ArticlePDF AvailableLiterature Review

Tipping the metabolic scales towards increased longevity in mammals

Authors:

Abstract and Figures

A hallmark of ageing is dysfunction in nutrient signalling pathways that regulate glucose homeostasis, negatively affecting whole-body energy metabolism and ultimately increasing the organism's susceptibility to disease. Maintenance of insulin sensitivity depends on functional mitochondrial networks, but is compromised by alterations in mitochondrial energy metabolism during ageing. Here we discuss metabolic paradigms that influence mammalian longevity, and highlight recent advances in identifying fundamental signalling pathways that influence metabolic health and ageing through mitochondrial perturbations.
Content may be subject to copyright.
SERIES ON METABOLISMSERIES ON METABOLISM
REVIEW SERIES ON METABOLISM
Tipping the metabolic scales towards increased
longevity in mammals
Celine E. Riera and Andrew Dillin
A hallmark of ageing is dysfunction in nutrient signalling pathways that regulate glucose homeostasis, negatively affecting whole-
body energy metabolism and ultimately increasing the organism’s susceptibility to disease. Maintenance of insulin sensitivity
depends on functional mitochondrial networks, but is compromised by alterations in mitochondrial energy metabolism during
ageing. Here we discuss metabolic paradigms that influence mammalian longevity, and highlight recent advances in identifying
fundamental signalling pathways that influence metabolic health and ageing through mitochondrial perturbations.
Life expectancy has been strikingly prolonged in developed coun-
tries as a positive outcome of medical progress and modern lifestyle.
Nonetheless, with old age comes a wide range of age-associated diseases
including neurodegenerative diseases, cardiovascular and metabolic
disorders, and higher susceptibility to cancer.
Although the clinical manifestations of ageing are well understood,
the genetic or environmental causes driving age-associated decline in
healthspan remain difficult to isolate. By identifying genetic perturba-
tions that can extend lifespan as well as retard the onset of physiological
decline in invertebrate model organisms, ageing research has started
to provide therapeutic strategies to prolong both healthspan and lifes-
pan1. Research over the last decade using the multitude of spatial and
temporal genetic tools available in mice has begun to identify points
of convergence between mammals and invertebrates. In comparison
to humans, mice are relatively short-lived mammals that suffer from
clinically relevant pathologies including metabolic syndrome, osteo-
porosis, cancer, sarcopenia, cardiovascular dysfunction and neurode-
generation2,3. Molecular and cellular pathways have been identified that
not only increase longevity of mammalian model organisms but also
generally promote their healthspan by delaying the onset of metabolic
decline. This delay is tightly coupled to an age-dependent deterio-
ration of insulin metabolism and subsequent disruption of nutrient
uptake by body tissues. Mitochondria play a key role in the utilization
of nutrient substrates for energy production. Alterations in mitochon-
drial biogenesis, dynamics and integrity render an organism unable
to maintain metabolic homeostasis or properly respond to metabolic
demand observed during ageing.
In this Review, we provide an overview of the metabolic signal-
ling pathways that modulate mammalian lifespan and discuss their
potential as therapeutic targets to treat age-associated loss in glucose
homeostasis and healthspan. Finally, we present key mitochondrial
pathways that have been implicated in age-dependent malfunction of
metabolic homeostasis.
Insulin action regulates metabolic health during ageing
The pathologies encountered in mouse longevity studies revealed a
dominant role for glucose homeostasis in regulating metabolic health
during ageing. In humans, ageing is also linked to increased visceral
fat mass, loss in lean mass and a deterioration in insulin sensitivity
resulting in glucose intolerance — all factors that favour the occur-
rence of metabolic syndrome and cardiovascular diseases4,5. Similarly,
in mice, signs of physiological decline are characterized by an increase
in fat mass and the progressive development of glucose intolerance,
reflecting a similar impairment of insulin sensitivity6,7. However, mild
insulin resistance has been observed in a variety of long-lived mice and
is therefore not necessarily an indicator of poor health or shortened
lifespan when paired with improved glucose tolerance7–10. Over the last
decade, elevated energy expenditure has emerged as a putative longev-
ity biomarker, as it has been associated with lifespan extension11. In old
animals, a decrease in energy expenditure is associated with reduced
activity and endurance linked to decreased oxygen consumption and
lipid accumulation in non-adipose tissue. The respiratory exchange
ratio (RER), obtained by indirect calorimetry, compares the volume
of carbon dioxide an organism produces to the volume of oxygen con-
sumed over a given time and varies inversely with lipid oxidation. In
young and healthy mice, the RER displays a youthful circadian shift
from night to day, reflecting the daily transition from carbohydrate
to lipid metabolism. Old mice, however, develop a substrate prefer-
ence towards lipids, losing the capacity to switch between fuel sources
(Fig.1). This is in accordance with a metabolic flexibility theory of
ageing, which predicts that longevity depends on metabolic health,
and lifespan extension is achieved by maintaining a youthful RER, thus
Celine E. Riera and Andrew Dillin are at the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
and Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA.
e-mail: dillin@berkeley.edu
196 NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW
SERIES ON METABOLISMSERIES ON METABOLISM REVIEW
protecting the organism against systemic damage associated with fat
metabolism and storage. In contrast, the onset of metabolic inflexibility
corresponds to aberrant glucose homeostasis and increased suscepti-
bility to age-onset diseases in response to an age-associated decline
in mitochondrial energy metabolism12. Another mechanism that may
contribute to the deterioration of metabolic health with age operates
through the oscillations of the circadian clock. Modulating the activity
of the major circadian regulators BMAL1 (also known as ARNTL; aryl
hydrocarbon receptor nuclear translocator-like) and CLOCK (clock
circadian regulator) results in premature ageing13, whereas enhanced
transcriptional activity of these factors protects against age-associated
decline in adapting to new circadian periods14. Most physiological,
endocrine and behavioural rhythms are governed by the molecular
clock, and accumulating evidence indicates that disruption of circadian
rhythms is associated with cancer15. Future research is required to shed
light on how the rhythmicity of nutrient homeostatic processes affects
metabolic flexibility and the ageing process.
Genetic manipulation of insulin metabolism in mouse lifespan
and healthspan
To date, dietary restriction is the most robust intervention to increase
healthspan and lifespan in multiple organisms including rodents and
primates, delaying the onset of age-related diseases2,16,17. In mice, dietary
restriction increases insulin sensitivity and induces a healthy transition
of the RER, due to a more dramatic shift from carbohydrate oxidation
and endogenous fatty acid synthesis in the feeding phase to fatty acid
oxidation during resting18. Similarly to dietary restriction, multiple
genetic manipulations in mice improve metabolic flexibility and lon-
gevity. Alterations of components in the insulin and insulin-like growth
factor signalling (IIS) pathway that dampen insulin signalling promote
longevity associated with improved insulin sensitivity. Some of these
long-lived mice include fat-specific insulin receptor knockout (FIRKO)
mice, hypomorphic PI3K mice, and mice overexpressing the liver hor-
mone FGF21 (fibroblast growth factor21), which antagonizes growth
hormone (GH) and insulin-like growth factor 1 (IGF-1)19–21. Reduction
of circulating IGF-1 positively correlates with increased longevity in
inbred mouse strains22, but is also linked to a reduced body size and
even dwarfism23.
Although Ames (Prop1df/Prop1df) and Snell (Pit1dw/Pit1dw) dwarf mice
are extremely long-lived presumably partially because of decreased IIS,
their metabolic characteristics must be interpreted with caution. This
is because profound deficiency in anterior pituitary function results in
other hormonal changes and negatively affects fertility, metabolic fitness,
adiposity, glucose tolerance and insulin secretion, despite considerable
life extension24,25. In humans, hypothyroidism is also characterized by
diminution of the acute insulin response, resulting in impaired glucose
tolerance26, but the effects of this condition on human longevity are
unclear. Thus, the lifespan extension of reduced IIS models must be
uncoupled from their growth delay or hormonal imbalances to isolate
clinically relevant therapeutic targets.
