Skip to main content
Cellular and Molecular Neurobiology logoLink to Cellular and Molecular Neurobiology
. 2016 Dec 10;37(7):1147–1160. doi: 10.1007/s10571-016-0452-2

Could Sirtuin Activities Modify ALS Onset and Progression?

Bor Luen Tang 1,2,
PMCID: PMC11482121  PMID: 27942908

Abstract

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a complex etiology. Sirtuins have been implicated as disease-modifying factors in several neurological disorders, and in the past decade, attempts have been made to check if manipulating Sirtuin activities and levels could confer benefit in terms of neuroprotection and survival in ALS models. The efforts have largely focused on mutant SOD1, and while limited in scope, the results were largely positive. Here, the body of work linking Sirtuins with ALS is reviewed, with discussions on how Sirtuins and their activities may impact on the major etiological mechanisms of ALS. Moving forward, it is important that the potentially beneficial effect of Sirtuins in ALS disease onset and progression are assessed in ALS models with TDP-43, FUS, and C9orf72 mutations.

Keywords: Amyotrophic lateral sclerosis (ALS), Motor neuron, Neuronal degeneration, Sirtuins

Introduction

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal motor neuron disease with the demise of cortical and spinal motor neurons, and death often results from the eventual loss of respiratory capacity (Zarei et al. 2015; Riva et al. 2016). There is no curative treatment, or any effective therapeutic intervention. Other than physical therapy and assisted ventilation at late stages, the only specific FDA-approved drug is the glutamate transmission moderating riluzole (Miller et al. 2012), which prolongs median survival by 2–3 months, but does not reverse motor neuron degeneration. ALS is a clinically complex disease (Turner and Swash 2015). Although largely sporadic and idiopathic, about 10% of ALS cases are hereditary, with more than 20 gene mutations now known to predispose individuals to familial ALS (Iguchi et al. 2013; Renton et al. 2014). The genetic and clinical view of ALS has changed drastically since the initial identification of mutations in Cu–Zn superoxide dismutase (or SOD1) (Rosen et al. 1993) as a prevalent cause of familial ALS. One major new understanding came with the recognition of a key pathological feature (ubiquitinated inclusions of the RNA/DNA-binding protein TAR DNA-binding protein 43 (TDP-43)) that is shared between familial/sporadic ALS and frontotemporal dementia (FTD) (Arai et al. 2006; Neumann et al. 2006; Mackenzie et al. 2007). This was followed by the identification of ALS causing mutations in another RNA-binding protein, Fused in Sarcoma (FUS) (Kwiatkowski et al. 2009; Vance et al. 2009). The ALS–FTD link and the renewed clinical perception of these as separate ends of a “disease spectrum” with multisystem disorder is further consolidated by the identification of the C9orf72 non-coding hexanucleotide repeat expansion as the most common mutation in familial ALS and FTD (DeJesus-Hernandez et al. 2011; Renton et al. 2011; Bennion Callister and Pickering-Brown 2014). Despite the identification of causative mutations and extensive investigations into the pathological mechanisms underlying ALS, there has been a paucity of advances made in terms of therapeutic interventions (Bartus et al. 2016; Stoica and Sena-Esteves 2016; Mazzini et al. 2016).

The class III histone deacetylases, or Sirtuins (named after its founding member Silent information regulator 2 (Sir2) (Imai et al. 2000) in S. cerevisiae) are NAD+-dependent protein deacetylases (Guarente 2007; Haigis and Sinclair 2010) with a large set of histone and non-histone substrates (Feige and Auwerx 2008). The human genome hosts seven Sirtuin paralogues (SIRT1-7) with differing primary subcellular localizations and sites of action (Haigis and Sinclair 2010). SIRT1, SIRT6, and SIRT7 are predominantly nuclear, while SIRT3, SIRT4, and SIRT5 are mitochondria localized (Gertz and Steegborn 2016). SIRT2, on the other hand, is largely cytoplasmic and is a tubulin deacetylase (Li and Yang 2015). Yeast Sir2 is a part of transcription repressor complexes functioning in expression silencing of multiple yeast gene loci (Imai et al. 2000), and the other Sirtuin orthologues likewise function in transcription regulation via deacetylation of histone, transcription factors, and as components of transcriptional complexes (Feige and Auwerx 2008; Bosch-Presegué and Vaquero 2015).

In the past two decades, extensive investigations in multiple cellular and animal models have implicated Sir2p and its homologues in a myriad of physiological and pathophysiological roles. Sirtuins regulate some key cellular and systemic processes that include specific cell/tissue survival (Hao and Haase 2010; Matsushima and Sadoshima 2015; Petegnief and Planas 2013), energy expenditure and metabolic control (Houtkooper et al. 2012; Nogueiras et al. 2012; Chang and Guarente 2014), as well as tissue homeostasis and repair (Tang 2011; Ng and Tang 2013; Poulose and Raju 2015). Sirtuins’ activities are dependent on the availability of NAD+, which changes according to cellular energy and redox status. Sirtuins are therefore energy and redox sensors, and have indeed been strongly associated with healthspan and lifespan extension effect of dietary or caloric restriction (Cohen et al. 2004; Guarente 2013). A prominent and somewhat controversial aspect of pharmacological Sirtuin regulation is its activation by resveratrol (Pallàs et al. 2013; Hubbard et al. 2013), a natural stilbenoid found in abundance in the skin of grapes and berries which has been shown to promote healthspan and lifespan in yeast (Howitz et al. 2003) and animal models (Wood et al. 2004; Lagouge et al. 2006; Baur et al. 2006). Sirtuin-activating compounds (STACs) with higher specificity and bioavailability than resveratrol have been synthesized and these have much touted clinical promise as therapeutics against aging-associated disorders (Milne et al. 2007; Bonkowski and Sinclair 2016).

Sirtuin activities are often associated with neuronal survival (Min et al. 2013; Herskovits and Guarente 2013; Ng et al. 2015). SIRT1, in particular, is highly expressed in the mammalian brain (Zakhary et al. 2010; Michán et al. 2010) and its activation was shown to confer survival benefits in acute physical trauma and ischemic injury (Zhao et al. 2012a, b; Koronowski and Perez-Pinzon 2015). SIRT1 and the mitochondrial SIRT3 have also been shown to confer benefits in a range of neurodegenerative diseases (Srivastava and Haigis 2011; Min et al. 2013; Ng et al. 2015), prominently Alzheimer’s disease (AD) (Wong and Tang 2016) and Parkinson’s disease (PD) (Tang 2016a, b). On the other hand, SIRT2 activity has been negatively implicated in experimental stroke (Krey et al. 2015), PD (Outeiro et al. 2007), and Huntington’s disease (HD) models. Like that of AD and PD, a number of in vitro and in vivo studies have linked Sirtuins to a potential role in modifying ALS neuropathology. These studies were, however, largely confined to the use of the human SOD1G93A mutant expressed in cells or transgenic mice. This has been a major limitation in the field. In the paragraphs that follow, we summarize the results obtained from these studies and discuss the potential of Sirtuins functioning as modifiers of ALS disease onset and progression.

Sirtuin Levels and Activity Changes in ALS Models and Post-mortem Samples

Transgenic overexpression of SOD1, such as SOD1G93A, recapitulates key neurodegenerative features of ALS, and is thus widely used in cellular and animal models for the disease (Jaarsma et al. 2000). Expression profiling analysis over the years has documented temporal and regional changes in a good number of genes during disease progression (Chen et al. 2004; Ferraiuolo et al. 2007; de Oliveira et al. 2013), and these include markers for other neurodegenerative diseases such as amyloid precursor protein (APP) (Koistinen et al. 2006; Rabinovich-Toidman et al. 2015), tau (Barańczyk-Kuźma et al. 2007), DJ-1 and PTEN-induced putative kinase 1 (PINK1) (Lev et al. 2009; Knippenberg et al. 2013), as well as cyclin-dependent kinase 5 (Cdk5) (Nguyen et al. 2001). SIRT1 is enriched in CNS neurons (Zakhary et al. 2010; Michán et al. 2010), and its levels are known to be reduced in brain tissues of neurodegenerative disease subjects such as Huntington’s disease (Pallàs et al. 2008) and AD (Julien et al. 2009; Lutz et al. 2014). SIRT1 level was found to be reduced in a SOD1G93A expressing motor neuron–neuroblastoma hybrid cell line, ventral spinal cord 4.1 (VSC 4.1), and resveratrol treatment appeared to elevate SIRT1 levels (Wang et al. 2011). Interestingly, a significant increase in SIRT1 expression was observed in the cerebral cortex, hippocampus, thalamus, and spinal cord of symptomatic SOD1G93A mice (Lee et al. 2012). SIRT1 levels in the spinal cords of mutant SOD1G37R mice correlated with disease progression, and become significantly upregulated when neurodegeneration was severe (Kim et al. 2007).

Another study had however found SIRT1 levels to be decreased in the spinal cord, but increased in muscles during disease progression in mice (Valle et al. 2014). SIRT2 transcripts are elevated in the spinal cord in both SOD1G93A and SOD1G86R mice, but this was not reflected in the protein levels (Valle et al. 2014). Analysis of transcript and protein levels of post-mortem human brain tissues instead revealed a significant reduction in SIRT1 and SIRT2 levels in the homogenates of the primary motor cortex (Körner et al. 2013). However, in situ hybridization and immunohistochemistry revealed neuron-specific upregulation of SIRT1, SIRT2, and SIRT5 in the spinal cord (Körner et al. 2013). These discrepancies observed are probably due to differences in Sirtuin level changes in different cell types that vary over time during disease progression. SIRT1, for example, may be transiently upregulated in neurons and glia during the early phase of ALS because of redox and toxic stress, but as neurons and accompanying cells are nearing their time of demise, the levels would likely be diminished.

