You are on page 1of 7

Review

SIRT3 regulates progression and


development of diseases of aging
Eoin McDonnell1*, Brett S. Peterson1*, Howard M. Bomze1, and
Matthew D. Hirschey1,2,3
1
Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701, USA
2
Department of Medicine, Duke University, Durham, NC 27710, USA
3
Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA

The mitochondrial sirtuin SIRT3 is a protein deacylase mitochondrial deacetylase [5] that is enriched in highly
that influences almost every major aspect of mitochon- metabolic tissues, such as the liver, heart, brain, and
drial biology, including nutrient oxidation, ATP genera- brown adipose tissue. SIRT3 has been reported to regulate
tion, reactive oxygen species (ROS) detoxification, almost every major aspect of mitochondrial biology, includ-
mitochondrial dynamics, and the mitochondrial unfold- ing ROS detoxification, ATP generation, mitochondrial
ed protein response (UPR). Interestingly, mice lacking dynamics, nutrient oxidation, and the mitochondrial
SIRT3 (SIRT3KO), either spontaneously or when crossed UPR [611]. SIRT3 regulates these processes by removing
with mouse models of disease, develop several diseases acetyl modifications from a growing list of mitochondrial
of aging at an accelerated pace, such as cancer, meta- proteins [5,12]. It has also been reported to remove other
bolic syndrome, cardiovascular disease, and neurode- acyl modifications, such as long-chain fatty acyl modifica-
generative diseases, and, thus, might be a valuable tions and histone crotonylation [13,14], although under-
model of accelerated aging. In this review, we discuss standing the biological relevance of these modifications
functions of SIRT3 in pathways involved in diseases of requires further work.
aging and how the lack of SIRT3 might accelerate the A major challenge in the field is to disentangle the direct
aging process. We also suggest that further studies on effects of SIRT3 from the indirect changes seen after SIRT3
SIRT3 will help uncover important new pathways driving manipulation and, ultimately, to identify SIRT3 targets of
the aging process. deacetylation. Interestingly, an emerging sense in the field
is that no single protein target can fully explain the bio-
The role of sirtuins in aging and longevity logical and physiological effects of SIRT3. For example,
Aging is a complex process, marked by numerous cellular SIRT3 is unlikely to regulate fatty acid oxidation (FAO)
changes throughout the lifespan of an organism. A major through deacetylation of a single enzyme [9]; instead, it
goal in the study of aging is to identify the genetic deter- likely imparts control over FAO at multiple enzymes in the
minants that regulate the aging process, influence the pathway [1517]. More work is needed to identify the
progression of the diseases of aging, and ultimately govern primary targets and activities of SIRT3, and to understand
longevity. The sirtuins are a conserved class of protein the mechanism(s) by which SIRT3 regulates proteins and
deacylases that were named after the founding member pathways, over a wide array of biology.
SIR2 (Sir2-ins) [1], and were among the first genes identi- Despite these challenges, one theme that emerges from
fied to extend lifespan in Saccharomyces cerevisiae its continued study is that SIRT3 attenuation or ablation is
[1,2]. An additional SIR2 allele can increase lifespan by associated with accelerated development of several dis-
30%, whereas ablation of SIR2 decreases lifespan in yeast eases of aging. Indeed, SIRT3KO mice alone or when
[2]. Consistent with this finding, the increased longevity crossed with mouse models of disease, develop nearly every
that is seen with calorie restriction in yeast requires SIR2 disease of aging at an accelerated pace (Figure 1). Thus, the
activation [3]. These early studies set the stage for a SIRT3KO mouse might be considered a model of acceler-
nascent field to investigate the role of sirtuins in longevity ated aging. Below, we review the roles for SIRT3 in cancer,
and the biology of aging. the metabolic syndrome, cardiovascular disease, and
SIR2 has seven mammalian orthologs (Sirtuin 17, aka neurodegenerative diseases (Table 1), and discuss what
SIRT17), three of which (SIRT3, 4, and 5) are localized to the study of SIRT3 might reveal about the aging process.
the mitochondria. Given the importance of mitochondria in
aging [4], the mitochondrial sirtuins are uniquely poised to SIRT3 and diseases of aging
regulate several aspects of the aging process. SIRT3 is a SIRT3 and cancer
Cancer is a major disease of aging, where almost 90% of all
Corresponding author: Hirschey, M.D. (matthew.hirschey@duke.edu).
cancer deaths occur in humans over 50 years of age
Keywords: mitochondria; sirtuins; SIRT3; aging; disease.
*
These authors contributed equally to this work. [18]. Cancer is thought to arise from an accumulation of
1043-2760/
mutations that occur slowly over time; as such, cancer
2015 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tem.2015.06.001 incidence increases dramatically with age. Further sup-
porting this idea, individual tumor biopsies show multiple
486 Trends in Endocrinology and Metabolism, September 2015, Vol. 26, No. 9
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

