Vous êtes sur la page 1sur 5

Leading Edge

Essay

Aging Research—Where Do We Stand


and Where Are We Going?
Leonard Guarente1,*
1Department of Biology, Koch Institute for Integrative Cancer Research and Paul F. Glenn Laboratories for the Science of Aging, MIT,

Cambridge, MA 02139, USA


*Correspondence: leng@mit.edu
http://dx.doi.org/10.1016/j.cell.2014.08.041

The 40th anniversary of Cell coincides with that of the National Institute on Aging (NIA). Indeed, Cell
papers on NIA-funded research helped move the field into a genetic and molecular era. Now is a fair
time to ask whether we are far down a trail leading to a deep understanding of aging or whether we
are still tiptoeing cautiously at the trailhead.

There is a general perception that the ef- idea has attracted much attention, this era, the most prominent ideas were
fects of aging have somehow been we must also consider that the complex- that aging was simply due to wear and
slowed during our lifetimes. Memories ities of aging processes likely exceed tear and, more recently, to the accumula-
and photographs of our grandparents those of specific diseases, and the chal- tion of oxidative damage in cells. The first
when they were in their 60s look more lenge of reigning in the global decline of genetic pathway implicated in aging was
like people in their 70s or even 80s today. cellular processes across many tissues found in C. elegans, where Tom Johnson
To address more rigorously whether ef- will be large. showed that hypomorphic mutations in a
fects of aging have indeed been slowed, At the cellular level, aging induces single gene age-1 could extend the life
we must first define what we mean by many potentially interconnected defects, span (Johnson, 2013), and Cynthia Ken-
aging. One metric comes from human including DNA damage in the nucleus yon and Gary Ruvkun defined a genetic/
actuarial data and from studies in lower and mitochondria, mitochondrial dys- molecular pathway involving insulin/
laboratory organisms like yeast, worms, function leading to increased production insulin-like growth factor (IGF) signaling,
and flies—the average and maximum life of reactive oxidative species (ROS) and which included age-1 (Kenyon, 2010).
spans. In mammalian systems, many decreased production of ATP, oxidative Downregulation of this pathway extended
more consequences of aging are readily damage to proteins and other macromol- the life span, but knocking it out entirely
measurable, such as functional decline ecules in cells, protein misfolding and ag- could be deleterious, and these effects
in performance tests, deterioration of indi- gregation, protein glycation, the induction are conserved in organisms ranging from
vidual tissues, and degradation in broad of proinflammatory cytokines, telomere worms to mice.
measures of metabolic health. These shortening, and cell senescense (López- My own lab was also interested in ag-
latter measures define the health span of Otı́n et al., 2013) (Figure 1). These will ing, and in 1991, two entering graduate
the organism, the maintenance of which impact mitotically active tissues over students, Brian Kennedy (now CEO of
I believe to be the most important goal time by triggering stem cell depletion by the Buck Institute) and Nicanor Austriaco
of ongoing aging research. Are we making senescence and apoptosis (e.g., intesti- (now a Franciscan priest and advisor to
significant progress? nal stem cells, hematopoietic stem cells, the Vatican) began studying the aging of
The magnitude of the challenge is illus- mesenchymal stem cells, etc.), and post- yeast mother cells, which senesce after
trated by considering known causes of mitotic tissues by causing cellular giving rise to 20–30 daughter cells. SIR2
aging. The good news is that many mech- dysfunction and loss (e.g., muscle, heart, emerged from these studies as an impor-
anisms causing aging, as well as path- and brain). Beyond tissue-autonomous tant gene combating yeast mother cell
ways that can mitigate effects of aging, aging, it is now clear that the brain helps aging, and it is noteworthy that three Cell
have been identified. This is also the bad govern aging of many organs (Satoh papers marked the trail of this research
news—aging processes and pathways et al., 2013; Chang and Guarente, 2014), (Kennedy et al., 1995; Smeal et al., 1996;
offering an ability to modify their effects i.e., dysfunction in the hypothalamus will Kennedy et al., 1997). In this case, the
(discussed below) are extremely com- exert systemic effects leading to func- modest upregulation of yeast SIR2
plex. It is widely assumed that aging is a tional decline and damage to cells and extended life span, but more prolific over-
major risk factor for most late-onset dis- organs. expression could be toxic. Indeed, a more
eases (cancer, cardiovascular disease, Offering some degree of hope, over the recent genome-wide quantitative trait lo-
diabetes, neurodegenerative diseases, past quarter century, numerous genetic cus (QTL) analysis identified SIR2 as the
etc.), and therefore interventions directed pathways in model organisms have been single most important yeast gene in deter-
at aging offer an opportunity to ameliorate identified that can confront at least some mining the difference in mother cell life
all these diseases at once. Although this of the villains of aging (Figure 1). Before span between a lab strain and a clinical

