Sie sind auf Seite 1von 8

Please cite this article in press as: Mitchell et al.

, The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Cell Reports

Report

The SIRT1 Activator SRT1720 Extends Lifespan


and Improves Health of Mice Fed a Standard Diet
Sarah J. Mitchell,1,2,3,8 Alejandro Martin-Montalvo,1,8 Evi M. Mercken,1 Hector H. Palacios,1 Theresa M. Ward,1
Gelareh Abulwerdi,1 Robin K. Minor,1 George P. Vlasuk,4 James L. Ellis,4 David A. Sinclair,5 John Dawson,6
David B. Allison,6 Yongqing Zhang,7 Kevin G. Becker,7 Michel Bernier,1 and Rafael de Cabo1,*
1Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore,

MD 21224, USA
2Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
3Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
4Sirtris, a GSK company, 200 Technology Square, Cambridge, MA 02139, USA
5Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA 02115, USA
6School of Public Health, University of Alabama at Birmingham, Birmingham, AL 35294, USA
7Gene Expression and Genomics Unit, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore,

MD 21224, USA
8These authors contributed equally to this work.

*Correspondence: decabora@grc.nia.nih.gov
http://dx.doi.org/10.1016/j.celrep.2014.01.031
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-No Derivative Works
License, which permits non-commercial use, distribution, and reproduction in any medium, provided the original author and source are
credited.

SUMMARY et al., 2012). SIRT1 has been proposed as an antiaging protein,


and its activation results in health benefits in multiple organisms
The prevention or delay of the onset of age-related (Baur et al., 2012). Pharmacological interventions aimed at
diseases prolongs survival and improves quality of increasing SIRT1 activity have been found to slow the onset of
life while reducing the burden on the health care aging and delay age-associated diseases. There is mounting
system. Activation of sirtuin 1 (SIRT1), an NAD+- evidence that overexpression of SIRT1 in mice and use of
dependent deacetylase, improves metabolism and small molecule activators of SIRT1, such as resveratrol and
SRT1720, enhance insulin sensitivity and protect against diet-
confers protection against physiological and cogni-
induced impairments in mitochondrial capacity and oxidative
tive disturbances in old age. SRT1720 is a specific
metabolism in laboratory animals (Banks et al., 2008; Feige
SIRT1 activator that has health and lifespan benefits et al., 2008; Milne et al., 2007; Pfluger et al., 2008). Even though
in adult mice fed a high-fat diet. We found extension SIRT1 activators improve health and extend lifespan of mice
in lifespan, delayed onset of age-related metabolic maintained on a high-fat diet (Baur et al., 2006; Minor et al.,
diseases, and improved general health in mice fed a 2011), there has yet to be a demonstration of both improvements
standard diet after SRT1720 supplementation. Inhibi- in lifespan and healthspan in mice fed a standard diet (SD). Re-
tion of proinflammatory gene expression in both liver sveratrol improves healthspan of SD-fed mice; yet, no effect on
and muscle of SRT1720-treated animals was noted. lifespan was noted in C57BL6/J mice (Pearson et al., 2008) or
SRT1720 lowered the phosphorylation of NF-kB in genetically heterogeneous mice (Strong et al., 2013). Other
pathway regulators in vitro only when SIRT1 was func- compounds that extend lifespan in mice include the immuno-
modulatory drug rapamycin (Harrison et al., 2009) and the insu-
tionally present. Combined with our previous work,
lin-sensitizing agent metformin. However, chronic administration
the current study further supports the beneficial ef-
of rapamycin induces glucose intolerance, insulin resistance, and
fects of SRT1720 on health across the lifespan in incidence of diabetogenic effects (Deblon et al., 2012; Houde
mice. et al., 2010; Lamming et al., 2012). Similarly, metformin is associ-
ated with adverse side effects, including lactic acidosis and kid-
INTRODUCTION ney dysfunction in a dose-dependent manner (Martin-Montalvo
et al., 2013). Here, we show that SRT1720 supplementation
Many of the chronic diseases that exist in older adults constitute a extends survival and improves health in mice fed a standard diet.
highly significant social and economic burden to the community.
Long-term illness, diminished quality of life, and increased health RESULTS AND DISCUSSION
care costs are all challenges that older adults are facing. Sirtuin 1
(SIRT1), an NAD+-dependent deacetylase, is one of seven To test the effect of SRT1720 on the health and lifespan of
mammalian sirtuins and is known to play an important role C57BL6/J mice, these animals were fed either a high-fat diet
in maintaining metabolic homeostasis in multiple tissues (Baur (HFD) or a standard AIN-93G diet (SD) supplemented with

Cell Reports 6, 18, March 13, 2014 2014 The Authors 1


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Figure 1. SRT1720 Extends Lifespan and


Improves Health in Mice Fed a Standard
Diet
(A) Kaplan-Meier survival curves for mice fed
either a standard diet (SD) or high-fat diet (HFD)
supplemented without or with SRT1720 (SD-
SRT1720, HFD-SRT1720).
(B) Average body weight over the study.
(C) Average daily caloric intake over the study.
(D) Percentage fat mass measured by nuclear
magnetic resonance spectroscopy at 13 months
of age.
(E) Respiratory exchange ratio (RER).
(F) Rotarod performance.
Data are shown as mean SEM. *p % 0.05 in
comparison to diet without SRT1720.

