the microtubule-associated protein tau. J Neurochem 2008;106:2119–2130. Perez M, Santa-Maria I, de Barreda EG, Zhu X, Cuadros R, Cabrero JR, Sánchez-Madrid F, Dawson HN, Vitek MP, Perry G, Smith MA, Avila J. Tau - an inhibitor of deacetylase HDAC6 function. J Neurochem 2009;109:1756–1766. Min S-W, Cho S-H, Zhou Y, Schroeder S, Haroutunian V, Seeley WW, Huang EJ, Shen Y, Masliah E, Mukherjee C. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 2010;67:953–966. De Vos KJ, Grierson AJ, Ackerley S, Miller CCJ. Role of axonal transport in neurodegenerative diseases. Ann Rev Neurosci 2008;31:151–173. Gao YS, Hubbert CC, Lu J, Lee YS, Lee JY, Yao TP. Histone deacetylase 6 regulates growth factor-induced actin remodeling and endocytosis. Mol Cell Biol 2007;27:8637–8647. Zhang X, Yuan Z, Zhang Y, Yong S, Salas-Burgos A, Koomen J, Olashaw N, Parsons JT, Yang XJ, Dent SR, Yao TP, Lane WS, Seto E. HDAC6 modulates cell motility by altering the acetylation level of cortactin. Mol Cell 2007; 27:197–213. Parmigiani RB, Xu WS, Venta-Perez G, Erdjument-Bromage H, Yaneva M, Tempst P, Marks PA. HDAC6 is a specific deacetylase of peroxiredoxins and is involved in redox regulation. Proc Natl Acad Sci U S A 2008;105:9633–9638. Kovacs J, Murphy P, Gaillard S, Zhao X, Wu J, Nicchitta C, Yoshida M, Toft D, Pratt W, Yao T. HDAC6 regulates Hsp90 acetylation and Chaperone-Dependent Activation of Glucocorticoid Receptor. Mol Cell 2005;18:601–607. Bali P. 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 2005;280:26729–26734. Scroggins BT, Robzyk K, Wang D, Marcu MG, Tsutsumi S, Beebe K, Cotter RJ, Felts S, Toft D, Karnitz L, Rosen N, Neckers L. An acetylation site in the middle domain of Hsp90 regulates chaperone function. Mol Cell 2007;25:151–159. de Zoeten EF, Wang L, Butler K, Beier UH, Akimova T, Sai H, Bradner JE, Mazitschek R, Kozikowski AP, Matthias P, Hancock WW. Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3+ T-regulatory cells. Mol Cell Biol 2011;31:2066–2078. Wang L, de Zoeten EF, Greene MI, Hancock WW. Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of FOXP3+ regulatory T cells. Nat Rev Drug Discov 2009;8:969–981. Boyault C, Zhang Y, Fritah S, Caron C, Gilquin B, Kwon SH, Garrido C, Yao TP, Vourc’h C, Matthias P, Khochbin S. HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates. Genes Dev 2007;21:2172–2181. Park J-H, Kim S-H, Choi M-C, Lee J, Oh D-Y, Im S-A, Bang Y-J, Kim T-Y. Class II histone deacetylases play pivotal roles in heat shock protein 90-mediated proteasomal degradation of vascular endothelial growth factor receptors. Biochem Biophys Res Commun 2008;368:318–322. 778 29. Storkebaum E, Lambrechts D, Dewerchin M, Moreno-Murciano M-P, Appelmans S, Oh H, van Damme P, Rutten B, Man WY, De Mol M, Wyns S, Manka D, Vermeulen K, Van Den Bosch L, Mertens N et al. Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat Neurosci 2004;8:85–92. 30. Van Den Bosch L, Storkebaum E, Vleminckx V, Moons L, Vanopdenbosch L, Scheveneels W, Carmeliet P, Robberecht W. Effects of vascular endothelial growth factor (VEGF) on motor neuron degeneration. Neurobiol Dis 2004;17:21–28. 31. Poesen K, Lambrechts D, van Damme P, Dhondt J, Bender F, Frank N, Bogaert E, Claes B, Heylen L, Verheyen A, Raes K, Tjwa M, Eriksson U, Shibuya M, Nuydens R et al. Novel role for vascular endothelial growth factor (VEGF) receptor-1 and its ligand VEGF-B in motor neuron degeneration. J Neurosci 2008;28:10451–10459. 32. Bogaert E, van Damme P, Poesen K, Dhondt J, Hersmus N, Kiraly D, Scheveneels W, Robberecht W, Van Den Bosch L. VEGF protects motor neurons against excitotoxicity by upregulation of GluR2. Neurobiol Aging 2010;31:2185–2191. 