Cell 159, September 25, 2014 ª2014 Elsevier Inc. 15


aging in primates. Two studies in
nonhuman primates came to opposite
conclusions as to whether CR extends
life span (Colman et al., 2009; Mattison
et al., 2012). Both studies, as well as
short-term human studies (Redman
et al., 2011), do agree that CR can
improve metabolic parameters, such as
glucose metabolism. Although it is
possible that CR could extend human
Figure 1. The Complexity of Aging life span, it is also possible that it would
Shown is a cell under the forces of aging (causes depicted inside and mitigating pathways depicted not, if it also exerts negative effects that
outside). Possible small-molecule interventions are also indicated.
cancel the metabolic benefit (e.g., unto-
ward effects on the immune system).
isolate (Stumpferl et al., 2012). SIR2 or- lin/IGF and sirtuins, both affected meta- Although difficult to conduct, more
thologs have also been shown to extend bolism offered the possibility that CR extended studies of the effects of CR on
life span in worms, flies, and mice (Guar- might extend life span by altering specific humans would be informative.
ente, 2013). pathways. For sirtuins, the data support- Another aspect of aging has recently
There was also a burning interest in that ing this idea are many (Guarente, 2013). emerged from the identification of NAD+
early period in what Sir2 protein’s First, CR induces the levels and activities as the sirtuin cosubstrate. We first sus-
biochemical activity might be. One clue of several sirtuins. Second, knocking out pected that changes in NAD+ or the
was that SIR2 was associated with yeast any one sirtuin gene abolishes at least NAD+/NADH ratio activate sirtuins and
genomic silencing, itself linked to the de- some of the phenotypes of CR, including help drive effects of CR (Guarente, 2000),
acetylation of histones (Braunstein et al., extended life span (SIRT1). Third, trans- a finding later supported by experimenta-
1996). But demonstrating any deacetyla- genic mice overexpressing SIRT1 in brain tion (Guarente, 2013). What we did not
tion activity by Sir2 protein in vitro initially or SIRT6 globally live longer and show a anticipate are the more recent findings
proved impossible. Another clue came better metabolic profile and retention of that NAD+ levels appear to decline during
from a somewhat baroque study showing tissue integrity as they age (Kanfi et al., aging across a broad spectrum of species
that Sir2 protein had a very weak ability to 2012; Satoh et al., 2013). Fourth, many (Ramsey et al., 2008; Mouchiroud et al.,
transfer ADP-ribose from NAD+ to the of the substrates deacylated and thus 2013). There are two main hypotheses to
irrelevant substrate bovine serum albumin activated by sirtuins are precisely those explain this decline (Figure 2). First, aging
in vitro (Frye, 1999). Through a series of mediating important physiological effects is associated with cumulative damage
serendipitous experiments, my then- of CR, i.e., induction of oxidative mito- to nuclear DNA, and the chronic acti-
postdoc Shin Imai and I were able to arrive chondrial metabolism and its accompa- vation of the DNA repair enzyme poly-
at the conclusion that yeast Sir2 protein nying stress resistance (Verdin et al., ADP-ribose polymerase (PARP) would
and its mammalian ortholog, SIRT1, 2010; Guarente 2013). Fifth, SIRT1 acti- consume NAD+ as it ADP-ribosylates pro-
were NAD+-dependent protein deacety- vator compounds (STACs) have been teins at sites of damage (Bai et al., 2011;
lases (Imai et al., 2000), thereby linking identified by screening for molecules Mouchiroud et al., 2013). Second, the
metabolism and aging. Sir2-related pro- that activate the enzyme in vitro, and gene encoding the NAD+ synthesis
teins are termed sirtuins, and in mam- these include polyphenols and more spe- enzyme nicotinamide phosphoribosyl
mals, the seven sirtuin proteins are desig- cific novel chemical entities (Sinclair and transferase (NAMPT) is activated by the
nated SIRT1–7. These seven proteins are Guarente, 2014). Importantly, STACs circadian clock and is a major link between
all encoded by nuclear genes but mainly have been shown to extend life span in the clock and the diurnal staging of meta-
reside in discrete cellular compart- mice on the normal diet (Mercken et al., bolic reactions (Nakahata et al., 2009;
ments—the nucleus (SIRT1, SIRT6, and 2014), slow the progression to tissue Ramsey et al., 2009). For example, fat
SIRT7), the cytosol (SIRT2), and the dysfunction in mice and nonhuman pri- oxidation in mitochondria is circadian due
mitochondria (SIRT3, SIRT4, and SIRT5) mates (Mattison et al., 2014), and induce to pulsatile NAD+ synthesis and SIRT3
(Verdin et al., 2010). patterns of gene expression resembling activation, and knocking out SIRT3 dis-
Metabolism and aging had already CR (Guarente, 2013). rupts this temporal organization (Peek
been linked in the 1930s by the diet CR may be viewed as high up on the et al., 2013). Because the amplitude of
termed calorie restriction (CR), which pyramid of health, with a potential to re- the central clock in the suprachiasmatic
became the gold standard to slow aging dress many of the cellular and tissue de- nucleus of the hypothalamus (and perhaps
and extend the life span in rodents. fects occurring during aging (Figure 1). cell-autonomous clocks in other tissues)
Many had assumed that CR worked by However, there are major issues to may decay during aging in mammals
generally slowing down metabolism and consider before this presumption can be (Chang and Guarente, 2013), a decline in
the presumed accompanying oxidative considered a certainty, at least as it might NAMPT from a reduction in the output of
damage in cells. But the fact that the first apply to humans. In particular, there is a the circadian clock may result in a decline
two longevity pathways described, insu- paucity of data on the effects of CR on in NAD+ synthesis with age.

