You are on page 1of 23

Biogerontology

Mitochondrial dysfunction in metabolism and ageing: shared


mechanisms and outcomes?
Guillermo Lo´pez-Lluch . Juan Diego Herna´ndez-Camacho . Daniel J. Moreno Ferna´ndez-Ayala .

Pla´cido Navas
G. Lo´pez-Lluch (&) J. D. Herna´ndez-Camacho D.
J. M. Ferna´ndez-Ayala P. Navas
Centro Andaluz de Biologı´a del Desarrollo, CABD-CSIC,
CIBERER, Instituto de Salud Carlos III, Universidad
Pablo de Olavide, Carretera de Utrera km. 1,
Received: 17 May 2018/Accepted: 21 August 2018 41013 Seville, Spain e-
Springer Nature B.V. 2018 mail: glopllu@upo.es

Abstract Mitochondria are key in the metabolism progression of age-related diseases and metabolic
of aerobic organisms and in ageing progression disease.
and agerelated diseases. Mitochondria are essential
for obtaining ATP from glucose and fatty acids but Keywords Mitochondria Metabolism Ageing
also in many other essential functions in cells Metabolic syndrome Fat Mitochondrial dynamics
including aminoacids metabolism, pyridine ROS
synthesis, phospholipid modifications and calcium
regulation. On the other hand, the activity of
mitochondria is also the principal source of
reactive oxygen species in cells. Ageing and Introduction
chronic age-related diseases are associated with
the deregulation of cell metabolism and Ageing is a natural and progressive process,
dysfunction of mitochondria. Cell metabolism is occurring in all the living organisms, characterized
controlled by three major nutritional sensors: by the deterioration of cells, tissues and organs
mTOR, AMPK and Sirtuins. These factors control affecting structure and physiology. Ageing is a
mitochondrial biogenesis and dynamics by multifactorial process involving several aspects of
regulating fusion, fission and turnover through the cells such as genomic instability, epigenetic
mito- and autophagy. A complex interaction alterations, telomere attrition, loss of proteostasis,
between the activity of these nutritional sensors, deregulation of nutrient sensing, mitochondrial
mitochondrial biogenesis rate and dynamics exists dysfunction, stem cell exhaustion, and alteration of
and affect ageing, age-related diseases including intracellular communication (Kennedy et al. 2014;
metabolic disease. Further, mitochondria maintain Lopez-Otin et al. 2013).
a constant communication with nucleus Mitochondria are the powerhouse of cells
modulating gene expression and modifying providing ATP for all the activities occurring in
epigenetics. In this review we highlight the aerobic organisms. Their dysfunction has been
importance of mitochondria in ageing and the associated with many age-related diseases
repercussion in the (sarcopenia, type 2 diabetes (T2DM), neurological

123
Biogerontology
disorders, cardiovascular disease, liver and kidney Barzilai 2009). In a transgenic mouse model,
disease or cancer) and the functional decline FIRKO, disruption of the insulin receptor in
associated with ageing (Boengler et al. 2017; adipose tissue is associated with the increase of
Bratic and Trifunovic 2010; Fabbri et al. 2017; longevity (Bluher et al. 2003). When compared
Lane et al. 2015; Payne and Chinnery 2015; Ryan with wild type mice, FIRKO animals show higher
et al. 2015; Wallace 2012). Further, mitochondria mitochondrial activity, elevated mitochondrial
are at the center of cell metabolism and their DNA content, modest increases in mitochondrial
regulation has been demonstrated to be important size and higher gene expression of proteins
in longevity in many different organisms from involved in oxidative metabolism. This mouse
yeast to primates (Lopez-Lluch et al. 2015). The model suggests that mitochondrial metabolism in
efficiency of mitochondrial activity has been white adipose tissue is important in the
considered essential in ageing and longevity and, progression of ageing and the development of
in fact, a ‘‘Mitochondrial Theory’’ was coined to metabolic diseases (Katic et al. 2007).
explain how ageing occurs (Harman 1972). In this article we discuss the importance of the
Mitochondrial theory of ageing was immediately maintenance of mitochondrial homeostasis during
associated with the widely known Free Radical ageing and MS and how the dysregulation of these
Theory of ageing that considers mitochondria as essential organelles affects the progression of both.
the central sources of reactive oxygen species Further, age-associated dysfunction of
(ROS) that cause the age-associated accumulation mitochondria is hypothesized to be the key factor
of damaged structures during ageing (Harman that associates the appearance of MS during
1956, 1972). ageing.
Activity, dynamics and turnover of
mitochondria depend on the energy intake and are
regulated by key factors of metabolism (Lopez- Mitochondria, structure and dynamics
Lluch 2017) One of these regulators is the
mammalian Target of Rapamycin (mTOR), which Mitochondria are dynamic organelles integrated by
is activated by high calorie intake or high levels of two membranes that define an intermembrane
aminoacids (Laplante and Sabatini 2009). The space and an inner matrix. The energy production
negative regulators of mTOR are AMPactivated in mitochondria relies on the electron transport
kinase (AMPK) and the NAD ?-dependent protein chain (ETC) that transfer high-energy electrons
deacetylases, Sirtuins, that are activated when from redox metabolic intermediaries such as
energy uptake is limited (Hardie 2011). NADH or FADH2 originated in glycolysis, fatty-
The metabolic syndrome (MS) is a high- acid oxidation and Krebs cycle, to oxygen. During
prevalence disorder that is defined by the presence this transfer, complexes I, III and IV of the ETC
of three or more of the following criteria: obesity act as proton pumps from mitochondrial matrix to
located in the abdominal region, high blood the intermembrane space. Proton pumping results
pressure, hypertriglyceridemia, low HDL in a proton gradient that generates a proton motive
cholesterol, and/or increased fasting glucose force used to synthesize ATP from ADP and Pi via
(Alberti et al. 2009; Shaw et al. 2005). MS has the ATP synthase (complex V). Both, ETC and
been associated with many ageingrelated chronic ATP synthase are located in the inner membrane
diseases such as cardiovascular disease (Mottillo of the mitochondria. For this reason, mitochondria
et al. 2010; Sundstrom et al. 2006), type 2 diabetes are the center of aerobic metabolism (Sousa et al.
mellitus (T2DM) (Kahn and Flier 2000) or cancer 2018).
(Cowey and Hardy 2006). Among the main The activity of mitochondria is regulated by the
contributors of the development of MS, insulin demand of energy in cells. When this demand
resistance and abdominal obesity are considered increases, respiration rate increases promoting a
the most significant (Alberti et al. 2009) and are higher ATP production (Bratic and Trifunovic
associated with ageing progression (Huffman and 2010). During this process, ETC complexes,