Ageing
Improved glucose homeostasisWild type
Youth:
metabolic health
Old age:
metabolic decline
0.7
1.0
Wild type
Ageing
RER
Decreased energy metabolism
• Insulin resistance
• Glucose intolerance
• Adipose tissue accumulation
Time
Survival (%)
Improved glucose homeostasis
Metabolic diseases
Neurodegenerative diseases
Cancer formation
Metabolic flexibility Metabolic inflexibility
Mitochondrial mass and integrity
0.7
1.0
a
b
c
100
50
0
Figure 1 Metabolic flexibility controls healthspan and lifespan. (a) A
schematic showing survival curves of a wild-type mouse (blue curve)
compared to a long-lived genetic model of improved glucose homeostasis
(green curve). The onset of metabolic decline precedes the appearance of
morbid pathologies associated with insulin resistance and nutrient uptake.
(b) Throughout lifespan, mitochondrial biogenesis, function and dynamics
decline. Accumulation of oversized abnormal mitochondria which result
from impaired degradation of dysfunctional mitochondria is observed after
the onset of metabolic inflexibility. (c) Metabolic flexibility correlates with
a healthy respiratory exchange ratio (RER) resulting from the circadian
shift between carbohydrate (value of 1.0) to lipid metabolism (value of
0.7). Metabolic inflexibility observed at old age correlates with a substrate
preference towards lipids, reflecting an inability to adapt fuel oxidation to
fuel availability.
SERIES ON METABOLISM
NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015 197
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW SERIES ON METABOLISMSERIES ON METABOLISM
REVIEW
Deletion of TRPV1 (transient receptor potential vanilloid1) pain
receptors in peripheral nerves innervating pancreatic βcells extends
mouse longevity by improving metabolic flexibility and in particu-
lar elevating prandial insulin secretion with age, without the corre-
sponding growth delay or altered body weight observed in decreased
IIS models7. In humans, metabolic syndrome is linked to dementia
and increased incidence of multiple cancers27,28. Strong correlations
between insulin sensitivity and cancer formation have also been found
in the mouse, and reduced cancer incidence is observed in mice with
improved energy expenditure at old age7,29. Mice carrying a TRPV1
mutation have both reduced cancer incidence and delayed onset of
cognitive decline with age. Other genetic manipulations that reduce
IIS meet these criteria to some extent. Long-lived Ames dwarf mice
and GHR-BP (growth-hormone receptor binding protein) knockout
mice display a lower incidence and delayed onset of certain cancers and
tumours, increased insulin sensitivity and improved cognitive func-
tion at old age3032. Additionally, prolonged rapamycin treatment delays
cancer formation in aged mice and extends lifespan through inhibition
of mTOR (mammalian target of rapamycin), a major cellular nutrient
sensing pathway that regulates cell growth33,34 and integrates insulin
signalling and cell stress signals35. The method by which rapamycin
regulates the rate of ageing is complex, because short-term treatment
results in immunodeficiency and favours glucose intolerance and insu-
lin resistance36,37. However, prolonged treatment results in improved
metabolic profiles, increased oxygen consumption and ketogenesis, and
markedly enhanced insulin sensitivity38. Robust genetic inhibition of
downstream mTOR complex1 (mTORC1) components, including dele-
tion of ribosomal protein S6K1 (S6 kinase1) and enhanced autophagy,
lead to increased longevity with improved glucose tolerance and insulin
sensitivity at mid-age39,40. However, despite the consensus that dampen-
ing mTORC1 activity extends lifespan, several studies report that this
beneficial effect is achieved mostly by slowing cellular growth and can-
cer incidence, without impacting insulin metabolism36,41. These reports
suggest that disruption of the other major mTOR complex, mTORC2,
mediates aspects of the impaired insulin metabolism associated with
rapamycin treatment, in particular the inability to suppress hepatic
gluconeogenesis following insulin release. It is therefore unclear how
prolonged rapamycin exposure promotes metabolic health, despite the
recent report that insulin sensitivity amelioration following rapamycin
treatment might rely on inhibitory effects of both mTOR complexes38.
Better resolution of the mechanism through which rapamycin positively
modulates glucose homeostasis is required to design drugs that can
recapitulate the longevity extension obtained by rapamycin treatment
without its detrimental side effects.
Mitochondria at the core of metabolic flexibility during ageing
Even though cellular energy can be rapidly generated through anaero-
bic glycolysis, it mostly originates from aerobic oxidation of carbohy-
drates and fatty acids in the mitochondria, highlighting this organelles
critical role in regulating global metabolic homeostasis. Not only is
insulin secretion from pancreatic βcells mitochondria-dependent, but
insulin signalling also relies on mitochondria to metabolize glucose
in energy-consuming cells. Mitochondrial activity regulates gluconeo-
genesis and triglyceride synthesis in the liver and lipolysis in adipose
tissue, as well as many other vital processes42. Accordingly, mitochon-
drial dysfunction or damage can greatly perturb metabolic flexibility
and insulin sensitivity, impacting metabolic human diseases and age-
ing12,43,44. During the ageing process, mitochondrial biogenesis, func-
tion and dynamics decline (Fig.2)45,46. This is particularly obvious in
skeletal muscle — a highly metabolically active tissue extremely rich in
mitochondria — which undergoes sarcopenia, a critical loss in mito-
chondrial volume density and decline in physical function with age47.
Accumulation of lipids in skeletal muscle, liver and adipose tissue is
indicative of decreased metabolic flexibility with ageing and therefore
demonstrates a global reduction in oxidative phosphorylation capacity
and mitochondrial biogenesis.
Molecular targets that can restore mitochondrial function and meta-
bolic homeostasis at old age have emerged over the past decade. A key
regulator of mitochondrial biogenesis and energy metabolism is the
transcriptional coactivator PGC1α (proliferator-activated receptor γ
coactivatorα)48,49. Decreased PGC1α expression has been implicated
in muscle insulin resistance in humans and diabetic mouse models50,51.
Increasing PGC1α content in mouse skeletal muscle preserves oxida-
tion phosphorylation, preventing muscle wasting and improving meta-
bolic fitness by inhibiting insulin resistance and fat accumulation with
age52. Mechanistically, PGC1α acts by regulating the activity of several
transcription factors — including the nuclear respiratory factors NRF1
and NRF2 and mitochondrial transcription factorA (TFAM) — that
in turn directly or indirectly induce genes related to mitochondrial
biogenesis and respiration53,54. AMP-activated protein kinase (AMPK)-
mediated phosphorylation activates PGC1α in conditions of low cel-
lular energy, which corresponds to a high AMP/ATP ratio, therefore
inhibiting anabolic processes consuming energy and activating cata-
bolic pathways producing energy, such as fatty acid oxidation and
respiration55,56. Strategies to directly manipulate AMPK to improve
mitochondrial function in skeletal muscle have proven successful and
offer important avenues to treat age-associated metabolic decline.
Pharmacological AMPK activation by AICAR (5-Aminoimidazole-
4-carboxamide ribonucleotide) treatment enhances exercise endur-
ance57 and increases mitochondrial biogenesis and respiration in
mouse models of cytochromec oxidase (COX) deficiency, rescuing
the limited exercise capacity of these mice58. Importantly, activat-
ing AMPK by metformin supplementation is also linked to lifespan
extension59, although metformin action on lifespan might depend on
other targets such as mTOR (ref.60). Under low energy conditions,
AMPK also increases the ratio of nicotinamide adenine dinucleotide
NAD+/NADH, a metabolic signal that triggers the NAD-dependent
deacetylase SIRT1 (sirtuin1) and is required for AMPK-dependent
PGC1α activation61. AMPK and SIRT1 can autoregulate each other, a
mechanism that has been postulated to improve the cellular response to
low-energy states62, and that orchestrates a complex catabolic response
that increases mitochondrial biogenesis, enhances antioxidant defence
and improves fatty-acid oxidation.
Mitochondrial oxidative stress and ageing
Mitochondrial networks are vulnerable to oxidative stress, resulting
in impaired cellular function and increasing the chance of cell death.
During ageing, mitochondrial electron transport chain (ETC) efficiency
decreases, reducing ATP generation through increased proton leakage.
Higher metabolic intensities correlated with higher proton conductance
across the inner membrane of skeletal muscle mitochondria were found
in long-lived mice, supporting the ‘uncoupling to survive’ theory11.
