You are on page 1of 12

Annals of Medicine

ISSN: 0785-3890 (Print) 1365-2060 (Online) Journal homepage: www.tandfonline.com/journals/iann20

Sirtuins: The ‘magnificent seven’, function,


metabolism and longevity

Nassim Dali‐Youcef, Marie Lagouge, Sébastien Froelich, Christian Koehl,


Kristina Schoonjans & Johan Auwerx

To cite this article: Nassim Dali‐Youcef, Marie Lagouge, Sébastien Froelich, Christian
Koehl, Kristina Schoonjans & Johan Auwerx (2007) Sirtuins: The ‘magnificent seven’,
function, metabolism and longevity, Annals of Medicine, 39:5, 335-345, DOI:
10.1080/07853890701408194

To link to this article: https://doi.org/10.1080/07853890701408194

Published online: 08 Jul 2009.

Submit your article to this journal

Article views: 5289

View related articles

Citing articles: 24 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iann20
Annals of Medicine. 2007; 39: 335–345

REVIEW ARTICLE

Sirtuins: The ‘magnificent seven’, function, metabolism and longevity

NASSIM DALI-YOUCEF1,3, MARIE LAGOUGE1, SÉBASTIEN FROELICH1,4,


CHRISTIAN KOEHL3, KRISTINA SCHOONJANS1 & JOHAN AUWERX1,2,3
1
Institut de Génétique et de Biologie Moléculaire et Cellulaire de Strasbourg (IGBMC), INSERM/CNRS/ULP, Illkirch,
France, 2Institut Clinique de la Souris, Genopole, Illkirch, France, 3Laboratoire de Biochimie Générale et Spécialisée,
Hôpitaux Universitaires de Strasbourg, Strasbourg, France, and 4Service de Neurochirurgie, Hôpital de Hautepierre, C.H.U.
de Strasbourg, Strasbourg, France

Abstract
The sirtuin family of histone deacetylases (HDACs) was named after their homology to the Saccharomyces cerevisiae gene
silent information regulator 2 (Sir2). In the yeast, Sir2 has been shown to mediate the effects of calorie restriction on the
extension of life span and high levels of Sir2 activity promote longevity. Like their yeast homologs, the mammalian sirtuins
(SIRT1-7) are class III HDACs and require NAD+ as a cofactor to deacetylate substrates ranging from histones to
transcriptional regulators. Through this activity, sirtuins are shown to regulate important biological processes ranging from
apoptosis, adipocyte and muscle differentiation, and energy expenditure to gluconeogenesis. We review here the current
knowledge regarding the role of sirtuins in metabolism, longevity, and discuss the possible therapeutic applications that
could result from the understanding of their function in different organs and pathologies.

Key words: Calorie restriction, longevity, metabolism, SIRT

Introduction yeast from gene silencing, DNA repair, progression


of the cell cycle, to the control of ageing.
Histone acetylation is the main type of covalent The Sir2 protein first deacetylates histones and
histone modification and is achieved by a class of then localizes with the yeast protein Sir4, to form a
enzymes termed histone acetyltransferases (HATs) tight silencing complex binding to the telomeres (6).
(1). HATs acetylate histones on lysines, whereas The localization of Sir2 complex to the histone tails,
histone deacetylation involves another family of initially identified to induce transcriptional repres-
enzymes, the histone deacetylases (HDACs) ((2), a sion of the silent mating type loci HML and HMR
review). These HDACs are classified in three groups (homothallic mating-type loci left and right, respec-
on the basis of their homology with the yeast tively) (7,8), produces chromatin silencing, and a
Saccharomyces cerevisiae HDACs: RPD3 (group I), recent study reported that the Sir2 complex reduces
HDA1 (group II), and Sir2 (group III). Class I and the promoter occupancy by the transcription factors
II enzymes are inhibited by trichostatin (TSA), IIB and IIE (TFIIB, TFIIE) and the RNA poly-
whereas class III HDACs are not inhibited by TSA merase II (Pol II) preventing hence the proper
and are NAD+-dependent (3,4), suggesting a reg- assembly of the preinitiation complex of the tran-
ulation of Sir2 molecules by the metabolic state of scriptional machinery (9). Most interesting was the
the cells (reviewed in (5)). Since the Sir2 family of fact that overexpression of the gene encoding the
proteins are also able to exert their enzymatic activity Sir2 protein leads to a decrease in histone acetylation
not only on histones but also on numerous other (10) and an increase in life span in yeast (11), in the
proteins, such as the transcriptional regulators, they nematode Caenorhabditis elegans (12) and in metazo-
are involved in many cellular processes, ranging in ans (13). Likewise, Sir2-activating compounds

Correspondence: Johan Auwerx, Institut de Génétique et de Biologie Moléculaire et Cellulaire de Strasbourg (IGBMC), INSERM/CNRS/ULP, 1 rue
Laurent Fries, BP 10142, 67404 Illkirch, France. E-mail: auwerx@igbmc.u-strasbg.fr
ISSN 0785-3890 print/ISSN 1365-2060 online # 2007 Taylor & Francis
DOI: 10.1080/07853890701408194
336 N. Dali-Youcef et al.