123
Biogerontology
mainly complexes I and III, can transfer electrons processes, depending on the bioenergetic state of
directly to oxygen and produce superoxide (ROS). the cell (Liesa et al. 2009). Mitochondrial fusion
For this reason, mitochondria have been also has been associated with active mitochondria
considered an important source of intracellular whereas fission is associated with damaged
ROS production in cells (Lambert and Brand mitochondria and their removal by mito/autophagy
2009). High rates of mitochondrial ROS have been (Lopez-Lluch et al. 2008). The main proteins
associated with the accumulation of most of the involved in fusion are large GTPases such as
oxidized mitochondrial macromolecules during mitofusins 1 and 2 (Mfn1 and Mfn2) and
ageing and in many chronic diseases (Page et al. autosomal dominant optic atrophy-1 (OPA1).
2010). Mitofusins are located at the surface of the outer
Coenzyme Q (CoQ) is a lipid present in the membrane of mitochondria and act in the fusion
inner mitochondrial membrane that mainly process of these membranes whereas OPA1 is an
transfers electrons from complexes I and II to intermembrane protein anchored to the inner
Complex III. Further, changes in the levels of CoQ membrane and acts in the fusion of inner
have been also associated with ageing progression membranes (Chen and Chan 2005; Olichon et al.
(Lopez-Lluch et al. 2010). In humans, CoQ 2002). Mfn2 also plays an important function in
decrease has been associate with several tethering mitochondria and endoplasmic reticulum
mitochondrial-linked diseases and with chronic (ER) playing a role in the regulation of
agerelated diseases such as obesity or type II communication between both organelles and
diabetes (Hernandez-Camacho et al. 2018). A endoplasmic reticulum (ER) stress and fat
significant reduction in the rate of CoQ accumulation in conditions of metabolic stress (de
biosynthesis has been proposed to occur during the Brito and Scorrano 2008, 2009; Naon et al. 2016).
ageing process and ageassociated diseases (Battino The main proteins involved in mitochondrial
et al. 1995; Beyer et al. 1985). Sedentary and fission are the dynamin related protein 1 (DRP1)
obese people show low levels of CoQ10 in plasma and the fission protein 1 (FIS1). DRP1 is also a
probably associated with a higher risk for GTPase, and is located mainly in the cytosol
cardiovascular disease (Del Pozo-Cruz et al. although a fraction is localized at punctate
2014). and the ratio of plasma cholesterol to CoQ10 structures onto the outer mitochondria membrane
has been recently suggested as biomarker of the (Smirnova et al. 2001). FIS1 is a protein inserted
development of early complications of obesity in in the outer mitochondrial membrane that recruits
children (Gvozdjakova et al. 2012). CoQ can be DRP1 (Santel and Frank 2008; Yoon et al. 2003).
found in a reduced form, ubiquinol (COQ10H2) and Prohibitin is a complex of two proteins, PHB-1
a oxidized form, ubiquinone (CoQ 10) and it has and PHB-2, which has been involved in C. elegans
been demonstrated that the ratio between longevity (Lourenco et al. 2015). Prohibitins are
reduced/oxidized CoQ10 (CoQ10H2/CoQ10) is ubiquitous and form a ring-like complex at the
essential for complex I stability and ROS inner membrane of the mitochondria (Artal-Sanz
production, which has been associated with the and Tavernarakis 2009). Prohibitins stabilize
extension of longevity (Guaras et al. 2016; Scialo OPA1 and then regulate mitochondrial fusion and
et al. 2016). For this reason, an unbalance of cristae morphogenesis. However, the exact
CoQ10 equilibrium in mitochondria could be also function of this complex in mitochondria has been
associated with mitochondrial dysfunction in not completely clarified yet although it has been
ageing and MS. associated with alterations of mitochondrial
In seems clear that during ageing and MS, less function, fat metabolism and oxidative stress
efficient mitochondria, producing less ATP and response (Lourenco et al. 2015; Theiss et al. 2009)
high ROS levels, accumulate (Lopez-Lluch 2017; (Fig. 1).
Son et al. 2004). Mitochondria are very dynamic
organelles that change their shape, biogenesis
ratio, turnover and dynamics, fusion/fission Mitochondrial are regulated by nutrient sensors