198 NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW
SERIES ON METABOLISMSERIES ON METABOLISM REVIEW
However, proton leakage in the ETC also diminishes the amount of reac-
tive oxygen species (ROS) produced, with ROS being by-products of
mitochondrial ATP generation resulting from oxygen reduction. Thus,
reduction of ROS levels seems to be beneficial by dampening oxida-
tive damage to mitochondria. The recruitment of uncoupling proteins
(UCPs) relieves mitochondrial ROS by uncoupling the proton gradi-
ent. In particular, UCP1, found in metabolically active tissues such as
brown fat, is activated by fatty acids and dissipates the proton gradient
as heat, with a low rate of ATP production allowing fast substrate oxida-
tion and thermogenesis63. The production of superoxide (O2
) — a toxic
by-product of respiration generated mostly by complexesI and III of the
ETC — may also induce the uncoupling of mitochondria by activating
UCP1 (or other UCPs) under certain conditions requiring fatty acids
and inhibited by purine nucleotides64. Accordingly, increasing energy
expenditure by UCP1 overexpression in mouse muscle has beneficial
effects on metabolic health, extending median but not maximal lifespan,
decreasing adiposity, increasing both core temperature and metabolic
rate, and lowering lymphoma incidence65.
The free radical’ theory of ageing proposes a causative role for
ROS in accumulated cellular damage over time, and in age-associated
pathologies and functional decline of tissues. Supporting this theory,
decreased ROS production from mitochondria has been observed in
response to dietary restriction and linked to improved mitochondrial
function in rodents. Despite the initial claim that increased respiration
following dietary restriction is achieved by higher mitochondrial bio-
genesis66, dietary restriction is now thought to preserve mitochondrial
function by maintaining the integrity of existing cellular components
and increasing the activity of oxidant scavengers such as catalase67.
Mice expressing human catalase in mitochondria are protected from
oxidative damage, cardiac dysfunction and cataract development, and
exhibit extended longevity69. However, a conflicting study reports that
overexpression of human catalase in all tissues is insufficient to extend
lifespan70. PGC1α also minimizes the build-up of ROS through the tran-
scriptional regulation of numerous ROS-detoxifying enzymes68. Other
genetic models challenge the theory that a linear relationship exists
between decreasing ROS and lifespan extension. Increasing mitochon-
drial ROS and oxidative damage do not cause progeria in mice71,72, and
multiple mouse models of increased antioxidant defences also do not
extend lifespan70. Furthermore, mice expressing a defective mitochon-
drial polymerase develop mitochondrial DNA (mtDNA) mutations
β-oxidation
mtDNA mutations
Triglycerides
Fatty Acyl-CoA
Diacylglycerol
ROS
Glucose uptake
Acetyl-CoA
TCA
cycle
ψm
OXPHOS
Stress kinases
PKC, IKK, JNK
IRS-1/2
ROS
Translation mitochondrial proteins
Damaged mitochondrial proteins
P
P
PPP
P
Ser/Thr
phosphorylation IRS-1/2
PI3K
AKT
Insulin receptor
PGC1α
activity
Biogenesis
Mitochondrial stress
NAD+
SIRT1
Dynamics
Aberrant fusion
Nucleus
Mitochondria
Cytoplasm
Mitophagy
Apoptosis
Mitochondrial quality
PGC1α
levels
III III IV
NAD+
NADH
H+
H+
H+
H+
V
ADP ATP
O
2.−
O
2
O
2
O
2.−
a
b
c
d
Figure 2 Effect of ageing on mitochondrial function. Multiple sources of
damage (orange bolts, ad) cause senescent mitochondrial phenotypes. (a)
Nutrient surplus promotes the accumulation of lipid metabolite intermediates
that overload and hyperpolarize the mitochondria. Incompletely oxidized
lipid products from the tricarboxylic acid cycle (TCA) and reactive oxygen
species (ROS) then build up, which causes oxidative stress to the respiratory
system (OXPHOS; simplified electron transport chain shown for illustrative
purposes). These intermediates also activate stress-sensitive kinases (protein
kinaseC, PKC; c-Jun N-terminal kinase, JNK; and IκB kinase, IKK), which
phosphorylate insulin receptor substrates (IRS-1/2). This disrupts the insulin
signalling pathway, and glucose uptake and glycolysis in response to insulin.
(b) ROS accumulation causes mutations to the mitochondrial DNA (mtDNA)
pool, affecting mitochondrial translation and damage to proteins vital to
OXPHOS and mitochondrial dynamics. (c) Impaired dynamics and decreased
mitophagy with age leads to aberrant mitochondria. (d) Mitochondrial
biogenesis, controlled by PGC1α, declines with age. Insulin resistance
contributes to the development of a vicious cycle of a negative feedback loop
by impairing biogenesis in addition to the TCA cycle and OXPHOS.
NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015 199
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW SERIES ON METABOLISMSERIES ON METABOLISM
REVIEW
that accelerate ageing by impairing mitochondrial function without
increasing ROS7375. Although the mtDNA mutator mouse studies ini-
tially challenged the free radical theory of ageing, recent conflicting
evidence suggests that oxidative damage actually occurs in skeletal
muscle of these mice76. Following these controversies, the role of ROS
in ageing is being revisited, mainly supported by evidence obtained
in Caenorhabditiselegans suggesting that high ROS levels may act as
an important signal that triggers a gene expression pattern promoting
survival under stressful conditions77.
Activation of the mitochondrial UPR and longevity
Long-lived mouse models provide supporting evidence for the role of
improving mitochondrial biogenesis and function in forestalling age-
ing. Paradoxically, in C.elegans, lifespan can be increased by reducing
mitochondrial respiration through reduced function of nuclear genes
encoding ETC components78,79. In particular, neuronal knockdown of
COX activates the mitochondrial unfolded protein response (UPRmt), a
mitochondrial proteostasis mechanism that confers increased longevity
to worms80,81. This pathway is part of the mitochondrial quality control
machinery that detects misfolded proteins in the mitochondria, initi-
ating a signalling cascade that leads to the transcription of protective
genes82. Notably, 13 subunits of the oxidative phosphorylation complex
are encoded by mtDNA, whereas the remaining mitochondrial proteins
(of which there are more than 1,000) are encoded by nuclear DNA
and imported into mitochondria to achieve proper folding. The UPRmt
senses imbalances in the stoichiometry of proteins in the mitochon-
drial compartment and relieves the stress by retrograde signalling to
the nucleus through the degradation of misfolded proteins into peptides
by the ClpP1 protease, consequently promoting transcriptional acti-
vation of nuclear-encoded chaperones residing in the mitochondrial
matrix, such as heat shock proteins60 (HSP60) and 70 (HSP70). This
surveillance pathway is conserved in mammalian cells83, but whether
it plays a pro-longevity role in mammals is unknown. In support of its
possible role in mouse lifespan regulation, low expression of a mouse
mitochondrial ribosomal protein (Mrps5) triggers the UPRmt and cor-
relates with increased lifespan of inbred progenies of a cross between
C57BL/6J and DBA/2J mice84.
Evidence from mouse longevity studies suggests that impairing
mitochondrial function can improve lifespan; however, it is unknown
whether the UPRmt is causal for the longevity increase. Constitutive
ablation of SURF1 (surfeit1), a putative assembly factor specific to
COX, results in mild deficiency of COX in mice and significantly
extended mouse longevity85. This is accompanied by a strong deple-
tion of PGC1α levels, mtDNA content, citrate synthase activity and
oxidative-phosphorylation-related genes in these mice, indicative of a
decrease in respiration and mitochondrial biogenesis58. Reduction of
COQ7 (coenzymeQ7, known as Mclk1 in mice), an enzyme responsible
for the biosynthesis of ubiquinone (an ETC component), also extends
mouse lifespan concomitant with ETC dysfunction and decreased
mitochondrial coupling of respiration and ATP synthesis86. Although
these studies confirm that decreased respiration has beneficial effects
on mouse longevity, it is unknown whether the UPRmt following mito-
chondrial dysfunction is required for longevity.
Strikingly, levels of NAD+ were also decreased in the Mclk1+/ mice.
It is well established that NAD+ plays crucial roles in mediating mito-
chondrial energy metabolism by donating electrons to the ETC or by
acting as a coenzyme for rate-limiting citric acid (TCA) cycle enzymes.