Studies on the Roles of Sirtuins in Cellular Models of ALS

While intervention-type experiments with either resveratrol/STACs or genetic overexpression of Sirtuin genes have been performed with the major neurodegenerative disorders AD and PD, ALS has received comparatively less attention. The reported works with cellular models are summarized in Table 1. One of the first investigations in this regard by Kim and colleagues showed that resveratrol treatment significantly attenuated SOD1G93A-mediated neurotoxicity in transfected primary neurons (Kim et al. 2007). This attenuation was also observed with the co-transfection of SIRT1, but not with a deacetylase-dead SIRT1-H363Y mutant. In another report, death of the motor neuron-like VSC 4.1 cells transfected with SOD1G39A was attenuated by resveratrol treatment (Wang et al. 2011). Interestingly, resveratrol elevated the SOD1G39A-depressed SIRT1 levels (compared with cells transfected with wild-type SOD1 or empty vector) in VSC 4.1 cells. While Sirtuin transcript and protein levels have been shown to be altered by resveratrol treatment in other contexts (Franco et al. 2010; Schirmer et al. 2012), it is possible that this effect may be indirect and at least partially due to the improved cell survival and health.

Table 1.

A summary of reports linking manipulation of Sirtuin levels or activities in ALS cellular and animal models

ALS model Intervention Reference
Neuronal culture models
 Rat cortical primary neurons transfected with SOD1G93A

1. Resveratrol attenuated mutant SOD1G93A-mediated neurotoxicity

2. Overexpression of SIRT1 (but not a deacetylase-dead mutant) protected against SOD1G93A toxicity

Kim et al. (2007)
 Motor neuron-like ventral spinal cord 4.1 cells (VSC 4.1) transfected with SOD1G93A Resveratrol treatment promoted neuronal viability, which was partially blocked by SIRT1 inhibition Wang et al. (2011)
 Exogenous expression of SIRT3 in motor neuron cultures from SOD1G93A mice SIRT3 and its substrate PGC-1α protected against SOD1G93A-induced mitochondrial defects and cell death Song et al. (2013)
 Co-culture of SOD1G93A spinal cord astrocytes with motor neurons Enhancing NAD+ salvage pathway and overexpression of SIRT3 (but not SIRT1) attenuated the neurotoxic phenotype of SOD1G93A astrocytes Harlan et al. (2016)
Mouse models
 Transgenic SOD1G93A mice Resveratrol (single dose) treatment did not improve survival or motor performance Markert et al. (2010)
 Transgenic SOD1G93A mice Resveratrol delayed disease onset and extended survival, evidence for HSP1 and p53 deacetylation Han et al. (2012)
 Transgenic SOD1G93A mice Resveratrol significantly extended lifespan and promoted survival of spinal motor neurons Mancuso et al. (2014)
 Transgenic SOD1G93A mice Resveratrol significantly delayed the disease onset and prolonged the lifespan of ALS mice Song et al. (2014)
 Transgenic SOD1G93A mice with low or high mutant protein expression Crossing mutant SOD1 mice with PrP-SIRT1 transgenics with overexpression of SIRT1 in the brain and spinal cord. Sirt1 transgene conferred longer lifespan for low levels of mutant SOD1 mice, but did not alter time of symptomatic onset Watanabe et al. (2014)

Other than SIRT1, the mitochondrial SIRT3 was also shown to promote neuronal survival against SOD1G93A. Expression of the mutant caused mitochondrial fragmentation and arrested mitochondrial transport in culture rat spinal cord motor neurons, and this defect is countered by the expression of both SIRT3 and its substrate Peroxisome proliferator-activated receptor γ Coactivator-1α (PGC-1α) (Song et al. 2013). Motor neuron demise in SOD1G93A-associated ALS is a complex phenomenon, and has non-cell autonomous components. It has been demonstrated that astrocytes are critical determinants of ALS progression (Yamanaka et al. 2008). Mutant SOD1 expressing astrocytes caused degeneration of co-cultured motor neurons (Nagai et al. 2007; Di Giorgio et al. 2007). In fact, astrocytes generated from post-mortem tissue (Haidet-Phillips et al. 2011) or by reprograming fibroblasts (Meyer et al. 2014) of both familial and sporadic ALS patients, exhibited non-cell autonomous toxicity towards co-cultured neurons. It was recently shown that enhancing the NAD+ salvage pathway (which would presumably activates Sirtuins) in astrocytes isolated from SOD1G93A mice or spinal cord-derived astrocytes from post-mortem ALS subjects attenuated their toxicities toward co-cultured motor neurons. Furthermore, overexpression of SIRT3 (but not SIRT1) rescued the motor neuron toxicity conferred by SOD1G93A astrocytes (Harlan et al. 2016).

On the other hand, the role of other Sirtuin paralogues in ALS is unclear. SIRT2, which is known to act negatively in promoting degeneration in other neurodegenerative disease models (Outeiro et al. 2007; Luthi-Carter et al. 2010), may not have a significant role in ALS. This was illustrated by the observation that the SIRT2 inhibitor AGK2 did not rescue cells from the toxicity of SOD1G93A (Valle et al. 2014). Furthermore, HDAC6 deletion, but not SIRT2 deletion, attenuated motor neuron degeneration in SOD1G93A mice (Taes et al. 2013).

Studies on the Roles of Sirtuins Using in Vivo Models of ALS

Multiple pathogenic processes contribute to symptoms in ALS (Ferraiuolo et al. 2011), and measurements of motor activity and overall survival in transgenic mice overexpressing mutant SOD1 (Mancuso and Navarro 2015) are common methods to assess disease progression in vivo. An early attempt using a single-dose resveratrol treatment of SOD1G93A mice did not result in any measurable benefit in terms of motor performance and survival extension (Markert et al. 2010). However, subsequent reports were more positive with regards to a beneficial effect of resveratrol that were administered in multiple doses (see Table 1). Han et al. reported both a delay in disease onset and extended survival using a continuous dosing regimen of resveratrol via peritoneal injection into SOD1G93A mice (Han et al. 2012). The authors observed a deacetylation of heat shock factor 1 (HSF-1) and consequential upregulation of Heat shock protein (HSP) 25 and HSP70 in spinal cord lysates. Deacetylation of p53, a major SIRT1 substrate (Vaziri et al. 2001; Luo et al. 2001), served as a proxy for SIRT1 activation (Han et al. 2012). In another report, Mancuso et al. gave SOD1G93A mice a resveratrol-enriched diet and observed delayed disease onset, improved survival of spinal motor neurons, and better preserved motor neuron function (Mancuso et al. 2014). Resveratrol appeared to increase SIRT1 levels and activity as indicated by increased p53 deacetylation, as well as normalizing autophagic flux and mitochondrial function. The latter observations thus linked known SIRT1 and SIRT3 activities in autophagy (Ng and Tang 2013) and mitochondria biogenesis (Tang 2016a, b) with resveratrol treatment. Song et al. made similar observations pertaining to ALS disease onset and survival also with daily peritoneal administration of resveratrol (Song et al. 2014), and found that resveratrol increased both SIRT1 and Peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) levels in the spinal cord.

In a complementary approach, Watanabe et al. crossed a SIRT1 overexpressing transgenic line (SIRT1 under the prion precursor protein (PrP) promoter, with high CNS levels) with two SOD1G93A lines with differing mutant SOD1 protein expression levels (Watanabe et al. 2014). High CNS SIRT1 expression was able to extend the lifespan and slowed disease progression in the line with lower levels of SOD1G93A, but not the one with a higher level. For the lower SOD1G93A expressing line, the authors observed that the SIRT1 overexpression caused hypoacetylation of HSF-1 and elevated the expression of an inducible heat shock protein, HSP70i, with the latter apparently reducing the formation of insoluble SOD1 dimers (Watanabe et al. 2014). In the mouse line with higher SOD1G93A, however, further upregulation of HSP70i by SIRT1 was not observed, and the expression of this chaperone protein appeared to be already at its maximum. Therefore, in agreement with the results of Han et al. (Han et al. 2012), reduction of mutant SOD1 toxicity via the enhanced expression of chaperone proteins of the HSP family could be an important mode of SIRT1’s neurotoxicity attenuation and survival promotion mechanism.

Another link between Sirtuins and ALS was demonstrated through an important SIRT1 substrate, namely PGC-1α (Rodgers et al. 2005; Lagouge et al. 2006), which could in turn regulate SIRT3 expression (Kong et al. 2010; Giralt et al. 2011). SIRT1’s deacetylation of PGC-1α is a key aspect of metabolic regulation and mitochondrial homeostatic activity exerted by the deacetylase (Rodgers et al. 2005; Lagouge et al. 2006; Gerhart-Hines et al. 2007; Tang 2016a, b). In this regard, Pasinetti’s group has shown that overexpression of PGC-1α in a double transgenic model significantly improved motor function and survival of SOD1G93A mice (Zhao et al. 2011). The authors also showed that a ketogenic diet enriched in medium chain fatty acids, the levels of which are reciprocally influenced by Sirtuin activities (Shimazu et al. 2010), improved mitochondrial respiration and alleviated motor symptoms of SOD1G93A mice (Zhao et al. 2012a, b).

Sirtuins as Modifiers of ALS Disease Onset and Progression: Potential Mechanisms

The investigations described above, albeit largely SOD1G93A based and therefore limited in scope, do provide tantalizing evidence that Sirtuin activities, particularly those of SIRT1 and SIRT3, could potentially act in modifying ALS disease onset and progress. Earlier perceptions of ALS pathology have largely focused on two main classes of etiological impairments that are interconnected, namely (1) mitochondrial dysfunction and oxidative stress, and (2) protein aggregation and impairment in the clearance of toxic proteins or their aggregates. These lead to, or are accompanied by defects in axonal transport, neuroinflammation, and excitotoxicity that cause neuronal death and axon–muscle detachment. With the findings of the involvement of mutations in the RNA-binding proteins TDP-43 and FUS, as well as the hexanucleotide repeat expansion in C9orf72 in familial and sporadic ALS, defects in RNA metabolism move into focus as an important etiological factor. In the following sections, we discuss how Sirtuin activities may confer benefit, or work against, these etiologically important impairments for ALS.