16 Cell 159, September 25, 2014 ª2014 Elsevier Inc.


The expression of SIRT3 and SIRT6, in
contrast, has also been associated with
beneficial effects that overlap CR,
including suppression of tumor growth. It
might be an important addition to the
tool kit of health span interventions if
small-molecule activators of these latter
two sirtuins can be found.
Fascinating, newer approaches to
study aging are also emerging and may
yield new strategies for intervention. Para-
biosis is a classic procedure in which the
circulations of two mice are connected
surgically. By thus fusing genetically iden-
tical old and young mice, some of the ef-
Figure 2. NAD+ Declines with Aging fects of aging in an old mouse can be
Shown are two possible mechanisms of decline. The left depicts a reduction in synthesis due to a decline ameliorated, a finding that would please
in the amplitude in the circadian clock with aging. The right depicts an increase in degradation due to
chronic DNA damage and PARP activation with aging. Bram Stoker (Conboy et al., 2013). This
approach has already led to the identifica-
tion of specific molecules from the young
Importantly, this aging-induced decline activates many aspects of mitochondria, mouse, which appear to rejuvenate
in NAD+ may be reversed in rodents by including biogenesis, oxidative meta- aspects of aging of the old mouse (Kat-
supplementing the diet with the NAD+ bolism, and an accompanying oxidative simpardi et al., 2014). How such circu-
precursors nicotinamide mononucleotide stress resistance. However, it is still not lating molecules identified by parabiosis
(NMN) (Ramsey et al., 2008; Yoshino clear how many of the vast number of might relate to the metabolic pathways
et al., 2011) or nicotinamide riboside degenerative changes in aging can be mentioned above is not clear at this point.
(NR) (Cantó et al., 2012). These com- influenced by this network of metabolic Another current approach in aging
pounds can ameliorate or even reverse pathways. research is to study centenarians (people
metabolic defects in mice, in part by With regard to intervening pharmaco- 100 years old or older), and especially
restoring proper stoichiometry in the logically in the above pathways, one families in which centenarians cluster
expression of nuclear- and mitochondri- must be cautious in concluding that significantly (Sebastiani and Perls, 2012;
ally encoded mitochondrial proteins via modulating any of them would be an Milman et al., 2013). The idea is to identify
SIRT1 reactivation (Gomes et al., 2013). effective way to mitigate effects of aging. genetic factors that explain the extreme
It will be interesting to see whether For example, downregulation of mTOR longevity of these rare individuals. This
combining NMN or NR with STACs exerts by rapamycin extends murine life span approach would seem to hold consider-
a synergistic effect in treating the decline (Shimobayashi and Hall, 2014). But an able promise, but we must keep in mind
associated with aging in rodents and pri- exhaustive examination of the tissues of that identical twin studies suggest that
mates. treated mice compared to untreated con- genes account for only about 25% of hu-
Since 2000, other pathways have also trols showed that, in some tissues, rapa- man life span.
been implicated in aging, including AMP mycin indeed preserved integrity in old In addition, we may have but scratched
kinase (AMPK) (Gowans and Hardie, animals, whereas in others, it had no ef- the surface of what bioinformatics can
2014), which is upregulated when energy fects, and in still others, it had negative ef- provide in identifying new genes and
is limiting, and target of rapamycin (TOR) fects (Neff et al., 2013). It remains possible pathways important in human aging, as
(Shimobayashi and Hall, 2014), a nutrient that at least some of the effect on life span well as allowing for the knowledge we
sensor that must be downregulated, by by rapamycin in mice is due to the slowing have already gained to be applied in a
rapamycin, for example, for beneficial ef- of cancer. Likewise, blockade of the IGF1 more effective, personalized way. Anal-
fects. It is now clear that these pathways receptor in humans would run the risk of ysis of the transcriptome, epigenome,
and the insulin/IGF and sirtuins pathways sarcopoenia and poor cardiac health and proteome of individuals spanning a
all interact in mammals to regulate a wide because muscle and heart depend on wide age range will provide the most
swath of metabolism in response to diet IGF1 signaling for their maintenance. In detailed phenotyping of human aging so
and also trigger mechanisms to relieve addition, blockade of the insulin receptor far. For example, it has already been
oxidative stress. Accordingly, these path- would trigger insulin resistance in muscle, possible to conduct transcriptome anal-
ways also regulate the functionality of liver, and adipose tissue. SIRT1 activation ysis of cortical regions obtained from hu-
mitochondria. For example, downregula- may actually aid the growth of certain pre- man brains of different ages preserved
tion of TOR stimulates mitophagy to existing tumors, perhaps due to its deace- at autopsy (Lu et al., 2004). This has
improve mitochondrial quality, and upre- tylation and downregulation of p53 (Guar- showed that a small fraction of genes
gulation of AMPK and sirtuins, partners ente, 2013), although in other cases, it change in their transcription levels in a
in a mutually reinforcing feedback loop, seems to repress the initiation of tumors. characteristic way correlated with the