SIRT3 regulates pathways in the diseases of aging cancers, including breast, liver, and gastric cancers [22,26
28], and low SIRT3 expression is associated with worse
overall survival in hepatocellular carcinoma [29]. These
studies collectively show the tumor suppressing potential
Neurodegenerave disease of SIRT3.
ROS detoxicaon Studies of the mechanisms linking SIRT3 ablation and
accelerated cancer development have found increased oxi-
Cancer
dative stress and changes in cellular metabolism, both
ROS detoxicaon likely contributing to tumor initiation and/or progression
Glucose metabolism [30]. Increased oxidative stress can promote genome insta-
bility and DNA damage, which may lead to increased
Cardiovascular disease
mutation frequency [31]. ROS are also important signaling
Fay acid oxidaon molecules that, when present at low levels, can alter gene
ROS detoxicaon transcription and promote cell proliferation [32]. Given
that mitochondria are the major source of ROS, SIRT3
Mitochondrial dynamics
may have a critical role in preventing tumor development
Metabolic syndrome by controlling ROS levels. Indeed, superoxide dismutase 2
Fay acid oxidaon
(SOD2) is a target of SIRT3 and SIRT3KO MEFs have
elevated levels of ROS [22,33]. In support of this model,
Glucose metabolism
overexpression of SOD2 (a mitochondrial enzyme that
ROS detoxicaon detoxifies superoxide) in SIRT3KO MEFs slowed their
Oxidave phosphorylaon
growth and prevented their transformation in the presence
of either Myc or Ras, indicating that mitochondrial ROS
were required for the transformative potential of SIRT3KO
cells [22]. SIRT3KO MEFs also had increased genomic
instability when challenged with a genotoxic stress that
was reduced by the overexpression of SOD2 [22]. Another
study found that stable knockdown of SIRT3 in colon
cancer cells led to larger tumor formation in mouse xeno-
graft models, which could be prevented by the addition of
the ROS scavenger N-acetyl cysteine (NAC) [34].
While the role of SIRT3 in controlling oxidative stress
TRENDS in Endocrinology & Metabolism has been well described, SIRT3 may also affect metabolic
reprogramming in tumor cells, an emerging hallmark of
Figure 1. Sirtuin 3 (SIRT3) regulates pathways in the diseases of aging. Loss of
SIRT3 leads to deregulation of several mitochondrial pathways, which contributes cancer [35]. Shifts in metabolism towards increased aero-
to the accelerated development of the disease of aging. See the main text for bic glycolysis have been well documented in cancer cells
details. Abbreviation: ROS, reactive oxygen species.
since Otto Warburg first reported that tumors metabolize
more glucose through glycolysis and produce more lactate
genetic lesions, indicative of the clonal evolution of cancer even in the presence of oxygen [36]. While Warburgs initial
cells during tumorigenesis [19]. Environmental or genetic hypothesis that mitochondrial defects cause cancer in only
factors that increase the frequency of mutations or support a few isolated settings [e.g., fumarate hydratase, succinate
cellular proliferation can increase the risk for, and devel- dehydrogenase, and Isocitrate dehydrogenase 2 (IDH2)
opment of, cancer. For example, a rise in oxidative stress mutations], altered mitochondrial metabolism seems to
can increase damage to DNA, leading to genomic instabili- be a common characteristic of all cancers. Many cancer
ty and increased mutagenesis, and, therefore, the risk of cells shift their metabolism towards anabolic processes,
oncogenic mutations [20]. Similarly, shifts in metabolism with reduced emphasis on ATP generation and increased
that promote anabolic growth may also support the growth generation of biosynthetic intermediates required for
of a tumor [21]. proliferation [21].
The role of SIRT3 in cancer was first shown by the Cancer cell lines with SIRT3 knockdown, and MEFs
discovery that SIRT3KO mouse embryonic fibroblasts from SIRT3KO mice have a shift in metabolism toward a
(MEFs) could be transformed into cancer cells by the Warburg-like phenotype [26], and have increased glucose
addition of a single oncogene (Myc or Ras) [22]. This single uptake and glycolysis, increased levels of biosynthetic
oncogene transformability in the setting of a gene knockout intermediates, increased ROS production, and proliferate
is characteristic of a tumor suppressor gene, because wild faster. Multiple studies have shown that SIRT3 ablation
type primary MEFs require the expression of an oncogene leads to increased hypoxia inducible factor 1 alpha (HIF1a)
in combination with either another oncogene (e.g., Myc or protein stabilization and a concomitant increase in mRNA
Ras) or the loss of a tumor suppressor gene (e.g., p53) to be levels of numerous HIF1a target genes, including genes
transformed [2325]. Seven out of 20 SIRT3KO mice spon- involved in glucose uptake and glycolysis [26,34]. These
taneously developed mammary tumors after 2 years of age, shifts in metabolism are due, at least in part, to the
compared with none of the wild-type mice [22]. Further- increase in ROS, which is known to lead to increased
more, SIRT3 expression is decreased in several human HIF1a protein levels by inhibiting prolyl hydroxlases
487
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

Table 1. SIRT3 regulates pathways involved in the diseases of aging


Disease Age-related pathway SIRT3KO molecular phenotype (Patho)physiology Refs
Cancer ROS detoxification " ROS; " oxidative stress; " Tumorigenesis; [22,2629,34,39,40,42]
" genomic instability " cellular transformation;
Glucose metabolism " Glucose uptake; " glycolysis, # SIRT3 expression in
" biosynthetic intermediates several human cancers
(" Warburg effect)
Metabolic disease Oxidative phosphorylation # Activity of oxidative " IR; " nonalcoholic [7,9,10,5153,55,60]
and/or IR phosphorylation machinery hepatosteatosis;
Glucose metabolism # Glucose uptake; # glucose oxidation # basal metabolic rate;
Fatty acid oxidation # Decreased fat metabolism # glucose-stimulated
insulin secretion
ROS detoxification " Oxidative stress
Cardiovascular Fatty acid oxidation # ATP production Cardiac hypertrophy [79,71,72]
disease ROS detoxification " ROS in cardiomyocytes
Mitochondrial dynamics " Mitochondrial permeability;
# mitochondrial fusion
Neurodegenerative ROS detoxification " Oxidative damage; Hearing loss [78,81,82]
disease # glutathione levels; elevated
ROS leads to increased
SIRT3 expression