123
Biogerontology
In order to maintain an equilibrated activity, factor of mTOR in the regulation of cell
mitochondrial physiology must respond to the bioenergetics by inhibiting its activity directly
needs of cell metabolism. Thus, mitochondrial (Gwinn et al. 2008) or indirectly (Inoki et al.
activity, biogenesis, turnover and dynamics 2003). In general, AMPK stimulates cellular
respond to main metabolism sensors in cells such catabolism of sugars, proteins and lipids and
as mTOR, AMPK and sirtuins [for a more inhibits many anabolic enzymes affecting
comprehensive, see (Lopez-Lluch 2017; Lopez- glycogen and lipid biosynthesis (Hardie et al.
Lluch et al. 2015)]. The ubiquitous serine/ 2012).
threonine kinase mTOR is activated in high calorie Sirtuins are a protein family found in all the
intake conditions inducing anabolic activities. organisms that regulates the activity of many
Activation of mTOR has been associated with enzymes by removing acetyl residues, except in
angiogenesis, tumor development, insulin the case of mammalian SIRT4 and SIRT5 that
resistance, adipogenesis and immune cells show ADP-ribosyl-transferase activity (Santa-
activation (Laplante and Sabatini 2009). The Cruz Calvo et al. 2012). Sirtuins act as metabolic
signaling pathway triggered by mTOR is sensors by detecting fluctuations in the
downregulated by AMPK and Sirtuins that are NAD?/NADH ratio such as in the case of reduced
activated when the amount of energy is low such nutrient availability. These proteins are involved
as in low food intake, starvation or calorie in different key aspects of cell physiology from
restriction (CR). AMPK acts as an antagonistic cell cycle control to metabolism and antioxidant
Fig. 1 Importance of the
equilibrium in
mitochondrial dynamics.
Mitochondria are
maintained in a balanced
status by their control via
biogenesis, dynamics and
removal. Mitochondrial
dynamics are controlled by
the fusion/fission balance
regulated by mitofusins
(Mfn1/2), OPA1 and
prohibitins (PHB) in the
case of fusion and DRP1
and FIS1 in the case of
fission. Dysfunction in the
equilibrium of these factors
can cause the accumulation
of giant mitochondria in
senescence or small and
damaged mitochondria in
obesity. In both cases, the
reduction of CoQ
biosynthesis, decrease of
mitochondrial biogenesis,
deterioration of
supercomplexes activities
and alteration of mito/
autophagy occur decreasing
mitochondria OXPHOS
activity and increasing ROS
production

123
Biogerontology
protection (Santa-Cruz Calvo et al. 2012). The modulation of the activity of Mn-SOD (Qiu et al.
effect of Sirtuins on the mitochondrial physiology 2010), regulation of autophagy by deacetylation of
and cell FOXO3 (Kume et al. 2010) and modulation of
metabolism has been associated with their effect mitochondrial dynamics by activating OPA1
on longevity and part of the benefits of CR can be (Samant et al. 2014).
explained by the activation of Sirtuins and Regarding turnover, AMPK and mTORC1
mitochondrial biogenesis (Guarente 2011, 2013; (mTOR complex 1) control the removal of
Imai and Guarente 2010). damaged mitochondria through the regulation of
All these nutrient sensor, mTOR, AMPK and autophagy and mitophagy. Whereas mTORC1
Sirtuins, regulate the activity of the peroxisome blocks autophagy by phosphorylating inhibitory
proliferator-activated receptor gamma coactivator sites of unc-51-like kinase 1/2 (ULK-1 and ULK-
1a (PGC1a), a key transcription factor involved in 2), initial mediators of this process (Chan 2009;
the activation of mitochondrial biogenesis. PGC1a Kim et al. 2011), AMPK induces autophagy by
stimulates mitochondrial biogenesis, regulates phosphorylating these same proteins in their
mitochondrial dynamics, modulates oxidative activation sites (Alers et al. 2012). In
phosphorylation, and controls mitochondrial mitochondrial removal, Forkhead box proteins
genome copy number (Gouspillou et al. 2014; (FoxO) proteins play an important role. FoxO are a
Handschin and Spiegelman 2006). PGC1a family of transcription factors involved in the
regulates the expression of many transcription regulation of cell growth, proliferation,
factors involved in mitochondrial biogenesis and differentiation and longevity. FoxO proteins
control of respiration such as nuclear respiratory induce several stress response genes indicating its
factors 1 and 2 (NRF1 and 2), important role in the induction of protective
peroxisomeproliferator activated receptor-c mechanisms in organisms (Goto and Takano
(PPARc) or mitochondrial transcription factor A 2009). Regulation of FoxO proteins during ageing
(TFAM) (MartinMontalvo and de Cabo 2013). and their modulation by metabolic regulators such
When cells are under nutrient-limited conditions, as mTOR, AMPK and Sirtuins associate these
AMPK and SIRT1 directly activate PGC1a proteins with the prevention of mitochondrial
through phosphorylation and deacetylation dysfunction by CR, polyphenols or exercise
respectively (Jager et al. 2007; Nemoto et al. (Lopez-Lluch and Navas 2016).
2005). SIRT1 not only activates transcriptional Sestrins are another highly conserved protein
activity of PGC1a by deacetylation (Rodgers et al. family encoded by genes known as Sens that are
2005), but also induces PGC1a expression since in upregulated after environmental stress, hypoxia
mice lacking SIRT1 PGC1a levels are also and DNA damage (Budanov and Karin 2008). In
reduced (Gerhart-Hines et al. 2007). On the other invertebrates, only one Sens gene is found whereas
hand, AMPK activates PGC1a by phosphorylation vertebrates express three different genes (Sens 1–
and induces its own transcription via a positive 3). Their importance in metabolism regulation
feedback loop (Jager et al. 2007) or by inducing its resides in their regulation of mTORC1 activity.
deacetylation through SIRT1 activation (Canto et Sestrins repress mTORC1 activity by activating
al. 2010). AMPK (Budanov and Karin 2008). Accordingly,
The activity of mitochondria is also regulated Sestrins have been recently proposed as
by Sirtuins. SIRT3, a Sirtuin family member prolongevity factors after the experiments
located in the mitochondria with deacetylase performed in dSens-deficient D. melanogaster
activity, is induced in skeletal muscle after CR and (Lee et al. 2010) and in C. elegans (Yang et al.
its levels decrease under high-fat diet indicating 2013). Further, inactivation of Sestrin genes in
calorie-dependent regulation (Palacios et al. 2009). invertebrates produces diverse metabolic
SIRT3 is involved in the catabolism of lipids and pathologies resembling accelerated aging such as
in the regulation of ETC complexes II, III and IV oxidative damage, fat accumulation, mitochondrial
activities by deacetylation (Kendrick et al. 2011),