Increasing NAD+ levels, by supplementation of a NAD+ precursor or
through a drug such as resveratrol, improves mitochondrial function
by activating SIRT1 in mice, and protects against the metabolic damage
of high-fat feeding8790. Thus, high NAD+ levels correspond to mito-
chondrial biogenesis and metabolic flexibility, ultimately improving
healthspan. Paradoxically, Mclk1+/ mice are long-lived despite their
depleted NAD+ levels. The mechanisms underlying this lifespan exten-
sion are unclear, and it is plausible that a different longevity paradigm
independent of mitochondrial biogenesis, such as UPRmt, is required
to promote lifespan of these mice. The relationship between UPRmt and
mitochondrial biogenesis is, however, still under debate. Rapamycin
and resveratrol treatment, which extend lifespan and metabolic fit-
ness, increased respiration in mammalian cells and induced the UPRmt
(ref.84). Thus, it is unclear whether UPRmt in mammals predicts longev-
ity and can be triggered as a cell survival mechanism following severe
mitochondrial impairment, or as a response to mitonuclear imbalance
that facilitates biogenesis. More investigation is necessary to elucidate
the physiological implication of the UPRmt in mammalian ageing.
Insulin resistance, mitochondrial perturbations and dynamics
Mitochondrial dysfunction has been implicated strongly in the onset
of metabolic diseases. Cells cope with nutrient supply by increasing
mitochondrial content, but persistent nutrient surplus can overload
the mitochondria and cause dysfunction, a phenomenon also observed
with ageing, as mitochondrial capacity declines (Fig.2). This functional
decline coincides with a global reduction in oxidative capacity of the
skeletal muscle of patients with type2 diabetes91, and contributes to
insulin resistance51. Energy excess overloads and hyperpolarizes mito-
chondria, leading to the accumulation of fatty-acyl-CoA and diacyl-
glycerol, causing excessive production of ROS. These mitochondrial
metabolites activate stress-sensing-kinases such as c-Jun N-terminal
kinase and protein kinaseC in the cytoplasm (which phosphorylate
insulin receptor substrates), impairing insulin signalling in glucose-
consuming tissues such as skeletal muscle and liver92,93. Insulin-
stimulated glucose transport is inhibited in these tissues and reduces
peripheral energy metabolism.
Furthermore, decreased brain insulin signalling is also observed
in ageing and contributes to neurodegenerative disorders, as a conse-
quence of insulin resistance and decreased insulin transport through
the blood–brain barrier94. Abnormal insulin metabolism in the brain
might originate from mitochondrial dysfunction, as the brain is highly
vulnerable to oxidative stress and elevated ROS levels can aggravate
insulin resistance95,96. Similar observations were made in the hypothala-
mus and hippocampus of mice fed a high-fat diet97,98. Downregulation
of the mitochondrial chaperone protein HSP60 by excessive adipokine
stimuli plays a central role in the onset of oxidative stress leading to
mitochondrial dysfunction and hypothalamic insulin resistance99.
Whether chaperone levels are also regulated by age-associated stress
in the brain and confer mitochondrial dysfunction and deficits in brain
energy metabolism remains undetermined.
Mitochondrial architecture is tightly regulated by the dynamic
opposing processes of fusion and fission. Mitochondrial dynam-
ics reflect bioenergetic adaptation to metabolic demand, facilitating
mixing and exchange of mitochondrial contents such as mtDNA or
metabolites100. Mitochondrial morphology and density are modulated
200 NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW
SERIES ON METABOLISMSERIES ON METABOLISM REVIEW
by variations in nutrient availability, with starvation triggering a fused
state that facilitates autophagy101. These changes in mitochondrial
architecture can affect oxidative phosphorylation complex assembly
and ATP synthesis, allowing mitochondria to reorganize and dispose
of damaged elements through mitophagy, an autophagic process that
eliminates damaged mitochondria102. Mitophagy prevents cell apop-
tosis or necrosis following excessive ROS accumulation, pro-inflam-
matory signals or permeabilization of mitochondrial membranes, but
the efficiency of autophagy declines with age103. Fission allows the
segregation of damaged mitochondria and their recycling through
autophagic processes, thereby ensuring mitochondria turnover and
cellular viability, whereas fusion counterbalances functional defects
and allows genetic complementation. Oversized abnormal mito-
chondria are present in senescent cells, as a result of dysfunctional
mitochondrial degradation104. Youthful mitochondrial fragmentation
can be restored in senescent cells by depleting critical mediators of
fusion, the dynamin-related proteins FIS1 (fission1 (mitochondrial
outer membrane) homolog) and OPA1 (optic atrophy1). Recently,
lack of the pro-fusion mitofusins Mfn1 and Mfn2 has been implicated
in regulation of whole-body energy homeostasis in distinct popula-
tions of hypothalamic neurons in mice105–107. Mfn2 deficiency pre-
vents changes in mitochondrial dynamics and communication with
the endoplasmic reticulum (ER), causing ER stress, insulin resistance
in skeletal muscle and liver tissues, and hypothalamic leptin resist-
ance, thus promoting obesity105–109. Mitochondrial health relies on a
tight contact with the ER that provides critical resources to the mito-
chondria such as mitochondrial lipids and calcium ions, which are
involved in mitochondrial dynamics and regulation of the TCA cycle.
Although gradual impairment of mitochondrial dynamics with age is
a proposed hypothesis for age-associated mitochondrial dysfunction,
additional layers of regulation may exist, including ER-mitochondria
tethering and impaired insulin signalling due to ER stress, resulting in
cell-autonomous inflammation (as observed in type2 diabetes)110,111.
One of the main pro-inflammatory transcriptional programs induced
following ER stress is the NFB (nuclear factorκB) signalling path-
way112. Interestingly, restoring hypothalamic immunity upon ageing
through the blockade of NF-κB signalling in microglia is sufficient to
extend lifespan in mice113. It will be critical to define whether meta-
bolic decline during ageing is mediated by the disruption of intricate
connections between mitochondria and the ER, and the onset of ER
stress and inflammation.
Conclusions and future perspectives
In light of the evidence reviewed here, progressive mitochondrial dys-
function is implicated in the deterioration of insulin sensitivity during
normal ageing, consequently impairing insulin signalling in central
and peripheral tissues, and contributing to a loss of metabolic flex-
ibility. Thus, restoring mitochondrial biogenesis is likely to provide
a therapeutic avenue to rejuvenate metabolism. However, in conflict-
ing studies, reduced respiration and biogenesis throughout life can
also increase lifespan, indicating an additional level of complexity.
Epigenetic regulation is now emerging as a potent factor influencing
mitochondrial biogenesis or integrity following the lack of exercise and
overfeeding. Notably, DNA hypermethylation suppresses the expres-
sion of Pgc1α and Tfam in mice, and microRNAs have the ability to
silence gene expression of Ucp2 and Mfn2, thus negatively affecting
biogenesis, oxidative phosphorylation and mitochondrial dynamics114.
Whether such epigenetic signatures are characteristic of the ageing
process remains poorly understood; thus, it will be essential to assess
the chain of events that leads to impaired transcriptional networks
that maintain biogenesis (nuclear and mitochondrial transcription) as
well as mitochondrial quality control machinery (dynamics, mitophagy
and the UPRmt). In particular, we are beginning to envision that these
processes are likely to co-regulate each other, as highlighted by the
reliance on fusion and fission events in order to remove damaged mito-
chondria through mitophagy and the dependence of biogenesis on
protein translocation resulting from mitochondrial dynamics. These
preliminary discoveries are identifying novel therapeutic perspectives
in the effort to delay the onset of metabolic decline and age-associated
metabolic diseases.
ACKNOWLEDGMENTS
We apologize to our colleagues for omitting numerous references due to space
limitations. We thank Carsten Merkwirth and Kristen Berendzen for their helpful
comments on the manuscript. C.E.R is supported by the American Diabetes
Association Pathway to Stop Diabetes Grant 1-15-INI-12.
COMPETING FINANCIAL INTERESTS
The authors declare no competing financial interests.
1. López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks
of aging. Cell 153, 1194–1217 (2013).
2. De Cabo, R., Carmona-Gutierrez, D., Bernier, M., Hall, M.N. & Madeo, F. The search for
antiaging interventions: from elixirs to fasting regimens. Cell 157, 1515–1526 (2014).