(PHDs) that target HIF1a for degradation [37,38]. Addition Accumulating evidence shows that mitochondrial dys-
of NAC or knockdown of HIF1a prevented this shift in function has a role in the metabolic syndrome and diabetes
metabolism in cells lacking SIRT3 [26]. [44]. For example, dysregulation of several mitochondrial
However, shifts in cancer cell metabolism in the absence pathways has been implicated in the pathogenesis of met-
of SIRT3 cannot be explained by ROS alone. Several abolic disease, including impaired FAO [45], oxidative
studies have found that pyruvate dehydrogenase (PDH) damage from ROS [46], inflammation [47], and loss of
is hyperacetylated and inhibited in the absence of SIRT3, metabolic flexibility [44,48]. Interestingly, the decline in
leading to alterations in pyruvate metabolism [39,40]. mitochondrial function during aging is concomitant with
PDH is inhibited in many cancers by phosphorylation by the development of hyperglycemia and hyperinsulinemia
pyruvate dehydrogenase kinases (PDKs), leading to de- [49], suggesting a mechanistic link between mitochondria
creased flux of glucose-derived carbon into the tricarboxylic and age-induced metabolic dysfunction.
acid (TCA) cycle and increased lactate production The first indication that SIRT3 had a role in fat metab-
[41]. Acetylation of PDH in the absence of SIRT3 also olism was the discovery that SIRT3KO mouse livers accu-
inhibits its activity, supports increased glycolysis, and, mulate lipid and acylcarnitine species and have impaired
thus, a Warburg-like shift in metabolism [39,40]. Another FAO [9]. Further studies demonstrated that hepatic SIRT3
recent study showed that the mitochondrial enzyme glu- expression and activity decrease in response to chronic
tamate oxaloacetate aminotransferase (GOT2) is deacety- high-fat diet (HFD) feeding, resulting in protein hypera-
lated by SIRT3; increased GOT2 acetylation promoted the cetylation, an impaired ability to protect against lipotoxic
growth of pancreatic tumor xenografts [42]. These exam- conditions, and hepatic steatosis [5052]. SIRT3KO mice
ples represent a growing list of metabolic pathways that fed a HFD showed accelerated development of several
are dysregulated upon loss of SIRT3, leading to increased hallmarks of the metabolic syndrome, including weight
tumor growth. gain, impaired glucose tolerance, and insulin resistance
Overall, loss of SIRT3 accelerates tumorigenesis in (IR) [51]. Thus, SIRT3 has a critical role in several key
mouse models and decreased SIRT3 expression is associ- pathways involved in whole-body energy homeostasis.
ated with several human cancers. Loss of SIRT3 leads to SIRT3KO mice display peripheral insulin resistance
the acetylation and dysregulation of several mitochondrial due, in part, to decreased PDH activity [10] and hexokinase
proteins, which can cause increased oxidative stress and 2 (HK2) activity in skeletal muscle [53]. Together, these
shifts in cellular metabolism, which are conditions that impairments lead to increased FAO and reduced glucose
favor accelerated tumorigenesis. uptake [10,53]. Furthermore, ROS is elevated in skeletal
muscle of SIRT3KO mice, which is implicated in the devel-
SIRT3 and metabolic disease opment of IR in metabolically active tissues, including the
Aging is recognized as a major risk factor for the develop- skeletal muscle, liver, and white adipose tissue [54
ment of diabetes [43]. The pervasiveness of diabetes and its 58]. Consistent with this idea, mice with heterozygous
related etiologies, including obesity, nonalcoholic fatty SOD2 deletion display impaired glucose tolerance on a
liver disease (NAFLD), and the metabolic syndrome, are low-fat diet (LFD), and exacerbated b cell dysfunction in
on the rise. Recent estimates report that 8.3% of the global the setting of a HFD regimen [54,59].
population (382 million people) currently have diabetes b cell failure is a critical event in the progression from
and this is expected to reach 10.1% of the global population insulin resistance to type 2 diabetes mellitus (T2DM).
(592 million people) by 2035 [43]. Interestingly, the current Interestingly, pancreatic islets from human diabetic
prevalence of diabetes is increased in people aged between donors have lower SIRT3 levels [60]. Whole-body
60 and 79 (18.6% or 134.6 million people) [43], suggesting a SIRT3KO mice fed a chronic HFD secreted less insulin
link in the development of metabolic disease and aging. in response to glucose, suggesting diet-induced pancreatic
488
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