Cell 159, September 25, 2014 ª2014 Elsevier Inc. 17


age of the subjects. Plotting these tran- overlap those associated with suscepti- diet-induced obesity leading to diabetes
script levels (molecular aging) versus bility to late-onset diseases impacting and pollution leading to reduced air and
chronological age at time of death thus those same tissues. Because drugs may water quality, as well as global warming.
yields a straight line for this set of genes. affect different tissues in different ways Changes in maximum human life span
Within a large enough cohort, there ought (e.g., rapamycin), this information would will, in my opinion, be quite difficult to
to be outliers for whom the molecular age guide application of the right drug for the achieve and will take many years to even
significantly deviates from the chronolog- right person. This would be formally anal- assess. From the point of view of eco-
ical age; i.e., they would show slower or ogous to genotyping tumors to identify nomic and societal benefits, striving to
faster transcriptional changes in many or markers, which will then guide therapy. make people healthier longer without
all of the genes sensitive to aging. Further Finally, IPS technology may also necessarily extending their maximum life
analysis of these outliers for enrichment in become important in combating effects span may be the wisest course. Put
SNPs defining specific haplotypes may of aging (Sánchez Alvarado and Yama- another way, the nightmare scenario
shine a light on genes and pathways that naka, 2014). If specific tissues vulnerable would be to extend maximum human life
favor slow or rapid aging (Glorioso et al., to aging in individuals can be identified span without extending health span.
2011). If SNPs passing significance for bioinformatically, then differentiated cells Second, bioinformatics will play a sub-
genome-wide discovery prove difficult to generated by IPS technology from that stantial role in the progress of aging
find, analyses of transcriptional (or epige- person may potentiate drug screening to research, especially as it applies to
netic) differences in the outliers (e.g., un- protect those cells from stressors and pro- humans. There may already be buried
usually high expression of gene X in all vide an avenue toward new therapeutics. in the sea of ever-increasing human
slow aging brains) may give clues about Beyond this approach, one can imagine genomic data novel clues about genes
genes and pathways that determine correcting the offending haplotype in IPS and pathways that govern aging in
slow or rapid aging. This latter approach cells by CRISPR/Cas9 technology and re- different tissues. In this regard, it remains
also has the advantage of identifying introducing genetically reprogrammed to be seen how much of aging will prove to
mechanisms that are not genetically cells into the patient, an approach that be systemic and affect all tissues simulta-
based. has been widely discussed for frank dis- neously emanating from brain signals, for
If the genes and pathways that seem to eases (Hsu et al., 2014). Unfortunate tis- example, and how much will be tissue
correlate with slow or fast aging can be sues bearing haplotypes that cause rapid autonomous.
thus identified by big data analysis, result- aging may thus be rescued by early inter- Third, aging and the genes and path-
ing hypotheses about brain aging may be ventions even before the onset of signifi- ways that govern its effects are complex.