Mitochondrial Dysfunction and Oxidative Stress

Although the pathology of ALS is complex (Rossi et al. 2016), one common and prominent pathological manifestation of the disease is mitochondrial dysfunction (Carrì et al. 2016). While oxidative stress could cause mitochondrial dysfunction, the mitochondria is itself a major source of cellular reactive oxygen species (ROS) and damaged mitochondria could produce more neuronal demise-promoting oxidative stress in many neurodegenerative diseases (Federico et al. 2012). Some proteins mutated in familial ALS (and also found in sporadic cases) are known to cause mitochondrial damage or dysfunction. The most obvious in this regard is mutant SOD1, whose association with mitochondria impairs mitochondrial dynamics and function (Magrané et al. 2009; Song et al. 2013; Tafuri et al. 2015). Mutation in a mitochondrial-localized protein, Coiled-coil-helix-coiled-coil-helix domain 10 (CHCHD10) which has an apparent role in stabilizing mitochondrial crista structure, has been recently associated with ALS/FTD (Bannwarth et al. 2014). The ER-resident Vesicle-associated membrane protein-associated protein B (VAPB), which is mutated in a familial form of ALS (Nishimura et al. 2004), functions in ER–mitochondria association by interacting with mitochondrial protein tyrosine phosphatase-interacting protein-51 (PTPIP51) to regulate calcium levels (De Vos et al. 2012). Interestingly, this interaction is disrupted by both mutant TDP-43 (Stoica et al. 2014) and FUS (Stoica et al. 2016). In general, therefore, ALS mutations and pathologies elicit an entangled web of reciprocal feedbacks between mitochondrial dysfunction and oxidative stress.

Sirtuins could counter mitochondrial dysfunction and oxidative stress in ALS, and it has been proposed to do also for AD, PD, and a number of other neurodegenerative disorders. SIRT1 and SIRT3 are known to be important regulators of mitochondrial biogenesis and function. As indicated above, SIRT1’s deacetylates and activates a key transcription factor, PGC-1α (Nemoto et al. 2005; Rodgers et al. 2005; Lagouge et al. 2006), thus regulating PGC-1α-controlled genes that in turn control metabolism and mitochondrial biogenesis (Tang 2016a, b; Brenmoehl and Hoeflich 2013; Yuan et al. 2016). PGC-1α is required for the induction of several ROS-detoxifying enzymes, and the SIRT1 activation of PGC-1α may thus also be important in countering mitochondrial oxidative stress (St-Pierre et al. 2006). SIRT3, on the other hand, appears to be an important factor downstream of PGC-1α in terms of mitochondrial function (Kong et al. 2010). A key regulator of mitochondrial metabolic enzymes (Hirschey et al. 2010), SIRT3 acts as well in mitochondria protective pathways such as the mitochondrial unfolded protein response (Papa and Germain 2014) and anti-oxidative damage responses (Someya et al. 2010). One way by which SIRT3 counters mitochondrial oxidative damage is through deacetylation of FOXO transcription factors and upregulation of genes that regulate mitochondrial homeostasis (Tseng et al. 2013). SIRT1, SIRT3, PGC-1α, and FOXO proteins may therefore constitute a transcription control network, whose levels and activities could modulate ALS mutant protein-induced mitochondrial dysfunction and oxidative damage. In this regard, it has been proposed that modulation of Sirtuin activities may influence ketogenic therapeutic approaches to improve mitochondrial function in ALS (Pasinetti et al. 2013). Collectively, these could essentially promote the survival of motor neurons in ALS.

Protein Misfolding, Aggregation Formation and Clearance Impairment

Another prominent pathological hallmark of ALS is misfolded protein accumulation and the formation of aggregates (Parakh and Atkin 2016). The presence of SOD1 mutant protein aggregates in transgenice mice, and neural as well as other human tissues, has been well documented (Watanabe et al. 2001; Jonsson et al. 2008). TDP-43 has an intrinsic propensity to aggregate, and this tendency is enhanced by ALS-associated mutations (Johnson et al. 2009; Nonaka et al. 2009). FUS is also known to form both cytoplasmic as well as nuclear aggregates (Sun et al. 2011; Nomura et al. 2014). The TDP-43/FUS family of proteins harbor prion-like domains that allow them to self-associate (Li et al. 2013) and to form functional ribonucleoprotein complexes (Kim et al. 2010), and TDP-43 mutations could promote its complex formation with FUS (Ling et al. 2010). The expanded nucleotide repeats within the C9ORF72 gene could generate abnormal dipeptide repeat proteins from repeat-associated non-AUG (RAN) translation of the repeat-containing transcripts (Cleary and Ranum 2013), and these form oligomers or aggregates (Wen et al. 2014; Porta et al. 2015; Schipper et al. 2016). The degree of ALS-associated protein misfolding and toxic oligomer formation (Fang et al. 2014) may therefore play a significant role in disease initiation. Insoluble aggregates, depending on its composition and nature, could however in some cases be neuroprotective (Cragnaz et al. 2014).

Accumulation of misfolded proteins and aggregate formation impair cellular regulation of protein biogenesis and turnover, or proteostasis, in multiple ways (Ruegsegger and Saxena 2016). For example, the aggregates could disrupt both axonal transport as well as membrane traffic within the soma, both of which would be detrimental to the motor neuron with a long axon. We focus here on two general ALS-linked proteostatic disruption mechanisms that could be helped by maintaining or elevating Sirtuin activities. Firstly, it is possible that global protein folding in the neuron is impaired by the sequestration of chaperone proteins by accumulating ALS protein aggregates. Mutant SOD1 interacts with HSP105 (Yamashita et al. 2007), and other chaperones such as HSP70 (Gifondorwa et al. 2007) and HSJ1a/DNAJB2 (Novoselov et al. 2013), confer survival benefits to SOD1G93A mouse. TDP-43 interacts with members of the HSP40/HSP70 family (Udan-Johns et al. 2014), and HSJ1a also acts to counter TDP-43 aggregation (Chen et al. 2016). FUS is known to interact with mitochondrial HSP60, which apparently aids pathogenic FUS entry into the mitochondria (Deng et al. 2015).

A second general way whereby ALS-associated protein aggregates could disrupt proteostasis is for these to impair or eventually overwhelm key cellular waste disposal mechanisms, namely the ubiquitin–proteasome pathway and macroautophagy. In fact, some ALS-associated mutations are components of these pathways. Mutations in a member of the ubiquitin family, ubiquilin-2, causes X-linked ALS (Deng et al. 2011). Protein aggregation in ALS is clearly connected to mitochondrial dysfunction and oxidative damage described in the section above, as damaged or aged mitochondria are cleared by a mitochondria-based process of autophagy, namely mitophagy (Hamacher-Brady and Brady 2016). Autophagy of damaged mitochondria requires their tagging with polyubiquitin chains via the activity of the E3 ubiquitin ligase Parkin (Durcan and Fon 2015). Mutations in optineurin, which harbors a polyubiquitin-binding domain and is an autophagy receptor for damaged mitochondria (Wong and Holzbaur 2014), causes familial ALS (Maruyama et al. 2010). In this connection, mutations in TANK-binding kinase 1 (TBK1), which enhances optineurin’s binding to ubiquitin chains to promote mitophagy of damaged mitochondria (Richter et al. 2016), also causes ALS (Freischmidt et al. 2015). Another autophagy receptor that is found to be linked to both familial and sporadic ALS is sequestosome-1 (SQSTM1)/p62 (Fecto et al. 2011), which is known to link mutant SOD1 to its clearance via autophagy (Gal et al. 2009). Autophagy-associated function has also recently been shown for C9orf72 (Yang et al. 2016; Sullivan et al. 2016; Webster et al. 2016; Sellier et al. 2016).

How could Sirtuin activities counter proteostasis impairment in ALS? Firstly, SIRT1 deacetylates an important transcriptional controller of the heat shock response, namely HSF-1 (Anckar and Sistonen 2007). HSF-1 deacetylation by SIRT1 promotes and stabilizes its binding to the promoter of HSP70 (Westerheide et al. 2009). HSF-1 activity and heat shock response are also regulated by SIRT1 activity regulators, and SIRT1-deficient mouse embryonic fibroblasts are defective in HSP70-dependent protein quality control (Tomita et al. 2015). HSF-1 augments the expression of synaptic proteins that are essential for synaptic function and stability (Hooper et al. 2016), and SIRT1-activated HSF-1 would therefore likely confer protection against neuromuscular junction failure in ALS (Fig. 1). A potential role for SIRT1 activity in modifying ALS onset and progression resulting from transgenic SOD1 mutants through HSF-1 was clearly illustrated by the benefits conferred by CNS SIRT1 overexpression (Watanabe et al. 2014). SIRT1 could of course act directly on some cytoplasmic chaperonins and enhance their modes of action, such as that documented for cytoplasmic chaperonin containing TCP-1 (CCT) (Gan and Tang 2010).

Fig. 1.

Fig. 1

A schematic diagram illustrating the potential mechanisms whereby Sirtuin activities could modify ALS disease onset and progression. SIRT1 and SIRT3 have roles in promoting autophagy and mitophagy through their deacetylation of multiple substrates, including FoXO family transcription factors. Through their interactions and influence on mitochondria biogenesis and the expression of anti-oxidative stress enzymes, these Sirtuins promote mitochondrial health and clearance of damaged mitochondria. SIRT deacetylation of HSF-1 enhances the expression of heat shock protein (HSP) family chaperones that would curb protein misfolding and aggregate formation in the soma, as well as the expression of synaptic components that could improve synaptic integrity. SIRT1’s decetylation of histone (H) and chromatin silencing effect may (speculatively) attenuate undesirable transcription of mutant C9orf72 and the generation of RAN-translated dipeptides. Taken as a whole, Sirtuin activities are likely to confer disease benefit in terms of promoting motor neuron survival, attenuating disruption of axonal transport, and preserving the integrity of the neuromuscular junction (NMJ) (cartoons in boxes). Disease-associated effects or damages are indicated by black arrows (→) while Sirtuin effects are indicated by red arrows (→) (enhancement) and (⊣) (inhibition) (Color figure online)

Secondly, Sirtuins are known modulators of autophagy (Ng and Tang 2013; Tang 2016a, b). SIRT1 regulates autophagy by directly deacetylating key autophagy proteins, including Atg5, Atg7, and Atg8 (Lee et al. 2008). It could also regulate autophagy indirectly through its deacetylation of key transcription factor substrates such as FoXO (Sengupta et al. 2009; Hariharan et al. 2010), p53 (Tasdemir et al. 2008), the RelA/p65 (Criollo et al. 2010), or via modulation of AMPK activation (Wu et al. 2011; Song et al. 2015) (Fig. 1). SIRT3 is also involved in autophagy regulation (Liang et al. 2013; Pi et al. 2015; Duan et al. 2016). Sirtuin activities have also been implicated in the induction or regulation of organelle autophagy of the mitochondria, i.e., mitophagy (Tang 2016a, b). Mitophagy is also induced by the NAD+ precursor nicotinamide, and this is likely due to SIRT1 activation (Kang and Hwang 2009; Jang et al. 2012). Silencing of mitochondrial acetyltransferase diminished mitochondrial protein acetylation levels and promoted accumulation of autophagy mediators in the mitochondria, a process that is impaired by SIRT3 depletion (Webster et al. 2013). SIRT3 deacetylates FoXO3 (Tseng et al. 2013), which leads to the upregulation of mitophagy (Das et al. 2014). Sirtuin activities’ promotion of autophagy and mitophagy could thus be potentially beneficial for ALS etiology by enhancing the clearance of protein aggregates and damaged mitochondria. On the whole, these activities should promote the survival of diseased motor neurons.