CR could affect metabolism and longevity in


Abbreviations humans. To better understand the mechanisms by
SIR2 silent information regulator 2 which CR extends life span, it is instructive to take a
HAT histone acetyltransferase close look at studies carried out in various species.
HDAC histone deacetylase
CR calorie restriction
PKB/AKT protein kinase B Studies based on non-mammalian models
GDP guanosine diphosphate Studies in Saccharomyces cerevisiae
GTP guanosine triphosphate
NAD+ nicotinamide adenine In yeast, reduction in glucose levels in the media,
dinucleotide mimicking CR, results in a substantial extension in
NADH reduced form of NAD+ life span, which is Sir2p- and NAD+-dependent,
Pol II RNA polymerase II since mutants for Sir2p and nicotinate phosphor-
TFIIB transcription factor IIB ibosyl transferase (NPT1), an enzyme required for
TFIIE transcription factor IIE NAD+ formation, failed to reproduce this effect
Daf-16 ‘dauer’ larvae transcription (19). In the same study, mutants for components of
factor-16 the glucose-signaling pathway, such as the GTP-
FOXO forkhead box subgroup ‘O’ GDP (guanosine triphosphate-guanosine dipho-
transcription factor sphate) exchange factor CDC25, the glucose sensing
HML homothallic mating-type loci left receptors gpr1 and gpa2, and hexokinase, the first
HMR homothallic mating-type loci enzyme in the glycolytic pathway, mimicked the CR-
right mediated increase in longevity. This life span
NFkB nuclear factor kappa B extension due to CR was attributed to an increase
transcription factor in respiration (20). Indeed a mutation in the gene
PGC-1a peroxisome proliferator-activated encoding the cytochrome c1 CYT1 is no longer able
receptor gamma (PPARc) to promote CR-mediated longevity, suggesting that
coactivator-1a blocking the mitochondrial electron transport chain
and respiration prevents life span extension.
Moreover, overexpression of Hap4 in yeast, a gene
known to cause a switch from fermentation to
(STACs), such as resveratrol, present in the skin of respiration, yielded a 35% extension in life span
red grapes, promote longevity in yeast (14) and other under normal glucose conditions. It is well estab-
organisms ranging from the worm and drosophila lished that respiration increases the NAD+/NADH
(15) to the mouse (16). On the other hand, both ratio through oxidation of NADH by the NADH
mutations of the Sir2 gene and pharmacological dehydrogenase (21).
inhibition of Sir2 protein by nicotinamide induces an The Sir2p-mediated conversion of NAD+ leads to
acceleration of ageing in yeast (17). Although the the formation of nicotinamide (NAM), a powerful
functions of Sir2 have been relatively well estab- Sir2/SIRT1 inhibitor (17) and O-acetyl-ADP-ribose
lished in yeast and C. elegans, their function in (O-AA-ribose) (22) (Figure 2A). Recent evidence
mammals remains rather elusive. This review will demonstrated an active role of O-AA-ribose in the
give an overview of the role of the sirtuin family of regulation of gene silencing by the Sir2/3/4 assembly
proteins in different species and their potential complex (23). In yeast, an alternative route to
contribution to disease. synthesize NAD+, other than its de novo production
from the amino acid tryptophan, and which starts
from nicotinic acid (NA), is present under the form
SIRT1/Sir2, caloric restriction, metabolism, of the NAD+ salvage pathway. In the NAD+ salvage
neurodegeneration, and longevity
pathway, NAM obtained from NAD+ cleavage is
In the last few years, an increasing number of studies deaminated to NA by the nicotinamidase PNC1.
from S. cerevisiae, C. elegans, Drosophila melanogaster, NA formed through the actions of PNC1 then
and mouse models have linked caloric restriction undergoes a series of reactions giving successively
(CR) and metabolism with longevity. The matter rise to nicotinate mononucleotide (NAMN), nicoti-
has been extensively reviewed by Bordone and nate adenine dinucleotide (NAAD), and NAD+, that
Guarente (18); in this section we give an overview are catalyzed by the enzymes nicotinate phosphor-
of how SIRT1, the mammalian Sir2 homolog, which ibosyl transferase (NPT1), nicotinate mononucleo-
is the best characterized sirtuin family member, and tide adenyltransferases (NMAT1/2), and NAD+
Sirtuins and metabolism 337

Figure 1. Sir2/SIRT1 and signaling pathways in different species. In yeast Sir2p activity is increased through increased expression of the
NAD+ salvage pathway enzyme, nicotinamidase PNC1, but also by increasing the NAD+/NADH ratio (or lowering NADH levels) in
response to calorie restriction (CR). Mutation of the glucose sensing receptors Gpr1 and Gpa2 mimics the CR effects on Sir2p, which
inhibit the formation of the deleterious extrachromosomal circular DNA repeats (ERCs) and contribute to life span extension in yeast. The
glucose and nutrients fermentation pathway activates the AKT-related kinase Sch9 which induces oxidative stress and participates in yeast
ageing. Inactivation of this pathway promotes longevity in yeast. The longevity pathway is conserved amongst worms, flies and mammals.
Activation of the insulin growth factor receptor IGFR (Daf-2/dIGFR) activates the insulin receptor substrate IRS (p65/CHICO), which
stimulates phosphoinositol-3 kinase PI3K (AGE-1/dPI3K), which in turn phosphorylates the PKB/AKT kinase. AKT phosphorylates and
inactivates FOXO (Daf-16/dFOXO). Inactivation of the insulin receptor pathway promotes longevity in worms, drosophila, and mammals
through increased activity of FOXO. Sir2/SIRT1 activation induces life span extension through interaction with Daf-16/FOXO factors.
Gpr-15G-protein coupled receptor-1; Gpa-25G-alpha protein-2; FGM5Fermentable growth medium; PNC15pyrazinamidase/
Nicotinamidase 1; Sir2p5yeast silent information regulator 2; NADH5reduced form of nicotinamide adenine dinucleotide;
Sch95Saccharomyces cerevisiae protein kinase 9; ERCs5extrachromosomal circular DNA repeats; Daf-165‘dauer’ larvae transcription
factor-16; AGE-15worm homolog of the phosphoinositol-3 kinase; AKT5protein kinase B; Rpd35reduced potassium dependence 3 (class
II histone deacetylase); IGF-15insulin growth factor-1; FOXO5forkhead box subgroup ‘O’ transcription factor; IRS5insulin receptor
substrate; CHICO5 drosophila homolog of IRS; SIRT15sirtuin 1.

synthetase (QNS1), respectively (24). The newly CR increased the expression of PNC1 and hence
recycled NAD+ then serves again as a cofactor for PNC1 activity, which is required and sufficient to
Sir2p-mediated deacetylation (Figure 2A). extend life span in yeast through Sir2p activation.
Multiple protagonists of the NAD+ salvage path- Conversely, mutations in the yeast PNC1 gene
way participate in life span extension in yeast. accelerate cellular ageing (25). These observations
Furthermore, an intense debate about whether hence suggested that NAM depletion, through the
Sir2p-dependent CR life span extension could be activation of PNC1, is sufficient to activate Sir2 and
due to a depletion in the noncompetitive Sir2p increase longevity in yeast. Moreover, mutation of
inhibitor NAM, rather than a modification in the PNC1 and overexpression of Nnt1, a NAM methyl-
NAD+/NADH ratio, is still ongoing. Interestingly, transferase which reduced the excess NAM levels,
338 N. Dali-Youcef et al.

Figure 2. A: yeast and mammalian NAD+ salvage pathways. Yeast (red) Sir2p utilizes the cofactor NAD+ to deacetylate proteins and in this
reaction produces nicotinamide (NAM) and O-acetyl-ADP-ribose (O-AA-ribose). NAM is deaminated by the nicotinamidase PNC1 to
form nicotinic acid (NA). NA will give rise successively to nicotinate mononucleotide (NAMN), nicotinate adenine dinucleotide (NAAD)
and nicotinamide adenine dinucleotide (NAD+) by the enzymes nicotinate phosphoribosyl-transferase (NPT1), nicotinate mononucleotide
adenyltransferase (NMAT), and nicotinamide adenine dinucleotide (NAD) synthetase (QNS), respectively (red arrows). In mammals
(blue) NAM is recycled to NAD+ in two steps through the formation of nicotinamide mononucleotide (NMN) by means of the NAM
phosphoribosyltransferase NamPT (PBEF/visfatin) and nicotinamide mononucleotide adenyltransferase (NMNAT) (blue arrows). B:
SIRT1 protective functions in metabolism and diseases. SIRT1 can be regulated positively by CR and SIRT-activating compounds and
negatively by SIRT inhibitors. SIRT1 activation induces survival of cardiomyocytes, protects neurons from cell death, and favors insulin
secretion by repressing the uncoupling protein 2 (UCP2). SIRT1 decreases white adipocyte tissue formation through repression of PPARc
and promotes gluconeogenesis in response to fasting through PGC-1a, and stimulates mitochondrial biogenesis in the brown adipose tissue
(BAT) and the muscle through activation of PGC-1a. NA5nicotinic acid; NAD+5nicotinamide adenine dinucleotide; NAM5
nicotinamide; NMN5nicotinamide mononucleotide; PNC15 pyrazinamidase/Nicotinamidase 1; NamPT15nicotinamide phosphoribosyl
transferase 1; PBEF5pre-B cell enhancing factor; NPT15nicotinate phosphoribosyl transferase 1; NMAT1/25nicotinate mononucleotide
adenyltransferase 1/2; QNS5NAD synthetase; NMNAT15nicotinamide mononucleotide adenyltransferase; O-AA-ribose5O-acetyl-
ADP-ribose; UCP-25uncoupling protein 2; NFkB5nuclear factor kappa B; PPARc5peroxisome proliferator-activated receptor gamma;
PGC-1a5peroxisome proliferator-activated receptor gamma coactivator-1 alpha; BAT5brown adipose tissue; SIRT15sirtuin 1.