tested by conducting field studies. cant physiological decline. Although IPS- It is not likely that there will be a silver bul-
Possible treasure troves are the various based approaches appear promising, let for aging any more than there will be a
long-term human longitudinal studies, they face many hurdles, including the suc- silver bullet for cancer. However, there will
which provide a cornucopia of health in- cessful incorporation of new cells into ex- likely be novel pharmaceutical interven-
formation spanning decades and, in isting organs. tions for the effects of aging emerging
some cases, provide access to genotyp- In closing, the past two centuries have directly from aging research. These inter-
ing. This may potentiate testing whether witnessed advances at many levels that ventions may need to be tissue specific,
genetic haplotypes that correlate with allow people to live longer and more pro- taking into account the personalized way
slow or rapid aging identified bio- ductive lives. I have attempted to place aging impacts an individual tissue-by-tis-
informatically exert predictable effects in current research on the biology of aging sue. Overall, it is an exciting, albeit uncer-
a human population over the course of a into this context and have arrived at a few tain, time to speculate how human health
lifetime. For example, one might test predictions. First, it will be more achiev- will be impacted in the decades to come
whether haplotypes correlating with slow able and desirable to extend human health by research on the biology of aging.
molecular brain aging protect against span rather than life span per se. Yes, a
cognitive decline or neurodegenerative historical perspective would indicate that ACKNOWLEDGMENTS
diseases in these longitudinal studies. average life span has been increasing in
For all diseases, there is a need to use the developed world for more than a cen- I want to thank lab members and Mark Collins for
comments and the NIH, The Jain Foundation,
haplotype data to stratify people by their tury, and this trend has not abated. Much
and Glenn Foundation for Medical Research for
DNA sequence to determine the best of this increase is due to improved sanita- funding. I apologize to those whose work I did
treatment options, i.e., personalized med- tion and reduction of insect-borne dis- not describe or cite due to space limitations. L.G.
icine. Such stratification would also guide eases, reduced mortality of infants, chil- consults for GSK, Chronos, and Segterra and is a
the selection of which humans to choose dren, and birthing mothers and advances founder and shareholder in Elysium Health.
for clinical trials of potential new drugs. in medicine including vaccines, antibi-
In the context of aging, extending bioin- otics, and antiviral drugs. Epidemiological REFERENCES
formatics studies on brain aging to other studies and medical research targeted at
Bai, P., Cantó, C., Oudart, H., Brunyánszki, A.,
tissues may reveal whether relevant hap- specific diseases has also continued to
Cen, Y., Thomas, C., Yamamoto, H., Huber, A.,
lotypes affect all tissues, or, more likely, push out average survival. On the other Kiss, B., Houtkooper, R.H., et al. (2011). PARP-1
affect aging in some tissues but not hand, certain environmental factors may inhibition increases mitochondrial metabolism
others. Tissue-specific haplotypes may be placing these gains at risk, such as through SIRT1 activation. Cell Metab. 13, 461–468.