Sirtuins Influencing ALS Genes/Proteins and Disease Progression: Extending the Investigations

Despite the connections shown above and the work summarized in Table 1, it is clear that there is much more to be learned with regard to the connection between Sirtuins and ALS genes, proteins, and pathology. All the investigations reported thus far had focused on the use of the SOD1G93A mutation, and nothing is yet known about how manipulating Sirtuin levels or activities will influence ALS disease onset and progression using other ALS models. An interesting connection that has surfaced recently is a link between TDP-43 and SIRT1 (Yu et al. 2012). SIRT1 expression could apparently be influenced by a complex between TDP-43, the fragile X mental retardation protein (FMRP) and Staufen, both components of ribonucleoprotein particles that mediate transport and translation of neuronal RNAs (Barbee et al. 2006). It would be interesting and important to check if SIRT1 levels are affected by ALS-associated TDP-43 mutations, and how SIRT1 activation may in turn influence TDP-43 pathology.

Examining the effect of Sirtuin level and activity manipulations in FUS (Nolan et al. 2016)- and C9orf72 (Liu et al. 2016)-based ALS mouse models would also be pertinent. Other than the major disease-modifying mechanisms discussed above, there may be related or new avenues of Sirtuin action that may confer neuroprotective and survival extension benefits. For example, given the histone deacetylation and chromatin silencing effect of SIRT1, transcription of mutant repeats of C9orf72 may be speculatively suppressed, with consequential reduction of RNA metabolism perturbations and RAN dipeptide generation (Fig. 1). A much-debated mode of SOD1 and TDP-43 pathology is a prion-like misfolding property and its propagation (Grad et al. 2015). In this regard, it is worth noting that SIRT1 has been shown to protect against PrP-induced neuronal death (Bizat et al. 2010; Seo et al. 2012; Jeong et al. 2013). Clearly, SIRT1’s induction of chaperones and autophagy could play significant roles in mitigating pathological propagation of toxically folded proteins as well as enhancing their clearance.

A particular interesting aspect of motor neuron death in ALS pertains to the non-cell autonomous effect of glia, namely microglia and astrocytes on motor neurons (Clement et al. 2003; Ferri et al. 2004; Boillée et al. 2006). SOD1G93A-expressing astrocytes exhibit secretory toxicity towards motor neurons in co-culture (Di Giorgio et al. 2007; Nagai et al. 2007; Di Giorgio et al. 2008; Marchetto et al. 2008). Although the mechanistic nature of this non-cell autonomous effect is not yet particularly clear, the concept of glia-mediated neuronal demise via neuroinflammation is well known. In a humanized co-culture model, death of motor neurons induced by primary astrocytes from either sporadic or familial ALS was shown to occur by necroptosis (Re et al. 2014). SIRT1 is a suppressor of inflammation (Hwang et al. 2013) and the pro-inflammatory senescence-associated secretory phenotype (Hayakawa et al. 2015). Moreover, it was recently shown that enhancing the NAD+ salvage pathway, which likely enhances Sirtuin activities, could attenuate neurotoxicity of astrocytes expressing mutant SOD1 (Harlan et al. 2016). Understanding the role of Sirtuins in astrocytes and how these modulate astrocyte-based motor neuron toxicity should thus be an important future pursuit.

Sirtuin-Based Therapeutics for ALS?

Clinical trial for ALS therapeutics in the past decade has been largely negative and disappointing, and riluzole is the only approved disease-modifying pharmacological intervention thus far (Mitsumoto et al. 2014). No Sirtuin or Sirtuin activator-based ALS trials have yet been reported. Given the tolerability of resveratrol and its significant modification of CNS biomarkers reported in a recent AD trial (Turner et al. 2015), conducting such a trial for ALS would be pertinent and justified if clear beneficial effects of Sirtuin activities are eventually demonstrated in other ALS animal models. Resveratrol has multiple targets such as AMPK (Dasgupta and Milbrandt 2007) and cAMP phosphodiesterases (Park et al. 2012), and more specific natural and synthetic STACs (Hubbard and Sinclair 2014) may provide a protective effect that could be more exclusively attributed to Sirtuin activities. Another cautionary note for the use of pan-Sirtuin activators is that not all Sirtuin activities are neuroprotective. SIRT2, for example, has been negatively implicated in neuronal survival in Parkinson’s disease (Outeiro et al. 2007) and Huntington’s disease models (Pallos et al. 2008; Chopra et al. 2012). The role of SIRT2 activity, if any, in motor neuron demise is yet unclear (Taes et al. 2013), and deserve further investigation.

Acknowledgement

The author is supported by the NUS Graduate School for Integrative Sciences and Engineering, and declares no conflict of interest. The author is grateful to the anonymous reviewers, whose constructive comments and suggestions have greatly improved the manuscript.