123
Biogerontology
dysfunction, and muscle degeneration (Lee et al. Mitochondrial activity and turnover are affected
2013). during ageing
Sestrins have been recently associated with
ageing by affecting mitochondrial turnover. In Many evidences indicate that mitochondria play
mice, Sestrins have been associated with the key roles in the pathophysiology of ageing and in
regulation of metabolism producing a prolongevity earlier stages of events leading to the ageing
effect. These proteins are regulated by oxidative phenotype (Gonzalez-Freire et al. 2015; Kennedy
stress through p53, NRF-2, AP-1 and FoxO and et al. 2014; Lopez-Otin et al. 2013). Damaged
they regulate several components that reduce ROS mitochondria showing low ATP production and
production, increase mitochondrial biogenesis and high release of ROS are associated with ageing
induce mitophagy increasing the efficiency of the and age-related diseases that show dysregulation
oxidative metabolism (Lee et al. 2013). Transgenic of mitochondrial dynamics and turnover
mice deficient in Sens2 and Sens3 genes have (Chistiakov et al. 2014; LopezLluch 2017; Merry
demonstrated the important role of these proteins 2002).
in the regulation of lipid metabolism and the A factor involved in mitochondrial dysfunction
suppression of age and obesity-associated during ageing is the rate of superassembly of the
mitochondrial disorders (Bae et al. 2013; Lee et al. respiratory chain complexes. ETC complexes can
2012). Sesn2 KO mice show higher glucose be organized in larger structural and functional
intolerance, lower insulin response and units named as ‘‘respirasome’’ already found in
development of hepatosteatosis (Lee et al. 2012). bacteria and in mitochondrial membranes. This
Sestrins-dependent mTOR Complex I (mTORC1) supra-organization permits the reduction of
inactivation through activating AMPK is critical in diffusion distances of substrates and a more
the maintenance of autophagy and the elimination effective flow of electrons through the ETC. It has
of dysfunctional mitochondria (Ishihara et al. been suggested that age-associated destabilization
2013). On the other hand, Sens3 has been recently of supercomplexes of ETC might be important for
associated with the activation of mTORC2 and the development of mitochondrial ageing
Akt phosphorylation enhancing insulin sensitivity phenotype and in particular in post-mitotic tissues
and glucose metabolism (Tao et al. 2015). During (Gomez and Hagen 2012). Mitochondrial ETC
aging Sestrin levels decrease in muscle and complexes can release ROS by transferring
aerobic physical exercise is able to restore Sestrin electrons directly to molecular oxygen (Goncalves
2 levels in mice muscle restoring the capacity to et al. 2015), and the assembly state of these
respond to insulin and autophagy (Lenhare et al. complexes has been associated with the rate of
2017), As Sestrins are regulated by many stress ROS production in mitochondria (Genova and
factors including ROS, these proteins emerge as Lenaz 2015; MorenoLoshuertos and Enriquez
interesting regulators of mitochondrial turnover 2016). Consequently, the incapacity to build
and ageing-related diseases by mild stress as supercomplexes or their progressive deterioration
hormetic response (Wang et al. 2017). In humans, could be one of the causes of the increase in
a recent article indicates that Sestrin 1 and 3 mitochondrial ROS production during ageing
decreased in aged muscle although mRNA levels (Dencher et al. 2007; Genova and Lenaz 2015).
of SESN1 (Zeng et al. 2018). A very recent study Maintenance of the quality control of
also demonstrate that muscle from frail elderly mitochondria is considered a crucial factor to
people show significant reductions of Sestrin1 and counteract ageing process (Romanello and Sandri
Sestrin2 indicating the importance of these factors 2015; Weber and Reichert 2010). Accumulation of
in the maintenance of muscle capacity during damaged mitochondria during ageing is associated
aging (Rai et al. 2018). with the decline of the mitochondrial turnover
through inhibiting mitophagy; a specific
autophagy process that removes damaged
mitochondria (Chistiakov et al. 2014). Then, it

123
Biogerontology
seems clear that removal of defective damaged mitochondria by inducing mito- and
mitochondria is essential to maintain cellular autophagy (Lopez-Lluch and Navas 2016) (Fig. 2).
homeostasis. In this role, the BCL-2 Interacting Several studies suggest that ageing affects the
Protein 3 (BNIP3) and BNIP3/ NIX homologue in equilibrium of mitochondrial dynamics affecting
C. elegans (DCT-1) is key in mediating mitophagy fusion and fission. Senescence has been associated