in agreement with our previous study (Mi-


nor et al., 2011), although maximum life-
span did not change (Figure 1A). Median
survival was significantly increased by
22 weeks in HFD-fed mice treated with
SRT1720 (c2 = 5.23, p = 0.022), indicating
an improvement in general health in
these mice (Figure 1A). The hazard ratio
for death was significantly reduced with
SRT1720 treatment (HR = 0.73, 95% con-
fidence interval [0.59, 0.90], p = 0.0034),
indicating a positive effect of SRT1720
on improving lifespan in mice. SRT1720
significantly reduced the average body
weight of HFD-fed mice (Figure 1B)
despite no difference in daily caloric
intake (Figure 1C) or food consumption
(Figure S1A). As expected, mice on HFD
weighed significantly more than their
SD-fed counterparts across the course
of the study (Figure S1B). SRT1720 sup-
plementation also reduced both average
body weight (Figure 1B) and body weight
of SD-fed animals (Figure S1B) without
100 mg/kg body weight of SRT1720 beginning at 6 months of impacting on daily caloric intake (Figure 1C). Treatment with
age for the remainder of their life. SRT1720 treatment signifi- SRT1720 significantly reduced body fat percentage in SD-fed
cantly extends mean lifespan of mice on both diets (Figure 1A). animals (Figure 1D), despite no difference in lean-to-fat ratio (Fig-
Survival among the SD-fed groups was significantly different ure S1C). However, the lack of difference for either parameter in
as assessed using the nonparametric log-rank (Mantel-Haens- HFD-treated mice is in contrast with our previous study (Minor
zel) test (c2 = 4.168, p = 0.0412). Mean lifespan was increased et al., 2011), which may be due to the time point at which the
8.8%, and we observed a trend toward an increase in median treatment was started (6 months versus 12 months).
lifespan of 5 weeks (Su and Wei, 1993) with SRT1720 supple- Recent studies have illustrated the beneficial cardioprotective
mentation (c2 = 2.77, p = 0.096). However, there was no differ- effects of SRT1720 against ischemia-reperfusion injury and
ence in 90th percentile survival (Wang et al., 2004) despite the the loss of cardiac function that occurs with aging (Tong et al.,
overall significant effect on lifespan. These data are consistent 2013). SRT1720 also confers protection from atherosclerosis
with the notion that in the absence of a metabolic insult, interven- in vitro (Zeng et al., 2013) via SIRT1 activation. The synchrony
tions such as SRT1720 will have a more pronounced effect in in circadian rhythms leads to increased longevity and improved
delaying the onset of age-associated diseases. Survival was health (Froy, 2011). Interestingly, SRT1720 is essential for
significantly lower in HFD-fed mice based on the nonparametric the proper regulation of circadian rhythm in the liver of SD-fed
log-rank (Mantel-Haenszel) test (c2 = 5.470, p = 0.0193), and mice through a SIRT1-dependent mechanism (Bellet et al.,
SRT1720 supplementation increased mean lifespan by 21.7%, 2013). Although some controversy exists concerning the