References

  1. Anckar J, Sistonen L (2007) Heat shock factor 1 as a coordinator of stress and developmental pathways. Adv Exp Med Biol 594:78–88 [DOI] [PubMed] [Google Scholar]
  2. Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T (2006) TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 351:602–611 [DOI] [PubMed] [Google Scholar]
  3. Bannwarth S et al (2014) A mitochondrial origin for frontotemporal dementia and amyotrophic lateral sclerosis through CHCHD10 involvement. Brain 137:2329–2345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Barańczyk-Kuźma A, Usarek E, Kuźma-Kozakiewcz M, Kaźmierczak B, Gajewska B, Schwalenstocker B, Ludolph AC (2007) Age-related changes in tau expression in transgenic mouse model of amyotrophic lateral sclerosis. Neurochem Res 32:415–421 [DOI] [PubMed] [Google Scholar]
  5. Barbee SA, Estes PS, Cziko AM, Hillebrand J, Luedeman RA, Coller JM, Johnson N, Howlett IC, Geng C, Ueda R, Brand AH, Newbury SF, Wilhelm JE, Levine RB, Nakamura A, Parker R, Ramaswami M (2006) Staufen- and FMRP-containing neuronal RNPs are structurally and functionally related to somatic P bodies. Neuron 52:997–1009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bartus RT, Bétourné A, Basile A, Peterson BL, Glass J, Boulis NM (2016) β2-Adrenoceptor agonists as novel, safe and potentially effective therapies for amyotrophic lateral sclerosis (ALS). Neurobiol Dis 85:11–24 [DOI] [PubMed] [Google Scholar]
  7. Baur JA et al (2006) Resveratrol improves health and survival of mice on a high-calorie diet. Nature 444:337–342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bennion Callister J, Pickering-Brown SM (2014) Pathogenesis/genetics of frontotemporal dementia and how it relates to ALS. Exp Neurol 262:84–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bizat N, Peyrin JM, Haïk S, Cochois V, Beaudry P, Laplanche JL, Néri C (2010) Neuron dysfunction is induced by prion protein with an insertional mutation via a Fyn kinase and reversed by sirtuin activation in Caenorhabditis elegans. J Neurosci 30:5394–5403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Boillée S, Yamanaka K, Lobsiger CS, Copeland NG, Jenkins NA, Kassiotis G, Kollias G, Cleveland DW (2006) Onset and progression in inherited ALS determined by motor neurons and microglia. Science 312:1389–1392 [DOI] [PubMed] [Google Scholar]
  11. Bonkowski MS, Sinclair DA (2016) Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol [DOI] [PMC free article] [PubMed]
  12. Bosch-Presegué L, Vaquero A (2015) Sirtuin-dependent epigenetic regulation in the maintenance of genome integrity. FEBS J 282:1745–1767 [DOI] [PubMed] [Google Scholar]
  13. Brenmoehl J, Hoeflich A (2013) Dual control of mitochondrial biogenesis by sirtuin 1 and sirtuin 3. Mitochondrion 13:755–761 [DOI] [PubMed] [Google Scholar]
  14. Carrì MT, D’Ambrosi N, Cozzolino M (2016) Pathways to mitochondrial dysfunction in ALS pathogenesis. Biochem Biophys Res Commun [DOI] [PubMed]
  15. Chang HC, Guarente L (2014) SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab 25:138–145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen LC, Smith A, Ben Y, Zukic B, Ignacio S, Moore D, Lee N (2004) Temporal gene expression patterns in G93A/SOD1 mouse. Amyotroph Lateral Scler Other Motor Neuron Disord 5:164–171 [DOI] [PubMed] [Google Scholar]
  17. Chen HJ, Mitchell JC, Novoselov S, Miller J, Nishimura AL, Scotter EL, Vance CA, Cheetham ME, Shaw CE (2016) The heat shock response plays an important role in TDP-43 clearance: evidence for dysfunction in amyotrophic lateral sclerosis. Brain 139:1417–1432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chopra V, Quinti L, Kim J, Vollor L, Narayanan KL, Edgerly C, Cipicchio PM, Lauver MA, Choi SH, Silverman RB, Ferrante RJ, Hersch S, Kazantsev AG (2012) The sirtuin 2 inhibitor AK-7 is neuroprotective in Huntington’s disease mouse models. Cell Rep 2:1492–1497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cleary JD, Ranum LPW (2013) Repeat-associated non-ATG (RAN) translation in neurological disease. Hum Mol Genet 22:R45–R51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Clement AM, Nguyen MD, Roberts EA, Garcia ML, Boillée S, Rule M, McMahon AP, Doucette W, Siwek D, Ferrante RJ, Brown RH, Julien JP, Goldstein LSB, Cleveland DW (2003) Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 302:113–117 [DOI] [PubMed] [Google Scholar]
  21. Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, Howitz KT, Gorospe M, de Cabo R, Sinclair DA (2004) Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 305:390–392 [DOI] [PubMed] [Google Scholar]
  22. Cragnaz L, Klima R, Skoko N, Budini M, Feiguin F, Baralle FE (2014) Aggregate formation prevents dTDP-43 neurotoxicity in the Drosophila melanogaster eye. Neurobiol Dis 71:74–80 [DOI] [PubMed] [Google Scholar]
  23. Criollo A et al (2010) The IKK complex contributes to the induction of autophagy. EMBO J 29:619–631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Das S, Mitrovsky G, Vasanthi HR, Das DK (2014) Antiaging properties of a grape-derived antioxidant are regulated by mitochondrial balance of fusion and fission leading to mitophagy triggered by a signaling network of Sirt1-Sirt3-Foxo3-PINK1-PARKIN. Oxid Med Cell Longev 2014:345105 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  25. Dasgupta B, Milbrandt J (2007) Resveratrol stimulates AMP kinase activity in neurons. Proc Natl Acad Sci USA 104:7217–7222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. de Oliveira GP, Alves CJ, Chadi G (2013) Early gene expression changes in spinal cord from SOD1(G93A) amyotrophic lateral sclerosis animal model. Front Cell Neurosci 7:216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. De Vos KJ, Mórotz GM, Stoica R, Tudor EL, Lau KF, Ackerley S, Warley A, Shaw CE, Miller CCJ (2012) VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet 21:1299–1311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. DeJesus-Hernandez M et al (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72:245–256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Deng HX et al (2011) Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 477:211–215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Deng J, Yang M, Chen Y, Chen X, Liu J, Sun S, Cheng H, Li Y, Bigio EH, Mesulam M, Xu Q, Du S, Fushimi K, Zhu L, Wu JY (2015) FUS Interacts with HSP60 to promote mitochondrial damage. PLoS Genet 11:e1005357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Di Giorgio FP, Carrasco MA, Siao MC, Maniatis T, Eggan K (2007) Non-cell autonomous effect of glia on motor neurons in an embryonic stem cell-based ALS model. Nat Neurosci 10:608–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Di Giorgio FP, Boulting GL, Bobrowicz S, Eggan KC (2008) Human embryonic stem cell-derived motor neurons are sensitive to the toxic effect of glial cells carrying an ALS-causing mutation. Cell Stem Cell 3:637–648 [DOI] [PubMed] [Google Scholar]
  33. Duan WJ, Li YF, Liu FL, Deng J, Wu YP, Yuan WL, Tsoi B, Chen JL, Wang Q, Cai SH, Kurihara H, He RR (2016) A SIRT3/AMPK/autophagy network orchestrates the protective effects of trans-resveratrol in stressed peritoneal macrophages and RAW 264.7 macrophages. Free Radic Biol Med 95:230–242 [DOI] [PubMed] [Google Scholar]
  34. Durcan TM, Fon EA (2015) The three ‘P’s of mitophagy: PARKIN, PINK1, and post-translational modifications. Genes Dev 29:989–999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Fang YS, Tsai KJ, Chang YJ, Kao P, Woods R, Kuo PH, Wu CC, Liao JY, Chou SC, Lin V, Jin LW, Yuan HS, Cheng IH, Tu PH, Chen YR (2014) Full-length TDP-43 forms toxic amyloid oligomers that are present in frontotemporal lobar dementia-TDP patients. Nat Commun 5:4824 [DOI] [PubMed] [Google Scholar]
  36. Fecto F, Yan J, Vemula SP, Liu E, Yang Y, Chen W, Zheng JG, Shi Y, Siddique N, Arrat H, Donkervoort S, Ajroud-Driss S, Sufit RL, Heller SL, Deng HX, Siddique T (2011) SQSTM1 mutations in familial and sporadic amyotrophic lateral sclerosis. Arch Neurol 68:1440–1446 [DOI] [PubMed] [Google Scholar]
  37. Federico A, Cardaioli E, Da Pozzo P, Formichi P, Gallus GN, Radi E (2012) Mitochondria, oxidative stress and neurodegeneration. J Neurol Sci 322:254–262 [DOI] [PubMed] [Google Scholar]
  38. Feige JN, Auwerx J (2008) Transcriptional targets of sirtuins in the coordination of mammalian physiology. Curr Opin Cell Biol 20:303–309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Ferraiuolo L, Heath PR, Holden H, Kasher P, Kirby J, Shaw PJ (2007) Microarray analysis of the cellular pathways involved in the adaptation to and progression of motor neuron injury in the SOD1 G93A mouse model of familial ALS. J Neurosci 27:9201–9219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ (2011) Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol 7:616–630 [DOI] [PubMed] [Google Scholar]
  41. Ferri A, Nencini M, Casciati A, Cozzolino M, Angelini DF, Longone P, Spalloni A, Rotilio G, Carrì MT (2004) Cell death in amyotrophic lateral sclerosis: interplay between neuronal and glial cells. FASEB J 18:1261–1263 [DOI] [PubMed] [Google Scholar]
  42. Franco JG, de Moura EG, Koury JC, Trotta PA, Cordeiro A, Souza LL, Almeida NADS, Lima NDS, Pazos-Moura CC, Lisboa PC, Passos MCF (2010) Resveratrol reduces lipid peroxidation and increases sirtuin 1 expression in adult animals programmed by neonatal protein restriction. J Endocrinol 207:319–328 [DOI] [PubMed] [Google Scholar]
  43. Freischmidt A et al (2015) Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat Neurosci 18:631–636 [DOI] [PubMed] [Google Scholar]
  44. Gal J, Ström AL, Kwinter DM, Kilty R, Zhang J, Shi P, Fu W, Wooten MW, Zhu H (2009) Sequestosome 1/p62 links familial ALS mutant SOD1 to LC3 via an ubiquitin-independent mechanism. J Neurochem 111:1062–1073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Gan BQ, Tang BL (2010) Sirt1’s beneficial roles in neurodegenerative diseases—a chaperonin containing TCP-1 (CCT) connection? Aging Cell 9:924–929 [DOI] [PubMed] [Google Scholar]
  46. Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH, Mostoslavsky R, Alt FW, Wu Z, Puigserver P (2007) Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J 26:1913–1923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gertz M, Steegborn C (2016) Using mitochondrial sirtuins as drug targets: disease implications and available compounds. Cell Mol Life Sci 73:2871–2896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gifondorwa DJ, Robinson MB, Hayes CD, Taylor AR, Prevette DM, Oppenheim RW, Caress J, Milligan CE (2007) Exogenous delivery of heat shock protein 70 increases lifespan in a mouse model of amyotrophic lateral sclerosis. J Neurosci 27:13173–13180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Giralt A, Hondares E, Villena JA, Ribas F, Díaz-Delfín J, Giralt M, Iglesias R, Villarroya F (2011) Peroxisome proliferator-activated receptor-gamma coactivator-1alpha controls transcription of the Sirt3 gene, an essential component of the thermogenic brown adipocyte phenotype. J Biol Chem 286:16958–16966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Grad LI, Fernando SM, Cashman NR (2015) From molecule to molecule and cell to cell: prion-like mechanisms in amyotrophic lateral sclerosis. Neurobiol Dis 77:257–265 [DOI] [PubMed] [Google Scholar]
