You are on page 1of 11

0888-8809/07/$15.

00/0
Printed in U.S.A.

Molecular Endocrinology 21(8):17451755


Copyright 2007 by The Endocrine Society
doi: 10.1210/me.2007-0079

MINIREVIEW

Sirtuin Functions in Health and Disease


Hiroyasu Yamamoto, Kristina Schoonjans, and Johan Auwerx
Institut de Genetique et de Biologie Moleculaire et Cellulaire (H.Y., K.S., J.A.), Centre National de la
Recherche Scientifique/Institut National de la Sante et de la Recherche Medicale/Universite Louis
Pasteur, 67404 Illkirch, France; and Institut Clinique de la Souris (J.A.), 67404 Illkirch, France
Sirtuins or Sir2 (silent information regulator 2)-related enzymes have originally been defined as a
family of nicotinamide adenine dinucleotide-dependent enzymes that deacetylate lysine residue
on various proteins. Certain sirtuins have in addition an ADP-ribosyltransferase activity. The sirtuins are remarkably conserved throughout evolution from archaebacteria to eukaryotes. The
mammalian sirtuins SIRT1SIRT7 are implicated in
a variety of cellular functions ranging from gene
silencing, over the control of the cell cycle and

apoptosis, to energy homeostasis. On a wholebody level, the wide range of cellular activities of
the sirtuins suggests that they could constitute
therapeutic targets to combat metabolic, neurodegenerative, and proliferative diseases. Here, we review some of the recent data related to the sirtuins
and discuss their mode of action, their biological
role in cellular and organismal models, and their
possible association to age-related human
diseases. (Molecular Endocrinology 21: 17451755,
2007)

thought to be one of the primary factors contributing to


this selective advantage.
Meanwhile, since the 1930s, it has been also well
known that caloric restriction (CR) can retard the aging
process and delay the onset of numerous aging-related diseases, such as cancer, cardiovascular diseases, and metabolic diseases. CR significantly expands lifespan in organisms ranging from yeast and
nematodes to rodents and monkeys (1, 2). Interestingly, the beneficial health outcomes of CR resemble
those that are induced by resveratrol in a number of
animal models, suggesting that the molecular pathways by which resveratrol acts are similar to those
activated by CR. Recently, it was suggested that the
sirtuins could be the common mediators that explain
both the effects of resveratrol and CR pathways. In
this review, we will discuss the molecular mechanism
that underlies the biological activity of these sirtuins,
their functional roles in whole-body physiology, and
their possible associations to human diseases.

HE RISING INCIDENCE of obesity-related diseases, such as diabetes, dyslipidemia, and cardiovascular and cerebrovascular diseases in industrialized countries has become a public health problem
of major importance. Many therapeutic and preventive
strategies to prevent or combat obesity have seen the
light of day, but few have survived the test of time. One
phenomenon that caught the interest in this context is
the so-called French paradox. First noted by Irish
physician Samuel Black in 1819, the French paradox
makes an allusion to the fact that the French are
perceived as having a relatively low incidence of cardiovascular and metabolic disease, although their diet
is rich in saturated fat. The high consumption of red
wine, which is rich in the polyphenol resveratrol, is

First Published Online April 24, 2007


Abbreviations: A, Amyloid-; AADPR, acetyl-ADP ribose;
AASIS, amino acid-stimulated insulin secretion; AceCS2,
acetyl coenzyme A synthetase 2; AD, Alzheimers disease;
AR, androgen receptor; BAT, brown adipose tissue; BER,
base excision repair; CoA, coenzyme A; CR, caloric restriction; FOXO, forkhead box type O transcription factor; GDH,
glutamate dehydrogenase; HD, Huntingtons disease; HDAC,
histone deacetylase; MEF, mouse embryonic fibroblast;
NAD, nicotinamide adenine dinucleotide; NADH, reduced
nicotinamide adenine dinucleotide; NFB, nuclear factor B;
Nmnat 1, nicotinamide mononucleotide adenyltransferase 1;
PGC-1, peroxisome proliferator-activated receptor coactivator 1; PolyQ, polyglutamine; PPAR, peroxisome proliferator-activated receptor ; Sir2p, silent information regulator
2 protein; UCP, uncoupling protein.
Molecular Endocrinology is published monthly by The
Endocrine Society (http://www.endo-society.org), the
foremost professional society serving the endocrine
community.

SIRTUINS ARE NICOTINAMIDE ADENINE


DINUCLEOTIDE-DEPENDENT HISTONE
DEACETYLASES OR ADP-RIBOSYL
TRANSFERASES
The founding member of the sirtuin protein family was
the silent information regulator 2 protein (Sir2p) of
Saccharomyces cervisiae, a nicotinamide adenine
dinucleotide (NAD)-dependent histone deacetylase
(HDAC) that regulates chromatin silencing (37). Yeast
strains with abnormal levels of Sir2p show defects in
1745

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

1746 Mol Endocrinol, August 2007, 21(8):17451755

many cellular functions, including transcriptional and


recombinational silencing, senescence, and DNA repair. In S. cervisiae, there are four sirtuins (NADdependent histone deacetylases Hst1Hst4) in addition to Sir2p, whereas in mammals seven homologs,
i.e. SIRT1SIRT7, have been identified (8, 9) (Table 1).
The remarkable conservation of members of the sirtuin
gene family from yeast to humans indicates that these
proteins play vital physiological roles (9).
Among the large HDAC protein family, sirtuins were
originally categorized as class III HDACs. Whereas
classes I and II HDACs use zinc as a cofactor and are
inhibited by trichostatin A (10), sirtuins are not inhibited
by trichostatin A and convert acetylated protein substrates in a reaction that uses NAD into a deacetylated protein, nicotinamide, and the acetyl ester metabolites 2-O- and 3-O-acetyl-ADP ribose (AADPR),
which are formed by the transfer of the acetyl group to
the ADP-ribose portion of NAD (6, 7, 1114) (Fig. 1).
The deacetylase activity of the sirtuins is controlled by
the cellular [NAD]/[NADH] ratio, i.e. NAD works as
an activator, whereas nicotinamide and reduced nicotinamide adenine dinucleotide (NADH) inhibit their
activity (1519).
Because sirtuins are class III HDACs, it was logical
that their function initially became associated with
transcriptional repression. Acetylated histones H1, H3,
and H4 are known to be physiological substrates for
the sirtuins, and lysine 16 in histone H4 appears to be
the most critical residue for sirtuin-mediated transcriptional silencing (20, 21). Afterwards, it has been recognized that a growing number of nonhistone proteins
are also deacetylated by the sirtuins, largely expanding their biological roles. These nonhistone sirtuin substrates include several transcriptional regulators, such
as the nuclear factor-B (NFB), forkhead box type O
transcription factors (FOXO), and the peroxisome proliferator-activated receptor (PPAR) coactivator 1
(PGC-1), but also enzymes, such as acetyl coenzyme
A (CoA) synthetase 2 (AceCS2), and structural proteins, such as -tubulin (Table 1).
Interestingly, AADPR, a product generated in the
deacetylation reaction catalyzed by the sirtuins, also
has a role as a second messenger because it is involved in establishing a transcriptionally silent and
functionally heterochromatic state (20, 22). AADPR
achieves this effect by two independent mechanisms

Yamamoto et al. Minireview

Fig. 1. Two Reactions Catalyzed by Sirtuins, i.e. Deacetylation and ADP-Ribosylation


Sirtuins (SIRT1SIRT3, SIRT5) catalyze a deacetylation reaction in which an acetyl group is transferred to the ADPribose (ADPR) moiety of NAD and 2-O-acetyl-ADPR is produced. 3-O-acetyl-ADPR is formed nonenzymatically from
2-O-acetyl-ADPR. In contrast, SIRT4 and SIRT6 catalyze
ADP-ribosylation of proteins rather than deacetylation.