b cell dysfunction in the absence of SIRT3, and supporting and an impairment in mitochondrial fusion [8]. Stressors
a model whereby SIRT3 serves to protect b cells from associated with heart failure, including cardiac hypertro-
nutritional stress [51]. Not surprisingly, transient knock- phy and obesity, also result in hyperacetylation of OPA1
down of SIRT3 in b cells in culture resulted in elevated [8].
ROS and impaired insulin secretion [60]. Furthermore, Taken together, these studies demonstrate that SIRT3
adenoviral overexpression of SIRT3 in primary rat islets regulates several different mitochondrial pathways that
rescued lipotoxic impairment glucose-stimulated insulin are critical to maintaining cardiac health. In the absence of
secretion [61]. Thus, SIRT3 is emerging as a critical player SIRT3, these pathways are dysregulated, which contrib-
in pancreatic b cell survival and/or compensation in the utes to cardiac hypertrophy and accelerated heart failure.
presence of a hyperglycemichyperlipidemic insult, similar
to its role in protecting against hepatic lipotoxicity. Taken SIRT3 and neurodegenerative disease
together, these results show that SIRT3 has a crucial role The onset of neurodegenerative diseases is a hallmark of
in whole-body metabolism, and its absence leads to accel- aging, and susceptibility to Alzheimers disease (AD), Par-
erated metabolic deregulation and development of the kinsons disease, Huntingtons disease, amyotrophic later-
metabolic syndrome. al sclerosis (ALS), and frontal temporal dementia increases
as humans age. Neurons are one of the highest energy-
SIRT3 and cardiac function consuming cell types, which make them particularly sus-
As humans age, cardiac function decreases, and under- ceptible to metabolic and oxidative stressors. Indeed, loss
standing the underlying molecular mechanisms driving of mitochondrial function has a major role in neurodegen-
this dysfunction is crucial for the development of future eration [73].
therapies. Mitochondria in the heart are highly oxidative, SIRT3 is expressed at high levels in the brain and other
and this capacity decreases with age in humans [62,63]. In nervous system tissues [7476]. Given that SIRT3 regu-
mice, decreased mitochondrial function is thought to con- lates several aspects of metabolic homeostasis, it is pre-
tribute to a decline in heart health and to accelerated dicted help protect against neurodegenerative diseases.
cardiac hypertrophy. The mechanisms implicated in cardi- SIRT3 can rescue neuronal loss in various neurodegenera-
ac hypertrophy and subsequent progression to heart fail- tive models. For example, primary cortical neurons grown
ure include impaired FAO, reduced oxidative in culture can be induced to undergo apoptosis in a manner
phosphorylation, increased ROS production, and dysregu- similar to AD after treatment with beta-amyloid (Ab)
lated mitochondrial dynamics [6365]. protein. In this model, co-treatment with pituitary adeny-
Not surprisingly, SIRT3 is proposed to have an impor- late cyclase-activating polypeptide (PACAP) protected pri-
tant role in regulating cardiac function. Mice lacking mary cultured neurons from Ab treatment by upregulating
SIRT3 have altered mitochondrial FAO in the heart SIRT3 expression [77]. The PACAP-mediated neuropro-
[9,66], and reduced oxidative phosphorylation (OXPHOS) tective effect was lost if SIRT3 expression was knocked
complex activity and ATP production [7]. Given that the down by short hairpin (sh)RNA, in primary cultured neu-
heart relies on FAO for most of its energy production, rons. Furthermore, SIRT3 overexpression conferred resis-
several mouse models of deficient lipid oxidation develop tance to oxidative stress and extended neuronal longevity
cardiac hypertrophy, including mice deficient in SIRT3, in primary neuron cultures [78].
AMP-activated protein kinase (AMPK) [67], peroxisome Similarly, ALS is associated with mitochondrial dys-
proliferator-activated receptor alpha (PPARa) [68], long- function [79], which can be modeled in cultured neurons by
chain acyl-CoA dehydrogenase (LCAD) [69], and oxidative expressing the G93A mutant of SOD1 [80]. Mutant SOD1
phosphorylation [70]. induces mitochondrial dysfunction and apoptosis, which
Consistent with these findings, increased ROS and can be reduced by overexpression of either SIRT3 or
subsequent oxidative damage can independently lead to PPARg coactivator-1 alpha (PGC1a), a transcription factor
cardiac hypertrophy [63]. Cardiomyocytes from SIRT3KO that regulates SIRT3 expression and other nuclear-
mice show increased ROS under basal conditions, as well encoded mitochondrial genes [80].
as after phenylephrine treatment, supporting the idea that Hearing also declines with age and is associated with
SIRT3 is required to prevent cellular ROS accumulation in the progressive loss of hair cells in the ear, which convert
the heart [71]. Further supporting the decline in antioxi- acoustic vibrations into nerve impulses. Interestingly, cal-
dant defense, cyclophilin D is thought to be a structural orie restriction can prevent age-related hearing loss, but
component of the mitochondrial permeability transition only with SIRT3 present [81]. Overexpression of SIRT3
pore and is a SIRT3 target. Cyclophilin D is hyperacety- leads to activation of IDH2 and confers protection from
lated in SIRT3KO hearts, resulting in increased mitochon- oxidative stress. Similarly, noise-induced hearing loss can
drial permeability, mitochondrial dysfunction, and altered be reduced by nicotinamide riboside, an NAD+ precursor
energetics [72]. thought to boost SIRT3 activity [82].
Impaired mitochondrial dynamics has also been associ- Based on this early evidence, we conclude that SIRT3
ated with several cardiac diseases, including cardiac hy- could have a neuroprotective role and loss of SIRT3 might
pertrophy and heart failure [64]. Generally, fusion is a accelerate the progression of neurodegenerative disease
mitochondrial stress response that promotes mitochondri- states. Even where a direct link between SIRT3 and a
al function and is regulated by a family of fusion proteins disease has not been established, growing evidence shows
that includes optic atrophy 1 (OPA1). OPA1 is a target of that mitochondrial dysfunction and oxidative stress have
SIRT3, and loss of SIRT3 leads to OPA1 hyperacetylation critical roles in the pathogenesis of neurodegenerative
489
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

diseases, highlighting a potentially broader role of SIRT3 Box 1. Outstanding questions


in regulating neuronal health.  What are the primary targets and/or pathways regulated by SIRT3?
 Do low stoichiometry sites of acetylation influence protein func-
SIRT3 and acetylation tion?
Much of the recent work on acetylation has been focused on  Does the reported in vitro deacylation activity of SIRT3, in addition
using large-scale proteomic approaches to identify proteins to deacetylation, contribute its regulatory role in vivo?
 Is the primary role of SIRT3-dependent deacetylation to restore
that are hyperacetylated in the absence of SIRT3, many of activity of metabolic enzymes?
which are in pathways with known roles in diseases of  Which other disease states or diseases of aging does SIRT3 influ-
aging [8387]. As the field moves towards more sophisti- ence?
cated proteomic technologies, an ever-increasing number  Does SIRT3 have a role in regulating longevity in model organisms
of acetylated proteins will be identified. As this number or human populations?