  51. Guarente L (2007) Sirtuins in aging and disease. Cold Spring Harb Symp Quant Biol 72:483–488 [DOI] [PubMed] [Google Scholar]
  52. Guarente L (2013) Calorie restriction and sirtuins revisited. Genes Dev 27:2072–2085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song S, Likhite S, Murtha MJ, Foust KD, Rao M, Eagle A, Kammesheidt A, Christensen A, Mendell JR, Burghes AHM, Kaspar BK (2011) Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat Biotechnol 29:824–828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Haigis MC, Sinclair DA (2010) Mammalian sirtuins: biological insights and disease relevance. Annu Rev Pathol 5:253–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hamacher-Brady A, Brady NR (2016) Mitophagy programs: mechanisms and physiological implications of mitochondrial targeting by autophagy. Cell Mol Life Sci 73:775–795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Han S, Choi JR, Soon Shin K, Kang SJ (2012) Resveratrol upregulated heat shock proteins and extended the survival of G93A-SOD1 mice. Brain Res 1483:112–117 [DOI] [PubMed] [Google Scholar]
  57. Hao CM, Haase VH (2010) Sirtuins and their relevance to the kidney. J Am Soc Nephrol 21:1620–1627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, Sadoshima J (2010) Deacetylation of FoxO by Sirt1 plays an essential role in mediating starvation-induced autophagy in cardiac myocytes. Circ Res 107:1470–1482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Harlan BA, Pehar M, Sharma DR, Beeson G, Beeson CC, Vargas MR (2016) Enhancing NAD+ salvage pathway reverts the toxicity of primary astrocytes expressing amyotrophic lateral sclerosis-linked mutant superoxide dismutase 1 (SOD1). J Biol Chem 291:10836–10846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Hayakawa T, Iwai M, Aoki S, Takimoto K, Maruyama M, Maruyama W, Motoyama N (2015) SIRT1 suppresses the senescence-associated secretory phenotype through epigenetic gene regulation. PLoS ONE 10:e0116480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Herskovits AZ, Guarente L (2013) Sirtuin deacetylases in neurodegenerative diseases of aging. Cell Res 23:746–758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hirschey MD, Shimazu T, Goetzman E, Jing E, Schwer B, Lombard DB, Grueter CA, Harris C, Biddinger S, Ilkayeva OR, Stevens RD, Li Y, Saha AK, Ruderman NB, Bain JR, Newgard CB, Farese RV, Alt FW, Kahn CR, Verdin E (2010) SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature 464:121–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hooper PL, Durham HD, Török Z, Hooper PL, Crul T, Vígh L (2016) The central role of heat shock factor 1 in synaptic fidelity and memory consolidation. Cell Stress Chaperones 21:745–753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Houtkooper RH, Pirinen E, Auwerx J (2012) Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol 13:225–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA (2003) Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425:191–196 [DOI] [PubMed] [Google Scholar]
  66. Hubbard BP, Sinclair DA (2014) Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci 35:146–154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hubbard BP et al (2013) Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science 339:1216–1219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hwang JW, Yao H, Caito S, Sundar IK, Rahman I (2013) Redox regulation of SIRT1 in inflammation and cellular senescence. Free Radic Biol Med 61:95–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Iguchi Y, Katsuno M, Ikenaka K, Ishigaki S, Sobue G (2013) Amyotrophic lateral sclerosis: an update on recent genetic insights. J Neurol 260:2917–2927 [DOI] [PubMed] [Google Scholar]
  70. Imai S, Armstrong CM, Kaeberlein M, Guarente L (2000) Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403:795–800 [DOI] [PubMed] [Google Scholar]
  71. Jaarsma D, Haasdijk ED, Grashorn JA, Hawkins R, van Duijn W, Verspaget HW, London J, Holstege JC (2000) Human Cu/Zn superoxide dismutase (SOD1) overexpression in mice causes mitochondrial vacuolization, axonal degeneration, and premature motoneuron death and accelerates motoneuron disease in mice expressing a familial amyotrophic lateral sclerosis mutant SOD1. Neurobiol Dis 7:623–643 [DOI] [PubMed] [Google Scholar]
  72. Jang SY, Kang HT, Hwang ES (2012) Nicotinamide-induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation. J Biol Chem 287:19304–19314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jeong JK, Moon MH, Lee YJ, Seol JW, Park SY (2013) Autophagy induced by the class III histone deacetylase Sirt1 prevents prion peptide neurotoxicity. Neurobiol Aging 34:146–156 [DOI] [PubMed] [Google Scholar]
  74. Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD (2009) TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral sclerosis-linked mutations accelerate aggregation and increase toxicity. J Biol Chem 284:20329–20339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Jonsson PA, Bergemalm D, Andersen PM, Gredal O, Brännström T, Marklund SL (2008) Inclusions of amyotrophic lateral sclerosis-linked superoxide dismutase in ventral horns, liver, and kidney. Ann Neurol 63:671–675 [DOI] [PubMed] [Google Scholar]
  76. Julien C, Tremblay C, Emond V, Lebbadi M, Salem N, Bennett DA, Calon F (2009) Sirtuin 1 reduction parallels the accumulation of tau in Alzheimer disease. J Neuropathol Exp Neurol 68:48–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kang HT, Hwang ES (2009) Nicotinamide enhances mitochondria quality through autophagy activation in human cells. Aging Cell 8:426–438 [DOI] [PubMed] [Google Scholar]
  78. Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, Puigserver P, Sinclair DA, Tsai LH (2007) SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J 26:3169–3179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kim SH, Shanware NP, Bowler MJ, Tibbetts RS (2010) Amyotrophic lateral sclerosis-associated proteins TDP-43 and FUS/TLS function in a common biochemical complex to co-regulate HDAC6 mRNA. J Biol Chem 285:34097–34105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Knippenberg S, Sipos J, Thau-Habermann N, Körner S, Rath KJ, Dengler R, Petri S (2013) Altered expression of DJ-1 and PINK1 in sporadic ALS and in the SOD1(G93A) ALS mouse model. J Neuropathol Exp Neurol 72:1052–1061 [DOI] [PubMed] [Google Scholar]
  81. Koistinen H, Prinjha R, Soden P, Harper A, Banner SJ, Pradat PF, Loeffler JP, Dingwall C (2006) Elevated levels of amyloid precursor protein in muscle of patients with amyotrophic lateral sclerosis and a mouse model of the disease. Muscle Nerve 34:444–450 [DOI] [PubMed] [Google Scholar]
  82. Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y (2010) Sirtuin 3, a new target of PGC-1alpha, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE 5:e11707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Körner S, Böselt S, Thau N, Rath KJ, Dengler R, Petri S (2013) Differential sirtuin expression patterns in amyotrophic lateral sclerosis (ALS) postmortem tissue: neuroprotective or neurotoxic properties of sirtuins in ALS? Neurodegener Dis 11:141–152 [DOI] [PubMed] [Google Scholar]
  84. Koronowski KB, Perez-Pinzon MA (2015) Sirt1 in cerebral ischemia. Brain Circ 1:69–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Krey L, Lühder F, Kusch K, Czech-Zechmeister B, Könnecke B, Fleming Outeiro T, Trendelenburg G (2015) Knockout of silent information regulator 2 (SIRT2) preserves neurological function after experimental stroke in mice. J Cereb Blood Flow Metab 35:2080–2088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kwiatkowski TJ et al (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323:1205–1208 [DOI] [PubMed] [Google Scholar]
  87. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J (2006) Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 127:1109–1122 [DOI] [PubMed] [Google Scholar]
  88. Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos M, Alt FW, Finkel T (2008) A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci USA 105:3374–3379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Lee JC, Shin JH, Park BW, Kim GS, Kim JC, Kang KS, Cha CI (2012) Region-specific changes in the immunoreactivity of SIRT1 expression in the central nervous system of SOD1(G93A) transgenic mice as an in vivo model of amyotrophic lateral sclerosis. Brain Res 1433:20–28 [DOI] [PubMed] [Google Scholar]
  90. Lev N, Ickowicz D, Barhum Y, Melamed E, Offen D (2009) DJ-1 changes in G93A-SOD1 transgenic mice: implications for oxidative stress in ALS. J Mol Neurosci 38:94–102 [DOI] [PubMed] [Google Scholar]
  91. Li L, Yang XJ (2015) Tubulin acetylation: responsible enzymes, biological functions and human diseases. Cell Mol Life Sci 72:4237–4255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Li YR, King OD, Shorter J, Gitler AD (2013) Stress granules as crucibles of ALS pathogenesis. J Cell Biol 201:361–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Liang Q, Benavides GA, Vassilopoulos A, Gius D, Darley-Usmar V, Zhang J (2013) Bioenergetic and autophagic control by Sirt3 in response to nutrient deprivation in mouse embryonic fibroblasts. Biochem J 454:249–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ling SC, Albuquerque CP, Han JS, Lagier-Tourenne C, Tokunaga S, Zhou H, Cleveland DW (2010) ALS-associated mutations in TDP-43 increase its stability and promote TDP-43 complexes with FUS/TLS. Proc Natl Acad Sci USA 107:13318–13323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Liu Y, Pattamatta A, Zu T, Reid T, Bardhi O, Borchelt DR, Yachnis AT, Ranum LPW (2016) C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron 90:521–534 [DOI] [PubMed] [Google Scholar]
  96. Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L, Gu W (2001) Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 107:137–148 [DOI] [PubMed] [Google Scholar]
  97. Luthi-Carter R, Taylor DM, Pallos J, Lambert E, Amore A, Parker A, Moffitt H, Smith DL, Runne H, Gokce O, Kuhn A, Xiang Z, Maxwell MM, Reeves SA, Bates GP, Neri C, Thompson LM, Marsh JL, Kazantsev AG (2010) SIRT2 inhibition achieves neuroprotection by decreasing sterol biosynthesis. Proc Natl Acad Sci USA 107:7927–7932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Lutz MI, Milenkovic I, Regelsberger G, Kovacs GG (2014) Distinct patterns of sirtuin expression during progression of Alzheimer’s disease. Neuromol Med 16:405–414 [DOI] [PubMed] [Google Scholar]
  99. Mackenzie IRA, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ, Kwong LK, Forman MS, Ravits J, Stewart H, Eisen A, McClusky L, Kretzschmar HA, Monoranu CM, Highley JR, Kirby J, Siddique T, Shaw PJ, Lee VMY, Trojanowski JQ (2007) Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol 61:427–434 [DOI] [PubMed] [Google Scholar]