and to increase longevity (Palikaras et al. 2015). In with the accumulation of giant dysfunctional
this role, ubiquitination of BNIP3 by PTEN mitochondria characterized by highly
Induced Putative Kinase 1 (PINK1)/ E3 Ubiquitin- interconnected networks and ultrastructural
Protein Ligase (PARKIN) pathway is essential for abnormalities (Beregi and Regius 1987)
the removal of damaged mitochondria by accompanied by higher production of ROS and
mitophagy (Harper et al. 2018). Further, damaged lower mitochondrial respiratory activity (Yoon et
mitochondria activate SKN-1/Nuclear factor al. 2006). Accordingly, blocking fission process by
erythroid-derived-like 2 (NRF-2) pathway in C. depleting FIS1 or inhibiting DRP1 leads
elegans (Fang et al. 2017), initiating a bipartite Fig. 2 Prolongevity factors prevent the accumulation of
retrograde signaling pathway that stimulates the damaged mitochondria by inducing the activity of
molecular regulators of mitochondrial turnover. A balanced
coordinated induction of both mitochondrial turnover of mitochondria is essential to avoid the
biogenesis and mitophagy (Palikaras et al. 2015), accumulation of damaged mitochondria. Accumulation of
indicating the importance of a balance between the damaged mitochondria is the main factor of mitochondrial
generation of new mitochondria and the removal dysfunction during aging and metabolic syndrome. Caloric
restriction, bioactive compounds such as polyphenols and
of damaged structures to increase longevity others and physical activity induce the activity of different
(Denzer et al. 2016). The renovation of factors which stimulate the biogenesis of new mitochondria
mitochondrial network plays also a key role in and the elimination of damaged structures by
health-span increase after CR (LopezLluch et al. mito/autophagy. Dysregulation of these molecular factors
by ageing or metabolic syndrome produces the
2008). Prolongevity effectors such as CR, exercise accumulation of damaged mitochondria and mitochondrial
and polyphenols prevent the accumulation of dysfunction
to cell senescence accompanied by the (Gomes and Scorrano 2008; Twig et al. 2008). A
accumulation of damaged mitochondria showing lower fission rate would allow mitochondria to
lower mitochondrial potential and higher ROS escape autophagic degradation and to maintain
production (Lee et al. 2007; Park et al. 2010). ATP production but with higher ROS production
These evidence highlight the importance of the during ageing (Gomes and Scorrano 2011).
presence of a functional fission machinery to Interestingly, fission proteins FIS1 and DRP1
remove damaged mitochondria by mito/autophagy levels increased in liver of a CR mouse model

123
Biogerontology
(40% CR for 6 months), whereas no changes were been also associated with ageing (Matsumoto
found in fusion-related proteins (Mfn1/2 and 2002). Mitochondrial dysfunction is also present in
OPA1) (Khraiwesh et al. 2013). Considering the obesity-related diseases such as insulin resistante
increase of mitochondrial biogenesis found in CR and type 2 diabetes mellitus, indicated by low
conditions (Lopez-Lluch et al. 2006; Nisoli et al. expression of mtDNA and reduction of the levels
2005), the increase in fission-related proteins as a of mitochondrial complexes affecting all the main
prolongevity mechanism would permit the organs involved in the development of metabolic
removal of damaged mitochondria and the syndrome such as muscle, liver and adipose tissue
regeneration of the mitochondrial network (Patti and Corvera 2010; Schottl et al. 2015).
(Khraiwesh et al. 2013; Lopez-Lluch et al. 2008). Mitochondrial dysfunction also reduces the
For more detailed information about the role of capacity to oxidize fatty acids (Patti and Corvera
changes of mitochondrial dynamics in longevity, 2010; Toledo and Goodpaster 2013). The
see (Sebastian et al. 2017). deficiency in the capacity of mitochondria to
In summary, multiple factors affects metabolize lipids has been proposed to predispose
mitochondrial activity and many of them have muscle, liver and adipose tissue to accumulate
been associated with cell senescence and ageing as lipids, especialy long-chain fatty acids (Hafizi Abu
has been recently reviewed (Ziegler et al. 2015). Bakar et al. 2015), and then, to aggravate insulin
The identification of these factors will be essential resistance (Lowell and Shulman 2005). Recently,
to understand how their deregulation affects the it has been proposed that lowering serum FFAs
progression of ageing and many pathological levels and improving mitochondrial function may
situations. Among them, PGC-1b emerges as one improve insulin response in youth with T2D.
interesting candidate by controling basal Obesity is also associated with the metabolic
mitochondrial biogenesis and mitochondrial fusion impairment in the capacity to transit between fatty
through the modulation of Mfn2 (Liesa et al. acid and glucose metabolism, known as
2008). For example, in a heart-selective PGC- ‘‘metabolic inflexibility’’ (Griffin et al. 2015), that
1bdeficient murine model, age-dependent cardiac is present in obese people since they oxidize lipids
phenotypes have been associated with the increase instead of glucose in insulin-stimulated conditions
of mitochondrial dysfunction (Valli et al. 2017) (Storlien et al. 2004). So, the accumulation of
(Fig. 2). damaged mitochondria would explain the increase
of ROS in skeletal muscle associated with insulin
resistance in obese individuals and the appearance
Mitochondrial dysfunction is associated of T2DM and muscle dysfunction during normal
with metabolic syndrome ageing (Barbieri et al. 2013).
Mitochondrial dynamics are also affected in
One of the characteristics of ageing is the obesity-related diseases. Reduced expression of
accumulation of visceral adiposity in elderly Mfn2 has been found in skeletal muscle in obese
people (Matsumoto 2002). Accumulation of and T2DM patients (Bach et al. 2005) and has
visceral fat is associated with greater morbidity been associated with the fragmentation of the
due to the increase of risk for cardiovascular mitochondrial network and the decrease of
disease, hypertension and T2DM. Obesity and membrane potential in mitochondria (Pich et al.
T2DM are also considered agerelated diseases. 2005). Mitochondrial fragmentation and down
Obesity is associated with the dysfunction of regulation of fusion have been also detected in
adipose tissue characterized by adipocyte T2DM and obese subjects associated with
hypertrophy, proinflammatory conditions, and impaired bioenergetics (Bach et al. 2005). The
insulin resistance. Additionally, the incapacity to reduction of fusion together with the increase in
respond to insulin induces ectopic fat deposition in fission indicates that there is a shift toward fission
other organs producing lipotoxicity in muscle, of mitochondria in obesity and obesity-related
liver and also pancreas, a phenomenon that has diseases. Decrease of Mfn2 plays an essential role