restored the CR-induced life span extension in decreased NADH levels without changing NAD+
PNC1 mutants, suggesting that other factors (such levels, but still increased yeast life span both under
as NADH lowering) might play a role in CR- normal glucose culture conditions (2%) and CR
mediated life span extension (26). Other compo- conditions (0.5%), suggesting that reducing the
nents of the NAD+ salvage pathway, like NPT1, the quantity of the competitive Sir2p inhibitor NADH
rate-limiting enzyme in the NAD+ biosynthesis, have might also contribute to the CR-induced increase in
also been shown to regulate life span in yeast. yeast longevity (26). Interestingly, Hst2, a Sir2
Increased dosage of NPT1, but not of NMAT, homolog that contributes to rDNA stability, is
enhanced the total cellular NAD+ levels and another mediator of CR-induced life span extension
enhanced the transcriptional activity of the catalytic in yeast, indicating that other Sir2 homologs could
domain of Sir2p thereby extending yeast life span regulate longevity under CR conditions (29). Taken
(27), whereas NPT1 mutations yielded a defect in together, these data suggest that Sir2 requires NAD+
silencing at silent mating-type loci, telomeres, and to deacetylate proteins, and manipulations of the
rDNA (28). Overexpression of two related mito- salvage pathway that converge on enhanced NAD+
chondrial NADH dehydrogenases, Nde1 and Nde2, availability induce Sir2-dependent life span
Sirtuins and metabolism 339

extension in the budding yeast (24). Under CR other larvae before reproductive maturation (reviewed in
pathways, such as a decrease in the competitive Sir2 (37)). Activation of Daf-2/AGE-1 signaling results in
inhibitor NADH (by increasing Nde activity) or the the phosphorylation of AKT kinase that sequesters
noncompetitive Sir2 inhibitor NAM (by increasing Daf-16 in the cytoplasm resulting in its inactivation
PNC1 activity), could contribute to control Sir2 (38,39). The long-lived mutants require the entry of
activity. Furthermore, a Sir2-independent CR- Daf-16 into the nucleus to activate target genes
mediated life span extension mechanism exists that necessary for dauer formation. Duplication of
is mediated via Hst2. chromosomal regions containing Sir2.1 in the
Three important factors are thought to participate nematode extends life span by up to 50%.
in Sir2-mediated longevity. First, one of the con- Interestingly, the Sir2.1-duplicated strains carrying
sequences of increased Sir2 activity is the significant a mutated Daf-16 displayed the same decreased life
reduction in the number of extrachromosomal span as observed in Daf-16 mutants, proving that
circular DNA repeats (ERCs) (11), whose accumu- Sir2.1 acts upstream of Daf-16. In addition, Sir2.1-
lation is deleterious to yeast and accelerates ageing duplicated strains do not further extend life span of
(30), probably through sequestration of transcrip- Daf-2 mutants indicating that extra copies of Sir2.1
tion factors essential for their replication. The promote longevity through the Daf-2/Daf-16 signal-
second mechanism that contributes to the yeast life ing pathway (12). A direct interaction between
span acts via the fermentable growth medium- Sir2.1 and Daf-16, facilitated by the nematode 14-
induced (FGM) pathway. Under fermentation con- 3-3 protein, furthermore suggests that these three
ditions, glucose and other nutrients activate a yet proteins physically interact (40,41).
unknown pathway that activates Sch9, a kinase Also dSir2, the drosophila Sir2 ortholog, controls
related to the mammalian protein kinase B (PKB)/ life span extension under CR conditions, and
AKT, a negative regulator of the stress resistance mutants that remove or decrease dSir2 levels are
genes. This results in the subsequent accumulation no longer able to promote the CR-mediated long-
of reactive oxygen species (ROS), thereby accelerat- evity. The dSir2-mediated life extension seems to
ing ageing (31). Interestingly, mutation of Sch9 work in the same pathway as the Rpd3 histone
yields a substantial increase in yeast life span deacetylase, since Rpd3 is thought to negatively
probably through increasing stress resistance regulate dSir2, and CR induces a decrease in Rpd3
(32,33) (Figure 1). It is at present not known expression (13). Moreover, long-lived Rpd3 mutant
whether this pathway is similar to Daf-16/FOXO flies display high dSir2 expression levels (42).
signaling, which participates in the control of ageing Interestingly, flies deficient in the homolog of the
in other organisms, and whether Sch9 mutations mammalian insulin receptor substrate (IRS) protein,
affect ERCs formation. Finally, yeast evolving in a CHICO, show a significant increase in life span
high osmolarity live much longer than those in a (43). Recent studies showed that activation of
balanced osmolarity medium. A high osmolarity dFOXO in the fly brain and/or fat body extends life
shock activates a subset of osmotic responsive genes span and inhibits the endogenous insulin-dependent
that change the metabolism to favor glycerol signaling in the fat body (44,45). Although both CR
biosynthesis thereby generating more NAD+, the and downregulation of the insulin pathway partici-
Sir2 cofactor necessary to mediate longevity (34). pate in life span extension, it is still not proven that
the two pathways work in concert and converge on
dFOXO to promote longevity in drosophila.
Studies in Caenorhabditis elegans and Drosophila
melanogaster
Studies in mammals
In C. elegans, the yeast Sir2p ortholog, Sir2.1,
extends life span through the forkhead transcription The mammalian NAD+ salvage pathway is different
factor Daf-16 (homolog of mammalian FOXO) from the yeast pathway. NAM is directly trans-
signaling pathway. Loss of function studies of formed to nicotinamide mononucleotide (NMN) by
components of the Daf-2 (homolog of the mamma- the enzyme nicotinamide phosphoribosyltransferase
lian insulin receptor) signaling pathway, such as in (NamPT) (46), which then yields NAD+ through
Daf-2 or the homolog of the mammalian phospha- the action of nicotinamide mononucleotide adenyl
tidyl-inositol-3 kinase (PI3K), AGE-1, extend the transferase (NMNAT) (Figure 2A). NamPT was
worm’s life span (Figure 1) (35,36). This pathway found to be the equivalent of the pre-B-cell-enhancing
controls the entry of worms in ‘dauer’, a larval factor (PBEF), a protein that stimulated B-cell colony
developmental state of growth arrest that is induced formation (47), and visfatin, an adipokine expressed in
upon food limitation, and which is a feature of young visceral fat with glucose-lowering properties (48).
340 N. Dali-Youcef et al.