  100. Magrané J, Hervias I, Henning MS, Damiano M, Kawamata H, Manfredi G (2009) Mutant SOD1 in neuronal mitochondria causes toxicity and mitochondrial dynamics abnormalities. Hum Mol Genet 18:4552–4564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Mancuso R, Navarro X (2015) Amyotrophic lateral sclerosis: Current perspectives from basic research to the clinic. Prog Neurobiol 133:1–26 [DOI] [PubMed] [Google Scholar]
  102. Mancuso R, del Valle J, Modol L, Martinez A, Granado-Serrano AB, Ramirez-Núñez O, Pallás M, Portero-Otin M, Osta R, Navarro X (2014) Resveratrol improves motoneuron function and extends survival in SOD1(G93A) ALS mice. Neurotherapeutics 11:419–432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Marchetto MCN, Muotri AR, Mu Y, Smith AM, Cezar GG, Gage FH (2008) Non-cell-autonomous effect of human SOD1 G37R astrocytes on motor neurons derived from human embryonic stem cells. Cell Stem Cell 3:649–657 [DOI] [PubMed] [Google Scholar]
  104. Markert CD, Kim E, Gifondorwa DJ, Childers MK, Milligan CE (2010) A single-dose resveratrol treatment in a mouse model of amyotrophic lateral sclerosis. J Med Food 13:1081–1085 [DOI] [PubMed] [Google Scholar]
  105. Maruyama H et al (2010) Mutations of optineurin in amyotrophic lateral sclerosis. Nature 465:223–226 [DOI] [PubMed] [Google Scholar]
  106. Matsushima S, Sadoshima J (2015) The role of sirtuins in cardiac disease. Am J Physiol Heart Circ Physiol 309:H1375–H1389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Mazzini L, Vescovi A, Cantello R, Gelati M, Vercelli A (2016) Stem cells therapy for ALS. Expert Opin Biol Ther 16:187–199 [DOI] [PubMed] [Google Scholar]
  108. Meyer K, Ferraiuolo L, Miranda CJ, Likhite S, McElroy S, Renusch S, Ditsworth D, Lagier-Tourenne C, Smith RA, Ravits J, Burghes AH, Shaw PJ, Cleveland DW, Kolb SJ, Kaspar BK (2014) Direct conversion of patient fibroblasts demonstrates non-cell autonomous toxicity of astrocytes to motor neurons in familial and sporadic ALS. Proc Natl Acad Sci USA 111:829–832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Michán S, Li Y, Chou MMH, Parrella E, Ge H, Long JM, Allard JS, Lewis K, Miller M, Xu W, Mervis RF, Chen J, Guerin KI, Smith LEH, McBurney MW, Sinclair DA, Baudry M, de Cabo R, Longo VD (2010) SIRT1 is essential for normal cognitive function and synaptic plasticity. J Neurosci 30:9695–9707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Miller RG, Mitchell JD, Moore DH (2012) Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). The Cochrane database of systematic reviews CD001447 [DOI] [PubMed]
  111. Milne JC et al (2007) Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450:712–716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Min SW, Sohn PD, Cho SH, Swanson RA, Gan L (2013) Sirtuins in neurodegenerative diseases: an update on potential mechanisms. Front Aging Neurosci 5:53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Mitsumoto H, Brooks BR, Silani V (2014) Clinical trials in amyotrophic lateral sclerosis: why so many negative trials and how can trials be improved? Lancet Neurol 13:1127–1138 [DOI] [PubMed] [Google Scholar]
  114. Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, Przedborski S (2007) Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat Neurosci 10:615–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Nemoto S, Fergusson MM, Finkel T (2005) SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem 280:16456–16460 [DOI] [PubMed] [Google Scholar]
  116. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VMY (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314:130–133 [DOI] [PubMed] [Google Scholar]
  117. Ng F, Tang BL (2013) Sirtuins’ modulation of autophagy. J Cell Physiol 228:2262–2270 [DOI] [PubMed] [Google Scholar]
  118. Ng F, Wijaya L, Tang BL (2015) SIRT1 in the brain-connections with aging-associated disorders and lifespan. Front Cell Neurosci 9:64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Nguyen MD, Larivière RC, Julien JP (2001) Deregulation of Cdk5 in a mouse model of ALS: toxicity alleviated by perikaryal neurofilament inclusions. Neuron 30:135–147 [DOI] [PubMed] [Google Scholar]
  120. Nishimura AL, Mitne-Neto M, Silva HCA, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JRM, Gillingwater T, Webb J, Skehel P, Zatz M (2004) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822–831 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Nogueiras R, Habegger KM, Chaudhary N, Finan B, Banks AS, Dietrich MO, Horvath TL, Sinclair DA, Pfluger PT, Tschöp MH (2012) Sirtuin 1 and sirtuin 3: physiological modulators of metabolism. Physiol Rev 92:1479–1514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Nolan M, Talbot K, Ansorge O (2016) Pathogenesis of FUS-associated ALS and FTD: insights from rodent models. Acta Neuropathol Commun 4:99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Nomura T, Watanabe S, Kaneko K, Yamanaka K, Nukina N, Furukawa Y (2014) Intranuclear aggregation of mutant FUS/TLS as a molecular pathomechanism of amyotrophic lateral sclerosis. J Biol Chem 289:1192–1202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Nonaka T, Kametani F, Arai T, Akiyama H, Hasegawa M (2009) Truncation and pathogenic mutations facilitate the formation of intracellular aggregates of TDP-43. Hum Mol Genet 18:3353–3364 [DOI] [PubMed] [Google Scholar]
  125. Novoselov SS, Mustill WJ, Gray AL, Dick JR, Kanuga N, Kalmar B, Greensmith L, Cheetham ME (2013) Molecular chaperone mediated late-stage neuroprotection in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. PLoS ONE 8:e73944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Outeiro TF, Kontopoulos E, Altmann SM, Kufareva I, Strathearn KE, Amore AM, Volk CB, Maxwell MM, Rochet JC, McLean PJ, Young AB, Abagyan R, Feany MB, Hyman BT, Kazantsev AG (2007) Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson’s disease. Science 317:516–519 [DOI] [PubMed] [Google Scholar]
  127. Pallàs M, Pizarro JG, Gutierrez-Cuesta J, Crespo-Biel N, Alvira D, Tajes M, Yeste-Velasco M, Folch J, Canudas AM, Sureda FX, Ferrer I, Camins A (2008) Modulation of SIRT1 expression in different neurodegenerative models and human pathologies. Neuroscience 154:1388–1397 [DOI] [PubMed] [Google Scholar]
  128. Pallàs M, Porquet D, Vicente A, Sanfeliu C (2013) Resveratrol: new avenues for a natural compound in neuroprotection. Curr Pharm Des 19:6726–6731 [DOI] [PubMed] [Google Scholar]
  129. Pallos J, Bodai L, Lukacsovich T, Purcell JM, Steffan JS, Thompson LM, Marsh JL (2008) Inhibition of specific HDACs and sirtuins suppresses pathogenesis in a Drosophila model of Huntington’s disease. Hum Mol Genet 17:3767–3775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Papa L, Germain D (2014) SirT3 regulates the mitochondrial unfolded protein response. Mol Cell Biol 34:699–710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Parakh S, Atkin JD (2016) Protein folding alterations in amyotrophic lateral sclerosis. Brain Res 1648:633–649 [DOI] [PubMed] [Google Scholar]
  132. Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, Ke H, Rehmann H, Taussig R, Brown AL, Kim MK, Beaven MA, Burgin AB, Manganiello V, Chung JH (2012) Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell 148:421–433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Pasinetti GM, Bilski AE, Zhao W (2013) Sirtuins as therapeutic targets of ALS. Cell Res 23:1073–1074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Petegnief V, Planas AM (2013) SIRT1 regulation modulates stroke outcome. Cell 4:663–671 [DOI] [PubMed] [Google Scholar]
  135. Pi H, Xu S, Reiter RJ, Guo P, Zhang L, Li Y, Li M, Cao Z, Tian L, Xie J, Zhang R, He M, Lu Y, Liu C, Duan W, Yu Z, Zhou Z (2015) SIRT3-SOD2-mROS-dependent autophagy in cadmium-induced hepatotoxicity and salvage by melatonin. Autophagy 11:1037–1051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Porta S, Kwong LK, Trojanowski JQ, Lee VMY (2015) Drosha inclusions are new components of dipeptide-repeat protein aggregates in FTLD-TDP and ALS C9orf72 expansion cases. J Neuropathol Exp Neurol 74:380–387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Poulose N, Raju R (2015) Sirtuin regulation in aging and injury. Biochim Biophys Acta 1852:2442–2455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Rabinovich-Toidman P, Rabinovich-Nikitin I, Ezra A, Barbiro B, Fogel H, Slutsky I, Solomon B (2015) Mutant SOD1 increases APP expression and phosphorylation in cellular and animal models of ALS. PLoS ONE 10:e0143420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Re DB, Le Verche V, Yu C, Amoroso MW, Politi KA, Phani S, Ikiz B, Hoffmann L, Koolen M, Nagata T, Papadimitriou D, Nagy P, Mitsumoto H, Kariya S, Wichterle H, Henderson CE, Przedborski S (2014) Necroptosis drives motor neuron death in models of both sporadic and familial ALS. Neuron 81:1001–1008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Renton AE et al (2011) A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72:257–268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Renton AE, Chiò A, Traynor BJ (2014) State of play in amyotrophic lateral sclerosis genetics. Nat Neurosci 17:17–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Richter B, Sliter DA, Herhaus L, Stolz A, Wang C, Beli P, Zaffagnini G, Wild P, Martens S, Wagner SA, Youle RJ, Dikic I (2016) Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci USA 113:4039–4044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Riva N, Agosta F, Lunetta C, Filippi M, Quattrini A (2016) Recent advances in amyotrophic lateral sclerosis. J Neurol 263:1241–1254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P (2005) Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 434:113–118 [DOI] [PubMed] [Google Scholar]
  145. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O’Regan JP, Deng HX (1993) Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362:59–62 [DOI] [PubMed] [Google Scholar]
  146. Rossi S, Cozzolino M, Carrì MT (2016) Old versus new mechanisms in the pathogenesis of ALS. Brain Pathol 26:276–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Ruegsegger C, Saxena S (2016) Proteostasis impairment in ALS. Brain Res 1648:571–579 [DOI] [PubMed] [Google Scholar]
  148. Schipper LJ, Raaphorst J, Aronica E, Baas F, de Haan R, de Visser M, Troost D (2016) Prevalence of brain and spinal cord inclusions, including dipeptide repeat proteins, in patients with the C9ORF72 hexanucleotide repeat expansion: a systematic neuropathological review. Neuropathol Appl Neurobiol 42:547–560 [DOI] [PubMed] [Google Scholar]
  149. Schirmer H, Pereira TCB, Rico EP, Rosemberg DB, Bonan CD, Bogo MR, Souto AA (2012) Modulatory effect of resveratrol on SIRT1, SIRT3, SIRT4, PGC1α and NAMPT gene expression profiles in wild-type adult zebrafish liver. Mol Biol Rep 39:3281–3289 [DOI] [PubMed] [Google Scholar]
  150. Sellier C, Campanari ML, Julie Corbier C, Gaucherot A, Kolb-Cheynel I, Oulad-Abdelghani M, Ruffenach F, Page A, Ciura S, Kabashi E, Charlet-Berguerand N (2016) Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J 35:1276–1297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Sengupta A, Molkentin JD, Yutzey KE (2009) FoxO transcription factors promote autophagy in cardiomyocytes. J Biol Chem 284:28319–28331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Seo JS, Moon MH, Jeong JK, Seol JW, Lee YJ, Park BH, Park SY (2012) SIRT1, a histone deacetylase, regulates prion protein-induced neuronal cell death. Neurobiol Aging 33:1110–1120 [DOI] [PubMed] [Google Scholar]