18 Cell 159, September 25, 2014 ª2014 Elsevier Inc.


Braunstein, M., Sobel, R.E., Allis, C.D., Turner, Kanfi, Y., Naiman, S., Amir, G., Peshti, V., Zinman, Neff, F., Flores-Dominguez, D., Ryan, D.P.,
B.M., and Broach, J.R. (1996). Efficient transcrip- G., Nahum, L., Bar-Joseph, Z., and Cohen, H.Y. Horsch, M., Schröder, S., Adler, T., Afonso, L.C.,
tional silencing in Saccharomyces cerevisiae re- (2012). The sirtuin SIRT6 regulates lifespan in Aguilar-Pimentel, J.A., Becker, L., Garrett, L.,
quires a heterochromatin histone acetylation male mice. Nature 483, 218–221. et al. (2013). Rapamycin extends murine lifespan
pattern. Mol. Cell. Biol. 16, 4349–4356. Katsimpardi, L., Litterman, N.K., Schein, P.A., but has limited effects on aging. J. Clin. Invest.
Cantó, C., Houtkooper, R.H., Pirinen, E., Youn, Miller, C.M., Loffredo, F.S., Wojtkiewicz, G.R., 123, 3272–3291.
D.Y., Oosterveer, M.H., Cen, Y., Fernandez-Mar- Chen, J.W., Lee, R.T., Wagers, A.J., and Rubin, Peek, C.B., Affinati, A.H., Ramsey, K.M., Kuo, H.Y.,
cos, P.J., Yamamoto, H., Andreux, P.A., Cettour- L.L. (2014). Vascular and neurogenic rejuvenation Yu, W., Sena, L.A., Ilkayeva, O., Marcheva, B., Ko-
Rose, P., et al. (2012). The NAD(+) precursor of the aging mouse brain by young systemic fac- bayashi, Y., Omura, C., et al. (2013). Circadian
nicotinamide riboside enhances oxidative meta- tors. Science 344, 630–634. clock NAD+ cycle drives mitochondrial oxidative
bolism and protects against high-fat diet-induced Kennedy, B.K., Austriaco, N.R., Jr., Zhang, J., and metabolism in mice. Science 342, 1243417.
obesity. Cell Metab. 15, 838–847. Guarente, L. (1995). Mutation in the silencing gene Ramsey, K.M., Mills, K.F., Satoh, A., and Imai, S.
Chang, H.C., and Guarente, L. (2013). SIRT1 medi- SIR4 can delay aging in S. cerevisiae. Cell 80, (2008). Age-associated loss of Sirt1-mediated
ates central circadian control in the SCN by a 485–496. enhancement of glucose-stimulated insulin secre-
mechanism that decays with aging. Cell 153, Kennedy, B.K., Gotta, M., Sinclair, D.A., Mills, K., tion in beta cell-specific Sirt1-overexpressing
1448–1460. McNabb, D.S., Murthy, M., Pak, S.M., Laroche, (BESTO) mice. Aging Cell 7, 78–88.
Chang, H.C., and Guarente, L. (2014). SIRT1 and T., Gasser, S.M., and Guarente, L. (1997). Redistri- Ramsey, K.M., Yoshino, J., Brace, C.S., Abrassart,
other sirtuins in metabolism. Trends Endocrinol. bution of silencing proteins from telomeres to the D., Kobayashi, Y., Marcheva, B., Hong, H.K.,
Metab. 25, 138–145. nucleolus is associated with extension of life Chong, J.L., Buhr, E.D., Lee, C., et al. (2009). Circa-
span in S. cerevisiae. Cell 89, 381–391. dian clock feedback cycle through NAMPT-medi-
Colman, R.J., Anderson, R.M., Johnson, S.C.,
Kastman, E.K., Kosmatka, K.J., Beasley, T.M., Al- Kenyon, C.J. (2010). The genetics of ageing. Na- ated NAD+ biosynthesis. Science 324, 651–654.