Although some evidence exists that NamPT activates mice. CR animals showed an increase in mitochon-
SIRT1 transcriptional activity (27), NamPT over- drial biogenesis that correlated with an increase in
expression in mice would be necessary to determine the expression of SIRT1, PGC-1a and eNOS in
whether NamPT is the functional homolog of yeast various tissues. Interestingly, these effects were
PNC1 in life span extension. completely inhibited in eNOS2/2 animals in which
The role of the insulin-signaling pathway in the life span was significantly reduced (55).
determination of mammalian life span has been SIRT1 has also been demonstrated to augment
studied in insulin-like growth factor receptor (IGF- insulin secretion in response to glucose in pancreatic
R)-deficient mice. Although IGF-R2/2 mice die b-cells of b-cell-specific SIRT1-overexpressing
shortly after birth, IGF-R+/2 animals are viable, (BESTO) transgenic mice (56). This response was
display reduced IGF-R levels, and live longer than accompanied by a decrease in the expression of the
IGF-R+/+ littermates. IGF-R+/2 animals exhibit low uncoupling protein-2 (UCP-2) that could increase
AKT kinase activity suggesting an increase in FOXO ATP production in the b-cells of BESTO transgenic
activity, reproducing features of the Daf-16- mice. UCPs uncouple oxygen consumption from
mediated longevity pathway in C. elegans. In addi- ATP generation by allowing leakage of protons (H+),
tion, IGF-R+/2 mice are more resistant to oxidative thereby participating in the reduction of ROS
stress. This phenotype was, however, gender-depen- generation. The SIRT1-mediated decrease in
dent since only IGF-R+/2 female mice displayed a UCP-2 expression impedes H+ ‘leakage’ and allows
significant increase in life span, whereas IGF-R+/2 a more efficient coupling of electron transport with
males showed a modest, non-significant increase in the ATP production. Likewise, UCP-22/2 mice also
longevity (49). Mice in which the insulin receptor was exhibit a similar phenotype with improved glucose
specifically inactivated in the adipose tissue also live tolerance and enhanced insulin secretion (57).
longer and are protected against age-related obesity SIRT1 achieves this effect on UCP-2 expression by
and its subsequent metabolic abnormalities (50). directly binding to and repressing UCP-2 gene
In mammals, SIRT1 has been linked with meta- transcription in pancreatic b-cells (58). SIRT1 can
bolic control. The importance of SIRT1 in the also form a complex with FOXO1 and the promye-
nutrient control of glucose homeostasis was demon- locytic leukemia protein PML to activate two insulin
strated to involve the modulation of the acetylation transcription factors, NeuroD and MafA, which may
status and hence the stimulation of the activity of the protect the pancreatic b-cell pathway from oxidative
metabolic coregulator peroxisome proliferator-acti- damage (59). An approach to increase SIRT1
vated receptor gamma (PPARc) coactivator-1a dosage in pancreatic b-cells could hence be of
(PGC-1a). In the liver, SIRT1 in a complex potential interest to maintain a healthy b-cell
including the hepatocyte nuclear factor-4 (HNF-4) function in diabetic patients.
deacetylates and activates PGC-1a, promoting glu- SIRT1 and SIRT3 deacetylate acetyl coenzyme A
coneogenesis following fasting (51). SIRT1 also synthetase (AceCS). AceCS catalyzes the formation
functions together with PGC-1a, beyond the liver. of acetyl-CoA from acetate, coenzyme A, and ATP.
Indeed, activated PGC-1a promotes mitochondrial Whereas SIRT1 has been shown to deacetylate the
function in the skeletal muscle and the brown cytoplasmic AceCS1, whose activity controls acetyl-
adipose tissue, leading to enhanced energy expendi- CoA levels in the cytoplasm for fatty acid synthesis,
ture, increased exercise performance, and protection SIRT3 deacetylates the mitochondrial AceCS2
from diet-induced insulin resistance and hepatostea- which regulates acetyl-CoA-requiring pathways in
tosis (16,52). Although these studies suggest that the mitochondria such as the tricarboxylic acid cycle
PGC-1a is a privileged partner for SIRT1, other (60). Since acetate metabolism is impaired in
signaling pathways are also solicited (Figure 2B). diabetes and ageing, it is legitimate to speculate on
For instance, in white adipose tissue, where the the potential role of SIRT1 and SIRT3 in the
activation of SIRT1 by resveratrol decreased fat pathophysiology of these diseases through the
accumulation and its inhibition resulted in triglycer- regulation of AceCS1 and AceCS2. Further valida-
ide accumulation, it was suggested that SIRT1 tion of these observations in in vivo models is still
activation repressed the nuclear receptor PPARc required.
(53), an effect that, in addition to SIRT1, also A central role for SIRT1 in disease of the central
involved the nuclear receptor corepressor (NcoR) nervous system has also been highlighted in animal
and silencing mediator SMRT (54). A recent study models. In the Wallerian degeneration slow (Wlds)
also emphasized the role of the endothelial nitric mouse model, SIRT1 activation protects axons
oxide synthase (eNOS) signaling in CR-mediated against neuronal injury. This Wlds mouse bears
increases in mitochondrial biogenesis and life span in in fact a dominant mutation producing an
Sirtuins and metabolism 341