that involve on the one hand a conformational change


in SIRT1, which in a feedforward loop potentiates the
gene-silencing effects of the Sir complex (20), and on
the other hand, by binding to the histone variant macro
H2A1.1, which is present in inactive heterochromatic
regions (22). Because cellular [NAD]/[NADH] ratio,
nicotinamide, and AADPR levels are governed by cellular energetics, SIRT1 may be an extremely versatile
energy sensor that enables transcription to sense the
metabolic state of the cell.
Two sirtuins, SIRT4 and SIRT6, are lacking important deacetylase activity but instead have a robust

Table 1. Main Characteristic of Mammalian Sirtuins


Targets

Biological Function

SIRT1

Nucleus (nuclei)

Intracellular Localization

Deacetylase

PGC-1, FOXOs, NFB

SIRT2
SIRT3
SIRT4
SIRT5
SIRT6

Cytoplasm
Nucleus and mitochondria
Mitochondria (matrix)
Mitochondria
Nucleus (heterochromatic
region)
Nucleus (nucleoli)

Deacetylase
Deacetylase
ADP-ribosyl transferase
Deacetylase
ADP-ribosyl transferase

H4, -tubulin
AceCS2
GDH
Unknown
DNA polymerase

Metabolism/inflammation/
neurodegeneration
Cell cycle/tumorigenesis
Metabolism
Insulin secretion
Unknown
DNA repair

Unknown

RNA polymerase I

rDNA transcription

SIRT7

Activity

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

Yamamoto et al. Minireview

NAD-dependent ADP-ribosyl transferase activity


(Fig. 1). The ADP-ribosyl transferase activities of these
two SIRTs are not a complete surprise in view of the
initial report on the enzymatic activity of yeast Sir2p,
which described a mono-ADP-ribosyl transferase activity (23). Posttranslational modification of protein
substrates by mono-ADP-ribosylation involves the
creation of an N- or S-glycosidic linkage between a
specific amino acid (such as arginine or cysteine) on
the acceptor protein and the ADP-ribose residue of
NAD.
The seven mammalian sirtuins show significant sequence homology and contain conserved catalytic
and NAD binding domains (Fig. 2 and Table 1). Although based on sequence similarities, eukaryotic sirtuins have been divided into four broad phylogenetic
groups, with SIRT1, SIRT2, and SIRT3 composing
class I, SIRT4 constituting class II, SIRT5 forming class
III, and SIRT6 and SIRT7 forming class IV (9), there is
no obvious correlation between this classification and
the specific biological functions of the sirtuins. Another
more relevant way to functionally classify the sirtuins is
based on their intracellular localizations (24) (Table 1).
Four sirtuins, SIRT1, SIRT3, SIRT6, and SIRT7, are
nuclear proteins, but their subnuclear localizations are
distinct. SIRT1 is detected in the nuclei but is excluded
from the nucleoli, whereas SIRT6 and SIRT7, are associated with heterochromatic regions and nucleoli,
respectively (24, 25). SIRT2 is generally localized in the
cytoplasm, but, during the G2/M phase, it binds chromatin in the nucleus (26). SIRT3, SIRT4, and SIRT5 are
present in the mitochondria. Although initially described as a mitochondrial protein, recent studies suggest that SIRT3 can also be a nuclear protein that
transfers to the mitochondria during cellular stress
(119). The exact localization of the SIRT3SIRT5 in the
mitochondria has, however, not yet been defined
experimentally.
All seven sirtuins are ubiquitously expressed in human tissues, although higher levels of mRNA expres-

Fig. 2. Schematic of the Structural Domains of the Mammalian Sirtuins

Mol Endocrinol, August 2007, 21(8):17451755 1747

sion are detected in the brain and testis for most


sirtuins (8, 24). Except for SIRT2 and SIRT5, the expression for the sirtuins is higher in fetal relative to
adult brain, which might indicate the possibility that
they play important roles in the development of the
neuronal system.

SIRTUINS AND THE CONTROL OF CELL


PROLIFERATION, STRESS RESISTANCE, AND
CANCER
Many factors that control cell proliferation and apoptosis are identified as sirtuin substrates, such as p53
(2729). SIRT1 is reported to be associated with the
tumor suppressor protein p53. p53 has several acetylation sites, and its hyperacetylation stabilizes and
activates it to trigger apoptosis and cell-cycle arrest
(3032). Conversely, the deacetylation of p53 by
SIRT1 is predicted to induce its destruction by the
MDM2 (mouse double minute 2)-dependent ubiquitinmediated pathway. In fact, overexpression of SIRT1
inhibits p53 transcriptional activity and p53-dependent apoptosis in response to DNA damage and oxidative stress, whereas overexpression of dominantnegative SIRT1 protein can potentiate these cellular
stress responses (28, 29). In thymocytes from SIRT1deficient mice, the levels of p53 acetylation were significantly up-regulated after exposure to ionizing radiation (33), indicating that SIRT1 has a role in increasing
the stress resistance of cells. Increased p53 acetylation has also been associated with senescence (34).
Interestingly, SIRT1 was shown recently to promote
replicative senescence, through a process that implicates p19ARF, which positively regulates p53 through
inhibiting MDM2 (35). This effect is in marked contrast
to Sir2p function in yeast, which extends replicative
lifespan (12, 36, 37).
Sirtuins also affect the activity of the FOXO family of
transcription factors (38, 39). Genetic epistasis in Caenorhabditis elegans and metabolic studies in mice indicate that FOXO genes regulate cell differentiation,
transformation, and metabolism (40). In C. elegans,
mutation of the FOXO ortholog Daf16 (abnormal dauer
formation) rescues the dauer state, caused by mutations of the insulin/IGF receptor ortholog Daf2 (4143).
In mammalian cells, growth factor-induced activation
of phosphatidylinositol 3-kinase leads to an increase
in the activity of the serine/threonine kinase AKT/protein kinase B (44, 45), which in turn leads to phosphorylation and inactivation of the FOXO proteins by their
retention in the cytoplasm (4649). The translocation
of FOXO3a from the cytoplasm to the nucleus is induced by its deacetylation by SIRT1 in response to
oxidative stress (50). In the nucleus, SIRT1 and
deacetylated FOXO3a form a complex that induces
cell-cycle arrest and resistance to oxidative stress but
inhibits the ability of FOXO3a to induce apoptosis.
SIRT1-mediated deacetylation also affects FOXO1 nu-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