123
Biogerontology
in mitochondrial dysfunction associated with fat of fission in the accumulation of damaged
accumulation. A screen of differentially expressed mitochondria (Wang et al. 2015).
genes from skeletal muscle of obese rats has Evidence suggests that Sestrins could be
identified Mfn2 as a suppressor of obesity (Bach et considered key factors in the control of
al. 2003). Mfn2 KO cells show lower mitochondrial homeostasis during ageing and
bioenergetics efficiency indicated by lower obesity. Inactivation of Sestrins in invertebrates
glucose oxidation and mitochondrial respiration results in metabolic pathologies that include
(Bach et al. 2003) that can be associated with the oxidative damage, mitochondrial dysfunction and
progression of obesity by reducing energy fat accumulation (Lee et al. 2013). However, to
expenditure and increasing fat store (Liesa et al. date, there is no information available about the
2009). On the contrary, conditions that increase activity of Sestrins in adipose tissue and its
energy expenditure such as cold exposure, chronic behavior during ageing. Indirect evidences
exercise or nutraceuticals as resveratrol induce indicate a putative role of this family of proteins in
Mfn2 expression in skeletal muscle and brown fat accumulation. Resveratrol (RSV) prevents lipid
adipose tissue (Cartoni et al. 2005; Dolinsky et al. accumulation in liver avoiding Liver X receptor
2013; Soriano et al. 2006). Interestingly, all these (LXRa)-mediated hepatic lipogenesis by inducing
conditions are known to induce SIRT1 that has Sens2 gene (Jin et al. 2013). LXRa is a
been recently shown to suppress mitochondrial transcription factor that regulates lipid synthesis in
dysfunction in a model of ischemia by modulating liver (Joseph et al. 2002) and insulinstimulated
Mfn2 activity by deacetylation indicating a lipogenesis (Chen et al. 2004). LXRa induces the
connection between SIRT1dependent expression of fatty acid synthesis (FAS), acetyl-
mitochondrial regulation and mitochondrial fusion CoA carboxylase (ACC) and sterol regulatory
(Biel et al. 2016). Further, Mfn2 also participates element binding protein 1c (SREBP-1c) (Grefhorst
in the maintenance of CoQ10 synthesis and could et al. 2002; Repa et al. 2000). Then, the
be also important in the regulation of CoQ 10 levels Sens2dependent repression of LXRa in liver or
during ageing and obesity (Mourier et al. 2015). adipose tissue by RSV would indicate a key role of
However, to date no data are available about its these proteins in fat accumulation during ageing.
relationship in obesity or ageing indicating the On the other hand, in old mice, Sens2 is induced in
need of further research. muscle by acute aerobic exercise accompanied by
On the other hand, higher mitochondrial fission an increase in autophagy and probably
has been associated with fat accumulation in contributing to the improvement of insulin
genetically induced obesity (ob/ob) mice or in sensitivity (Lenhare et al. 2017).
high-fat induced obesity (Jheng et al. 2012) and Among the regulators of metabolism, AMPK
with insulin resistance in another mice model of plays an important role in the progression of MS
obesity (db/db) (Holmstrom et al. 2012). These and insulin resistance in white adipose tissue
studies indicate that obesity is associated with the (WAT). Phosphorylated AMPK controls the
accumulation of small and inefficient activity of many downstream substrates involved
mitochondria that are unable to metabolize lipids in the increase in lipid oxidation and the uptake of
and are, at least in part, responsible of the glucose (Jager et al. 2007). Thus, deficiency in
dysregulation of insulin signaling in cells. In a AMPK results in glucose intolerance, lower
vicious circle, insulin resistance can increase physical capacity and obesity (Steinberg et al.
mitochondrial dysfunction and aggravate obesity 2010). Taken into consideration the central role of
as has been recently demonstrated in a double KO AMPK in the control of metabolism and
mice model for insulin and insulin-growth factor 1 mitochondria (Burkewitz et al. 2014), the decrease
receptors (Viana-Huete et al. 2018). On the other of its levels and activity during ageing could
hand, disruption of mitochondrial fission in liver explain the deterioration of the physiology of
can protect mice from diet induced obesity and adipose and other tissues during aging in the
metabolic deterioration indicating the importance elderly (Ruiz et al. 2016). Regarding this aspect, a