3. Selman, C. & Withers, D.J. Mammalian models of extended healthy lifespan. Philos.
Trans. R.Soc. Lond. Ser.B.Biol. Sci. 366, 99–107 (2011).
4. Rowe, J.W., Minaker, K.L., Pallotta, J.A. & Flier, J.S. Characterization of the insulin
resistance of aging. J.Clin. Invest. 71, 1581–1587 (1983).
5. Fink, R.I., Kolterman, O. G., Griffin, J. & Olefsky, J. M. Mechanisms of insulin
resistance in aging. J.Clin. Invest. 71, 1523–1535 (1983).
6. Houtkooper, R.H. etal. The metabolic footprint of aging in mice. Sci. Rep. 1, 134
(2011).
7. Riera, C. E. et al. TRPV1 pain receptors regulate longevity and metabolism by
neuropeptide signaling. Cell 157, 1023–1036 (2014).
8. Kurosu, H. etal. Suppression of aging in mice by the hormone klotho. Science 309,
1829–1833 (2005).
9. Selman, C. etal. Evidence for lifespan extension and delayed age-related biomarkers
in insulin receptor substrate1 null mice. FASEB J. 22, 807–818 (2008).
10. Taguchi, A., Wartschow, L.M. & White, M.F. Brain IRS2 signaling coordinates life span
and nutrient homeostasis. Science 317, 369–372 (2007).
11. Speakman, J.R. etal. Uncoupled and surviving: individual mice with high metabolism
have greater mitochondrial uncoupling and live longer. Aging Cell 3, 87–95 (2004).
12. Galgani, J.E., Moro, C. & Ravussin, E. Metabolic flexibility and insulin resistance. Am.
J.Physiol. Endocrinol. Metab. 295, 1009–1017 (2008).
13. Kondratov, R.V., Kondratova, A.A., Gorbacheva, V.Y., Vykhovanets, O.V. & Antoch,
M.P. Early aging and age-related pathologies in mice deficient in BMAL1, the core
component of the circadian clock. Genes Dev. 20, 1868–1873 (2006).
14. Chang, H-C. & Guarente, L. SIRT1 mediates central circadian control in the SCN by a
mechanism that decays with aging. Cell 153, 1448–1460 (2013).
15. Sahar, S. & Sassone-Corsi, P. Metabolism and cancer: the circadian clock connection.
Nat. Rev. Cancer 9, 886–896 (2009).
16. Fontana, L., Partridge, L. & Longo, V.D. Dietary restriction, growth factors and aging:
from yeast to humans. Science 328, 321–326 (2010).
17. Mair, W. & Dillin, A. Aging and survival: the genetics of life span extension by dietary
restriction. Annu. Rev. Biochem. 77, 727–754 (2008).
18. Bruss, M.D., Khambatta, C.F., Ruby, M.A., Aggarwal, I. & Hellerstein, M.K. Calorie
restriction increases fatty acid synthesis and whole body fat oxidation rates. Am.
J.Physiol. Endocrinol. Metab. 298, E108–E116 (2010).
19. Bluher, M. Extended longevity in mice lacking the insulin receptor in adipose tissue.
Science 299, 572–574 (2003).
20. Foukas, L.C. etal. Long-term p110? PI3K inactivation exerts a beneficial effect on
metabolism. EMBO Mol. Med. 5, 563–571 (2013).
21. Zhang, Y. etal. The starvation hormone, fibroblast growth factor-21, extends lifespan
in mice. eLife 1, e00065 (2012).
22. Yuan, R. etal. Aging in inbred strains of mice: study design and interim report on
median lifespans and circulating IGF1 levels. Aging Cell 8, 277–287 (2009).
23. Berryman, D.E., Christiansen, J.S., Johannsson, G., Thorner, M.O. & Kopchick, J.J.
Role of the GH/IGF-1 axis in lifespan and healthspan: lessons from animal models.
Growth Horm. IGF Res. 18, 455–471 (2008).
24. Hsieh, C-C., DeFord, J. H., Flurkey, K., Harrison, D. E. & Papaconstantinou, J.
Implications for the insulin signaling pathway in Snell dwarf mouse longevity: a similarity
with the C.elegans longevity paradigm. Mech. Ageing Dev. 123, 1229–1244 (2002).
NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015 201
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW SERIES ON METABOLISMSERIES ON METABOLISM
REVIEW
25. Dominici, F.P., Hauck, S., Argentino, D.P., Bartke, A. & Turyn, D. Increased insulin
sensitivity and upregulation of insulin receptor, insulin receptor substrate (IRS)-1 and
IRS-2 in liver of Ames dwarf mice. J.Endocrinol. 173, 81–94 (2002).
26. Shah, J.H. & Cerchio, G.M. Hypoinsulinemia of hypothyroidism. Arch. Intern. Med.
132, 657–661 (1973).
27. Tsugane, S. & Inoue, M. Insulin resistance and cancer: epidemiological evidence.
Cancer Sci. 101, 1073–1079 (2010).
28. Naderali, E.K., Ratcliffe, S.H. & Dale, M.C. Obesity and Alzheimer’s disease: a link
between body weight and cognitive function in old age. Am. J.Alzheimers Dis. Other
Demen. 24, 445–449 (2009).
29. Ortega-Molina, A. et al. Pten positively regulates brown adipose function, energy
expenditure, and longevity. Cell Metab. 15, 382–394 (2012).
30. Coschigano, K. T., Clemmons, D., Bellush, L.L. & Kopchick, J. J. Assessment of
growth parameters and life span of GHR/BP gene-disrupted mice. Endocrinology 141,
2608–2613 (2000).
31. Ikeno, Y., Bronson, R.T., Hubbard, G.B., Lee, S. & Bartke, A. Delayed occurrence
of fatal neoplastic diseases in ames dwarf mice: correlation to extended longevity.
J.Gerontol. A.Biol. Sci. Med. Sci. 58, 291–296 (2003).
32. Ikeno, Y. etal. Reduced incidence and delayed occurrence of fatal neoplastic diseases
in growth hormone receptor/binding protein knockout mice. J.Gerontol. A.Biol. Sci.
Med. Sci. 64, 522–529 (2009).
33. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically
heterogeneous mice. Nature 460, 392–395 (2009).
34. Miller, R.A. etal. Rapamycin, but not resveratrol or simvastatin, extends life span of
genetically heterogeneous mice. J.Gerontol. A.Biol. Sci. Med. Sci. 66A, 191–201
(2011).
35. Corradetti, M.N. & Guan, K-L. Upstream of the mammalian target of rapamycin: do
all roads pass through mTOR? Oncogene 25, 6347–6360 (2006).
36. Lamming, D.W. etal. Rapamycin-induced insulin resistance is mediated by mTORC2
loss and uncoupled from longevity. Science 335, 1638–1643 (2012).
37. Roy, J., Paquette, J-S., Fortin, J.-F. & Tremblay, M.J. The immunosuppressant
rapamycin represses human immunodeficiency virus type1 replication. Antimicrob.
Agents Chemother. 46, 3447–3455 (2002).
38. Fang, Y. etal. Duration of rapamycin treatment has differential effects on metabolism
in mice. Cell Metab. 17, 456–462 (2013).
39. Pyo, J-O. etal. Overexpression of Atg5 in mice activates autophagy and extends
lifespan. Nat. Commun. 4, 2300 (2013).
40. Selman, C. etal. Ribosomal protein S6 kinase1 signaling regulates mammalian life
span. Science 326, 140–144 (2009).
41. Wu, J. J. et al. Increased mammalian lifespan and a segmental and tissue-specific
slowing of aging after genetic reduction of mTOR expression. Cell Rep. 4, 913–920
(2013).
42. Saltiel, A.R. & Kahn, C.R. Insulin signalling and the regulation of glucose and lipid
metabolism. Nature 414, 799–806 (2001).
43. Bratic, A. & Larsson, N-G. The role of mitochondria in aging. J.Clin. Invest. 123,
951–957 (2013).
44. Schapira, A.H.V. Mitochondrial diseases. Lancet 379, 1825–1834 (2012).
45. Herbener, G.H. A morphometric study of age-dependent changes in mitochondrial
population of mouse liver and heart. J.Gerontol. 31, 8–12 (1976).