lison, D.B., Cruzen, C., Simmons, H.A., Kemnitz, ture 464, 504–512. Redman, L.M., Huffman, K.M., Landerman, L.R.,
J.W., and Weindruch, R. (2009). Caloric restriction López-Otı́n, C., Blasco, M.A., Partridge, L., Pieper, C.F., Bain, J.R., Muehlbauer, M.J., Ste-
delays disease onset and mortality in rhesus mon- Serrano, M., and Kroemer, G. (2013). The hall- vens, R.D., Wenner, B.R., Kraus, V.B., Newgard,
keys. Science 325, 201–204. marks of aging. Cell 153, 1194–1217. C.B., et al. (2011). Effect of caloric restriction with
Conboy, M.J., Conboy, I.M., and Rando, T.A. Lu, T., Pan, Y., Kao, S.Y., Li, C., Kohane, I., Chan, and without exercise on metabolic intermediates
(2013). Heterochronic parabiosis: historical J., and Yankner, B.A. (2004). Gene regulation and in nonobese men and women. J. Clin. Endocrinol.
perspective and methodological considerations DNA damage in the ageing human brain. Nature Metab. 96, E312–E321.
for studies of aging and longevity. Aging Cell 12, 429, 883–891. Sánchez Alvarado, A., and Yamanaka, S. (2014).
525–530. Mattison, J.A., Roth, G.S., Beasley, T.M., Tilmont, Rethinking differentiation: stem cells, regeneration,
Frye, R.A. (1999). Characterization of five human E.M., Handy, A.M., Herbert, R.L., Longo, D.L., Alli- and plasticity. Cell 157, 110–119.
cDNAs with homology to the yeast SIR2 gene: son, D.B., Young, J.E., Bryant, M., et al. (2012). Satoh, A., Brace, C.S., Rensing, N., Cliften, P.,
Sir2-like proteins (sirtuins) metabolize NAD and Impact of caloric restriction on health and survival Wozniak, D.F., Herzog, E.D., Yamada, K.A., and
may have protein ADP-ribosyltransferase activity. in rhesus monkeys from the NIA study. Nature Imai, S. (2013). Sirt1 extends life span and delays
Biochem. Biophys. Res. Commun. 260, 273–279. 489, 318–321. aging in mice through the regulation of Nk2 homeo-
Glorioso, C., Oh, S., Douillard, G.G., and Sibille, E. Mattison, J.A., Wang, M., Bernier, M., Zhang, J., box 1 in the DMH and LH. Cell Metab. 18, 416–430.
(2011). Brain molecular aging, promotion of neuro- Park, S.S., Maudsley, S., An, S.S., Santhanam, Sebastiani, P., and Perls, T.T. (2012). The genetics
logical disease and modulation by sirtuin 5 L., Martin, B., Faulkner, S., et al. (2014). Resveratrol of extreme longevity: lessons from the New
longevity gene polymorphism. Neurobiol. Dis. 41, prevents high fat/sucrose diet-induced central England centenarian study. Front Genet. 3, 277.
279–290. arterial wall inflammation and stiffening in
Shimobayashi, M., and Hall, M.N. (2014). Making
Gomes, A.P., Price, N.L., Ling, A.J., Moslehi, J.J., nonhuman primates. Cell Metab. 20, 183–190.
new contacts: the mTOR network in metabolism
Montgomery, M.K., Rajman, L., White, J.P., Teo- Mercken, E.M., Mitchell, S.J., Martin-Montalvo, A., and signalling crosstalk. Nat. Rev. Mol. Cell Biol.
doro, J.S., Wrann, C.D., Hubbard, B.P., et al. Minor, R.K., Almeida, M., Gomes, A.P., Scheibye- 15, 155–162.