33. Zhang Y, Kwon S, Yamaguchi T, Cubizolles F, Rousseaux S, Kneissel M, Cao C, Li N, Cheng HL, Chua K, Lombard D, Mizeracki A, Matthias G, Alt FW, Kochbin S et al. Mice lacking histone deacetylase 6 have hyperacetylated tubulin but are viable and develop normally. Mol Cell Biol 2008;28:1688–1701. 34. Lee J-Y, Koga H, Kawaguchi Y, Tang W, Wong E, Gao Y-S, Pandey UB, Kaushik S, Tresse E, Lu J, Taylor JP, Cuervo AM, Yao TP. HDAC6 controls autophagosome maturation essential for ubiquitin-selective quality-control autophagy. EMBO J 2010;29:969–980. 35. Lee J-A. Autophagy in neurodegeneration: two sides of the same coin. BMB Rep 2009;42:324–330. 36. 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 et al. HDAC6 rescues neurodegeneration and provides an essential link between autophagy and the UPS. Nature 2007;447:859–863. 37. Iwata A, Riley BE, Johnston JA, Kopito RR. HDAC6 and microtubules are required for autophagic degradation of aggregated huntingtin. J Biol Chem 2005;280:40282–40292. 38. Ding W-X, Yin X-M. Sorting, recognition and activation of the misfolded protein degradation pathways through macroautophagy and the proteasome. Autophagy 2008;4:141–150. 39. Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao T-P. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003;115:727–738. 40. Seigneurin-Berny D, Verdel A, Curtet S, Lemercier C, Garin J, Rousseaux S, Khochbin S. Identification of components of the murine histone deacetylase 6 complex: link between acetylation and ubiquitination signaling pathways. Mol Cell Biol 2001;21:8035–8044. 41. Ju J-S, Miller SE, Hanson PI, Weihl CC. Impaired protein aggregate handling and clearance underlie the pathogenesis of p97/VCPassociated disease. J Biol Chem 2008;283:30289–30299. 42. Kwon SH, Zhang Y, Matthias P. The deacetylase HDAC6 is a novel critical component of stress granules involved in the stress response. Genes Dev 2007;21:3381–3394. 43. Mazroui R, Di Marco S, Kaufman RJ, Gallouzi I-E. Inhibition of the ubiquitin-proteasome system induces stress granule formation. Mol Biol Cell 2007;18:2603–2618. 44. Kim SH, Shanware NP, Bowler MJ, Tibbetts RS. Amyotrophic lateral sclerosis-associated proteins TDP-43 and FUS/TLS function in a common biochemical complex to co-regulate HDAC6 mRNA. J Biol Chem 2010;285:34097–34105. 45. Fiesel FC, Voigt A, Weber SS, van den Haute C, Waldenmaier A, Görner K, Walter M, Anderson ML, Kern JV, Rasse TM, Schmidt T, Springer W, Kirchner R, Bonin M, Neuamnn M et al. Knockdown of transactive response DNA-binding protein (TDP-43) downregulates histone deacetylase 6. EMBO J 2009;29:209–221. 46. Loktev AV, Zhang Q, Beck JS, Searby CC, Scheetz TE, Bazan JF, Slusarski DC, Sheffield VC, Jackson PK, Nachury MV. A BBSome subunit links ciliogenesis, microtubule stability, and acetylation. Dev Cell 2008;15:854–865. 47. Chang J, Baloh RH, Milbrandt J. The NIMA-family kinase Nek3 regulates microtubule acetylation in neurons. J Cell Sci 2009;122:2274–2282. Traffic 2012; 13: 771–779 Histone Deacetylase 6 in Neurodegeneration 48. Li Y, Wei Q, Zhang Y, Ling K, Hu J. The small GTPases ARL-13 and ARL-3 coordinate intraflagellar transport and ciliogenesis. J Cell Biol 2010;189:1039–1051. 49. Su M, Shi J-J, Yang Y-P, Li J, Zhang Y-L, Chen J, Hu L-F, Liu C-F. HDAC6 regulates aggresome-autophagy degradation pathway of α-synuclein in response to MPP+ -induced stress. J Neurochem 2011;117:112–120. 50. Miki Y, Mori F, Tanji K, Kakita A, Takahashi H, Wakabayashi K. Accumulation of histone deacetylase 6, an aggresome-related protein, is specific to Lewy bodies and glial cytoplasmic inclusions. Neuropathology 2011;31:561–568. 51. Lee JY, Nagano Y, Taylor JP, Lim KL, Yao TP. Disease-causing mutations in Parkin impair mitochondrial ubiquitination, aggregation, and HDAC6-dependent mitophagy. J Cell Biol 2010;189:671–679. 