2 Cell Reports 6, 18, March 13, 2014 2014 The Authors


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

2013). Overall these data indicate that SRT1720 improves mus-


cle function and motor coordination over the life of SD-fed mice.
For the remainder of the study, we focused on the effects of
SRT1720 on various metabolic variables in SD-fed mice.
Because aging is associated with functional and metabolic de-
clines, we assessed the effects of SRT1720 on several variables
of aging. The increase in cataract formation with age has been
attributed, in part, to the accumulation of reactive oxygen spe-
cies (Wolf et al., 2005). Here, a marked reduction in cataract
opacity was observed in SRT1720-treated SD-fed mice (Fig-
ure 2A), which is consistent with our previous study on resvera-
trol (Pearson et al., 2008).
Another hallmark feature of aging is the increase in blood
glucose and insulin levels, which leads to type 2 diabetes,
cardiovascular disease, and nonalcoholic fatty liver disease.
In this study, an oral glucose tolerance test was performed
to determine whether SRT1720 supplementation improved
glucose disposal in SD-fed mice (Figure 2B). Our results indi-
cated significant reduction in area under the curve values,
which are consistent with greater glucose disposal in response
to SRT1720 (Figure 2B, inset). Although not statistically signifi-
Figure 2. SRT1720 Improves the Quality of Life of SD-Fed Mice
cant, the reduction in the Homeostatic Model Assessment of In-
(A) Cataract formation as assessed by lens opacity classification.
(B) Oral glucose tolerance test with area under the curve (inset). sulin Resistance (HOMA-IR) index was indicative of improved
(C) The homeostatic model assessment calculation of insulin resistance insulin sensitivity in SRT1720-treated animals (Figure 2C).
(HOMA-IR) and serum biochemical markers. Recent work has reported that SRT1720 lowers insulin resis-
Data are shown as mean SEM. *p % 0.05 in comparison to SD diet without tance in mouse models of obesity (Minor et al., 2011; Yamazaki
SRT1720. et al., 2009), which may be due to an enhancement of glucose-
stimulated insulin secretion by SRT1720. This biological
specificity of SRT1720 toward SIRT1 (Huber et al., 2010; Pacho- response was not measured in the current study. Our results
lec et al., 2010), the notion that SIRT1 is a target of SRT1720 are consistent with two previous studies showing that lifelong
is supported by recent data showing that it activates SIRT1 via resveratrol supplementation and short-term SRT1720 treatment
a direct allosteric mechanism (Hubbard et al., 2013). Taken of HFD-fed mice both elicit beneficial effects on insulin sensi-
together, these results illustrate the involvement of multiple tivity and protect against diet-induced impairments in mito-
SIRT1-dependent mechanisms in the biological actions of chondrial capacity and oxidative metabolism in vivo (Feige
SRT1720. et al., 2008; Pearson et al., 2008). Total cholesterol and low-
We assessed metabolism using the Comprehensive Lab density lipoprotein levels were lower in SRT1720-treated mice
Animal Monitoring System (CLAMS) and found that SRT1720 fed an SD (Figure 2C), which may be due to reduced expression
supplementation significantly lowered respiratory exchange ra- of lipogenic enzymes that occur with SRT1720 treatment
tio (RER) in SD-fed animals during the light cycle, with a trend to- (Yamazaki et al., 2009). The ability of SRT1720 to influence
ward a reduction during the dark cycle (Figure 1E). No difference serum lipid profile may contribute to an improvement in risk fac-
in locomotor activity between groups was observed (Figure S1D). tors for metabolic syndrome that occurs as part of the normal
The RER for HFD-fed mice was close to 0.7, indicating a prefer- progression of aging. Thus, mechanisms of lifespan and health-
ence for fatty acid oxidation (Figure 1E). Interestingly, increased span extension by SRT1720 could be linked to protection
fatty acid oxidation in SRT1720-treated SD-fed mice, as evi- against age-associated metabolic and functional decline in
denced by a decrease of RER, could explain the lower percent- laboratory mice.
age of fat mass in these animals (Figure 1D). It was important to SRT1720 treatment did not alter the number of pathologies in
ascertain whether there was improvement in the quality of life of SD-fed mice on gross postmortem pathology screens (Table
SRT1720-treated mice through assessment of balance and mo- S1). Although the incidence of hepatocellular carcinoma and
tor coordination. SRT1720 significantly improved rotarod perfor- glomerulonephritis appeared higher in the SRT1720-treated
mance at 13 and 18 months of age in SD-fed mice, with a nearly group (Table S1), the mean age at which necropsy was per-
significant (p = 0.08) improvement in performance at 24 months formed was 10 weeks later, consistent with the notion that
of age (Figure 1F). No beneficial effects of SRT1720 were SRT1720 delays the onset of pathologies allowing mice to live
observed in HFD-fed mice (Figure 1F). Although it is recognized a longer and healthier life. The incidence of lymphoma was not
that there is both a learning and body weight component to the assessed in these mice. Upon further histological analysis of tis-
rotarod performance test, a recent study has shown that sues by a blinded pathologist (Table S2; Figure S2), we observed
improvement in rotarod performance can be independent of a significant reduction in steatosis in the SRT1720-treated mice
body weight and that diet may have a larger effect on neurobe- relative to the SD-fed animals, despite a small but significant
havior than either learning or body weight (Kovacs and Pearce, increase in lymphocyte infiltration in the kidneys (Table S2).