grows, it is becoming clear that regulation of a single


protein at a single site by SIRT3 cannot explain the role
of SIRT3 across a wide-array of biology and diseases of used to study longevity (i.e., S. cerevisiae, Caenorhabditis
aging. elegans, and Drosophila melanogaster) have sirtuin genes,
Supporting this idea are recent studies investigating but no SIRT3 ortholog has been identified. Furthermore, a
the stoichiometry of acetylation sites. Early estimates of SIRT3 transgenic mouse, which was generated recently,
the stoichiometry of any given site of acetylation are has been used to only study SIRT3 in the context of noise-
thought to be low [88,89]. If these findings hold, then again induced hearing loss [82]. Thus, while some studies show
the acetylation status of a single protein at one site is that SIRT3 is a compelling regulator of aging [95], future
unlikely to account for the changes in physiology or disease studies should focus on directly testing the role of SIRT3 in
observed. longevity.
Instead, an emerging sense in the field is that multiple
enzymes of a given pathway that are acetylated, even at Concluding remarks
low stoichiometry, could have a measurable effect on the Even in the absence of understanding the complete molec-
activity of the entire pathway [88]. In this setting, SIRT3 ular mechanisms of SIRT3 action (Box 1), it is clear that
might influence the pathway through sites of deacetyla- SIRT3 ablation leads to accelerated aging in several mod-
tion. Thus, the overall picture emerging from these prote- els. SIRT3KO mice have decreased time to development
omic studies is that the sum of all acetyl modifications, or and/or severity of age-related diseases. Thus, identifying
the sum of a set of SIRT3-regulated modifications in a the pathways that are affected when SIRT3 is ablated may
given pathway, could be critical for the regulation of me- aid in identifying pathways that become dysfunctional
tabolism. Future proteomic and physiological studies will during the aging process. From this perspective, continued
test this idea. work on SIRT3 may uncover important new pathways of
the aging process.
Does SIRT3 have a role in longevity?
Recently, nine hallmarks of aging were described that Acknowledgments
We would like to thank the Hirschey lab for thoughtful feedback. We
contribute to the aging process [4]. Current thinking on would also like to acknowledge funding support for the Hirschey lab from
aging places time-dependent accumulation of cellular dam- the American Heart Association grants 12SDG8840004 and
age at the core of the aging process. Based on this model, 12IRG9010008, Friedreichs Ataxia Research Alliance, The Ellison
future work should focus on the sources of damage and the Medical Foundation, the National Institutes of Health (NIH) grants
compensatory processes in response to damage. Along R01AA022146, R01AG045351, and R24DK085610-05; The Duke Pepper
Older Americans Independence Center (OAIC) Program in Aging
these lines, the regulation by SIRT3 of ROS and ROS- Research supported by the National Institute of Aging (P30AG028716-
induced damage could be an important mechanism by 01); E.M. is supported by an NIH training grant to Duke University
which SIRT3 influences aging and the diseases of aging. Cancer Biology Training Program (5T32-CA059365); B.S.P. is supported
Furthermore, protein acetylation and acylation could rep- by an NIH/NIGMS training grant to Duke University Pharmacological
Sciences Training Program (5T32GM007105-40).
resent a type of protein damage induced by carbon stress
[90].
Early studies investigating a role for SIRT3 in longevity References
1 Frye, R.A. (1999) Characterization of five human cDNAs with homology
took a genomics approach by screening long-lived humans
to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD
for possible variations in the SIRT3 gene. While early and may have protein ADP-ribosyltransferase activity. Biochem.
studies reported a possible correlation between a unique Biophys. Res. Commun. 260, 273279
SIRT3 genotype and longevity [91] or a novel variable 2 Kaeberlein, M. et al. (1999) The SIR2/3/4 complex and SIR2 alone
number tandem repeat (VNTR) enhancer within SIRT3 promote longevity in Saccharomyces cerevisiae by two different
mechanisms. Genes Dev. 13, 25702580
[92], a later study by the same group reported no associa- 3 Lin, S.J. et al. (2000) Requirement of NAD and SIR2 for life-span
tion [93]. Furthermore, an independent large-scale inves- extension by calorie restriction in Saccharomyces cerevisiae. Science
tigation into the genetics of longevity found no role for 289, 21262128
SIRT3 [94]. Thus, a direct link between SIRT3 and lon- 4 Lopez-Otin, C. et al. (2013) The hallmarks of aging. Cell 153, 11941217
gevity has not yet been established. 5 Lombard, D.B. et al. (2007) Mammalian Sir2 homolog SIRT3 regulates
global mitochondrial lysine acetylation. Mol. Cell. Biol. 27, 88078814
Additionally, no studies in model organisms have for- 6 Jacobs, K.M. et al. (2008) SIRT3 interacts with the daf-16 homolog
mally tested the role of SIRT3 in longevity. Part of this FOXO3a in the mitochondria, as well as increases FOXO3a dependent
could be due to the fact that some model organisms often gene expression. Int. J. Biol. Sci. 4, 291299