1748 Mol Endocrinol, August 2007, 21(8):17451755

cleocytoplasmic shuttling, leading to the expression of


FOXO1 target genes, hence inducing gluconeogenesis
and glucose release from hepatocytes (51).
SIRT1 is also reported to play an important role
during myocyte differentiation. The levels of SIRT1 and
the [NAD]/[NADH] ratio decrease during muscle differentiation. Overexpression of SIRT1 retards muscle
differentiation via formation of a complex with the
acetyltransferase PCAF (p300/CBP-associated factor)
and MyoD, whereas in cells with reduced SIRT1 expression, muscle gene expression and differentiation
are enhanced (52). In addition, the muscle cell transcription factor, myocyte enhancer factor MEF2 is inactivated through deacetylation by SIRT1 (53). SIRT1
also was reported to bind and deacetylate the androgen receptor (AR) at a conserved lysine motif, thereby
repressing the ligand-induced AR transcriptional activity by the inhibition of coactivator-induced interactions between the AR amino and carboxyl termini (54).
Hst2, the yeast ortholog of SIRT2 can induce Sir2pindependent lifespan extension and rDNA silencing in
yeast (10), highlighting the redundancies of the SIRTs
in the control of lifespan in yeast. As to the mammalian
SIRT2, it deacetylates a number of substrates, including -tubulin and histone H4K16Ac (55, 56). In mammalian cell culture systems, SIRT2 was shown to play
an important role in the control of the cell cycle (26,
57). The global levels of H4K16 acetylation peak at the
S and G2 phase, dropping before cells enter mitosis,
coinciding with the increased expression of SIRT2, its
nuclear translocation, and association with chromatin
(26). In SIRT2-deficient mouse embryonic fibroblasts
(MEFs), H4K16 acetylation remains high during mitosis, delaying S-phase entry. This suggests that the
SIRT2-mediated conversion of H4K16Ac to its
deacetylated form may be pivotal to the formation of
condensed chromatin. SIRT2 has also been suggested to act as a tumor suppressor gene in human
gliomas (58). Down-regulation of SIRT2 gene expression and/or deletion of the chromosomal region harboring the SIRT2 gene is frequently observed in gliomas. SIRT2 expression might hence serve as a
potential diagnostic molecular marker for gliomas, and
modulation of its activity might be of interest for the
management of gliomas.
The nuclear protein SIRT6 is a weak deacetylase but
is endowed with a robust ADP-ribosyltransferase activity. SIRT6/ MEFs have an increased frequency of
various chromosomal aberrations, which indicates
that SIRT6 is involved in maintaining genome integrity
(25). SIRT6 deficiency also impairs the proliferation of
these MEFs and enhances their sensitivity to DNAdamaging agents. This regulation of genomic stability
by SIRT6 is related to its function in base excision
repair (BER) of single-stranded DNA breaks. Interestingly, overexpression of the DNA polymerase involved
in BER, Pol, rescues these defects (25, 59, 60).
SIRT6/ mice die prematurely subsequent to several
rather acute degenerative processes, including loss of
sc fat, reduction of bone mineral density, colitis, and

Yamamoto et al. Minireview

lymphopenia associated with increased lymphocyte


apoptosis (25). SIRT6 may also control metabolism,
because SIRT6/ mice exhibits low levels of serum
IGF-I and a gradual decrease of serum glucose. It
remains, however, unclear how SIRT6 influences BER
and whether the altered serum IGF and insulin levels of
SIRT6/ mice directly contribute to aging-like phenotypes or, alternatively, reflect compensatory
changes.
SIRT7 is a nucleolar protein that is associated with
active rRNA genes in which it interacts with RNA polymerase I (61). SIRT7 overexpression increases rRNA
transcription, whereas its down-regulation decreases
rRNA transcription. Interestingly, SIRT7 expression is
enriched in tissues with a high proliferation potential,
such as liver, spleen, and testis. This is in contrast to
tissues with a low cellular turnover rate, such as skeletal and heart muscle and brain that express low levels
of SIRT7. SIRT7 seems hence to drive ribosome biogenesis in dividing cells, and it has been associated
with thyroid and breast cancer (62, 63). SIRT7 gene
expression is up-regulated in these cancers, and,
moreover, its levels are closely related to tumor development and disease progression of breast cancer.
Additional study will, however, be required to identify
the mechanism underlying enhanced SIRT7 gene expression in these cancers.
All of these studies combined suggest important
roles of the sirtuins in the control of cell proliferation:
SIRT1 inhibits p53 and modulates FOXO activity,
SIRT2 controls chromosome condensation during the
cell cycle, SIRT6 acts in BER, whereas SIRT7 activates
rRNA transcription.

SIRTUINS CONTROL METABOLIC ACTIVITY


The fact that several of the protein substrates, such as
AceCS2 and PGC-1, which are deacetylated by the
sirtuins, are involved in metabolism indicated a metabolic role for this protein family (6467). This hypothesis was substantiated through studies that used both
cell-based approaches as well as a combination of
whole
animal
genetic
and
pharmacological
approaches.
Two groups reported the phenotypes of germ-line
SIRT1/ mice, which showed some similarities but
also revealed differences, potentially the result from
the methods used to generate the mice (33, 68). In
general, SIRT1/ mice were smaller at birth and
showed an elevated postnatal lethality attributable to
developmental problems that are not observed in
yeast, C. elegans, or Drosophila. In an outbred background, some of the SIRT1/ mice survived to adulthood, but they have fertility problems and display a
variety of other problems, including skeletal, eye, and
cardiac defects. The study of some of these genetically engineered SIRT1 mouse models revealed a role
of SIRT1 in pancreatic homeostasis. In the pancreas,

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

Yamamoto et al. Minireview

SIRT1 was preferentially localized in the islets of Langerhans. In the SIRT1/ mice, insulin secretion in response to glucose was lower compared with wild-type
littermates, indicating that SIRT1 positively regulates
insulin secretion in pancreatic -cells (69). Conversely,
-cell-specific SIRT1-overexpressing transgenic mice
exhibit an improved glucose tolerance and an enhanced glucose-stimulated insulin secretion (70).
From the microarray analysis comparing gene expression patterns in SIRT1-overexpressing- and knockdown pancreatic -cell lines, uncoupling protein 2
(UCP2), a protein that negatively regulates insulin secretion in pancreatic -cells, was identified as a target
that was repressed by SIRT1. SIRT1 decreases UCP2
gene expression by directly binding to the UCP2 promoter, leading to a better coupling of mitochondrial
respiration and ATP synthesis, which will induce insulin secretion (69, 70).
PPAR is a key regulator in adipogenesis and fat
storage through the control of the expression of many
adipocyte-specific genes (71). SIRT1 represses
PPAR actively via docking with two of its corepressors, NcoR (nuclear receptor corepressor) and SMRT
(silencing mediator of retinoid and thyroid hormone
receptor). Hence, SIRT1 was suggested to act as a
corepressor of PPAR-mediated transcription. From a
functional point of view, the repression of PPAR by
SIRT1 attenuates adipogenesis, and up-regulation of
SIRT1 triggers lipolysis and loss of fat in differentiated
fat cells (72). Conversely, the reduction in SIRT1 expression in SIRT1/ mice hence compromises the
mobilization of fatty acids from adipose tissue during
fasting.
Perhaps the most relevant target of SIRT1 in the
metabolic arena is the cofactor PGC-1, the master
regulator of mitochondrial biogenesis. PGC-1 is activated by SIRT1-mediated deacetylation (65, 66). In
the liver, the activation of PGC-1 will facilitate the
gluconeogenic activity of hepatocyte nuclear factor 4
and stimulate hepatic glucose output (66). In the muscle and brown adipose tissue (BAT), the SIRT1-mediated deacetylation of PGC-1 is translated into enhanced mitochondrial activity, which translated in
increased exercise tolerance and thermogenesis,
leading to protection against the onset of obesity and
associated metabolic dysfunction (73). For its
deacetylase activity, SIRT1 is strictly dependent on
cellular NAD levels, which reflect cellular energy status. The changes in cellular NAD levels that affect
SIRT1 deacetylase activity hence seem to inform
PGC-1 about the cellular energy status. PGC-1 can
then adapt cellular energy production through its commanding role on mitochondrial biogenesis and function. These studies place SIRT1, which acts as cellular
energy sensor, upstream of PGC-1 as an important
regulator of mitochondrial activity.
It is clear that many of these studies, which focused
on a given tissue type, indicated potential links between metabolic homeostasis and SIRT1 action. As
discussed, SIRT1 enhances insulin secretion in re-