123
Biogerontology
new regulator of AMPK, the prolyl isomerase, Schubeler 2015). One of the most crucial factors
Pin1, has been found to increase in high fat that modify DNA methylation patterns is the
conditions (Nakatsu et al. 2011). Pin1 KO mice environment (Feil and Fraga 2012; Leenen et al.
show a high fat diet-induced obesity resistant 2016), although the mechanisms behind this
phenotype (Nakatsu et al. 2015). Interestingly, influence must be clarified. DNA methylation
deregulation of Pin1 has been also associated with changes are associated with pollutants, UV
ageing and age-related diseases (Lee et al. 2011) radiations and temperature, viral infections and
and downregulation of its levels are associated microbiota, mood disorders, lifestyle, overeating,
with senescence in human tendon stem cells (Chen high-fat diet, famine, caloric restriction, physical
et al. 2015) although more research is needed to activity and circadian rhythms (Feil and Fraga
associate reduction of activity of Pin1 with ageing 2012; Grazioli et al. 2017; Leenen et al. 2016).
and age-related diseases. Comparative analyses carried out in twin pairs
All these studies suggest that mitochondrial demonstrated that the methylation status gradually
dysfunction is the common denominator in ageing diverges with age as a consequence of different
and fat accumulation. Metabolic dysfunction environments and lifestyles (Fraga et al. 2005).
associated with the accumulation of damaged Recently it has been proposed the ‘‘Epigenetic
mitochondria is one of the main factors in the Theory of Ageing’’, which is the result of two
evolution of T2DM, fat accumulation in muscle, opposing phenomena: (1) baseline DNA
WAT or liver and the increase of oxidative stress. methylation levels progressively diverge during
The study of factors involved in the improvement ageing (Fraga et al. 2005), and (2) some locus-
of mitochondrial dynamics in order to increase specific DNA methylation changes are highly
mitochondrial turnover and to improve reproducible across aged people, independently of
mitochondrial architecture and function is gender and tissue type (Horvath 2013). While
essential to develop therapeutic strategies in order environmental factors are responsible of such
to improve fat accumulation-related diseases. DNA methylation pattern divergence (Feil and
Fraga 2012; Leenen et al. 2016), it seems that in
later stages of life the whole process paradoxically
Metabolic activity can be regulated by epigenetic culminates in a convergence of epigenomes,
mechanisms leading to a programmed epigenetic
reconfiguration during ageing that had been
Gene expression is regulated by modifications in postulated as the ‘‘epigenetic clock’’ theory
DNA and histones. The addition of a methyl group (Horvath 2013).
to cytosine results in the formation of 5- Therefore, the environment and the signals we
methylcytosine (5-mC), which is one of the receive during development determine the
heritable epigenetic marks. DNA methylation has epigenetic marks which will make us more prone
been proposed as the most promising molecular to suffer certain diseases and to determine the
marker for monitoring ageing and predict life process by which we will age. Many of these
expectancy (Horvath 2013). However, the signals are related with metabolism. Dietary habits
mechanisms underlying age-related DNA during pregnancy strongly determine the genome
methylation changes are still unknown. methylation status of offspring (Aagaard-Tillery et
The initial establishment of DNA methylation al. 2008). The offspring of a mother exposed to
patterns takes place during development just after famine had an increased incidence of several
the massive demethylation that takes part on the health problems associated with metabolism such
preimplantation embryo genome re-establish cell as obesity, hypertension, diabetes and
totipotency (Feng et al. 2010). DNA methylation cardiovascular diseases, whereas children from
patterns are maintained or change along life by mothers with depression seems to be predisposed
methylation and demethylation processes to higher risk of chronic diseases during adulthood
depending on different factors (Clark 2015; (Ciccarone et al. 2017). On the other hand, some

123
Biogerontology
environmental or behavioral factors induce of mitochondrial activity in stem cell maintenance,
beneficial epigenetic outcomes via changes of immune response, metabolic adaptations to
DNA methylation patterns. Long-term physical physiological changes and ageing (Lin and Haynes
activity changes the methylation of several genes 2016).
involved in metabolism, muscle growth,
haematopoiesis and inflammation, reducing cancer
risk and mortality (Ciccarone et al. 2017; Grazioli Mitochondria and metabolism are regulated by
et al. 2017; Lapinska et al. 2018). prolongevity factors
A number of genes are hypermethylated with
age and with other age-related diseases indicating Much evidence indicates that the maintenance of
their relationship with gene regulation during mitochondrial activity is crucial to delay ageing
ageing (Lapinska et al. 2018; Zampieri et al. process. In fact, the decline in mitochondrial
2015). Many of these genes are involved in turnover has been associated with the impairment
metabolism regulation related with cell survival as of mitochondrial biogenesis during ageing,
has been demonstrated in mitochondrial diseases especially in high energy demanding tissues such
independently of the etiology and the organism. as muscle, brain or heart (Conley et al. 2000; Short
Interestingly, this response is the same in human et al. 2005). Several interventions such as CR,
fibroblasts (Fernandez-Ayala et al. 2013) and in bioactive compounds such as polyphenols or
the fruit fly Drosophila melanogaster (Fernandez- physical activity increase mitochondrial turnover
Ayala et al. 2010) indicating the importance of inducing the synthesis of new mitochondrial
metabolism regulation during ageing. structures while eliminate damaged elements by
Mitochondria has been associated with the autophagy highlighting the important role of the
epigenetic regulation during development mainly maintenance of the mitochondrial physiology
affecting protective genes such as mitochondrial during ageing (Baur et al. 2010; de Cabo et al.
chaperones, control proteases and xenobiotic 2014; Feige et al. 2008; Rodriguez-Bies et al.
response components that comprise the 2015). Further, one recent review emphasized the
mitochondrial unfolded protein response (UPR mt) role of autophagy in the prolongevity effect of CR
(Tatar and Sedivy 2016). Mild mitochondrial or bioactive compounds such resveratrol,
stress has been associated with changes in the spermidine and rapamycin (Das et al. 2017). This
epigenome affecting chromatin reorganization effect has been associated with the inhibition of
involved in the maintenance of UPRmt and mTOR and activation of Sirtuins (Szafranski and
increasing longevity in C. elegans (Tian et al. Mekhail 2014). Further, the inductor of
2016). Epigenetic modifications involved mito/autophagy FOXO3 is also induced in CR
methylation of histones (H3K9me1/2) have been conditions regulating different mechanisms
associated with the regulation of energy involved in cell survival (Willcox and Willcox
expenditure and fat storage in these animals 2014). Studies performed in non-obese humans
(Meister et al. 2011) indicating a communication under 25% CR has highlighted the importance of
between mitochondrial activity and energy the regulation of metabolism as indicated by
regulation and the control of the expression of weight loss, reduction of resting metabolic rate
genes involved in longevity. Therefore, a and many other markers (Ravussin et al. 2015).
reduction in the activity of the UPR mt can be The benefits from CR in ageing and associated
responsible of the accumulation of damaged comorbidities has been directly associated to
proteins in dysfunctional mitochondria in ageing changes in metabolism during fasting periods
and in age-related neurodegenerative diseases (Mattson et al. 2018). Intermittent metabolic
(Moehle et al. 2018). However, it seems that switching is defined as fasting or exercise periods
UPRmt activation alone is not sufficient to prolong when the transition from glucose to fatty acids and
life-span (Bennett et al. 2014; Rauthan et al. ketone bodies. Mattson et al. 2018 propose that
2013), although it is essential in the maintenance switching between negative energy balance