490
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

7 Ahn, B-H. et al. (2008) A role for the mitochondrial deacetylase Sirt3 in 34 Bell, E.L. et al. (2011) SirT3 suppresses hypoxia inducible factor 1a and
regulating energy homeostasis. Proc. Natl. Acad. Sci. U.S.A. 105, tumor growth by inhibiting mitochondrial ROS production. Oncogene
1444714452 30, 29862996
8 Samant, S.A. et al. (2014) SIRT3 deacetylates and activates OPA1 to 35 Hanahan, D. and Weinberg, R.A. (2011) Hallmarks of cancer: the next
regulate mitochondrial dynamics during stress. Mol. Cell. Biol. 34, generation. Cell 144, 646674
807819 36 Warburg, O. (1956) On respiratory impairment in cancer cells. Science
9 Hirschey, M.D. et al. (2010) SIRT3 regulates mitochondrial fatty-acid 124, 269270
oxidation by reversible enzyme deacetylation. Nature 464, 121125 37 Chandel, N.S. (2000) Reactive oxygen species generated at
10 Jing, E. et al. (2013) Sirt3 regulates metabolic flexibility of skeletal mitochondrial complex iii stabilize hypoxia-inducible factor-1alpha
muscle through reversible enzymatic deacetylation. Diabetes 62, during hypoxia: a mechanism of O2 sensing. J. Biol. Chem. 18,
34043417 2513025138
11 Papa, L. and Germain, D. (2014) SirT3 regulates a novel arm of the 38 Bell, E.L. et al. (2007) The Qo site of the mitochondrial complex III is
mitochondrial unfolded protein response. Mol. Cell. Biol. 34, 699710 required for the transduction of hypoxic signaling via reactive oxygen
12 Onyango, P. et al. (2002) SIRT3, a human SIR2 homologue, is an NAD- species production. J. Cell Biol. 177, 10291036
dependent deacetylase localized to mitochondria. Proc. Natl. Acad. Sci. 39 Ozden, O. et al. (2014) SIRT3 deacetylates and increases pyruvate
U.S.A. 99, 1365313658 dehydrogenase activity in cancer cells. Free Radic. Biol. Med. 76,
13 Feldman, J.L. et al. (2013) Activation of the protein deacetylase SIRT6 163172
by long-chain fatty acids and widespread deacylation by mammalian 40 Fan, J. et al. (2014) Tyr phosphorylation of PDP1 toggles recruitment
sirtuins. J. Biol. Chem. 288, 3135031356 between ACAT1 and SIRT3 to regulate the pyruvate dehydrogenase
14 Bao, X. et al. (2014) Identification of erasers for lysine crotonylated complex. Mol. Cell 53, 534548
histone marks using a chemical proteomics approach. Elife 2014, 3 41 McFate, T. et al. (2008) Pyruvate dehydrogenase complex activity
15 Peek, C.B. et al. (2013) Circadian clock NAD+ cycle drives controls metabolic and malignant phenotype in cancer cells. J. Biol.
mitochondrial oxidative metabolism in mice. Science 342, 1243417 Chem. 283, 2270022708
16 Zhang, Y. et al. (2015) SIRT3 and SIRT5 regulate the enzyme activity 42 Yang, H. et al. (2015) SIRT3-dependent GOT2 acetylation status affects
and cardiolipin binding of very long-chain acyl-CoA dehydrogenase. the malate-aspartate NADH shuttle activity and pancreatic tumor
PLoS ONE 10, e0122297 growth. EMBO J. 34, 11101125
17 Bharathi, S.S. et al. (2013) SIRT3 Regulates long-chain acyl-CoA 43 International Diabetes Federation (2013) IDF Diabetes Atlas (6th edn),
dehydrogenase by deacetylating conserved lysines near the active International Diabetes Federation
site. J. Biol. Chem. 288, 3383733847 44 Gao, A.W. et al. (2014) Mitochondrial response to nutrient availability
18 Altekruse, S. et al. (2015) SEER Cancer Statistics Review 19752007, and its role in metabolic disease. EMBO Mol. Med. 6, 580589
National Cancer Institute 45 Ji, H. and Friedman, M.I. (2007) Reduced capacity for fatty acid
19 Nowell, P.C. (1976) The clonal evolution of tumor cell populations. oxidation in rats with inherited susceptibility to diet-induced
Science 194, 2328 obesity. Metabolism 56, 11241130
20 Jackson, A.L. and Loeb, L.A. (2001) The contribution of endogenous 46 Bonomini, F. et al. (2015) Metabolic syndrome, aging and involvement
sources of DNA damage to the multiple mutations in cancer. Mutat. of oxidative stress. Aging Dis. 6, 109120
Res. 477, 721 47 Hotamisligil, G.S. (2006) Inflammation and metabolic disorders.