overexpressed chimeric Wlds protein composed of gliomas, which are amongst the most frequent
the ubiquitin assembly protein Ufd2a and the NAD+ malignant brain tumors (67). The SIRT2 gene maps
salvage pathway enzyme NMNAT1. Decreasing at chromosome 19q13.2, a region frequently deleted
SIRT1 activity reduces the axonal protection origin- in human gliomas. Furthermore, ectopic expression
ally observed, whereas SIRT1 activation by resver- of SIRT2 in a glioma cell line decreased colony
atrol decreases the axonal degeneration after formation suggesting a potential tumor suppressor
neuronal injury. This suggests that the neuroprotec- role of SIRT2. This could be explained by the fact
tion in the Wlds mouse model is achieved by that SIRT2 plays an important role in the control of
increasing the neuronal NAD+ reserve and/or mitotic exit in the cell cycle where increased SIRT2
SIRT1 activity (61). Furthermore it has been activity severely delays cell cycle progression through
reported that the SIRT1 agonist resveratrol protects mitosis (68), but also by the fact that SIRT2 acts as a
C. elegans neurons expressing a fragment of the mitotic checkpoint protein that prevents chromoso-
Huntington disease-associated protein huntingtin mal instability and the formation of hyperploid cells
and mammalian neurons from mutant polygluta- in the early metaphase (69). Further studies such as
mine cytotoxicity in a HdhQ111 knock-in mouse SIRT2 brain-specific inactivation in genetically
model of Huntington disease (62). In addition, engineered mice should bring an insight into the
SIRT1 activation significantly decreases neuronal mechanism and the role of SIRT2 in the pathophy-
cell death induced by amyloid-beta (Ab) peptides siology of this aggressive type of brain cancer. Very
through inhibition of NFkB signaling (63). Specific recently, SIRT2 was described as an oligodendro-
brain hSIRT1 overexpression in transgenic animals glial cytoplasmic protein, localized to the outer and
induces a significant increase in the a-secretase juxtanodal loops in the myelin sheath, that decreases
activity, an enzyme that cleaves the amyloid pre- cell differentiation through a-tubulin deacetylation
cursor peptide (APP) within the Ab peptide, suggesting a potential implication in myelinogenesis
favoring thereby the nonamyloidogenic pathway of (70).
the APP processing (64). In addition, a recent study
demonstrated the protective effect of CR against
Alzheimer’s disease-type brain amyloidosis in mon- SIRT3
keys. Monkeys maintained on CR diet had signifi- The mitochondrial SIRT3 deacetylase (71,72) has
cantly reduced contents of Ab peptides in the been linked with adaptative thermogenesis. SIRT3
temporal cortex that correlated with enhanced expression is induced in mice in both white and
SIRT1 concentrations as compared to monkeys brown adipose tissue (BAT) by CR and in BAT
under normal diet (65). From these studies, it upon cold exposure. SIRT3 furthermore activates
became clear that pharmacological strategies aiming known mitochondrial genes such as PGC-1a and
at activating SIRT1 would impede Ab peptide
UCP-1 suggesting an important role of SIRT3 in
deposition in the brain and prevent the development
thermogenesis (73). In addition, the SIRT3 gene has
of Alzheimer’s disease.
been linked to longevity. In fact, the genotype
From the animal studies discussed above, it was
defined by the G477T polymorphism in exon 3,
suggested that SIRT1 could contribute to the
was associated with survivorship (74). Furthermore,
pathogenesis of some complex diseases. SIRT1
a VNTR (a 72-bp repeat core) polymorphism in
could hence be considered as a serious candidate
intron 5 of the SIRT3 gene, that acts as an allele-
target for therapeutic interventions in metabolic and
specific enhancer activity on a reporter gene, was
neurodegenerative disorders. In line with this
associated with longevity in male subjects (75). This
hypothesis, genetic variants (SNPs) in the human
VNTR might represent the functional variant that
SIRT1 gene have been shown to be tightly asso-
accounts for the association between the silent
ciated with energy expenditure (52). We predict that
marker G477T and longevity in old male subjects.
future human studies will link SIRT1 even more
tightly with metabolic and neurodegenerative dis-
eases, stimulating the development of therapeutics SIRT4
for their treatment.
Although the mitochondrial expressed protein
SIRT4 has a conserved sirtuin domain, it seems
Emerging functions for the other sirtuins not to possess in vitro deacetylase activity (76).
However, SIRT4 ADP-ribosylates and inhibits the
SIRT2
mitochondrial glutamate dehydrogenase (GDH).
The tubulin-deacetylase protein SIRT2 (66) has GDH controls amino acid-stimulated insulin secre-
been demonstrated to be downregulated in human tion by regulating glutamine and glutamate oxidative
342 N. Dali-Youcef et al.

Table I. Expression pattern, cellular distribution, and function of sirtuin deacetylases. Sirtuins are differentially expressed in different
organs based on their transcripts.

Expression levels
Cellular Putative Potential link
Gene High Low compartmentation target genes with diseases

SIRT1 Br, Te, Sk, Li, Sp, He Lu, Nuclear p53,Ku70, NFkB, ageing, obesity, Insulin
Ki (+++), Ov, BM PGC-1a, MEF2D, resistance, inflammation,
Th, Ut (++) MyoD, PPARc, diabetes, heart failure, axonal
FOXO, p300, degeneration, AIDS
AceCS1, tat
SIRT2 Br, Sk (+++), Li, Th, Lu, BM, Ut, Cytoplasmic a-tubulin downregulated in human gliomas
Te, Ki, He (++) Ov, Sp
SIRT3 Ov (+++), most Mitochondrial PGC-1a, AceCS2 adaptive thermogenesis,
other organs (++) overexpressed in node-positive
breast cancer
SIRT4 Br, Te, He, BM, Sp Mitochondrial Glutamate inhibits amino acid-stimulated
Lu (+++), Li, dehydrogenase insulin secretion
Sk, Ki, Th, Ut,
Ov (++)
SIRT5 Br, Te, Sk, Ki, He Sp Mitochondrial unknown unknown
(+++) Li, Ov, Lu,
Th, Ut, BM (++)
SIRT6 Fetal Br (+++), Br, Sp, Th, Ut, BM, Lu Nuclear DNA polb age-related diseases
Li, Te, Sk, Ki, He,
Ov (++)
SIRT7 Br, Te, Ki Lower in other organs Nuclear RNA polymerase highly expressed in thyroid
Sp, Te, Li, Ki, Br, Sk, He (proteins) Pol I cancers, overexpressed in node-
Pa (proteins) positive breast cancer

Abbreviations: Br5Brain; He5heart; Ki5kidney; Sk5skeletal muscle; Li5liver; Te5testis; Lu5lung; Sp5spleen; Ov5ovary; Ut5uterus;
BM5bone marrow; Th5thyroid.

metabolism. Inhibition of GDH activity by SIRT4 SIRT6 deacetylates histones and the DNA repair
decreases insulin secretion in mouse pancreatic b- enzyme, DNA polymerase b (polb) in vitro (77). An
cells in response to amino acids (76). Interestingly, effect on DNA repair was hence proposed to explain
the two different sirtuins SIRT1 and SIRT4 seem to the phenotype in SIRT62/2 mice, and it was
work in opposite directions to control insulin suggested that SIRT6 could play an essential role
secretion. Furthermore SIRT4 expression is down- in maintaining organ integrity as animals age.
regulated in response to CR in b-cells, which is
opposite to the regulation of SIRT1 during CR.
SIRT7
More studies are still needed to see whether SIRT4
(and SIRT1) can be integrated in the pathophysiol- SIRT7 is the only sirtuin to be localized in the
ogy of type 1 and 2 diabetes that both are nucleolus and is a component of the RNA poly-
characterized by alterations in insulin secretion. merase I (Pol I) transcriptional machinery. SIRT7
interacts with RNA Pol I and histones, and positively
regulates the transcription of rDNA during tran-
SIRT6
scriptional elongation, which accounts for about
Whereas little is known about the activity of SIRT5, 60% of total transcription in metabolically active
SIRT6 is suggested to control genomic DNA cells in mammals (79). SIRT7 overexpression
stability and DNA repair. SIRT62/2 mice die increases Pol I-mediated transcription in a NAD+-
prematurely and display severe defects including dependent manner, whereas SIRT7 inhibition
important lymphopenia, loss of subcutaneous fat, induces a decrease in the transcription of Pol I and
decreased bone mineral density, metabolic defects its association with rDNA (80). Depletion of
with a profound imbalance in glucose homeostasis SIRT7 stopped cell proliferation and triggered
and decreased IGF-1 levels. The phenotype of the apoptosis. It was suggested that SIRT7 may regu-
SIRT62/2 mice mimics multiple pathologies found late rDNA transcription by sensing cellular NAD+
in elderly humans (77). Although SIRT6 was levels and that diet-induced changes in NAD+/
originally described as an exclusive ADP-ribosyl- NADH ratio might modulate SIRT7 to link the
transferase (78), it was recently demonstrated that cellular energy status with rRNA synthesis and
Sirtuins and metabolism 343