Mol Endocrinol, August 2007, 21(8):17451755 1749

sponse to glucose in the pancreas through the repression of UCP2 (69, 70); in the liver, SIRT1 induces
gluconeogenesis and represses glycolysis (66); in adipose tissue, SIRT1 inhibits fat storage and increases
lipolysis via repression of PPAR (72). These pleiotropic, often opposing, metabolic effects of SIRT1 in
different tissues complicated the elucidation of the
impact of SIRT1 on whole-body metabolic homeostasis. Two recent studies using the SIRT1 activator resveratrol shed more light on this complex role of SIRT1
in metabolism (73, 74). In one study, it was shown that
resveratrol mimics several aspects of CR in mice on a
high-calorie diet, by prolonging lifespan, improving insulin sensitivity, and enhancing motor function (74).
This study hence extends previous work that SIRT1
activation by resveratrol mimics CR and delays aging
in a wide range of organisms going from S. cerevisiae
(75) over C. elegans to Drosophila (76). In a second
independent study, treatment of mice with a higher
dose of resveratrol was also shown to protect them
against diet-induced obesity and the associated insulin resistance (73). This study demonstrated that the
amelioration of insulin sensitivity was linked to an enhanced mitochondrial function subsequent to activation of PGC-1 by SIRT1-mediated PGC-1 deacetylation (73). The enhanced mitochondrial activity
furthermore led to an increase in oxidative type-muscle fibers and enhanced resistance to muscle fatigue.
Moreover, a significant association between three single-nucleotide polymorphisms in the SIRT1 gene and
energy homeostasis in humans indicated that SIRT1
constitutes an attractive and validated target to regulate energy and metabolic homeostasis in man (73).
SIRT3 was originally thought to be a mitochondrial
protein, but recently it was demonstrated that mitochondrial transfer from its normal nuclear location was
induced during cellular stress (77, 78, 119). The expression of SIRT3 is finely regulated. In mice, caloric
restriction (CR) up-regulates SIRT3 expression levels
in white adipose tissue and BAT. Furthermore, cold
exposure also induces SIRT3 in BAT (79). Interestingly,
the constitutive expression of SIRT3 promotes the
expression of PGC-1, UCP1, and other genes involved in mitochondrial functions, indicating that
SIRT3 modulates adaptive thermogenesis in BAT, a
process that most likely involves both nuclear and
mitochondrial activities. One mitochondrial activity of
SIRT3 is the deacetylation and activation of the mitochondrial form of AceCS2, an enzyme that catalyzes
the formation of acetyl CoA from acetate (64, 67).
Deacetylation of AceCS2 hence increases the conversion of acetate into acetyl CoA, an intermediate of the
tricarboxylic acid cycle. AceCS2 is abundantly expressed in heart and skeletal muscle but absent from
liver, and its expression is induced when energy becomes limiting, as during CR and ketogenesis (80).
Because SIRT3 facilitates the metabolic use of acetate, it may hence be especially important to ensure
energy production under conditions when ATP is
scarce (64, 67, 80). In analogy to this function of

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

1750 Mol Endocrinol, August 2007, 21(8):17451755

SIRT3, SIRT1 deacetylates and activates the cytoplasmic AceCS1 to provide acetyl CoA, which acts as a
building block for fatty acid and cholesterol synthesis
(64). For only one of the human sirtuins, i.e. SIRT3, a
direct genetic link with longevity has been established.
In fact, mutations in the SIRT3 gene enhancer, which
up-regulate its expression, were enriched in long-lived
individuals (81).
Another mitochondrial SIRT protein, SIRT4, was
shown recently to interact with glutamate dehydrogenase (GDH) (82). Glutamate formed from glutamine is
converted to the tricarboxylic acid cycle intermediate
-ketoglutarate by GDH in the mitochondria (82, 83).
This promotes mitochondrial activation and increases
the ATP/ADP ratio, which subsequently activates insulin secretion in pancreatic -cells. SIRT4 uses NAD
to ADP-ribosylate and decrease the activity of GDH,
consequently reducing the production of -ketoglutarate and the generation of ATP (82). In SIRT4-deficient
pancreatic -cells, GDH activity increases, leading to a
stimulation of insulin secretion in response to glutamine. SIRT4 therefore has an inhibitory effect on
amino acid-stimulated insulin secretion (AASIS). It
seems reasonable to speculate that AASIS is activated
during chronic CR, because protein turnover is increased and amino acids are used as carbon and
energy sources to drive gluconeogenesis in this condition. Consistent with this, SIRT4 repression of GDH
is alleviated during long-term CR, resulting in activation of AASIS in -cells and potentially gluconeogenesis in liver. CR hence decreases SIRT4 activity, which
is opposed to the induction of SIRT1 activity during
CR. Furthermore, SIRT4 and SIRT1 exert, respectively,
a negative and positive control on insulin secretion,
which is striking given that their activities are both
controlled by a single metabolite, i.e. NAD.

Yamamoto et al. Minireview

zyme required for both the de novo and salvage pathways of NAD biosynthesis (9092) (Fig. 3), and a
short region of a ubiquitin fusion degradation protein
2a. In a recent study, overexpression of Nmnat 1 alone
could prevent axonal degeneration, indicating that the
protective effect of Nmnat 1 could be mediated by an
increase of neuronal NAD reserve and/or SIRT1 activity (93).
It is well known that CR protects neurons from degeneration in mouse models of AD and Parkinsons
disease, and SIRT1 might facilitate neuronal survival
(9497). Although it is reported that caloric intake and
insulin sensitivity are linked to AD, the mechanism
underlying these connections are not fully clarified as
of yet (98, 99). The pathology of AD is characterized by
the presence of amyloid plaques, intracellular neurofibrillary tangles, and pronounced cell death (100). The
amyloid plaque is composed of amyloid- (A) peptide, which is cleaved from the amyloid precursor protein sequentially by -secretase and -secretase (101
103). This abnormal A peptide deposition within the
brain is the hallmark of AD neuropathology, and ac-

SIRTUINS IN NEURAL PROTECTION AND


NEURODEGENERATIVE DISEASES
Axonal degeneration is a major morphological characteristic observed in both peripheral neuropathies and
neurodegenerative diseases, such as Alzheimers disease (AD) and amyotrophic lateral sclerosis (84, 85).
Axonal degeneration usually occurs in the early stage
in degenerative processes and often precedes or correlates closely with clinical symptoms such as cognitive decline. There are several reports that support an
axonal protective role for SIRT1 in the neuronal system. A self-destructive degeneration process is observed at the distal portion of a transected axon, which
is called Wallerian degeneration (86). Wallerian degeneration slow (wlds) is a mouse line with delayed axonal
degeneration in response to axonal injury (8789). This
phenomenon is thought to be derived from overexpression of a chimeric nuclear molecule (Wlds protein)
that corresponds to the full-length nicotinamide
mononucleotide adenyltransferase 1 (Nmnat 1), an en-

Fig. 3. Superpathway of NAD Biosynthesis in Mammals


NAD is synthesized via two major pathways, the de novo
and salvage pathways, and these two pathways converge at
nicotinic acid mononucleotide. In the de novo pathway, the
nicotinic acid moiety of NAD is synthesized from tryptophan
via the kynurenine pathway. In the NAD salvage pathway,
NAD is generated through the recycling of its degradation
product such as nicotinamide. ART, ADP-ribosyl transferase;
NA, nicotinic acid; NAAD, nicotinic acid adenine dinucleotide;
NAM, nicotinamide; NaMN, nicotinic acid mononucleotide;
Nampt, nicotinamide phosphoribosyltransferase; NMN, nicotinamide mononucleotide; NPT, nicotinic acid phosphoribosyltransferase; NR, nicotinamide riboside; Nrk, nicotinamide
riboside kinase; PARP, poly-ADP-polymerases; PBEF, pre-B
cell colony-enhancing factor.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