Cell Reports 6, 18, March 13, 2014 2014 The Authors 3


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

SRT1720 has been shown to be effective in reducing HFD- inducible transcription factor, Ankyrin repeat domain 1 (Ankrd1),
induced pathologies (Minor et al., 2011), atherosclerosis (Zeng was observed in the muscle following SRT1720 supplementation
et al., 2013), and myocardial infarction (Tong et al., 2013). (Table S3).
Although overexpression of SIRT1 confers a multidrug resis- The effect of SRT1720 on gene expression was further inves-
tance phenotype in various cancer cell lines in vitro (de Jong tigated using parametric analysis of gene set enrichment (PAGE)
et al., 2011; Oh et al., 2010; Zhu et al., 2012), pharmacological (Figure 3C). Interestingly, the negative transcriptional effect of
activation of SIRT1 by SRT1720 has been associated with reduc- SRT1720 was stronger in muscle than in liver. The expanded
tion in multiple myeloma tumor growth (Chauhan et al., 2011), but list of top up- and downregulated pathways in the liver and mus-
increased lung metastasis of breast cancer cells in a mouse cle of SRT1720-treated mice are presented in Tables S4 and S5.
xenograft model (Suzuki et al., 2012). Harmful effects of this Notably, ribosomal proteins were the most upregulated path-
kind warrant further studies on dosage, timing, and actions in ways in liver (Table S4), and alterations in ribosomal biology
various tissues of SRT1720 relevant to the development of ther- and translation are tightly linked to longevity in other organisms
apeutically useful SIRT1 activators. In the study herein, low levels (Heeren et al., 2009; Houtkooper et al., 2013; Rogers et al.,
of serum markers of hepatic and renal functionAST, creatinine, 2011), which may provide another mechanism through which
and CKwere found with SRT1720 treatment (Figure 2C), indi- SRT1720 exerts its prolongevity effects. One of the top path-
cating that the compound was well tolerated. To ensure that ways to be altered by SRT1720 supplementation in both liver
the ingested SRT1720 reached adequate circulating concentra- and muscle was the inflammation pathway, and quantitative
tions, the serum levels of SRT1720 were measured in the morn- RT-PCR analysis showed significant reduction in select proin-
ing and evening and were found to be 193 30.0 and 339 flammatory mediators in both tissues when compared to SD-fed
23.0 ng/ml, respectively, consistent with the feeding cycle of control animals (Figure 3D). Consistent with this idea, circulating
the mice and with our previous study of HFD-fed mice (Minor tumor necrosis factor (TNF)-a levels were lower in the SRT1720
et al., 2011). To date, there has been no correlation between cohort (Figure 3E), and a number of NF-kB target genes were
STAC concentrations (resveratrol or SRT1720) in vivo and acti- among the top downregulated genes following SRT1720 supple-
vation of SIRT1 in vitro, the reason being that there is controversy mentation (Table S3). Similarly, western blotting of liver lysates
surrounding the methods for measuring SIRT1 enzymatic activity illustrated the significant reduction in protein levels encoding
(Dominy et al., 2013). Thus far, the measurement of the steady- for p65Rel and two known targets of NF-kB, COX2, and BAX
state acetylation status of select sirtuin substrates in vivo (e.g., (Figure 3F). No alteration in SIRT1 protein expression was
PGC-1a, p53, NF-kB, and others) remains an important tool for observed. Although the acetylation status of canonical SIRT1
evaluating changes in sirtuin activity (Dominy et al., 2013). Based targets was not directly assessed, these results led us to hypoth-
on our NF-kB data (see below), we propose that the measured esize that SRT1720 may inhibit NF-kB signaling via SIRT1
plasma concentration of SRT1720 in treated mice is sufficient activation.
to activate SIRT1 in vivo. To explore the molecular mechanisms underlying SRT1720
Whole-genome DNA microarrays were performed on liver and control of inflammatory signaling, murine embryonic fibroblasts
muscle samples of SD-fed mice supplemented or not with (MEFs) derived from SIRT1-knockout mice were used for
SRT1720. Principal component analysis showed a distinct sep- cDNA microarray analysis (Figure 4; Table S6) in conjunction
aration of both treatment groups, with the effect of SRT1720 with phosphoantibody microarray (Table S7). Using the latter
supplementation being more pronounced in the muscle tissue method, it has previously been demonstrated that SIRT1 dele-
(Figure 3A versus 3B). The top ten up- and downregulated genes tion is associated with constitutive activation of the proinflamma-
in both tissues are shown in Table S3, with the complete data set tory NF-kB pathway (Bernier et al., 2011). Overexpression of
made available at http://ncbi.nlm.nih.gov/geo/. Among the top SIRT1 is associated with reduced NF-kB activity in mice (Pfluger
upregulated genes, members of the cytokine-induced STAT in- et al., 2008), whereas SIRT1 activation by SRT1720 reduces
hibitor (CIS) family (e.g., suppressor of cytokine signaling 2 expression of NF-kB and its targets in foam cells (Zeng et al.,
[SOCS2] and cytokine-inducible SH2-containing protein 2013) and multiple myeloma cells (Chauhan et al., 2011). Here,
[CISH]), which are associated with suppression of the inflamma- it was found that the expression level of a few genes was influ-
tory response (Kubo et al., 2003), were significantly higher in the enced by SRT1720 in a SIRT1-independent manner (e.g., Id2),
liver and muscle of SRT1720-treated mice. Similarly, a 7-fold in- whereas the expression of other genes, including Has2,
crease in hepatic expression of cytochrome P450 1A2, which Gadd45a, Ddit3, Thbs1, and Ifitm3, required SIRT1 following
has a role in xenobiotic, cholesterol, and lipid metabolism (Haf- MEF stimulation with SRT1720 (Table S6). Importantly,
ner et al., 2011), was observed in SRT1720-treated mice and SRT1720 induced significant changes in the phosphorylation
may explain the lower circulating serum cholesterol and LDL status of several proteins involved in NF-kB activation in wild-
levels in these animals. These results are consistent with a recent type MEFs, but not Sirt1-KO MEFs (Table S7). Given that low-
report showing that SRT1720 ameliorates fatty liver through a grade chronic inflammation is believed to contribute to aging
reduction in lipogenic enzyme expression in monosodium gluta- and age-related diseases (Franceschi et al., 2000), our results
mate-treated mice (Yamazaki et al., 2009). Lipocalin 2 (Lcn2) and indicate that SRT1720 may keep nonobese mice healthier and
serum amyloid A1 and A2 (Saa1, Saa2), which are involved in extend their mean lifespan by spurring robust inflammatory de-
innate immunity and the acute phase response, respectively, fense. Indeed, SRT1720 exerts antiinflammatory properties in
were the top three most downregulated genes in the liver of mouse models of asthma (Ichikawa et al., 2013) and emphysema
SRT1720-treated animals. Downregulation of the cytokine- (Yao et al., 2012).

4 Cell Reports 6, 18, March 13, 2014 2014 The Authors


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Figure 3. SRT1720 Elicits Differential Gene Expression Profiles in the Liver and Muscle of SD-Fed Mice
(A and B) Principal component analysis (PCA) was performed on liver (A) and skeletal (B) muscle of SD-fed mice supplemented without and with SRT1720.
(C) Parametric analysis of gene-set enrichment (PAGE) analysis was performed on microarray data. Columns show significantly up- (red) and downregulated
(blue) pathways following SRT1720 supplementation.
(D) mRNA expression analysis in liver and skeletal muscle by quantitative real-time PCR. Relative expression values were normalized to those of SD-fed control mice.
(E) Serum tumor necrosis factor alpha (TNF-a) concentrations in SD-fed mice supplemented or not with SRT1720. Data are shown as mean SEM.
(F) Western blotting of liver lysates from SD-fed mice supplemented or not with SRT1720. Upper-left panel, representative blots; remaining panels, Signals
associated with bands of interest were normalized to GAPDH and plotted.
*p % 0.05; **p % 0.01; ***p % 0.001 in comparison to SD-fed control animals.