ribosome production. It is worth noting that SIRT1 the Centre National de la Recherche Scientifique
negatively regulates RNA Pol I through deacetyla- (CNRS); Institut National pour la Science et la
tion of TAFI68 (81), which goes opposite of SIRT7 Recherche Médicale (INSERM); National Institutes
action. of Health (NIH); the European Union (EU) and the
Hôpitaux Universitaires de Strasbourg.
Concluding remarks
References
In the last decade, sirtuin biology has come a long
way from their original description as yeast NAD+- 1. Chen H, Lin RJ, Xie W, Wilpitz D, Evans RM. Regulation of
dependent class III HDACs, that control yeast life hormone-induced histone hyperacetylation and gene activa-
tion via acetylation of an acetylase. Cell. 1999;98:675–86.
span. In mammals, seven orthologs of Sir2 have 2. Courey AJ, Jia S. Transcriptional repression: the long and the
been identified, SIRT1 to 7, and the exact biological short of it. Genes Dev. 2001;15:2786–96.
function of most of these sirtuins still remains only 3. Imai S, Johnson FB, Marciniak RA, McVey M, Park PU,
partially characterized. Of particular interest is the Guarente L. Sir2: an NAD-dependent histone deacetylase
fact that SIRT1 not only deacetylates histones to that connects chromatin silencing, metabolism, and aging.
Cold Spring Harb Symp Quant Biol. 2000;65:297–302.
mediate gene silencing, but is able to interact with
4. Imai S, Armstrong CM, Kaeberlein M, Guarente L.
and deacetylate some well known transcriptional Transcriptional silencing and longevity protein Sir2 is an
regulators thereby modulating specifically various NAD-dependent histone deacetylase. Nature. 2000;403:
biological processes. Hence modulating the expres- 795–800.
sion of SIRT1 or its activity, by using sirtuin- 5. Lin SJ, Guarente L. Nicotinamide adenine dinucleotide, a
activating compounds such as resveratrol, will have metabolic regulator of transcription, longevity and disease.
Curr Opin Cell Biol. 2003;15:241–6.
pleiotropic effects. SIRT1 activation reduces fat 6. Hoppe GJ, Tanny JC, Rudner AD, Gerber SA, Danaie S,
accumulation and adipocyte differentiation through Gygi SP, et al. Steps in assembly of silent chromatin in yeast:
repression of the activity of the adipogenic nuclear Sir3-independent binding of a Sir2/Sir4 complex to silencers
receptor PPARc. SIRT1 also promotes mitochon- and role for Sir2-dependent deacetylation. Mol Cell Biol.
drial function and energy expenditure and conse- 2002;22:4167–80.
7. Ivy JM, Klar AJ, Hicks JB. Cloning and characterization of
quently protects mice from diet-induced obesity,
four SIR genes of Saccharomyces cerevisiae. Mol Cell Biol.
through deacetylation and subsequent activation of 1986;6:688–702.
PGC-1a in the skeletal muscle and in the brown 8. Rine J, Herskowitz I. Four genes responsible for a position
adipose tissue. The SIRT1/PGC-1a interaction is effect on expression from HML and HMR in Saccharomyces
also important in the liver, where SIRT1 activation cerevisiae. Genetics. 1987;116:9–22.
upon fasting induces gluconeogenesis and prevents 9. Chen L, Widom J. Mechanism of transcriptional silencing in
yeast. Cell. 2005;120:37–48.
against hepatosteatosis. In addition, SIRT1 signifi-
10. Braunstein M, Rose AB, Holmes SG, Allis CD, Broach JR.
cantly enhances insulin secretion in the pancreatic b- Transcriptional silencing in yeast is associated with reduced
cells. In combination, these studies illustrate that nucleosome acetylation. Genes Dev. 1993;7:592–604.
SIRT1 is a major modulator of metabolism. SIRT1 11. Kaeberlein M, McVey M, Guarente L. The SIR2/3/4
activation also seems to be endowed with neuropro- complex and SIR2 alone promote longevity in
Saccharomyces cerevisiae by two different mechanisms.
tective activities as suggested from the study of
Genes Dev. 1999;13:2570–80.
models of Huntington or Alzheimer’s disease. 12. Tissenbaum HA, Guarente L. Increased dosage of a sir-2
Furthermore, other sirtuins might play important gene extends lifespan in Caenorhabditis elegans. Nature.
roles in some diseases, as illustrated by SIRT2, 2001;410:227–30.
which is downregulated in human gliomas. 13. Rogina B, Helfand SL. Sir2 mediates longevity in the fly
Obviously, more studies, in animal models and through a pathway related to calorie restriction. Proc Natl
Acad Sci U S A. 2004;101:15998–6003.
humans, are still needed to define the exact role of
14. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW,
sirtuins in the pathophysiology of human diseases. It Lavu S, Wood JG, et al. Small molecule activators of sirtuins
can, however, be predicted that therapeutic inter- extend Saccharomyces cerevisiae lifespan. Nature. 2003;425:
ventions aiming at activating or blocking sirtuins, 191–6.
depending on the context, will one day become 15. Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL,
Tatar M, et al. Sirtuin activators mimic caloric restriction
helpful in the treatment of human diseases.
and delay ageing in metazoans. Nature. 2004;430:686–9.
16. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C,
Kalra A, et al. Resveratrol improves health and survival of
Acknowledgements mice on a high-calorie diet. Nature. 2006;444:337–42.
17. Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M,
We thank greatly members of the Auwerx laboratory Sinclair DA. Inhibition of silencing and accelerated aging by
for critical reading of the manuscript and helpful nicotinamide, a putative negative regulator of yeast sir2 and
discussions. This work was supported by grants of human SIRT1. J Biol Chem. 2002;277:45099–107.
344 N. Dali-Youcef et al.