Yamamoto et al. Minireview

cumulation of aggregated A is hypothesized to initiate a pathological cascade resulting in the onset and
progression of AD (104). A peptides can induce NFB
activity in microglia via TNF-receptor type 1 or receptor of advanced glycation end product (105, 106).
SIRT1 activation or the administration of the SIRT1
activator resveratrol markedly reduces this NFB signaling (107). This strongly suggests that SIRT1 can
attenuate A-stimulated neurotoxicity and AD-related
inflammatory responses via inhibition of microglial
NFB signaling. SIRT1 is furthermore supposed to
prevent A peptide generation through promotion of
the nonamyloidogenic processing of amyloid precursor protein by the inhibition of Rho kinase 1 expression
(108).
In addition to this inflammatory cascade leading to
neuronal cell death, another intrinsic cell death pathway, i.e. mitochondria-based cell death pathway, is
attracting attention in the context of AD. In fact, A
peptides, which can directly enter the mitochondrial
inner membrane, are able to bind to a mitochondrialmatrix protein termed A-binding alcohol dehydrogenase and localize to the mitochondria (109). This reduces ATP production and increases the generation of
oxygen radicals (110), which subsequently may induce
mitochondria-dependent cell death because damaged
mitochondria are unable to maintain the energy demands of the cells. Consistent with these observations, in AD mouse models, a strong association between A and the inner mitochondrial membrane
together with increased free-radical generation and
decreased cytochrome c oxidase activity has been
reported (111, 112). SIRT1 could, through its stimulating activity on mitochondria (73), contribute to this
process in AD.
Also in Huntingtons disease (HD), another neurodegenerative disease, mitochondrial insufficiency is
observed. HD patients are characterized by marked
reductions in glucose metabolism and increased levels of lactate in the basal ganglia, by a reduced activity
of several key components of the oxidative phosphorylation pathways in the mitochondria of the striatal
neurons (113) and by pronounced morphological abnormalities, including derangement of the mitochondrial matrix and cristae (114). These mitochondrial
dysfunctions are supposed to be associated with dysregulation of PGC-1 transcription and/or activity by
the mutant huntingtin protein (115117). Because recent reports show that PGC-1 activity is regulated by
SIRT1 (66, 73) and because some aspects of mitochondrial metabolism are controlled by some of the
sirtuins, the modulation of sirtuin activity could be an
interesting approach for the therapy of these neurodegenerative diseases. In fact, the potential of such a
strategy was validated in nematode HD models and in
mouse neuronal cell lines (118). In these models, the
expanded polyglutamine (PolyQ) track in HD-associated protein huntingtin (htt) were shown to induce
PolyQ-dependent neuronal dysfunction. This abnormality caused by the mutant PolyQ could be rescued

Mol Endocrinol, August 2007, 21(8):17451755 1751

by overexpression of SIRT1 or by resveratrol treatment


(118). This favorable effect was suppressed by sirtuin
inhibitors, such as nicotinamide or sirtinol, directly
proving that SIRT1 activation could be useful in HD.

PERSPECTIVES
The founding member of the sirtuin family, Sir2p in
yeast or SIRT1 in mammals, has now been well established as a key molecule that affects longevity
within the context of CR in several model organisms
ranging from yeast to mouse, although the mechanisms involved may be distinct in the different species.
The vital role that the sirtuins play in cellular metabolic
control indicated that they could be important determinants of whole-body metabolism and protect
against many chronic diseases associated with metabolic dysfunction. Likewise, potential applications of
the sirtuins in neuronal cell survival and response to
stress and cell-cycle control hint to eventual importance of this gene family in the pathogenesis of neurodegenerative diseases and cancer. Additional insight into the biological actions of the sirtuins will
require the definition of the exact roles of each of the
gene family members in vivo with appropriate genetic,
pharmacological, and physiological tools. Once this is
achieved, it is expected that a select member of the
sirtuins could become potential interesting targets for
future therapies against age-related diseases.
Acknowledgments
Received February 12, 2007. Accepted April 18, 2007.
Address all correspondence and requests for reprints to:
Johan Auwerx, M.D., Ph.D., Institut de Genetique et de Biologie Moleculaire et Cellulaire, 1 Rue Laurent Fries, Bote
Postale 10142, 67404 Illkirch, France. E-mail: auwerx@
igbmc.u-strasbg.fr.
Disclosure Statement: The authors have nothing to
disclose.

REFERENCES
1. Barger JL, Walford RL, Weindruch R 2003 The retardation of aging by caloric restriction: its significance in the
transgenic era. Exp Gerontol 38:13431351
2. Bordone L, Guarente L 2005 Calorie restriction, SIRT1
and metabolism: understanding longevity. Nat Rev Mol
Cell Biol 6:298305
3. Denu JM 2003 Linking chromatin function with metabolic networks: Sir2 family of NAD-dependent
deacetylases. Trends Biochem Sci 28:4148
4. Gasser SM, Cockell MM 2001 The molecular biology of
the SIR proteins. Gene 279:116
5. Imai S, Armstrong CM, Kaeberlein M, Guarente L 2000
Transcriptional silencing and longevity protein Sir2 is an
NAD-dependent histone deacetylase. Nature 403:
795800
6. Landry J, Sutton A, Tafrov ST, Heller RC, Stebbins J,
Pillus L, Sternglanz R 2000 The silencing protein SIR2

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

1752 Mol Endocrinol, August 2007, 21(8):17451755

7.

8.

9.

10.

11.

12.
13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

and its homologs are NAD-dependent protein deacetylases. Proc Natl Acad Sci USA 97:58075811
Smith JS, Brachmann CB, Celic I, Kenna MA, Muhammad S, Starai VJ, Avalos JL, Escalante-Semerena JC,
Grubmeyer C, Wolberger C, Boeke JD 2000 A phylogenetically conserved NAD-dependent protein deacetylase activity in the Sir2 protein family. Proc Natl Acad Sci
USA 97:66586663
Frye RA 1999 Characterization of five human cDNAs
with homology to the yeast SIR2 gene: Sir2-like proteins
(sirtuins) metabolize NAD and may have protein ADPribosyltransferase activity. Biochem Biophys Res Commun 260:273279
Frye RA 2000 Phylogenetic classification of prokaryotic
and eukaryotic Sir2-like proteins. Biochem Biophys Res
Commun 273:793798
Lamming DW, Latorre-Esteves M, Medvedik O, Wong
SN, Tsang FA, Wang C, Lin SJ, Sinclair DA 2005 HST2
mediates SIR2-independent life-span extension by calorie restriction. Science 309:18611864
Smith BC, Denu JM 2006 Sir2 protein deacetylases:
evidence for chemical intermediates and functions of a
conserved histidine. Biochemistry 45:272282
Blander G, Guarente L 2004 The Sir2 family of protein
deacetylases. Annu Rev Biochem 73:417435
Tanner KG, Landry J, Sternglanz R, Denu JM 2000
Silent information regulator 2 family of NAD-dependent
histone/protein deacetylases generates a unique product, 1-O-acetyl-ADP-ribose. Proc Natl Acad Sci USA
97:1417814182
Borra MT, Langer MR, Slama JT, Denu JM 2004 Substrate specificity and kinetic mechanism of the Sir2
family of NAD-dependent histone/protein deacetylases. Biochemistry 43:98779887
Anderson RM, Bitterman KJ, Wood JG, Medvedik O,
Sinclair DA 2003 Nicotinamide and PNC1 govern lifespan extension by calorie restriction in Saccharomyces
cerevisiae. Nature 423:181185
Lin SJ, Defossez PA, Guarente L 2000 Requirement of
NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science 289:
21262128
Lin SJ, Ford E, Haigis M, Liszt G, Guarente L 2004
Calorie restriction extends yeast life span by lowering
the level of NADH. Genes Dev 18:1216
Lin SJ, Kaeberlein M, Andalis AA, Sturtz LA, Defossez
PA, Culotta VC, Fink GR, Guarente L 2002 Calorie restriction extends Saccharomyces cerevisiae lifespan by
increasing respiration. Nature 418:344348
Revollo JR, Grimm AA, Imai S 2004 The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian
cells. J Biol Chem 279:5075450763
Liou GG, Tanny JC, Kruger RG, Walz T, Moazed D 2005
Assembly of the SIR complex and its regulation by
O-acetyl-ADP-ribose, a product of NAD-dependent histone deacetylation. Cell 121:515527
Vaquero A, Scher M, Lee D, Erdjument-Bromage H,
Tempst P, Reinberg D 2004 Human SirT1 interacts with
histone H1 and promotes formation of facultative heterochromatin. Mol Cell 16:93105
Kustatscher G, Hothorn M, Pugieux C, Scheffzek K,
Ladurner AG 2005 Splicing regulates NAD metabolite
binding to histone macroH2A. Nat Struct Mol Biol 12:
624625
Tanny JC, Dowd GJ, Huang J, Hilz H, Moazed D 1999
An enzymatic activity in the yeast Sir2 protein that is
essential for gene silencing. Cell 99:735745
Michishita E, Park JY, Burneskis JM, Barrett JC,
Horikawa I 2005 Evolutionarily conserved and nonconserved cellular localizations and functions of human
SIRT proteins. Mol Biol Cell 16:46234635