In summary, we report data showing that SRT1720 elicits life- (drug/kg body weight). Study diets were purchased from Dyets. SRT1720
span extension and improved healthspan in mice maintained was provided by Sirtris Pharmaceuticals. All groups had ad libitum access
to their prescribed diet and water throughout the study. Body weight and
on a standard diet. This work highlights the importance of exam-
food intake were monitored biweekly. Animal rooms were maintained at 20
ining the therapeutic value of small molecule activators of SIRT1 22 C with 30%70% relative humidity and a 12 hr light/dark cycle. All animal
in general health and longevity. protocols were approved by the Animal Care and Use Committee (352-LEG-
2012) of the National Institute on Aging.
EXPERIMENTAL PROCEDURES
Survival Study
Animals and Diets Moribund animals were euthanized, and every animal found dead or eutha-
Male C57BL/6J mice were obtained from the Jackson Laboratory at 15 weeks nized was necropsied. Additional information is available in the Supplemental
of age and housed at the Gerontology Research Center, in Baltimore, MD, in Information.
cages of four. Diets were started at 28 weeks of age after randomization into
four groups of 100 mice per group. Mice were fed one of four diets: standard Histology
diet (SD) of AIN-93G (carbohydrate:protein:fat ratio of 64:19:17 percent of Organs were fixed in 4% paraformaldehyde then stained with hematoxylin and
kcal), an SD supplemented with SRT1720 (SD-SRT1720), a high-fat diet eosin for scoring of pathology by a histopathologist blinded to the treatment
consisting of AIN-93G modified to provide 60% of calories from fat (HFD; group.
carbohydrate:protein:fat ratio of 16:23:61), or a HFD supplemented with
SRT1720 (HFD-SRT1720). SRT1720 was included in diets at a concentration Determination of Serum Marker Concentrations and HOMA-IR
of 1.33 g/kg (SD) and 2 g/kg (HFD; g drug/kg chow), respectively, which was Calculation
formulated to provide mice with approximate daily doses of 100 mg/kg Detailed information can be found in the Supplemental Information.

Cell Reports 6, 18, March 13, 2014 2014 The Authors 5


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Z-normalization, as described elsewhere (Cheadle et al., 2003; Lee et al.,


2012). Additional information is available in the Supplemental Information.

Quantitative Real-Time RT-PCR


Total RNA was extracted from frozen tissue samples or cells using the RNeasy
kit (QIAGEN). Additional information can be found in the Supplemental
Information.

Phosphoprotein Profiling
The Phospho Explorer antibody microarray from Full Moon Biosystems was
used to survey the phosphorylation state of select proteins involved in the
IKK/NF-kB activation pathway, according to the protocol outlined in Bernier
et al. (2011). In brief, wild-type and Sirt1-KO murine embryonic fibroblasts
(gift of Raul Mostoslavsky, Harvard Medical School) were incubated with
vehicle or 3 mM SRT1720 for 18 hr, after which cell lysates were prepared
and biotinylated. The antibody array experiment was performed by Full
Moon Biosystems and the data were analyzed with GenePix Pro 6.0 (Molecular
Devices). Each of the 1,318 antibodies had two replicates printed on a coated
microscope slide, along with multiple positive and negative controls.