  153. Shimazu T, Hirschey MD, Hua L, Dittenhafer-Reed KE, Schwer B, Lombard DB, Li Y, Bunkenborg J, Alt FW, Denu JM, Jacobson MP, Verdin E (2010) SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metab 12:654–661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Someya S, Yu W, Hallows WC, Xu J, Vann JM, Leeuwenburgh C, Tanokura M, Denu JM, Prolla TA (2010) Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 143:802–812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Song W, Song Y, Kincaid B, Bossy B, Bossy-Wetzel E (2013) Mutant SOD1G93A triggers mitochondrial fragmentation in spinal cord motor neurons: neuroprotection by SIRT3 and PGC-1α. Neurobiol Dis 51:72–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Song L, Chen L, Zhang X, Li J, Le W (2014) Resveratrol ameliorates motor neuron degeneration and improves survival in SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Biomed Res Int 2014:483501 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  157. Song YM, Lee YH, Kim JW, Ham DS, Kang ES, Cha BS, Lee HC, Lee BW (2015) Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy 11:46–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Srivastava S, Haigis MC (2011) Role of sirtuins and calorie restriction in neuroprotection: implications in Alzheimer’s and Parkinson’s diseases. Curr Pharm Des 17:3418–3433 [DOI] [PubMed] [Google Scholar]
  159. Stoica L, Sena-Esteves M (2016) Adeno associated viral vector delivered RNAi for gene therapy of SOD1 amyotrophic lateral sclerosis. Front Mol Neurosci 9:56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau KF, Vizcay-Barrena G, Lin WL, Xu YF, Lewis J, Dickson DW, Petrucelli L, Mitchell JC, Shaw CE, Miller CCJ (2014) ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun 5:3996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Stoica R, Paillusson S, Gomez-Suaga P, Mitchell JC, Lau DH, Gray EH, Sancho RM, Vizcay-Barrena G, De Vos KJ, Shaw CE, Hanger DP, Noble W, Miller CC (2016) ALS/FTD-associated FUS activates GSK-3β to disrupt the VAPB-PTPIP51 interaction and ER-mitochondria associations. EMBO Rep 17:1326–1342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jäger S, Handschin C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, Spiegelman BM (2006) Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127:397–408 [DOI] [PubMed] [Google Scholar]
  163. Sullivan PM, Zhou X, Robins AM, Paushter DH, Kim D, Smolka MB, Hu F (2016) The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol Commun 4:51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Sun Z, Diaz Z, Fang X, Hart MP, Chesi A, Shorter J, Gitler AD (2011) Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS. PLoS Biol 9:e1000614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Taes I, Timmers M, Hersmus N, Bento-Abreu A, Van Den Bosch L, Van Damme P, Auwerx J, Robberecht W (2013) Hdac6 deletion delays disease progression in the SOD1G93A mouse model of ALS. Hum Mol Genet 22:1783–1790 [DOI] [PubMed] [Google Scholar]
  166. Tafuri F, Ronchi D, Magri F, Comi GP, Corti S (2015) SOD1 misplacing and mitochondrial dysfunction in amyotrophic lateral sclerosis pathogenesis. Front Cell Neurosci 9:336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Tang BL (2011) Sirt1’s systemic protective roles and its promise as a target in antiaging medicine. Transl Res 157:276–284 [DOI] [PubMed] [Google Scholar]
  168. Tang BL (2016a) Sirt1 and the mitochondria. Mol Cells 39:87–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Tang BL (2016) Sirtuins as modifiers of Parkinson’s disease pathology. J Neurosci Res [DOI] [PubMed]
  170. Tasdemir E et al (2008) Regulation of autophagy by cytoplasmic p53. Nat Cell Biol 10:676–687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Tomita T, Hamazaki J, Hirayama S, McBurney MW, Yashiroda H, Murata S (2015) Sirt1-deficiency causes defective protein quality control. Sci Rep 5:12613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Tseng AHH, Shieh SS, Wang DL (2013) SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative damage. Free Radic Biol Med 63:222–234 [DOI] [PubMed] [Google Scholar]
  173. Turner MR, Swash M (2015) The expanding syndrome of amyotrophic lateral sclerosis: a clinical and molecular odyssey. J Neurol Neurosurg Psychiatry 86:667–673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Turner RS, Thomas RG, Craft S, van Dyck CH, Mintzer J, Reynolds BA, Brewer JB, Rissman RA, Raman R, Aisen PS, Alzheimer’s Disease Cooperative Study (2015) A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 85:1383–1391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Udan-Johns M, Bengoechea R, Bell S, Shao J, Diamond MI, True HL, Weihl CC, Baloh RH (2014) Prion-like nuclear aggregation of TDP-43 during heat shock is regulated by HSP40/70 chaperones. Hum Mol Genet 23:157–170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Valle C, Salvatori I, Gerbino V, Rossi S, Palamiuc L, René F, Carrì MT (2014) Tissue-specific deregulation of selected HDACs characterizes ALS progression in mouse models: pharmacological characterization of SIRT1 and SIRT2 pathways. Cell Death Dis 5:e1296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Vance C et al (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323:1208–1211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, Guarente L, Weinberg RA (2001) hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 107:149–159 [DOI] [PubMed] [Google Scholar]
  179. Wang J, Zhang Y, Tang L, Zhang N, Fan D (2011) Protective effects of resveratrol through the up-regulation of SIRT1 expression in the mutant hSOD1-G93A-bearing motor neuron-like cell culture model of amyotrophic lateral sclerosis. Neurosci Lett 503:250–255 [DOI] [PubMed] [Google Scholar]
  180. Watanabe M, Dykes-Hoberg M, Culotta VC, Price DL, Wong PC, Rothstein JD (2001) Histological evidence of protein aggregation in mutant SOD1 transgenic mice and in amyotrophic lateral sclerosis neural tissues. Neurobiol Dis 8:933–941 [DOI] [PubMed] [Google Scholar]
  181. Watanabe S, Ageta-Ishihara N, Nagatsu S, Takao K, Komine O, Endo F, Miyakawa T, Misawa H, Takahashi R, Kinoshita M, Yamanaka K (2014) SIRT1 overexpression ameliorates a mouse model of SOD1-linked amyotrophic lateral sclerosis via HSF1/HSP70i chaperone system. Mol Brain 7:62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Webster BR, Scott I, Han K, Li JH, Lu Z, Stevens MV, Malide D, Chen Y, Samsel L, Connelly PS, Daniels MP, McCoy JP, Combs CA, Gucek M, Sack MN (2013) Restricted mitochondrial protein acetylation initiates mitochondrial autophagy. J Cell Sci 126:4843–4849 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Webster CP, Smith EF, Bauer CS, Moller A, Hautbergue GM, Ferraiuolo L, Myszczynska MA, Higginbottom A, Walsh MJ, Whitworth AJ, Kaspar BK, Meyer K, Shaw PJ, Grierson AJ, De Vos KJ (2016) The C9orf72 protein interacts with Rab1a and the ULK1 complex to regulate initiation of autophagy. EMBO J 35:1656–1676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Wen X, Tan W, Westergard T, Krishnamurthy K, Markandaiah SS, Shi Y, Lin S, Shneider NA, Monaghan J, Pandey UB, Pasinelli P, Ichida JK, Trotti D (2014) Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84:1213–1225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Westerheide SD, Anckar J, Stevens SM, Sistonen L, Morimoto RI (2009) Stress-inducible regulation of heat shock factor 1 by the deacetylase SIRT1. Science 323:1063–1066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Wong Y, Holzbaur E (2014) Optineurin is an autophagy receptor for damaged mitochondria in parkin-mediated mitophagy that is disrupted by an ALS-linked mutation. Proc Natl Acad Sci USA 111:E4439–E4448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Wong SY, Tang BL (2016) SIRT1 as a therapeutic target for Alzheimer’s disease. Rev Neurosci [DOI] [PubMed]
  188. Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, Tatar M, Sinclair D (2004) Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430:686–689 [DOI] [PubMed] [Google Scholar]
  189. Wu Y, Li X, Zhu JX, Xie W, Le W, Fan Z, Jankovic J, Pan T (2011) Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson’s disease. Neurosignals 19:163–174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW (2008) Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat Neurosci 11:251–253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Yamashita H, Kawamata J, Okawa K, Kanki R, Nakamizo T, Hatayama T, Yamanaka K, Takahashi R, Shimohama S (2007) Heat-shock protein 105 interacts with and suppresses aggregation of mutant Cu/Zn superoxide dismutase: clues to a possible strategy for treating ALS. J Neurochem 102:1497–1505 [DOI] [PubMed] [Google Scholar]
  192. Yang M, Liang C, Swaminathan K, Herrlinger S, Lai F, Shiekhattar R, Chen JF (2016) A C9ORF72/SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. Sci Adv 2:e1601167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Yu Z, Fan D, Gui B, Shi L, Xuan C, Shan L, Wang Q, Shang Y, Wang Y (2012) Neurodegeneration-associated TDP-43 interacts with fragile X mental retardation protein (FMRP)/Staufen (STAU1) and regulates SIRT1 expression in neuronal cells. J Biol Chem 287:22560–22572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Yuan Y, Cruzat VF, Newsholme P, Cheng J, Chen Y, Lu Y (2016) Regulation of SIRT1 in aging: roles in mitochondrial function and biogenesis. Mech Ageing Dev 155:10–21 [DOI] [PubMed] [Google Scholar]
  195. Zakhary SM, Ayubcha D, Dileo JN, Jose R, Leheste JR, Horowitz JM, Torres G (2010) Distribution analysis of deacetylase SIRT1 in rodent and human nervous systems. Anat Rec (Hoboken) 293:1024–1032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, Pagani W, Lodin D, Orozco G, Chinea A (2015) A comprehensive review of amyotrophic lateral sclerosis. Cell 6:171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Zhao W, Varghese M, Yemul S, Pan Y, Cheng A, Marano P, Hassan S, Vempati P, Chen F, Qian X, Pasinetti GM (2011) Peroxisome proliferator activator receptor gamma coactivator-1alpha (PGC-1α) improves motor performance and survival in a mouse model of amyotrophic lateral sclerosis. Mol Neurodegener 6:51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Zhao W, Varghese M, Vempati P, Dzhun A, Cheng A, Wang J, Lange D, Bilski A, Faravelli I, Pasinetti GM (2012a) Caprylic triglyceride as a novel therapeutic approach to effectively improve the performance and attenuate the symptoms due to the motor neuron loss in ALS disease. PLoS ONE 7:e49191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Zhao Y, Luo P, Guo Q, Li S, Zhang L, Zhao M, Xu H, Yang Y, Poon W, Fei Z (2012b) Interactions between SIRT1 and MAPK/ERK regulate neuronal apoptosis induced by traumatic brain injury in vitro and in vivo. Exp Neurol 237:489–498 [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Neurobiology are provided here courtesy of Springer

RESOURCES