46. Lanza, I.R. & Nair, K.S. Mitochondrial function as a determinant of life span. Pflüg.
Arch. Eur. J.Physiol. 459, 277–289 (2010).
47. Conley, K.E., Jubrias, S.A. & Esselman, P.C. Oxidative capacity and ageing in human
muscle. J.Physiol. 526, 203–210 (2000).
48. Lin, J. etal. Transcriptional co-activator PGC-1α drives the formation of slow-twitch
muscle fibres. Nature 418, 797–801 (2002).
49. Puigserver, P. etal. A cold-inducible coactivator of nuclear receptors linked to adaptive
thermogenesis. Cell 92, 829–839 (1998).
50. Mootha, V.K. etal. PGC-1α-responsive genes involved in oxidative phosphorylation
are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).
51. Patti, M.E. etal. Coordinated reduction of genes of oxidative metabolism in humans
with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc. Natl Acad.
Sci. USA 100, 8466–8471 (2003).
52. Wenz, T., Rossi, S.G., Rotundo, R.L., Spiegelman, B.M. & Moraes, C.T. Increased
muscle PGC-1α expression protects from sarcopenia and metabolic disease during
aging. Proc. Natl Acad. Sci. USA 106, 20405–20410 (2009).
53. Anderson, R. & Prolla, T. PGC-1α in aging and anti-aging interventions. Biochim.
Biophys. Acta 1790, 1059–1066 (2009).
54. Austin, S. & St-Pierre, J. PGC1α and mitochondrial metabolism — emerging concepts
and relevance in ageing and neurodegenerative disorders. J.Cell Sci. 125, 4963–4971
(2012).
55. Cantó, C. etal. AMPK regulates energy expenditure by modulating NAD+ metabolism
and SIRT1 activity. Nature 458, 1056–1060 (2009).
56. Jäger, S., Handschin, C., St-Pierre, J. & Spiegelman, B.M. AMP-activated protein
kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proc.
Natl Acad. Sci. USA 104, 12017–12022 (2007).
57. Narkar, V.A. et al. AMPK and PPARδ agonists are exercise mimetics. Cell 134,
405–415 (2008).
58. Viscomi, C. etal. Invivo correction of COX deficiency by activation of the AMPK/PGC-1α
axis. Cell Metab. 14, 80–90 (2011).
59. Martin-Montalvo, A. etal. Metformin improves healthspan and lifespan in mice. Nat.
Commun. 4, 2192 (2013).
60. Kalender, A. etal. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-
dependent manner. Cell Metab. 11, 390–401 (2010).
61. Cantó, C. etal. Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting
and exercise in skeletal muscle. Cell Metab. 11, 213–219 (2010).
62. Price, N.L. etal. SIRT1 is required for AMPK activation and the beneficial effects of
resveratrol on mitochondrial function. Cell Metab. 15, 675–690 (2012).
63. Nicholls, D.G., Bernson, V.S.M. & Heaton, G.M. in Effectors of Thermogenesis (eds.
Girardier, L. & Seydoux, J.) 89–93 (Birkhäuser Basel, 1978).
64. Echtay, K.S. etal. Superoxide activates mitochondrial uncoupling proteins. Nature
415, 96–99 (2002).
65. Gates, A. C. et al. Respiratory uncoupling in skeletal muscle delays death and
diminishes age-related disease. Cell Metab. 6, 497–505 (2007).
66. Nisoli, E. etal. Calorie restriction promotes mitochondrial biogenesis by inducing the
expression of eNOS. Science 310, 314–317 (2005).
67. Lanza, I.R. etal. Chronic caloric restriction preserves mitochondrial function in senescence
without increasing mitochondrial biogenesis. Cell Metab. 16, 777–788 (2012).
68. St-Pierre, J. etal. Bioenergetic analysis of peroxisome proliferator-activated receptorγ
coactivators 1α and 1β (PGC-1α and PGC-1β) in muscle cells. J.Biol. Chem. 278,
26597–26603 (2003).
69. Schriner, S.E. etal. Extension of murine life span by overexpression of catalase targeted
to mitochondria. Science 308, 1909–1911 (2005).
70. Pérez, V.I. etal. The overexpression of major antioxidant enzymes does not extend the
lifespan of mice. Aging Cell 8, 73–75 (2009).
71. Van Remmen, H. etal. Life-long reduction in MnSOD activity results in increased
DNA damage and higher incidence of cancer but does not accelerate aging. Physiol.
Genomics 16, 29–37 (2003).
72. Zhang, Y. etal. Mice deficient in both Mn superoxide dismutase and glutathione
peroxidase-1 have increased oxidative damage and a greater incidence of pathology
but no reduction in longevity. J.Gerontol.A.Biol. Sci. Med. Sci. 64, 1212–1220
(2009).
73. Edgar, D. etal. Random point mutations with major effects on protein-coding genes
are the driving force behind premature aging in mtDNA mutator mice. Cell Metab. 10,
131–138 (2009).
74. Trifunovic, A. etal. Premature ageing in mice expressing defective mitochondrial DNA
polymerase. Nature 429, 417–423 (2004).
75. Vermulst, M. etal. DNA deletions and clonal mutations drive premature aging in
mitochondrial mutator mice. Nat. Genet. 40, 392–394 (2008).
76. Kolesar, J.E. etal. Defects in mitochondrial DNA replication and oxidative damage in
muscle of mtDNA mutator mice. Free Radic. Biol. Med. 75, 241–251 (2014).
77. Yee, C., Yang, W. & Hekimi, S. The intrinsic apoptosis pathway mediates the
pro-longevity response to mitochondrial ROS in C.elegans. Cell 157, 897–909 (2014).
78. Dillin, A. etal. Rates of behavior and aging specified by mitochondrial function during
development. Science 298, 2398–2401 (2002).
79. Lee, S.S. etal. A systematic RNAi screen identifies a critical role for mitochondria in
C.elegans longevity. Nat. Genet. 33, 40–48 (2003).
80. Durieux, J., Wolff, S. & Dillin, A. The cell-non-autonomous nature of electron transport
chain-mediated longevity. Cell 144, 79–91 (2011).
81. Yoneda, T. etal. Compartment-specific perturbation of protein handling activates genes
encoding mitochondrial chaperones. J.Cell Sci. 117, 4055–4066 (2004).
82. Mottis, A., Jovaisaite, V. & Auwerx, J. The mitochondrial unfolded protein response in
mammalian physiology. Mamm. Genome 25, 424–433 (2014).
83. Zhao, Q. etal. A mitochondrial specific stress response in mammalian cells. EMBO J.
21, 4411–4419 (2002).
84. Houtkooper, R.H. et al. Mitonuclear protein imbalance as a conserved longevity
mechanism. Nature 497, 451–457 (2013).
85. Dell’agnello, C. etal. Increased longevity and refractoriness to Ca2+-dependent
neurodegeneration in Surf1 knockout mice. Hum. Mol. Genet. 16, 431–444
(2007).
86. Lapointe, J. & Hekimi, S. Early mitochondrial dysfunction in long-lived Mclk1+/– mice.
J.Biol. Chem. 283, 26217–26227 (2008).
87. Bai, P. etal. PARP-2 regulates SIRT1 expression and whole-body energy expenditure.
Cell Metab. 13, 450–460 (2011).
88. Bai, P. et al. PARP-1 inhibition increases mitochondrial metabolism through SIRT1
activation. Cell Metab. 13, 461–468 (2011).
89. Lagouge, M. etal. Resveratrol improves mitochondrial function and protects against
metabolic disease by activating SIRT1 and PGC-1α. Cell 127, 1109–1122 (2006).
90. Yoshino, J., Mills, K.F., Yoon, M.J. & Imai, S. Nicotinamide mononucleotide, a key
NAD+ intermediate, treats the pathophysiology of diet- and age-induced diabetes in
mice. Cell Metab. 14, 528–536 (2011).
91. Kelley, D.E., He, J., Menshikova, E.V. & Ritov, V.B. Dysfunction of mitochondria in
human skeletal muscle in type2 diabetes. Diabetes 51, 2944–2950 (2002).
92. Fisher-Wellman, K.H. & Neufer, P.D. Linking mitochondrial bioenergetics to insulin
resistance via redox biology. Trends Endocrinol. Metab. 23, 142–153 (2012).