Statistics
Figure 4. SIRT1 Dependence in SRT1720-Mediated Gene Expres-
Data are expressed as means SEM. Students t tests were used for all
sion in MEF Cells
comparisons unless otherwise stated. Mortality during the survival study
Microarray data collected from untreated (UT) and SRT1720-treated wild-type
was assessed through the use of the nonparametric log-rank (Mantel-
(WT) and Sirt1-KO MEFs were used to illustrate enrichment of select genes. A
Haenszel) test to compare the differences in Kaplan-Meier survival curves.
larger list of genes can be found in Table S6.
Median survival was assessed using the nonparametric chi-square test (Su
and Wei, 1993). Ninetieth percentile survival estimates were used as surrogate
Oral Glucose Tolerance Test measures of maximum survival times according to the methods of Wang et al.
Following an overnight fast (18 hr), mice received an oral gavage of 30% (2004). The survival analyses were implemented in R (The R Development Core
glucose solution (Sigma-Aldrich) at a dose of 2 g/kg body weight. Blood Team) from scratch using the methodological references given. Comparisons
glucose was measured using an Ascensia Elite glucose meter at 0, 15, 30, for the histopathology data were performed using Fishers exact test. Analyses
60, and 120 min following gavage (n = 6 SD, n = 8 SD-SRT1720, 73 weeks were performed using Excel 2010 (Microsoft), IBM SPSS Statistics, or
age, 46 weeks on diet). SigmaStat 3.0 (Aspire Software International). A p value of % 0.05 was consid-
ered statistically significant.
Body Composition
Measurements of lean body mass, fat, and fluid mass in live mice were
ACCESSION NUMBERS
acquired by nuclear magnetic resonance spectroscopy using the Minispec
LF90 (Bruker Optics) (n = 84 SD, n = 93 SD-SRT1720, n = 51 HFD, n = 73
All raw data were deposited to the NCBI Gene Expression Omnibus under
HFD-SRT1720, 76 weeks age, 49 weeks on diet).
accession number GSE50987.
Metabolic Assessment
Mouse metabolic rate was assessed by indirect calorimetry in open-circuit SUPPLEMENTAL INFORMATION
Oxymax chambers with CLAMS (Columbus Instruments) as previously
described (Minor et al., 2011) (n = 8 SD, n = 8 SD-SRT1720, n = 7 HFD, Supplemental Information includes Supplemental Experimental Procedures,
n = 8 HFD-SRT1720, 52 weeks of age, 25 weeks on diet). two figures, and seven tables and can be found with this article online at
http://dx.doi.org/10.1016/j.celrep.2014.01.031.
Rotarod
All mice were acclimated 15 min before testing. Mice were tested at the same ACKNOWLEDGMENTS
time of day during their light cycle over a 5-day period. Mice were given a habit-
uation trial at a constant speed of 4 rpm for 1 min before the first trial. On the This research was conducted under a Cooperative Research and Develop-
same day, there was a total of three trials given, separated by 30 min rest ment Agreement (CRADA) between Sirtris, a GSK Company, and the National
periods, during which the rotarod accelerated from 4 to 40 rpm over a period Institute on Aging, National Institutes of Health (NIA/NIH). Funding was pro-
of 5 min. Latency to fall was recorded and averaged over all three trials. This vided by the Intramural Research Program of the NIA/NIH. We are grateful
test was completed on the same mice at 13, 18, and 24 months of age. to Dawn Nines, Justine Lucas, and Dawn Phillips-Boyer for their excellent an-
imal care. We thank Dr. Elin Lehrmann for microarray assistance. We thank
Cataract Formation Drs. Hua-Jun He (National Institute of Standards and Technology, Gaithers-
Lens opacity reading was performed using a Kowa SL14 hand-held slit lamp burg, MD), Yaping Zong (Full Moon Biosystems, Inc., Sunnyvale, CA), and
(Kowa) by an experienced pathologist blinded to the experimental group, Xiong Li (Department of Mathematics and Computer Science, Emory Univer-
as described previously (Wolf et al., 2005) (n = 138 eyes SD, n = 142 eyes sity, Atlanta, GA) for their contribution in carrying out the phosphoantibody
SD-SRT1720, 105 weeks age, 78 weeks diet). array experiment and data analysis. We thank Dr. Norm Wolf for the assess-
ment of cataracts in the mice and Olga Carlson for technical assistance with
Western Blotting the multiplex assay. S.J.M. was supported by a National Medical Health and
Detailed information can be found in the Supplemental Information. Research Council of Australia CJ Martin Early Career Fellowship (RGMS ID
2010-01671). J.D. is supported by a University of Alabama at Birmingham Sta-
Microarray tistical Genetics Postdoctoral Training Program Grant (T32HL072757). D.A.S.
RNA from tissues was isolated using the RNeasy kit (QIAGEN) and then consults for and G.P.V. and J.L.E. are employed by Sirtris, a GSK Company
hybridized to BD-202-0202 Illumina BeadChips. Raw data were subjected to that has a commercial interest in developing SIRT1 activators.

6 Cell Reports 6, 18, March 13, 2014 2014 The Authors


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Received: July 18, 2013 Houtkooper, R.H., Mouchiroud, L., Ryu, D., Moullan, N., Katsyuba, E., Knott,
Revised: December 16, 2013 G., Williams, R.W., and Auwerx, J. (2013). Mitonuclear protein imbalance as
Accepted: January 23, 2014 a conserved longevity mechanism. Nature 497, 451457.
Published: February 27, 2014 Hubbard, B.P., Gomes, A.P., Dai, H., Li, J., Case, A.W., Considine, T., Riera,
T.V., Lee, J.E., e, S.Y., Lamming, D.W., et al. (2013). Evidence for a common
REFERENCES mechanism of SIRT1 regulation by allosteric activators. Science 339, 1216
1219.
Banks, A.S., Kon, N., Knight, C., Matsumoto, M., Gutierrez-Juarez, R., Ros-
Huber, J.L., McBurney, M.W., Distefano, P.S., and McDonagh, T. (2010).
setti, L., Gu, W., and Accili, D. (2008). SirT1 gain of function increases energy
SIRT1-independent mechanisms of the putative sirtuin enzyme activators
efficiency and prevents diabetes in mice. Cell Metab. 8, 333341.
SRT1720 and SRT2183. Future Med Chem 2, 17511759.
Baur, J.A., Pearson, K.J., Price, N.L., Jamieson, H.A., Lerin, C., Kalra, A.,
Ichikawa, T., Hayashi, R., Suzuki, K., Imanishi, S., Kambara, K., Okazawa, S.,
Prabhu, V.V., Allard, J.S., Lopez-Lluch, G., Lewis, K., et al. (2006). Resveratrol
Inomata, M., Yamada, T., Yamazaki, Y., Koshimizu, Y., et al. (2013). Sirtuin 1
improves health and survival of mice on a high-calorie diet. Nature 444, 337342.
activator SRT1720 suppresses inflammation in an ovalbumin-induced mouse
Baur, J.A., Ungvari, Z., Minor, R.K., Le Couteur, D.G., and de Cabo, R. (2012). model of asthma. Respirology 18, 332339.
Are sirtuins viable targets for improving healthspan and lifespan? Nat. Rev.
Drug Discov. 11, 443461. Kovacs, A.D., and Pearce, D.A. (2013). Location- and sex-specific differences
in weight and motor coordination in two commonly used mouse strains. Sci
Bellet, M.M., Nakahata, Y., Boudjelal, M., Watts, E., Mossakowska, D.E.,
Rep 3, 2116.
Edwards, K.A., Cervantes, M., Astarita, G., Loh, C., Ellis, J.L., et al. (2013).
Pharmacological modulation of circadian rhythms by synthetic activators of Kubo, M., Hanada, T., and Yoshimura, A. (2003). Suppressors of cytokine
the deacetylase SIRT1. Proc. Natl. Acad. Sci. USA 110, 33333338. signaling and immunity. Nat. Immunol. 4, 11691176.