21 DeBerardinis, R.J. et al. (2008) The biology of cancer: metabolic Nature 444, 860867
reprogramming fuels cell growth and proliferation. Cell Metab. 7, 48 Andreux, P.A. et al. (2013) Pharmacological approaches to restore
1120 mitochondrial function. Nat. Rev. Drug Discov. 12, 465483
22 Kim, H-S. et al. (2010) SIRT3 is a mitochondria-localized tumor 49 Petersen, K.F. et al. (2003) Mitochondrial dysfunction in the elderly:
suppressor required for maintenance of mitochondrial integrity and possible role in insulin resistance. Science 300, 11401142
metabolism during stress. Cancer Cell 17, 4152 50 Bao, J. et al. (2010) SIRT3 is regulated by nutrient excess and
23 Hinds, P. et al. (1989) Mutation is required to activate the p53 gene for modulates hepatic susceptibility to lipotoxicity. Free Radic. Biol.
cooperation with the ras oncogene and transformation. J. Virol. 63, Med. 49, 12301237
739746 51 Hirschey, M.D. et al. (2011) SIRT3 deficiency and mitochondrial
24 Land, H. et al. (1983) Tumorigenic conversion of primary embryo protein hyperacetylation accelerate the development of the
fibroblasts requires at least two cooperating oncogenes. Nature 304, metabolic syndrome. Mol. Cell 44, 177190
596602 52 Kendrick, A.A. et al. (2011) Fatty liver is associated with reduced
25 Sun, H. and Taneja, R. (2007) Analysis of transformation and SIRT3 activity and mitochondrial protein hyperacetylation.
tumorigenicity using mouse embryonic fibroblast cells. Methods Mol. Biochem. J. 433, 505514
Biol. 383, 303310 53 Lantier, L. et al. (2015) SIRT3 is crucial for maintaining skeletal
26 Finley, L.W.S. et al. (2011) SIRT3 opposes reprogramming of cancer muscle insulin action and protects against severe insulin resistance
cell metabolism through HIF1alpha destabilization. Cancer Cell 19, in high fat fed mice. Diabetes Published online May 6, 2015. http://
416428 dx.doi.org/10.2337/db14-1810
27 Zhang, Y-Y. and Zhou, L-M. (2012) Sirt3 inhibits hepatocellular 54 Hoehn, K.L. et al. (2009) Insulin resistance is a cellular antioxidant
carcinoma cell growth through reducing Mdm2-mediated p53 defense mechanism. Proc. Natl. Acad. Sci. U.S.A. 106, 1778717792
degradation. Biochem. Biophys. Res. Commun. 423, 2631 55 Jing, E. et al. (2011) Sirtuin-3 (Sirt3) regulates skeletal muscle
28 Yang, B. et al. (2014) Aberrant expression of SIRT3 is conversely metabolism and insulin signaling via altered mitochondrial
correlated with the progression and prognosis of human gastric oxidation and reactive oxygen species production. Proc. Natl. Acad.
cancer. Biochem. Biophys. Res. Commun. 443, 156160 Sci. U.S.A. 108, 1460814613
29 Zhang, C.Z. et al. (2012) Low SIRT3 expression correlates with poor 56 Matsuzawa, N. et al. (2007) Lipid-induced oxidative stress causes
differentiation and unfavorable prognosis in primary hepatocellular steatohepatitis in mice fed an atherogenic diet. Hepatology 46,
carcinoma. PLoS ONE 7, e51703 13921403
30 Haigis, M.C. et al. (2012) SIRT3 is a mitochondrial tumor suppressor: a 57 Nakamura, S. et al. (2009) Palmitate induces insulin resistance in
scientific tale that connects aberrant cellular ROS, the Warburg effect, H4IIEC3 hepatocytes through reactive oxygen species produced by
and carcinogenesis. Cancer Res. 72, 24682472 mitochondria. J. Biol. Chem. 284, 1480914818
31 Tudek, B. et al. (2010) Involvement of oxidatively damaged DNA and 58 Paglialunga, S. et al. (2015) In adipose tissue, increased mitochondrial
repair in cancer development and aging. Am. J. Transl. Res. 2, 254284 emission of reactive oxygen species is important for short-term high-fat
32 Schieber, M. and Chandel, N.S. (2014) ROS function in redox signaling diet-induced insulin resistance in mice. Diabetologia 58, 10711080
and oxidative stress. Curr. Biol. 24, R453R462 59 Kang, L. et al. (2014) Heterozygous SOD2 deletion impairs glucose-
33 Qiu, X. et al. (2010) Calorie restriction reduces oxidative stress by stimulated insulin secretion, but not insulin action, in high-fat-fed
SIRT3-mediated SOD2 activation. Cell Metab. 12, 662667 mice. Diabetes 63, 36993710