Yamamoto et al. Minireview

25. Mostoslavsky R, Chua KF, Lombard DB, Pang WW,


Fischer MR, Gellon L, Liu P, Mostoslavsky G, Franco S,
Murphy MM, Mills KD, Patel P, Hsu JT, Hong AL, Ford
E, Cheng HL, Kennedy C, Nunez N, Bronson R, Frendewey D, Auerbach W, Valenzuela D, Karow M, Hottiger
MO, Hursting S, Barrett JC, Guarente L, Mulligan R,
Demple B, Yancopoulos GD, Alt FW 2006 Genomic
instability and aging-like phenotype in the absence of
mammalian SIRT6. Cell 124:315329
26. Vaquero A, Scher MB, Lee DH, Sutton A, Cheng HL, Alt
FW, Serrano L, Sternglanz R, Reinberg D 2006 SirT2 is
a histone deacetylase with preference for histone H4
Lys 16 during mitosis. Genes Dev 20:12561261
27. Langley E, Pearson M, Faretta M, Bauer UM, Frye RA,
Minucci S, Pelicci PG, Kouzarides T 2002 Human SIR2
deacetylates p53 and antagonizes PML/p53-induced
cellular senescence. EMBO J 21:23832396
28. Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A,
Guarente L, Gu W 2001 Negative control of p53 by Sir2
promotes cell survival under stress. Cell 107:137148
29. 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 107:149159
30. Appella E, Anderson CW 2001 Post-translational modifications and activation of p53 by genotoxic stresses.
Eur J Biochem 268:27642772
31. Beumer TL, Roepers-Gajadien HL, Gademan IS, van
Buul PP, Gil-Gomez G, Rutgers DH, de Rooij DG 1998
The role of the tumor suppressor p53 in spermatogenesis. Cell Death Differ 5:669677
32. Brooks CL, Gu W 2003 Ubiquitination, phosphorylation
and acetylation: the molecular basis for p53 regulation.
Curr Opin Cell Biol 15:164171
33. Cheng HL, Mostoslavsky R, Saito S, Manis JP, Gu Y,
Patel P, Bronson R, Appella E, Alt FW, Chua KF 2003
Developmental defects and p53 hyperacetylation in Sir2
homolog (SIRT1)-deficient mice. Proc Natl Acad Sci
USA 100:1079410799
34. Pearson M, Carbone R, Sebastiani C, Cioce M, Fagioli
M, Saito S, Higashimoto Y, Appella E, Minucci S, Pandolfi PP, Pelicci PG 2000 PML regulates p53 acetylation
and premature senescence induced by oncogenic Ras.
Nature 406:207210
35. Chua KF, Mostoslavsky R, Lombard DB, Pang WW,
Saito S, Franco S, Kaushal D, Cheng HL, Fischer MR,
Stokes N, Murphy MM, Appella E, Alt FW 2005 Mammalian SIRT1 limits replicative life span in response to
chronic genotoxic stress. Cell Metab 2:6776
36. Kaeberlein M, McVey M, Guarente L 1999 The SIR2/3/4
complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms.
Genes Dev 13:25702580
37. Sinclair DA, Guarente L 1997 Extrachromosomal rDNA
circlesa cause of aging in yeast. Cell 91:10331042
38. Daitoku H, Hatta M, Matsuzaki H, Aratani S, Ohshima T,
Miyagishi M, Nakajima T, Fukamizu A 2004 Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity. Proc Natl
Acad Sci USA 101:1004210047
39. van der Horst A, Tertoolen LG, de Vries-Smits LM, Frye
RA, Medema RH, Burgering BM 2004 FOXO4 is acetylated upon peroxide stress and deacetylated by the
longevity protein hSir2 (SIRT1). J Biol Chem 279:
2887328879
40. Accili D, Arden KC 2004 FoxOs at the crossroads of
cellular metabolism, differentiation, and transformation.
Cell 117:421426
41. Kenyon C 2001 A conserved regulatory system for aging. Cell 105:165168
42. Lin K, Dorman JB, Rodan A, Kenyon C 1997 daf-16: an
HNF-3/forkhead family member that can function to

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

Yamamoto et al. Minireview

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

double the life-span of Caenorhabditis elegans. Science


278:13191322
Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L,
Tissenbaum HA, Ruvkun G 1997 The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature 389:
994999
Anderson KE, Coadwell J, Stephens LR, Hawkins PT
1998 Translocation of PDK-1 to the plasma membrane
is important in allowing PDK-1 to activate protein kinase
B. Curr Biol 8:684691
Stephens L, Anderson K, Stokoe D, Erdjument-Bromage H, Painter GF, Holmes AB, Gaffney PR, Reese
CB, McCormick F, Tempst P, Coadwell J, Hawkins PT
1998 Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of
protein kinase B. Science 279:710714
Brunet A, Kanai F, Stehn J, Xu J, Sarbassova D, Frangioni JV, Dalal SN, DeCaprio JA, Greenberg ME, Yaffe
MB 2002 14-3-3 transits to the nucleus and participates
in dynamic nucleocytoplasmic transport. J Cell Biol 156:
817828
Cahill CM, Tzivion G, Nasrin N, Ogg S, Dore J, Ruvkun
G, Alexander-Bridges M 2001 Phosphatidylinositol 3-kinase signaling inhibits DAF-16 DNA binding and function via 14-3-3-dependent and 14-3-3-independent
pathways. J Biol Chem 276:1340213410
Rena G, Prescott AR, Guo S, Cohen P, Unterman TG
2001 Roles of the forkhead in rhabdomyosarcoma
(FKHR) phosphorylation sites in regulating 14-3-3 binding, transactivation and nuclear targeting. Biochem J
354:605612
Tzivion G, Shen YH, Zhu J 2001 14-3-3 proteins; bringing new definitions to scaffolding. Oncogene 20:
63316338
Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL,
Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu
LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA,
Alt FW, Greenberg ME 2004 Stress-dependent regulation of FOXO transcription factors by the SIRT1
deacetylase. Science 303:20112015
Frescas D, Valenti L, Accili D 2005 Nuclear trapping of
the forkhead transcription factor FoxO1 via Sirt-dependent deacetylation promotes expression of glucogenetic genes. J Biol Chem 280:2058920595
Fulco M, Schiltz RL, Iezzi S, King MT, Zhao P, Kashiwaya Y, Hoffman E, Veech RL, Sartorelli V 2003 Sir2
regulates skeletal muscle differentiation as a potential
sensor of the redox state. Mol Cell 12:5162
Zhao X, Sternsdorf T, Bolger TA, Evans RM, Yao TP
2005 Regulation of MEF2 by histone deacetylase 4- and
SIRT1 deacetylase-mediated lysine modifications. Mol
Cell Biol 25:84568464
Fu M, Liu M, Sauve AA, Jiao X, Zhang X, Wu X, Powell
MJ, Yang T, Gu W, Avantaggiati ML, Pattabiraman N,
Pestell TG, Wang F, Quong AA, Wang C, Pestell RG
2006 Hormonal control of androgen receptor function
through SIRT1. Mol Cell Biol 26:81228135
Finnin MS, Donigian JR, Pavletich NP 2001 Structure of
the histone deacetylase SIRT2. Nat Struct Biol
8:621625
North BJ, Marshall BL, Borra MT, Denu JM, Verdin E
2003 The human Sir2 ortholog, SIRT2, is an NADdependent tubulin deacetylase. Mol Cell 11:437444
Dryden SC, Nahhas FA, Nowak JE, Goustin AS, Tainsky
MA 2003 Role for human SIRT2 NAD-dependent
deacetylase activity in control of mitotic exit in the cell
cycle. Mol Cell Biol 23:31733185
Hiratsuka M, Inoue T, Toda T, Kimura N, Shirayoshi Y,
Kamitani H, Watanabe T, Ohama E, Tahimic CG, Kurimasa A, Oshimura M 2003 Proteomics-based identification of differentially expressed genes in human