18. Bordone L, Guarente L. Calorie restriction, SIRT1 and diapause in Caenorhabditis elegans. Nature. 1996;382:
metabolism: understanding longevity. Nat Rev Mol Cell Biol. 536–9.
2005;6:298–305. 37. Burnell AM, Houthoofd K, O’Hanlon K, Vanfleteren JR.
19. Lin SJ, Defossez PA, Guarente L. Requirement of NAD and Alternate metabolism during the dauer stage of the nematode
SIR2 for life-span extension by calorie restriction in Caenorhabditis elegans. Exp Gerontol. 2005;40:850–6.
Saccharomyces cerevisiae. Science. 2000;289:2126–8. 38. Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L,
20. Lin SJ, Kaeberlein M, Andalis AA, Sturtz LA, Defossez PA, Tissenbaum HA, et al. The Fork head transcription factor
Culotta VC, et al. Calorie restriction extends Saccharomyces DAF-16 transduces insulin-like metabolic and longevity
cerevisiae lifespan by increasing respiration. Nature. signals in C. elegans. Nature. 1997;389:994–9.
2002;418:344–8. 39. Lin K, Dorman JB, Rodan A, Kenyon C. daf-16: An HNF-3/
21. Bakker BM, Overkamp KM, van Maris AJ, Kotter P, forkhead family member that can function to double the life-
Luttik MA, van Dijken JP, et al. Stoichiometry and span of Caenorhabditis elegans. Science. 1997;278:1319–22.
compartmentation of NADH metabolism in Saccharomyces 40. Berdichevsky A, Viswanathan M, Horvitz HR, Guarente L.
cerevisiae. FEMS Microbiol Rev. 2001;25:15–37. C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate
22. Tanner KG, Landry J, Sternglanz R, Denu JM. Silent DAF-16 and extend life span. Cell. 2006;125:1165–77.
information regulator 2 family of NAD-dependent histone/ 41. Wang Y, Oh SW, Deplancke B, Luo J, Walhout AJ,
protein deacetylases generates a unique product, 1-O-acetyl- Tissenbaum HA. C. elegans 14-3-3 proteins regulate life
ADP-ribose. Proc Natl Acad Sci U S A. 2000;97:14178–82. span and interact with SIR-2.1 and DAF-16/FOXO. Mech
23. Liou GG, Tanny JC, Kruger RG, Walz T, Moazed D. Ageing Dev. 2006;127:741–7.
Assembly of the SIR complex and its regulation by O-acetyl- 42. Rogina B, Helfand SL, Frankel S. Longevity regulation by
ADP-ribose, a product of NAD-dependent histone deacetyla- Drosophila Rpd3 deacetylase and caloric restriction. Science.
tion. Cell. 2005;121:515–27. 2002;298:1745.
24. Anderson RM, Bitterman KJ, Wood JG, Medvedik O, 43. Clancy DJ, Gems D, Harshman LG, Oldham S, Stocker H,
Cohen H, Lin SS, et al. Manipulation of a nuclear NAD+ Hafen E, et al. Extension of life-span by loss of CHICO, a
salvage pathway delays aging without altering steady-state Drosophila insulin receptor substrate protein. Science.
NAD+ levels. J Biol Chem. 2002;277:18881–90. 2001;292:104–6.
25. Anderson RM, Bitterman KJ, Wood JG, Medvedik O, 44. Hwangbo DS, Gershman B, Tu MP, Palmer M, Tatar M.
Sinclair DA. Nicotinamide and PNC1 govern lifespan Drosophila dFOXO controls lifespan and regulates insulin
extension by calorie restriction in Saccharomyces cerevisiae. signalling in brain and fat body. Nature. 2004;429:562–6.
Nature. 2003;423:181–5. 45. Giannakou ME, Goss M, Junger MA, Hafen E, Leevers SJ,
26. Lin SJ, Ford E, Haigis M, Liszt G, Guarente L. Calorie Partridge L. Long-lived Drosophila with overexpressed
restriction extends yeast life span by lowering the level of dFOXO in adult fat body. Science. 2004;305:361.
NADH. Genes Dev. 2004;18:12–6. 46. Rongvaux A, Shea RJ, Mulks MH, Gigot D, Urbain J, Leo O,
27. Revollo JR, Grimm AA, Imai S. The NAD biosynthesis et al. Pre-B-cell colony-enhancing factor, whose expression is
pathway mediated by nicotinamide phosphoribosyltransferase up-regulated in activated lymphocytes, is a nicotinamide
regulates Sir2 activity in mammalian cells. J Biol Chem. phosphoribosyltransferase, a cytosolic enzyme involved in
2004;279:50754–63. NAD biosynthesis. Eur J Immunol. 2002;32:3225–34.
28. Sandmeier JJ, Celic I, Boeke JD, Smith JS. Telomeric and 47. Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I.
rDNA silencing in Saccharomyces cerevisiae are dependent Cloning and characterization of the cDNA encoding a novel
on a nuclear NAD(+) salvage pathway. Genetics. 2002;160: human pre-B-cell colony-enhancing factor. Mol Cell Biol.
877–89. 1994;14:1431–7.
29. Lamming DW, Latorre-Esteves M, Medvedik O, Wong SN, 48. Fukuhara A, Matsuda M, Nishizawa M, Segawa K,
Tsang FA, Wang C, et al. HST2 mediates SIR2-independent Tanaka M, Kishimoto K, et al. Visfatin: a protein secreted
life-span extension by calorie restriction. Science. 2005;309: by visceral fat that mimics the effects of insulin. Science.
1861–4. 2005;307:426–30.
30. Sinclair DA, Guarente L. Extrachromosomal rDNA circles— 49. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A,
a cause of aging in yeast. Cell. 1997;91:1033–42. Even PC, et al. IGF-1 receptor regulates lifespan and
31. Longo VD. The Ras and Sch9 pathways regulate stress resistance to oxidative stress in mice. Nature. 2003;421:
resistance and longevity. Exp Gerontol. 2003;38:807–11. 182–7.
32. Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD. 50. Bluher M, Kahn BB, Kahn CR. Extended longevity in mice
Regulation of longevity and stress resistance by Sch9 in yeast. lacking the insulin receptor in adipose tissue. Science.
Science. 2001;292:288–90. 2003;299:572–4.
33. Fabrizio P, Liou LL, Moy VN, Diaspro A, 51. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM,
SelverstoneValentine J, Gralla EB, et al. SOD2 functions Puigserver P. Nutrient control of glucose homeostasis
downstream of Sch9 to extend longevity in yeast. Genetics. through a complex of PGC-1alpha and SIRT1. Nature.
2003;163:35–46. 2005;434:113–8.
34. Kaeberlein M, Andalis AA, Fink GR, Guarente L. High 52. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H,
osmolarity extends life span in Saccharomyces cerevisiae by a Lerin C, Daussin F, et al. Resveratrol improves mitochondrial
mechanism related to calorie restriction. Mol Cell Biol. function and protects against metabolic disease by activating
2002;22:8056–66. SIRT1 and PGC-1alpha. Cell. 2006;127:1109–22.
35. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. daf-2, an 53. Picard F, Auwerx J. PPAR(gamma) and glucose homeostasis.
insulin receptor-like gene that regulates longevity and Annu Rev Nutr. 2002;22:167–97.
diapause in Caenorhabditis elegans. Science. 1997;277: 54. Picard F, Kurtev M, Chung N, Topark-Ngarm A,
942–6. Senawong T, Machado De Oliveira R, et al. Sirt1 promotes
36. Morris JZ, Tissenbaum HA, Ruvkun G. A phosphatidylino- fat mobilization in white adipocytes by repressing PPAR-
sitol-3-OH kinase family member regulating longevity and gamma. Nature. 2004;429:771–6.
Sirtuins and metabolism 345

55. Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R, deacetylase activity in control of mitotic exit in the cell cycle.
Tedesco L, et al. Calorie restriction promotes mitochondrial Mol Cell Biol. 2003;23:3173–85.
biogenesis by inducing the expression of eNOS. Science. 69. Inoue T, Hiratsuka M, Osaki M, Yamada H, Kishimoto I,
2005;310:314–7. Yamaguchi S, et al. SIRT2, a tubulin deacetylase, acts to
56. Moynihan KA, Grimm AA, Plueger MM, Bernal-Mizrachi E, block the entry to chromosome condensation in response to
Ford E, Cras-Meneur C, et al. Increased dosage of mitotic stress. Oncogene. 2007;26:945–57.
mammalian Sir2 in pancreatic beta cells enhances glucose- 70. Li W, Zhang B, Tang J, Cao Q, Wu Y, Wu C, et al. Sirtuin 2,
stimulated insulin secretion in mice. Cell Metab. a mammalian homolog of yeast silent information regulator-2
2005;2:105–17. longevity regulator, is an oligodendroglial protein that
57. Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, decelerates cell differentiation through deacetylating alpha-
et al. Uncoupling protein-2 negatively regulates insulin tubulin. J Neurosci. 2007;27:2606–16.
secretion and is a major link between obesity, beta cell 71. Onyango P, Celic I, McCaffery JM, Boeke JD, Feinberg AP.
dysfunction, and type 2 diabetes. Cell. 2001;105:745–55. SIRT3, a human SIR2 homologue, is an NAD-dependent
58. Bordone L, Motta MC, Picard F, Robinson A, Jhala US, deacetylase localized to mitochondria. Proc Natl Acad Sci U
Apfeld J, et al. Sirt1 regulates insulin secretion by repressing S A. 2002;99:13653–8.
UCP2 in pancreatic beta cells. PLoS Biol. 2006;4:e31. 72. Schwer B, North BJ, Frye RA, Ott M, Verdin E. The human
59. Kitamura YI, Kitamura T, Kruse JP, Raum JC, Stein R, silent information regulator (Sir)2 homologue hSIRT3 is a
Gu W, et al. FoxO1 protects against pancreatic beta cell mitochondrial nicotinamide adenine dinucleotide-dependent
failure through NeuroD and MafA induction. Cell Metab. deacetylase. J Cell Biol. 2002;158:647–57.
2005;2:153–63. 73. Shi T, Wang F, Stieren E, Tong Q. SIRT3, a mitochondrial
60. Hallows WC, Lee S, Denu JM. Sirtuins deacetylate and
sirtuin deacetylase, regulates mitochondrial function and
activate mammalian acetyl-CoA synthetases. Proc Natl Acad
thermogenesis in brown adipocytes. J Biol Chem.
Sci U S A. 2006;103:10230–5.
2005;280:13560–7.
61. Araki T, Sasaki Y, Milbrandt J. Increased nuclear NAD
74. Rose G, Dato S, Altomare K, Bellizzi D, Garasto S, Greco V,
biosynthesis and SIRT1 activation prevent axonal degenera-
et al. Variability of the SIRT3 gene, human silent information
tion. Science. 2004;305:1010–3.
regulator Sir2 homologue, and survivorship in the elderly.
62. Parker JA, Arango M, Abderrahmane S, Lambert E,
Exp Gerontol. 2003;38:1065–70.
Tourette C, Catoire H, et al. Resveratrol rescues mutant
75. Bellizzi D, Dato S, Cavalcante P, Covello G, Di Cianni F,
polyglutamine cytotoxicity in nematode and mammalian
Passarino G, et al. Characterization of a bidirectional
neurons. Nat Genet. 2005;37:349–50.
promoter shared between two human genes related to aging:
63. Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi S,
SIRT3 and PSMD13. Genomics. 2007;89:143–50.
et al. SIRT1 protects against microglia-dependent amyloid-
beta toxicity through inhibiting NF-kappaB signaling. J Biol 76. Haigis MC, Mostoslavsky R, Haigis KM, Fahie K,
Chem. 2005;280:40364–74. Christodoulou DC, Murphy AJ, et al. SIRT4 inhibits
64. Qin W, Yang T, Ho L, Zhao Z, Wang J, Chen L, et al. glutamate dehydrogenase and opposes the effects of calorie
Neuronal SIRT1 activation as a novel mechanism underlying restriction in pancreatic beta cells. Cell. 2006;126:941–54.
the prevention of Alzheimer disease amyloid neuropathology 77. Mostoslavsky R, Chua KF, Lombard DB, Pang WW,
by calorie restriction. J Biol Chem. 2006;281:21745–54. Fischer MR, Gellon L, et al. Genomic instability and aging-
65. Qin W, Chachich M, Lane M, Roth G, Bryant M, de Cabo R, like phenotype in the absence of mammalian SIRT6. Cell.
et al. Calorie restriction attenuates Alzheimer’s disease type 2006;124:315–29.
brain amyloidosis in Squirrel monkeys (Saimiri sciureus). J 78. Liszt G, Ford E, Kurtev M, Guarente L. Mouse Sir2
Alzheimers Dis. 2006;10:417–22. homolog SIRT6 is a nuclear ADP-ribosyltransferase. J Biol
66. North BJ, Marshall BL, Borra MT, Denu JM, Verdin E. The Chem. 2005;280:21313–20.
human Sir2 ortholog, SIRT2, is an NAD+-dependent tubulin 79. Grummt I, Pikaard CS. Epigenetic silencing of RNA
deacetylase. Mol Cell. 2003;11:437–44. polymerase I transcription. Nat Rev Mol Cell Biol. 2003;4:
67. Hiratsuka M, Inoue T, Toda T, Kimura N, Shirayoshi Y, 641–9.
Kamitani H, et al. Proteomics-based identification of 80. Ford E, Voit R, Liszt G, Magin C, Grummt I, Guarente L.
differentially expressed genes in human gliomas: down- Mammalian Sir2 homolog SIRT7 is an activator of RNA
regulation of SIRT2 gene. Biochem Biophys Res Commun. polymerase I transcription. Genes Dev. 2006;20:1075–80.
2003;309:558–66. 81. Muth V, Nadaud S, Grummt I, Voit R. Acetylation of
68. Dryden SC, Nahhas FA, Nowak JE, Goustin AS, TAF(I)68, a subunit of TIF-IB/SL1, activates RNA poly-
Tainsky MA. Role for human SIRT2 NAD-dependent merase I transcription. EMBO J. 2001;20:1353–62.

You might also like