Bernier, M., Paul, R.K., Martin-Montalvo, A., Scheibye-Knudsen, M., Song, S., Lamming, D.W., Ye, L., Katajisto, P., Goncalves, M.D., Saitoh, M., Stevens,
He, H.J., Armour, S.M., Hubbard, B.P., Bohr, V.A., Wang, L., et al. (2011). D.M., Davis, J.G., Salmon, A.B., Richardson, A., Ahima, R.S., et al. (2012).
Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 Rapamycin-induced insulin resistance is mediated by mTORC2 loss and
protein. J. Biol. Chem. 286, 1927019279. uncoupled from longevity. Science 335, 16381643.
Chauhan, D., Bandi, M., Singh, A.V., Ray, A., Raje, N., Richardson, P., and Lee, C., Raffaghello, L., Brandhorst, S., Safdie, F.M., Bianchi, G., Martin-Mon-
Anderson, K.C. (2011). Preclinical evaluation of a novel SIRT1 modulator talvo, A., Pistoia, V., Wei, M., Hwang, S., Merlino, A., et al. (2012). Fasting
SRT1720 in multiple myeloma cells. Br. J. Haematol. 155, 588598. cycles retard growth of tumors and sensitize a range of cancer cell types to
chemotherapy. Sci. Translat. Med. 4, 124ra127.
Cheadle, C., Vawter, M.P., Freed, W.J., and Becker, K.G. (2003). Analysis of
microarray data using Z score transformation. J. Mol. Diagn. 5, 7381. Martin-Montalvo, A., Mercken, E.M., Mitchell, S.J., Palacios, H.H., Mote, P.L.,
de Jong, E., Winkel, P., Poelstra, K., and Prakash, J. (2011). Anticancer effects Scheibye-Knudsen, M., Gomes, A.P., Ward, T.M., Minor, R.K., Blouin, M.J.,
of 15d-prostaglandin-J2 in wild-type and doxorubicin-resistant ovarian cancer et al. (2013). Metformin improves healthspan and lifespan in mice. Nat Com-
cells: novel actions on SIRT1 and HDAC. PLoS ONE 6, e25192. mun 4, 2192.

Deblon, N., Bourgoin, L., Veyrat-Durebex, C., Peyrou, M., Vinciguerra, M., Milne, J.C., Lambert, P.D., Schenk, S., Carney, D.P., Smith, J.J., Gagne, D.J.,
Caillon, A., Maeder, C., Fournier, M., Montet, X., Rohner-Jeanrenaud, F., Jin, L., Boss, O., Perni, R.B., Vu, C.B., et al. (2007). Small molecule activators
and Foti, M. (2012). Chronic mTOR inhibition by rapamycin induces muscle in- of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450,
sulin resistance despite weight loss in rats. Br. J. Pharmacol. 165, 23252340. 712716.