93. Lowell, B.B. & Shulman, G.I. Mitochondrial dysfunction and type2 diabetes. Science
307, 384–387 (2005).
94. Cholerton, B., Baker, L.D. & Craft, S. Insulin resistance and pathological brain ageing.
Diabet. Med. 28, 1463–1475 (2011).
95. Evans, J.L., Maddux, B.A. & Goldfine, I.D. The molecular basis for oxidative stress-
induced insulin resistance. Antioxid. Redox Signal. 7, 1040–1052 (2005).
96. Halliwell, B. Reactive oxygen species and the central nervous system. J.Neurochem.
59, 1609–1623 (1992).
97. Pipatpiboon, N., Pratchayasakul, W., Chattipakorn, N. & Chattipakorn, S.C. PPARγ
agonist improves neuronal insulin receptor function in hippocampus and brain
mitochondria function in rats with insulin resistance induced by long term high-fat
diets. Endocrinology 153, 329–338 (2012).
202 NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEW
SERIES ON METABOLISMSERIES ON METABOLISM REVIEW
98. Thaler, J.P. etal. Obesity is associated with hypothalamic injury in rodents and humans.
J.Clin. Invest. 122, 153–162 (2012).
99. Kleinridders, A. et al. Leptin regulation of Hsp60 impacts hypothalamic insulin
signaling. J.Clin. Invest. 123, 4667–4680 (2013).
100. Chan, D.C. Fusion and fission: interlinked processes critical for mitochondrial health.
Annu. Rev. Genet. 46, 265–287 (2012).
101. Gomes, L.C., Di Benedetto, G. & Scorrano, L. During autophagy mitochondria
elongate, are spared from degradation and sustain cell viability. Nat. Cell Biol. 13,
589–598 (2011).
102. Green, D.R., Galluzzi, L. & Kroemer, G. Mitochondria and the autophagy-
inflammation-cell death axis in organismal aging. Science 333, 1109–1112
(2011).
103. Zhang, J. Autophagy and mitophagy in cellular damage control. Redox Biol. 1, 19–23
(2013).
104. Lee, S. etal. Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate
cellular senescence. J.Biol. Chem. 282, 22977–22983 (2007).
105. Dietrich, M.O., Liu, Z-W. & Horvath, T.L. Mitochondrial dynamics controlled by
mitofusins regulate Agrp neuronal activity and diet-induced obesity. Cell 155,
188–199 (2013).
106. Schneeberger, M. etal. Mitofusin2 in POMC neurons connects ER stress with leptin
resistance and energy imbalance. Cell 155, 172–187 (2013).
107. De Brito, O. M. & Scorrano, L. Mitofusin 2 tethers endoplasmic reticulum to
mitochondria. Nature 456, 605–610 (2008).
108. Sebastián, D. etal. Mitofusin2 (Mfn2) links mitochondrial and endoplasmic reticulum
function with insulin signaling and is essential for normal glucose homeostasis. Proc.
Natl Acad. Sci. USA 109, 5523–5528 (2012).
109. Muñoz, J.P. etal. Mfn2 modulates the UPR and mitochondrial function via repression
of PERK. EMBO J. 32, 2348–2361 (2013).
110. Ozcan, U. etal. Endoplasmic reticulum stress links obesity, insulin action, and type2
diabetes. Science 306, 457–461 (2004).
111. Garg, A.D. etal. ER stress-induced inflammation: does it aid or impede disease
progression? Trends Mol. Med. 18, 589–598 (2012).
112. Zhang, K. & Kaufman, R.J. From endoplasmic-reticulum stress to the inflammatory
response. Nature 454, 455–462 (2008).
113. Zhang, G. etal. Hypothalamic programming of systemic ageing involving IKK-β, NF-κB
and GnRH. Nature 497, 211–216 (2013).
114. Cheng, Z. & Almeida, F.A. Mitochondrial alteration in type2 diabetes and obesity:
an epigenetic link. Cell Cycle 13, 890–897 (2014).
NATURE CELL BIOLOGY VOLUME 17 | NUMBER 3 | MARCH 2015 203
© 2015 Macmillan Publishers Limited. All rights reserved
... Another hallmark of aging is the process of controlling the coordination between mitochondrial and nuclear activities that are essential for overall cellular respiration. Disruption of such coordination leads to mitochondrial dysfunction, affecting overall aging [74,83]. Signaling pathways drive the crosstalk between the mitochondria and the nucleus, which in turn affects nuclear gene expression to maintain mitochondrial homeostasis [84,85]. ...
Article
Full-text available
The mammalian target of rapamycin (mTOR) is a pivotal regulator, integrating diverse environmental signals to control fundamental cellular functions, such as protein synthesis, cell growth, survival, and apoptosis. Embedded in a complex network of signaling pathways, mTOR dysregulation is implicated in the onset and progression of a range of human diseases, including metabolic disorders such as diabetes and cardiovascular diseases, as well as various cancers. mTOR also has a notable role in aging. Given its extensive biological impact, mTOR signaling is a prime therapeutic target for addressing these complex conditions. The development of mTOR inhibitors has proven advantageous in numerous research domains. This review delves into the significance of mTOR signaling, highlighting the critical components of this intricate network that contribute to disease. Additionally, it addresses the latest findings on mTOR inhibitors and their clinical implications. The review also emphasizes the importance of developing more effective next-generation mTOR inhibitors with dual functions to efficiently target the mTOR pathways. A comprehensive understanding of mTOR signaling will enable the development of effective therapeutic strategies for managing diseases associated with mTOR dysregulation.
Article
Full-text available
Tumor immunotherapy has significantly transformed the field of oncology over the past decade. An optimal tumor immunotherapy would ideally elicit robust innate and adaptive immune responses within tumor immune microenvironment (TIME). Unfortunately, immune system experiences functional decline with chronological age, a process termed “immunosenescence,” which contributes to impaired immune responses against pathogens, suboptimal vaccination outcomes, and heightened vulnerability to various diseases, including cancer. In this context, we will elucidate hallmarks and molecular mechanisms underlying immunosenescence, detailing alterations in immunosenescence at molecular, cellular, organ, and disease levels. The role of immunosenescence in tumorigenesis and senescence‐related extracellular matrix (ECM) has also been addressed. Recognizing that immunosenescence is a dynamic process influenced by various factors, we will evaluate treatment strategies targeting hallmarks and molecular mechanisms, as well as methods for immune cell, organ restoration, and present emerging approaches in immunosenescence for tumor immunotherapy. The overarching goal of immunosenescence research is to prevent tumor development, recurrence, and metastasis, ultimately improving patient prognosis. Our review aims to reveal latest advancements and prospective directions in the field of immunosenescence research, offering a theoretical basis for development of practical anti‐immunosenescence and anti‐tumor strategies.
Article
Cellular senescence is a stress-induced irreversible cell cycle arrest involved in tumor suppression and aging. Many stresses, such as telomere shortening and oncogene activation, induce senescence by damaging nuclear DNA. However, the mechanisms linking DNA damage to senescence remain unclear. Here, we show that DNA damage response (DDR) signaling to mitochondria triggers senescence. A genome-wide small interfering RNA screen implicated the outer mitochondrial transmembrane protein BNIP3 in senescence induction. We found that BNIP3 is phosphorylated by the DDR kinase ataxia telangiectasia mutated (ATM) and contributes to an increase in the number of mitochondrial cristae. Stable isotope labeling metabolomics indicated that the increase in cristae enhances fatty acid oxidation (FAO) to acetyl–coenzyme A (acetyl-CoA). This promotes histone acetylation and expression of the cyclin-dependent kinase inhibitor p16 INK4a . Notably, pharmacological activation of FAO alone induced senescence both in vitro and in vivo. Thus, mitochondrial energy metabolism plays a critical role in senescence induction and is a potential intervention target to control senescence.
Article
The gut microbiota is pivotal in maintaining health, with most microorganisms being beneficial, except for a few pathogens. Emerging evidence suggests a link between the gut microbiome and aging, hinting at its potential role in longevity. However, understanding the relationship is challenging due to the microbiota's complexity. This perspective summarizes the mechanisms by which gut microbes regulate host lifespan and explores genetic manipulation strategies to promote healthy aging in the elderly.