(2013). Declining NAD(+) induces a pseudohypoxic Knudsen, M., Palacios, H.H., Licata, J.J., Zhang,
Sinclair, D.A., and Guarente, L. (2014). Small-mole-
state disrupting nuclear-mitochondrial communi- Y., et al. (2014). SRT2104 extends survival of male
cule allosteric activators of sirtuins. Annu. Rev.
cation during aging. Cell 155, 1624–1638. mice on a standard diet and preserves bone and
Pharmacol. Toxicol. 54, 363–380.
Gowans, G.J., and Hardie, D.G. (2014). AMPK: a muscle mass. Aging Cell. Published online June
16, 2014. http://dx.doi.org/10.1111/acel.12220. Smeal, T., Claus, J., Kennedy, B., Cole, F., and
cellular energy sensor primarily regulated by
Guarente, L. (1996). Loss of transcriptional
AMP. Biochem. Soc. Trans. 42, 71–75. Milman, S., Atzmon, G., Crandall, J., and Barzilai,
silencing causes sterility in old mother cells of
Guarente, L. (2000). Sir2 links chromatin silencing, N. (2013). Phenotypes and Genotypes of High
S. cerevisiae. Cell 84, 633–642.
metabolism, and aging. Genes Dev. 14, 1021–1026. Density Lipoprotein Cholesterol in Exceptional
Longevity. Curr. Vasc. Pharmacol. Published on- Stumpferl, S.W., Brand, S.E., Jiang, J.C., Korona,
Guarente, L. (2013). Calorie restriction and sirtuins line December 18, 2013. http://dx.doi.org/10. B., Tiwari, A., Dai, J., Seo, J.G., and Jazwinski,
revisited. Genes Dev. 27, 2072–2085. 2174/1570161111666131219101551. S.M. (2012). Natural genetic variation in yeast
Hsu, P.D., Lander, E.S., and Zhang, F. (2014). longevity. Genome Res. 22, 1963–1973.
Mouchiroud, L., Houtkooper, R.H., Moullan, N.,
Development and applications of CRISPR-Cas9 Katsyuba, E., Ryu, D., Cantó, C., Mottis, A., Jo, Verdin, E., Hirschey, M.D., Finley, L.W., and Haigis,
for genome engineering. Cell 157, 1262–1278. Y.S., Viswanathan, M., Schoonjans, K., et al. M.C. (2010). Sirtuin regulation of mitochondria: en-
Imai, S., Armstrong, C.M., Kaeberlein, M., and (2013). The NAD(+)/Sirtuin Pathway Modulates ergy production, apoptosis, and signaling. Trends
Guarente, L. (2000). Transcriptional silencing and Longevity through Activation of Mitochondrial Biochem. Sci. 35, 669–675.
longevity protein Sir2 is an NAD-dependent his- UPR and FOXO Signaling. Cell 154, 430–441. Yoshino, J., Mills, K.F., Yoon, M.J., and Imai, S.
tone deacetylase. Nature 403, 795–800. Nakahata, Y., Sahar, S., Astarita, G., Kaluzova, M., (2011). Nicotinamide mononucleotide, a key
Johnson, T.E. (2013). 25 years after age-1: genes, and Sassone-Corsi, P. (2009). Circadian control of NAD(+) intermediate, treats the pathophysiology
interventions and the revolution in aging research. the NAD+ salvage pathway by CLOCK-SIRT1. of diet- and age-induced diabetes in mice. Cell
Exp. Gerontol. 48, 640–643. Science 324, 654–657. Metab. 14, 528–536.

Cell 159, September 25, 2014 ª2014 Elsevier Inc. 19

Vous aimerez peut-être aussi