Dominy, J., Puigserver, P., and Canto, C. (2013). In vivo measurement of the Minor, R.K., Baur, J.A., Gomes, A.P., Ward, T.M., Csiszar, A., Mercken, E.M.,
acetylation state of sirtuin substrates as a proxy for sirtuin activity. Methods Abdelmohsen, K., Shin, Y.K., Canto, C., Scheibye-Knudsen, M., et al. (2011).
Mol. Biol. 1077, 217237. SRT1720 improves survival and healthspan of obese mice. Sci Rep 1, 70.
Feige, J.N., Lagouge, M., Canto, C., Strehle, A., Houten, S.M., Milne, J.C., Oh, W.K., Cho, K.B., Hien, T.T., Kim, T.H., Kim, H.S., Dao, T.T., Han, H.K.,
Lambert, P.D., Mataki, C., Elliott, P.J., and Auwerx, J. (2008). Specific SIRT1 Kwon, S.M., Ahn, S.G., Yoon, J.H., et al. (2010). Amurensin G, a potent natural
activation mimics low energy levels and protects against diet-induced meta- SIRT1 inhibitor, rescues doxorubicin responsiveness via down-regulation of
bolic disorders by enhancing fat oxidation. Cell Metab. 8, 347358. multidrug resistance 1. Mol. Pharmacol. 78, 855864.
Franceschi, C., Bonafe`, M., Valensin, S., Olivieri, F., De Luca, M., Ottaviani, E., Pacholec, M., Bleasdale, J.E., Chrunyk, B., Cunningham, D., Flynn, D., Garo-
and De Benedictis, G. (2000). Inflamm-aging. An evolutionary perspective on falo, R.S., Griffith, D., Griffor, M., Loulakis, P., Pabst, B., et al. (2010). SRT1720,
immunosenescence. Ann. N Y Acad. Sci. 908, 244254. SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J. Biol.
Froy, O. (2011). Circadian rhythms, aging, and life span in mammals. Physi- Chem. 285, 83408351.
ology (Bethesda) 26, 225235. Pearson, K.J., Baur, J.A., Lewis, K.N., Peshkin, L., Price, N.L., Labinskyy, N.,
Hafner, M., Rezen, T., and Rozman, D. (2011). Regulation of hepatic cyto- Swindell, W.R., Kamara, D., Minor, R.K., Perez, E., et al. (2008). Resveratrol
chromes p450 by lipids and cholesterol. Curr. Drug Metab. 12, 173185. delays age-related deterioration and mimics transcriptional aspects of dietary
restriction without extending life span. Cell Metab. 8, 157168.
Harrison, D.E., Strong, R., Sharp, Z.D., Nelson, J.F., Astle, C.M., Flurkey, K.,
Nadon, N.L., Wilkinson, J.E., Frenkel, K., Carter, C.S., et al. (2009). Rapamycin Pfluger, P.T., Herranz, D., Velasco-Miguel, S., Serrano, M., and Tschop, M.H.
fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, (2008). Sirt1 protects against high-fat diet-induced metabolic damage. Proc.
392395. Natl. Acad. Sci. USA 105, 97939798.
Heeren, G., Rinnerthaler, M., Laun, P., von Seyerl, P., Kossler, S., Klinger, H., Rogers, A.N., Chen, D., McColl, G., Czerwieniec, G., Felkey, K., Gibson, B.W.,
Hager, M., Bogengruber, E., Jarolim, S., Simon-Nobbe, B., et al. (2009). The Hubbard, A., Melov, S., Lithgow, G.J., and Kapahi, P. (2011). Life span exten-
mitochondrial ribosomal protein of the large subunit, Afo1p, determines sion via eIF4G inhibition is mediated by posttranscriptional remodeling of
cellular longevity through mitochondrial back-signaling via TOR1. Aging stress response gene expression in C. elegans. Cell Metab. 14, 5566.
(Albany, N.Y. Online) 1, 622636. Strong, R., Miller, R.A., Astle, C.M., Baur, J.A., de Cabo, R., Fernandez, E.,
Houde, V.P., Brule, S., Festuccia, W.T., Blanchard, P.G., Bellmann, K., Guo, W., Javors, M., Kirkland, J.L., Nelson, J.F., et al. (2013). Evaluation of
Deshaies, Y., and Marette, A. (2010). Chronic rapamycin treatment causes resveratrol, green tea extract, curcumin, oxaloacetic acid, and medium-chain
glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogen- triglyceride oil on life span of genetically heterogeneous mice. J. Gerontol. A
esis and impairing lipid deposition in adipose tissue. Diabetes 59, 13381348. Biol. Sci. Med. Sci. 68, 616.

Cell Reports 6, 18, March 13, 2014 2014 The Authors 7


Please cite this article in press as: Mitchell et al., The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet,
Cell Reports (2014), http://dx.doi.org/10.1016/j.celrep.2014.01.031

Su, J.Q., and Wei, L.J. (1993). Nonparametric estimation for the difference or Yamazaki, Y., Usui, I., Kanatani, Y., Matsuya, Y., Tsuneyama, K., Fujisaka, S.,
ratio of median failure times. Biometrics 49, 603607. Bukhari, A., Suzuki, H., Senda, S., Imanishi, S., et al. (2009). Treatment with
Suzuki, K., Hayashi, R., Ichikawa, T., Imanishi, S., Yamada, T., Inomata, M., SRT1720, a SIRT1 activator, ameliorates fatty liver with reduced expression
Miwa, T., Matsui, S., Usui, I., Urakaze, M., et al. (2012). SRT1720, a SIRT1 acti- of lipogenic enzymes in MSG mice. Am. J. Physiol. Endocrinol. Metab. 297,
vator, promotes tumor cell migration, and lung metastasis of breast cancer in E1179E1186.
mice. Oncol. Rep. 27, 17261732. Yao, H., Chung, S., Hwang, J.W., Rajendrasozhan, S., Sundar, I.K., Dean,
Tong, C., Morrison, A., Mattison, S., Qian, S., Bryniarski, M., Rankin, B., Wang, D.A., McBurney, M.W., Guarente, L., Gu, W., Ronty, M., et al. (2012). SIRT1
J., Thomas, D.P., and Li, J. (2013). Impaired SIRT1 nucleocytoplasmic shut- protects against emphysema via FOXO3-mediated reduction of premature
tling in the senescent heart during ischemic stress. FASEB J. 27, 43324342. senescence in mice. J. Clin. Invest. 122, 20322045.
Wang, C., Li, Q., Redden, D.T., Weindruch, R., and Allison, D.B. (2004). Statis- Zeng, H.T., Fu, Y.C., Yu, W., Lin, J.M., Zhou, L., Liu, L., and Wang, W. (2013).
tical methods for testing effects on maximum lifespan. Mech. Ageing Dev. SIRT1 prevents atherosclerosis via liver-X-receptor and NF-kB signaling in a
125, 629632. U937 cell model. Mol Med Rep 8, 2328.
Wolf, N., Penn, P., Pendergrass, W., Van Remmen, H., Bartke, A., Rabinovitch, Zhu, H., Xia, L., Zhang, Y., Wang, H., Xu, W., Hu, H., Wang, J., Xin, J., Gang, Y.,
P., and Martin, G.M. (2005). Age-related cataract progression in five mouse Sha, S., et al. (2012). Activating transcription factor 4 confers a multidrug
models for anti-oxidant protection or hormonal influence. Exp. Eye Res. 81, resistance phenotype to gastric cancer cells through transactivation of
276285. SIRT1 expression. PLoS ONE 7, e31431.

8 Cell Reports 6, 18, March 13, 2014 2014 The Authors

Das könnte Ihnen auch gefallen