52. Du G, Liu X, Chen X, Song M, Yan Y, Jiao R, Wang C-C. Drosophila histone deacetylase 6 protects dopaminergic neurons against {alpha}synuclein toxicity by promoting inclusion formation. Mol Biol Cell 2010;21:2128–2137. 53. Bobrowska A, Paganetti P, Matthias P, Bates GP. Hdac6 knock-out increases tubulin acetylation but does not modify disease progression in the R6/2 mouse model of Huntington’s disease. PLoS ONE 2011 ;6:e20696. 54. Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 2010;68:857–864. 55. Custer SK, Neumann M, Lu H, Wright AC, Taylor JP. Transgenic mice expressing mutant forms VCP/p97 recapitulate the full spectrum of IBMPFD including degeneration in muscle, brain and bone. Hum Mol Genet 2010;19:1741–1755. 56. Chang Y-C, Hung W-T, Chang Y-C, Chang HC, Wu C-L, Chiang A-S, Jackson GR, Sang T-K. Pathogenic VCP/TER94 alleles are dominant actives and contribute to neurodegeneration by altering cellular ATP level in a Drosophila IBMPFD model. PLoS Genet 2011;7:e1001288. 57. Deng HX, Chen W, Hong ST, Boycott KM, Gorrie GH, Siddique N, Yang Y, Fecto F, Shi Y, Zhai H, Jiang H, Hirano M, Rampersaud E, Jansen GH, Donkervoort S et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 2011;477:211–215. 58. Yoo Y-E, Ko C-P. Treatment with trichostatin A initiated after disease onset delays disease progression and increases survival Traffic 2012; 13: 771–779 59. 60. 61. 62. 63. 64. 65. 66. 67. in a mouse model of amyotrophic lateral sclerosis. Exp Neurol 2011;231:147–159. Bilsland LG, Sahai E, Kelly G, Golding M, Greensmith L, Schiavo G. Deficits in axonal transport precede ALS symptoms in vivo. Proc Natl Acad Sci U S A 2010;107:20523–20528. Evgrafov OV, Mersiyanova I, Irobi J, Van Den Bosch L, Dierick I, Leung CL, Schagina O, Verpoorten N, Van Impe K, Fedotov V, Dadali E, Auer-Grumbach M, Windpassinger C, Wagner K, Mitrovic Z et al. Mutant small heat-shock protein 27 causes axonal Charcot-MarieTooth disease and distal hereditary motor neuropathy. Nat Genet 2004;36:602–606. Fortun J, Go JC, Li J, Amici SA, Dunn WA Jr, Notterpek L. Alterations in degradative pathways and protein aggregation in a neuropathy model based on PMP22 overexpression. Neurobiol Dis 2006;22:153–164. Ackerley S, James PA, Kalli A, French S, Davies KE, Talbot K. A mutation in the small heat-shock protein HSPB1 leading to distal hereditary motor neuronopathy disrupts neurofilament assembly and the axonal transport of specific cellular cargoes. Hum Mol Genet 2006;15:347–354. Zhai J, Lin H, Julien J-P, Schlaepfer WW. Disruption of neurofilament network with aggregation of light neurofilament protein: a common pathway leading to motor neuron degeneration due to Charcot-MarieTooth disease-linked mutations in NFL and HSPB1. Hum Mol Genet 2007;16:3103–3116. Fortun J, Verrier JD, Go JC, Madorsky I, Dunn WA, Notterpek L. The formation of peripheral myelin protein 22 aggregates is hindered by the enhancement of autophagy and expression of cytoplasmic chaperones. Neurobiol Dis 2007;25:252–265. Goryunov D, Nightingale A, Bornfleth L, Leung C, Liem RKH. Multiple disease-linked myotubularin mutations cause NFL assembly defects in cultured cells and disrupt myotubularin dimerization. J Neurochem 2007;104:1536–1552. Rivieccio MA, Brochier C, Willis DE, Walker BA, D’Annibale MA, McLaughlin K, Siddiq A, Kozikowski AP, Jaffrey SR, Twiss JL, Ratan RR, Langley B. HDAC6 is a target for protection and regeneration following injury in the nervous system. Proc Natl Acad Sci U S A 2009;106:19599–19604. Boyault C, Sadoul K, Pabion M, Khochbin S. HDAC6, at the crossroads between cytoskeleton and cell signaling by acetylation and ubiquitination. Oncogene 2007;26:5468–5476. 779