Article
Full-text available
Age is a prominent risk factor for cardiometabolic disease, often leading to heart structural and functional changes. However, precise molecular mechanisms underlying cardiac remodeling and dysfunction exclusively resulting from physiological aging remain elusive. Previous research demonstrated age‐related functional alterations in baboons, analogous to humans. The goal of this study is to identify early cardiac molecular alterations preceding functional adaptations, shedding light on the regulation of age‐associated changes. Unbiased transcriptomics of left ventricle samples are performed from female baboons aged 7.5–22.1 years (human equivalent ≈30–88 years). Weighted‐gene correlation network and pathway enrichment analyses are performed, with histological validation. Modules of transcripts negatively correlated with age implicated declined metabolism‐oxidative phosphorylation, tricarboxylic acid cycle, glycolysis, and fatty‐acid β‐oxidation. Transcripts positively correlated with age suggested a metabolic shift toward glucose‐dependent anabolic pathways, including hexosamine biosynthetic pathway (HBP). This shift is associated with increased glycosaminoglycan synthesis, modification, precursor synthesis via HBP, and extracellular matrix accumulation, verified histologically. Upregulated extracellular matrix‐induced signaling coincided with glycosaminoglycan accumulation, followed by cardiac hypertrophy‐related pathways. Overall, these findings revealed a transcriptional shift in metabolism favoring glycosaminoglycan accumulation through HBP before cardiac hypertrophy. Unveiling this metabolic shift provides potential targets for age‐related cardiac diseases, offering novel insights into early age‐related mechanisms.
Article
A surge of public interest in NMN supplementation has been observed in recent years. However, whether NMN supplements are effective in improving metabolic health remains unclear. The objective of the review was to assess the effects of NMN supplementation on fasting glucose, triglycerides, total cholesterol, LDL-C, and HDL-C in adults. Studies were located by searching four databases (PubMed, Embase, Cochrane, and Web of Science). Two reviewers independently conducted title/abstract and full-text screening, data extraction, and risk-of-bias assessment. Of the 4049 records reviewed, 12 studies with a total of 513 participants met the criteria for analysis. Random-effects meta-analyses found an overall significant effect of NMN supplementation in elevating blood NAD levels. However, most of the clinically relevant outcomes were not significantly different between NMN supplementation and control group. Risk-of-bias assessment using RoB2 showed some concerns in seven studies and high risk of bias in the other five studies. Together, our findings suggest that an exaggeration of the benefits of NMN supplementation may exist in the field. Although the limited number of eligible studies was sufficiently powered to detect changes in the abovementioned primary outcomes, more studies are needed to conclude about the exact effects of NMN supplementation.
Article
Energetic demand and nutrient supply fluctuate as a function of time-of-day, in alignment with sleep-wake and fasting-feeding cycles. These daily rhythms are mirrored by 24-hour oscillations in numerous cardiovascular functional parameters, including blood pressure, heart rate, and myocardial contractility. It is, therefore, not surprising that metabolic processes also fluctuate over the course of the day, to ensure temporal needs for ATP, building blocks, and metabolism-based signaling molecules are met. What has become increasingly clear is that in addition to classic signal-response coupling (termed reactionary mechanisms), cardiovascular-relevant cells use autonomous circadian clocks to temporally orchestrate metabolic pathways in preparation for predicted stimuli/stresses (termed anticipatory mechanisms). Here, we review current knowledge regarding circadian regulation of metabolism, how metabolic rhythms are synchronized with cardiovascular function, and whether circadian misalignment/disruption of metabolic processes contribute toward the pathogenesis of cardiovascular disease.
Article
Circadian rhythms govern a number of fundamental physiological functions in almost all organisms, from prokaryotes to humans. Disruption of these rhythms can have a profound influence to human health and has been linked to depression, insomnia, jet lag, coronary heart disease and a variety of neurodegenerative disorders. The complex program of gene expression that characterizes circadian physiology is possible through dynamic changes in chromatin transitions. A central element of the clock machinery, the protein CLOCK, has HAT enzymatic properties. It directs acetylation of histone H3 and of the non‐histonic protein BMAL1, its own dimerization partner, at K537, an event that is essential for circadian function. We show that the HDAC activity of the NAD ⁺ ‐dependent SIRT1 enzyme is regulated in a circadian manner. SIRT1 associates with CLOCK and is recruited to the CLOCK:BMAL1 chromatin complex at circadian gene promoters. Genetic ablation of the Sirt1 gene or pharmacological inhibition of SIRT1 leads to disturbances in the circadian cycle. Finally, using liver‐specific SIRT1 mutant mice we show that SIRT1 contributes to circadian control in vivo . We propose that SIRT1 functions as an enzymatic rheostat of circadian function, transducing signals originated by cellular metabolites to the clock machinery. One question however arose: how acetylation can be oscillatory if the intracellular levels of CLOCK and SIRT1 do not cycle? We demonstrate that intracellular NAD ⁺ levels cycle with a 24 h rhythm, an oscillation driven by the circadian clock. CLOCK:BMAL1 regulate the circadian expression of Nampt (nicotinamide phosphoribosyltransferase), a rate limiting step enzyme in the NAD ⁺ salvage pathway. Our findings reveal an interlocked transcriptional‐enzymatic feedback loop that governs the interplay between cellular metabolism and circadian rhythms.
Article
Mutation analyses in the nematode, Caenorhabditis elegans, and mice have identified genes that increase their life-span via hormonal signal transduction, i.e. the insulin/insulin-like growth factor-1 (IGF-1) pathway in nematodes, and the growth hormone (GH)-thyriod stimulating hormone (TSH)-prolactin system in Snell dwarf mouse mutants. We have shown that the GH deficiency due to Pit1 mutation in the long-lived Snell dwarf mice may decrease circulating insulin levels, thereby resulting in a decreased activity of the insulin/IGF-1 signaling pathway. The data presented are consistent with our hypothesis that the decreased circulating insulin levels resulting from the Pit1 mutation mimics a physiological state similar to that proposed to occur in the long-lived C. elegons, daf-2 mutant. Our studies demonstrate a series of changes in components of the insulin/IGF-1-signaling pathway that suggest a reduction-of-function of this pathway in the aged dwarf. These include a decreased IRS-2 pool level, a decrease in PI3K activity and its association with IRS-2 and decreased docking of p85alpha to IRS-2. Our data also suggest a preferential docking of IRS-2-p85alpha-p110alpha in the aged dwarf liver and IRS-2-p85alpha-p110beta in the aged control. We speculate that the preference for the p110alpha-containing complex may be a specific characteristic of a downstream segment of the longevity-signaling cascade. We conclude that the Pit1 mutation may result in physiological homeostasis that favors longevity, and that the Snell dwarf mutant conforms to the nematode longevity paradigm.
Article
In the present study we have used hypopituitary Ames dwarf mice, which lack GH, prolactin and TSH, to investigate the consequences of the deficiency of these hormones on glucose homeostasis and on the initial components of the insulin signal transduction pathway in the liver. Ames dwarf mice displayed hypersensitivity to insulin since they maintained lower fasting glucose concentrations (73% of control values), had significantly reduced amounts of insulin (58% of control values), and exhibited an increased hypoglycemic response to exogenous insulin. Probably as a result of reduced insulin production, Ames dwarf mice displayed intolerance to glucose. The insulin-stimulated phosphorylation of the insulin receptor (IR) tended to be increased in the liver of Ames dwarf mice, while IR receptor protein content was increased by 38%. Insulin-stimulated phosphorylation of insulin receptor substrate (IRS)-1 and IRS-2 was increased by 61 and 72% respectively, while IRS-1 and IRS-2 protein levels were increased by 76 and 95%. The insulin-stimulated association of the p85 regulatory subunit of phosphatidylinositol (PI) 3-kinase with IRS-1 was increased by 28%, but unaltered with IRS-2. Interestingly, while the insulin-stimulated phosphotyrosinederived PI 3-kinase activity was not changed, insulin-stimulated protein kinase B activation was increased by 41% in this tissue. These alterations may account for the insulin hypersensitivity exhibited by these animals. The present findings in long-lived Ames dwarf mice add to the evidence that insulin signaling is importantly related to the regulation of aging and life span.