Mol Endocrinol, August 2007, 21(8):17451755 1753

59.

60.

61.

62.

63.

64.
65.

66.

67.

68.

69.

70.

71.
72.

73.

74.

gliomas: down-regulation of SIRT2 gene. Biochem Biophys Res Commun 309:558566


Sobol RW, Horton JK, Kuhn R, Gu H, Singhal RK,
Prasad R, Rajewsky K, Wilson SH 1996 Requirement of
mammalian DNA polymerase- in base-excision repair.
Nature 379:183186
Sobol RW, Prasad R, Evenski A, Baker A, Yang XP,
Horton JK, Wilson SH 2000 The lyase activity of the
DNA repair protein -polymerase protects from DNAdamage-induced cytotoxicity. Nature 405:807810
Ford E, Voit R, Liszt G, Magin C, Grummt I, Guarente L
2006 Mammalian Sir2 homolog SIRT7 is an activator of
RNA polymerase I transcription. Genes Dev 20:
10751080
de Nigris F, Cerutti J, Morelli C, Califano D, Chiariotti L,
Viglietto G, Santelli G, Fusco A 2002 Isolation of a
SIR-like gene, SIR-T8, that is overexpressed in thyroid
carcinoma cell lines and tissues. Br J Cancer 86:
917923
Ashraf N, Zino S, Macintyre A, Kingsmore D, Payne AP,
George WD, Shiels PG 2006 Altered sirtuin expression
is associated with node-positive breast cancer. Br J
Cancer 95:10561061
Hallows WC, Lee S, Denu JM 2006 Sirtuins deacetylate
and activate mammalian acetyl-CoA synthetases. Proc
Natl Acad Sci USA 103:1023010235
Nemoto S, Fergusson MM, Finkel T 2005 SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1. J Biol Chem 280:
1645616460
Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM,
Puigserver P 2005 Nutrient control of glucose homeostasis through a complex of PGC-1 and SIRT1.
Nature 434:113118
Schwer B, Bunkenborg J, Verdin RO, Andersen JS,
Verdin E 2006 Reversible lysine acetylation controls the
activity of the mitochondrial enzyme acetyl-CoA synthetase 2. Proc Natl Acad Sci USA 103:1022410229
McBurney MW, Yang X, Jardine K, Hixon M, Boekelheide K, Webb JR, Lansdorp PM, Lemieux M 2003 The
mammalian SIR2 protein has a role in embryogenesis
and gametogenesis. Mol Cell Biol 23:3854
Bordone L, Motta MC, Picard F, Robinson A, Jhala US,
Apfeld J, McDonagh T, Lemieux M, McBurney M, Szilvasi A, Easlon EJ, Lin SJ, Guarente L 2006 Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic
beta cells. PLoS Biol 4:e31
Moynihan KA, Grimm AA, Plueger MM, Bernal-Mizrachi
E, Ford E, Cras-Meneur C, Permutt MA, Imai S 2005
Increased dosage of mammalian Sir2 in pancreatic
cells enhances glucose-stimulated insulin secretion in
mice. Cell Metab 2:105117
Picard F, Auwerx J 2002 PPAR and glucose homeostasis. Annu Rev Nutr 22:167197
Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, Machado De Oliveira R, Leid M, McBurney
MW, Guarente L 2004 Sirt1 promotes fat mobilization in
white adipocytes by repressing PPAR-. Nature 429:
771776
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-1. Cell 127:11091122
Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C,
Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K,
Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D,
Wang M, Ramaswamy S, Fishbein KW, Spencer RG,
Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA 2006
Resveratrol improves health and survival of mice on a
high-calorie diet. Nature 444:337342

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

1754 Mol Endocrinol, August 2007, 21(8):17451755

75. 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 425:191196
76. 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:
686689
77. Onyango P, Celic I, McCaffery JM, Boeke JD, Feinberg
AP 2002 SIRT3, a human SIR2 homologue, is an NADdependent deacetylase localized to mitochondria. Proc
Natl Acad Sci USA 99:1365313658
78. Schwer B, North BJ, Frye RA, Ott M, Verdin E 2002 The
human silent information regulator (Sir)2 homologue
hSIRT3 is a mitochondrial nicotinamide adenine dinucleotide-dependent deacetylase. J Cell Biol 158:
647657
79. Shi T, Wang F, Stieren E, Tong Q 2005 SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial
function and thermogenesis in brown adipocytes. J Biol
Chem 280:1356013567
80. Fujino T, Kondo J, Ishikawa M, Morikawa K, Yamamoto
TT 2001 Acetyl-CoA synthetase 2, a mitochondrial matrix enzyme involved in the oxidation of acetate. J Biol
Chem 276:1142011426
81. Bellizzi D, Rose G, Cavalcante P, Covello G, Dato S, De
Rango F, Greco V, Maggiolini M, Feraco E, Mari V,
Franceschi C, Passarino G, De Benedictis G 2005 A
novel VNTR enhancer within the SIRT3 gene, a human
homologue of SIR2, is associated with survival at oldest
ages. Genomics 85:258263
82. Haigis MC, Mostoslavsky R, Haigis KM, Fahie K,
Christodoulou DC, Murphy AJ, Valenzuela DM, Yancopoulos GD, Karow M, Blander G, Wolberger C, Prolla
TA, Weindruch R, Alt FW, Guarente L 2006 SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic cells. Cell
126:941954
83. Kelly A, Stanley CA 2001 Disorders of glutamate metabolism. Ment Retard Dev Disabil Res Rev 7:287295
84. Fischer LR, Culver DG, Tennant P, Davis AA, Wang M,
Castellano-Sanchez A, Khan J, Polak MA, Glass JD
2004 Amyotrophic lateral sclerosis is a distal
axonopathy: evidence in mice and man. Exp Neurol
185:232240
85. Stokin GB, Lillo C, Falzone TL, Brusch RG, Rockenstein
E, Mount SL, Raman R, Davies P, Masliah E, Williams
DS, Goldstein LS 2005 Axonopathy and transport deficits early in the pathogenesis of Alzheimers disease.
Science 307:12821288
86. Raff MC, Whitmore AV, Finn JT 2002 Axonal self-destruction and neurodegeneration. Science 296:868871
87. Conforti L, Tarlton A, Mack TG, Mi W, Buckmaster EA,
Wagner D, Perry VH, Coleman MP 2000 A Ufd2/
D4Cole1e chimeric protein and overexpression of Rbp7
in the slow Wallerian degeneration (WldS) mouse. Proc
Natl Acad Sci USA 97:1137711382
88. Lunn ER, Perry VH, Brown MC, Rosen H, Gordon S
1989 Absence of Wallerian degeneration does not
hinder regeneration in peripheral nerve. Eur J Neurosci
1:2733
89. Mack TG, Reiner M, Beirowski B, Mi W, Emanuelli M,
Wagner D, Thomson D, Gillingwater T, Court F, Conforti
L, Fernando FS, Tarlton A, Andressen C, Addicks K,
Magni G, Ribchester RR, Perry VH, Coleman MP 2001
Wallerian degeneration of injured axons and synapses
is delayed by a Ube4b/Nmnat chimeric gene. Nat Neurosci 4:11991206
90. Bieganowski P, Brenner C 2004 Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes
establish a Preiss-Handler independent route to NAD
in fungi and humans. Cell 117:495502