491
Review Trends in Endocrinology and Metabolism September 2015, Vol. 26, No. 9

60 Caton, P.W. et al. (2013) Sirtuin 3 regulates mouse pancreatic beta cell 77 Han, P. et al. (2014) Pituitary adenylate cyclase-activating polypeptide
function and is suppressed in pancreatic islets isolated from human protects against beta-amyloid toxicity. Neurobiol. Aging 35, 20642071
type 2 diabetic patients. Diabetologia 56, 10681077 78 Weir, H.J. et al. (2012) CNS SIRT3 expression is altered by reactive
61 Kim, M. et al. (2015) SIRT3 Overexpression attenuates palmitate- oxygen species and in Alzheimers disease. PLoS ONE 7, e48225
induced pancreatic beta-cell dysfunction. PLoS ONE 10, e0124744 79 Manfredi, G. and Xu, Z. (2005) Mitochondrial dysfunction and its role in
62 Jarreta, D. et al. (2000) Mitochondrial function in heart muscle from motor neuron degeneration in ALS. Mitochondrion 5, 7787
patients with idiopathic dilated cardiomyopathy. Cardiovasc. Res. 45, 80 Song, W. et al. (2013) Mutant SOD1G93A triggers mitochondrial
860865 fragmentation in spinal cord motor neurons: neuroprotection by
63 Dai, D.F. et al. (2012) Mitochondria and cardiovascular aging. Circ. SIRT3 and PGC-1alpha. Neurobiol. Dis. 51, 7281
Res. 110, 11091124 81 Someya, S. et al. (2010) Sirt3 mediates reduction of oxidative damage
64 Ikeda, Y. et al. (2015) Molecular mechanisms mediating mitochondrial and prevention of age-related hearing loss under caloric restriction.
dynamics and mitophagy and their functional roles in the Cell 143, 802812
cardiovascular system. J. Mol. Cell. Cardiol. 78, 116122 82 Brown, K.D. et al. (2014) Activation of SIRT3 by the NAD+ precursor
65 Hyyti, O.M. et al. (2010) Aging impairs myocardial fatty acid and nicotinamide riboside protects from noise-induced hearing loss. Cell
ketone oxidation and modifies cardiac functional and metabolic Metab. 20, 10591068
responses to insulin in mice. Am. J. Physiol. Heart Circ. Physiol. 83 Rardin, M.J. et al. (2013) Label-free quantitative proteomics of the
299, H868H875 lysine acetylome in mitochondria identifies substrates of SIRT3 in
66 Alrob, O.A. et al. (2014) Obesity-induced lysine acetylation increases metabolic pathways. Proc. Natl. Acad. Sci. U.S.A. 110, 66016606
cardiac fatty acid oxidation and impairs insulin signalling. Cardiovasc. 84 Hebert, A.S. et al. (2013) Calorie restriction and SIRT3 trigger global
Res. 103, 485497 reprogramming of the mitochondrial protein acetylome. Mol. Cell 49,
67 Ahmad, F. et al. (2005) Increased alpha2 subunit-associated 186199
AMPK activity and PRKAG2 cardiomyopathy. Circulation 112, 85 Sol, E.M. et al. (2012) Proteomic investigations of lysine acetylation
31403148 identify diverse substrates of mitochondrial deacetylase Sirt3. PLoS
68 Smeets, P.J.H. et al. (2008) Cardiac hypertrophy is enhanced in PPAR ONE 7, e50545
alpha-/- mice in response to chronic pressure overload. Cardiovasc. Res. 86 Dittenhafer-Reed, K.E. et al. (2015) SIRT3 mediates multi-tissue
78, 7989 coupling for metabolic fuel switching. Cell Metab. 21, 637646
69 Cox, K. et al. (2009) Cardiac hypertrophy in mice with long-chain acyl- 87 Still, A.J. et al. (2013) Quantification of mitochondrial acetylation
CoA dehydrogenase or very long-chain acyl-CoA dehydrogenase dynamics highlights prominent sites of metabolic regulation. J. Biol.
deficiency. Lab. Invest. 89, 13481354 Chem. 288, 2620926219
70 Suenaga, M. et al. (2003) Functional disorders of the oxidative 88 Baeza, J. et al. (2014) Stoichiometry of site-specific lysine acetylation in
phosphorylation system in the heart mitochondria of mice with an entire proteome. J. Biol. Chem. 289, 2132621338
juvenile visceral steatosis. Biol. Pharm. Bull. 26, 289294 89 Weinert, B.T. et al. (2014) Acetylation dynamics and stoichiometry in
71 Sundaresan, N.R. et al. (2009) Sirt3 blocks the cardiac hypertrophic Saccharomyces cerevisiae. Mol. Syst. Biol. 10, 716
response by augmenting Foxo3a-dependent antioxidant defense 90 Wagner, G. and Hirschey, M.D. (2014) Non-enzymatic protein
mechanisms in mice. J. Clin. Invest. 119, 27582771 acylation as a carbon stress regulated by sirtuin deacylases. Mol.
72 Hafner, A.V. et al. (2010) Regulation of the mPTP by SIRT3-mediated Cell 54, 516
deacetylation of CypD at lysine 166 suppresses age-related cardiac 91 Rose, G. et al. (2003) Variability of the SIRT3 gene, human silent
hypertrophy. Aging 2, 914923 information regulator Sir2 homologue, and survivorship in the elderly.
73 Beal, M.F. (2005) Mitochondria take center stage in aging and Exp. Gerontol. 38, 10651070
neurodegeneration. Ann. Neurol. 58, 495505 92 Bellizzi, D. et al. (2005) A novel VNTR enhancer within the SIRT3 gene,
74 Ban, N. et al. (2013) Light-dark condition regulates sirtuin mRNA a human homologue of SIR2, is associated with survival at oldest ages.
levels in the retina. Exp. Gerontol. 48, 12121217 Genomics 85, 258263
75 Sidorova-Darmos, E. et al. (2014) Differential expression of sirtuin 93 De Rango, F. et al. (2008) A novel sampling design to explore gene-
family members in the developing, adult, and aged rat brain. Front. longevity associations: the ECHA study. Eur. J. Hum. Genet. 16,
Aging Neurosci. 6, 333 236242
76 Zeng, L. et al. (2014) Age-related decrease in the mitochondrial sirtuin 94 Lescai, F. et al. (2009) Human longevity and 11p15.5: a study in
deacetylase Sirt3 expression associated with ROS accumulation in 1321 centenarians. Eur. J. Hum. Genet. 17, 15151519
the auditory cortex of the mimetic aging rat model. PLoS ONE 9, 95 Benigni, A. et al. (2009) Disruption of the Ang II type 1 receptor
e88019 promotes longevity in mice. J. Clin. Invest. 119, 524530

492

You might also like