Yamamoto et al. Minireview

91. Llorente B, Dujon B 2000 Transcriptional regulation of


the Saccharomyces cerevisiae DAL5 gene family and
identification of the high affinity nicotinic acid permease
TNA1 (YGR260w). FEBS Lett 475:237241
92. Grubisha O, Smith BC, Denu JM 2005 Small molecule
regulation of Sir2 protein deacetylases. FEBS J 272:
46074616
93. Araki T, Sasaki Y, Milbrandt J 2004 Increased nuclear
NAD biosynthesis and SIRT1 activation prevent axonal
degeneration. Science 305:10101013
94. Luchsinger JA, Tang MX, Shea S, Mayeux R 2002 Caloric intake and the risk of Alzheimer disease. Arch
Neurol 59:12581263
95. Maswood N, Young J, Tilmont E, Zhang Z, Gash DM,
Gerhardt GA, Grondin R, Roth GS, Mattison J, Lane MA,
Carson RE, Cohen RM, Mouton PR, Quigley C, Mattson
MP, Ingram DK 2004 Caloric restriction increases neurotrophic factor levels and attenuates neurochemical
and behavioral deficits in a primate model of Parkinsons disease. Proc Natl Acad Sci USA 101:
1817118176
96. Patel NV, Gordon MN, Connor KE, Good RA, Engelman
RW, Mason J, Morgan DG, Morgan TE, Finch CE 2005
Caloric restriction attenuates A-deposition in Alzheimer transgenic models. Neurobiol Aging 26:9951000
97. Wang J, Ho L, Qin W, Rocher AB, Seror I, Humala N,
Maniar K, Dolios G, Wang R, Hof PR, Pasinetti GM 2005
Caloric restriction attenuates -amyloid neuropathology in a mouse model of Alzheimers disease. FASEB J
19:659661
98. Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, Peng
Y, Cambareri G, Rocher A, Mobbs CV, Hof PR, Pasinetti
GM 2004 Diet-induced insulin resistance promotes
amyloidosis in a transgenic mouse model of Alzheimers
disease. FASEB J 18:902904
99. Rasgon N, Jarvik L 2004 Insulin resistance, affective
disorders, and Alzheimers disease: review and hypothesis. J Gerontol A Biol Sci Med Sci 59:178183; discussion 184192
100. Hardy JA, Higgins GA 1992 Alzheimers disease: the
amyloid cascade hypothesis. Science 256:184185
101. Haass C 2004 Take fiveBACE and the -secretase
quartet conduct Alzheimers amyloid -peptide generation. EMBO J 23:483488
102. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K 1985 Amyloid plaque core
protein in Alzheimer disease and Down syndrome. Proc
Natl Acad Sci USA 82:42454249
103. Selkoe DJ 2001 Alzheimers disease: genes, proteins,
and therapy. Physiol Rev 81:741766
104. Selkoe DJ 1996 Amyloid -protein and the genetics of
Alzheimers disease. J Biol Chem 271:1829518298
105. Li R, Yang L, Lindholm K, Konishi Y, Yue X, Hampel H,
Zhang D, Shen Y 2004 Tumor necrosis factor death
receptor signaling cascade is required for amyloid-
protein-induced neuron death. J Neurosci 24:
17601771
106. Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery
T, Zhao L, Nagashima M, Morser J, Migheli A, Nawroth
P, Stern D, Schmidt AM 1996 RAGE and amyloid-
peptide neurotoxicity in Alzheimers disease. Nature
382:685691
107. Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi
S, Mucke L, Gan L 2005 SIRT1 protects against microglia-dependent amyloid- toxicity through inhibiting
NF-B signaling. J Biol Chem 280:4036440374
108. Qin W, Yang T, Ho L, Zhao Z, Wang J, Chen L, Zhao W,
Thiyagarajan M, MacGrogan D, Rodgers JT, Puigserver
P, Sadoshima J, Deng H, Pedrini S, Gandy S, Sauve AA,
Pasinetti GM 2006 Neuronal SIRT1 activation as a novel
mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction.
J Biol Chem 281:2174521754

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

Yamamoto et al. Minireview

Mol Endocrinol, August 2007, 21(8):17451755 1755

109. Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang


N, Caspersen C, Chen X, Pollak S, Chaney M, Trinchese
F, Liu S, Gunn-Moore F, Lue LF, Walker DG, Kuppusamy P, Zewier ZL, Arancio O, Stern D, Yan SS, Wu
H 2004 ABAD directly links A to mitochondrial toxicity
in Alzheimers disease. Science 304:448452
110. McLellan ME, Kajdasz ST, Hyman BT, Bacskai BJ 2003
In vivo imaging of reactive oxygen species specifically
associated with thioflavin S-positive amyloid plaques
by multiphoton microscopy. J Neurosci 23:22122217
111. Crouch PJ, Blake R, Duce JA, Ciccotosto GD, Li QX,
Barnham KJ, Curtain CC, Cherny RA, Cappai R, Dyrks
T, Masters CL, Trounce IA 2005 Copper-dependent
inhibition of human cytochrome c oxidase by a dimeric
conformer of amyloid-142. J Neurosci 25:672679
112. Manczak M, Anekonda TS, Henson E, Park BS, Quinn J,
Reddy PH 2006 Mitochondria are a direct site of A
accumulation in Alzheimers disease neurons: implications for free radical generation and oxidative damage
in disease progression. Hum Mol Genet 15:14371449
113. Browne SE, Beal MF 2004 The energetics of Huntingtons disease. Neurochem Res 29:531546
114. Squitieri F, Cannella M, Sgarbi G, Maglione V, Falleni A,
Lenzi P, Baracca A, Cislaghi G, Saft C, Ragona G,
Russo MA, Thompson LM, Solaini G, Fornai F 2006
Severe ultrastructural mitochondrial changes in lym-

phoblasts homozygous for Huntington disease mutation. Mech Ageing Dev 127:217220
Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N,
Krainc D 2006 Transcriptional repression of PGC-1 by
mutant huntingtin leads to mitochondrial dysfunction
and neurodegeneration. Cell 127:5969
St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager 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 127:397408
Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield
TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler
TK, Strand AD, Cui L, Beyer RP, Easley CN, Smith AC,
Krainc D, Luquet S, Sweet IR, Schwartz MW, La Spada
AR 2006 Thermoregulatory and metabolic defects in
Huntingtons disease transgenic mice implicate
PGC-1 in Huntingtons disease neurodegeneration.
Cell Metab 4:349362
Parker JA, Arango M, Abderrahmane S, Lambert E,
Tourette C, Catoire H, Neri C 2005 Resveratrol rescues
mutant polyglutamine cytotoxicity in nematode and
mammalian neurons. Nat Genet 37:349350
Scher MB, Vaquero A, Reinberg D 2007 SirT3 is a
nuclear NAD-dependent histone deacetylase that
translocates to the mitochondria upon cellular stress.
Genes Dev 21:920928

115.

116.

117.

118.

119.

Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost
professional society serving the endocrine community.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 August 2014. at 14:59 For personal use only. No other uses without permission. . All rights reserved.

You might also like