123
Biogerontology
(exercise or fasting) and positive balance (eating intervention by modulating bioenergetics and
and resting) could improve health parameters such maintaining mitochondrial activity.
as insulin sensitivity, abdominal fat, resting heart In general, all the prolongevity interventions,
rate, blood pressure and muscle mass. In this CR, bioactive compounds, or exercise, induce
process mitochondrial physiology plays an modifications in mitochondrial physiology
important role since intermittent metabolic activating mitochondrial biogenesis at the same
switching incite mitochondrial biogenesis in time that induce the removal of mitochondrial
neurons through the activation of PGC1a. During structures through mito/ autophagy. These models
metabolic switching in fasting or vigorous indicate that activation of mitochondrial turnover
exercises states, mTOR activity tends to be is key in longevity.
reduced while AMPK is inducing autophagy.
Further, very recently, a low-carbohydrate,
ketogenic diet extended longevity in adult male Circadian rhythm and mitochondrial activity
mice preserving motor function, muscle mass and
memory in aged animals in an effect associated Circadian rhythm is affected during aging and
with the decrease of mTOR downstream signaling metabolic disease (Borengasser et al. 2014;
(Roberts et al. 2018). Weinert 2010). During aging, a reduction in the
CR can be mimicked by different bioactive amplitude of circadian body temperature rhythms
compounds. In a yeast model rapamycin occurs (Weinert 2010). On the other hand,
significantly increased lifespan by 87% and offspring fed with high fat diet from obese dams
resveratrol and dapsone showed a tendency to showed suppression in circadian and metabolic
increase lifespan (Choi et al. 2013). Resveratrol genes in comparison with offspring from lean
also showed capacity to increase lifespan of dams (Borengasser et al. 2014). These
middle-aged mice on a high calorie diet, perturbations of the circadian rhythm increase
improving insulin sensitivity (Baur et al. 2006). In oxidative damage to proteins and lipids, decrease
this model, resveratrol increased the activity of of the capacity
AMPK and PGC-1a, mitochondrial number and forfreeradicalscavengingandimpairedmitochondria
motor function. This same effect was found in l function (Jagota and Kalyani 2010; Weinert
muscle by activating SIRT1 and PGC-1a (Lagouge 2010). In this regulation administration of
et al. 2006). melatonin, an hormone known to control the
It is also clear that physical activity prevents circadian rhythm, affected diary rhythms of lipid
muscle changes involved in frailty in elderly peroxidation and antioxidant enzymes in male rat
people such as sarcopenia, although these liver indicating the link with mitochondrial
alterations can be also associated to chronic disuse activity (Manikonda and Jagota 2012).
instead to muscle ageing. It is clear that Biological rhythms are regulated by many
maintenance of muscle mass reduces the risk of nuclear transcription factors. One of these factors
fall, functional decline, feeling of weakness and is the circadian locomotor output cycles kaput
dependence (Garatachea et al. 2015; McKendry et (CLOCK)delta19. Dysruption of this gene in mice
al. 2018; Wroblewski et al. 2011). At the causes the reduction of PGC1a and decrease in the
molecular level, many research has demonstrated amount of the mitochondrial transcription factor-A
that exercise affects mitochondrial biogenesis accompanied by the decrease of mitochondria
(Vina et al. 2009), mitochondrial fragmentation (Pastore and Hood 2013). This mutation also
and autophagy in a mechanism dependent on affects mitochondrial gene expression in young
PGC-1a activity (Halling and Pilegaard 2017; mice livers (Gong et al. 2015). Further, changes in
Halling et al. 2017), and ETC activity and mitochondrial morphology, fusion and fission
mitochondrial content (Menshikova et al. 2006) processes as well as generation of new
among others. These evidence clearly indicate the mitochondria are dependent of circadian clock (de
potential of physical activity as prolongevity Goede et al. 2018). In cells, master clock gene

123
Biogerontology
Aryl Hydrocarbon Receptor Nuclear Translocator Concluding remarks
Like/ Brain and Muscle ARNT-Like 1
(ARNTL/BMA1) silencing affects the rhythm of Much evidence indicate that maintenance of
mitochondrial phosphorylation during the day bioenergetics homeostasis associated with
whereas mitochondrial dysfunction deregulates the mitochondrial activity is key to delay ageing and
rhythmic clock-gene expression in reciprocal prevent ageassociated diseases (Riera et al. 2016).
interplay (Scrima et al. 2016). On the other hand, Regulation of nutrient sensors such as mTOR,
mitochondrial alterations with 3-nitropropionic AMPK and Sirtuins are in the center of this effect.
acid induce severe circadian deficit in behavior These sensors are key in the regulation of the
(Kudo et al. 2014). All these studies indicate an complex equilibrium between mitochondrial
important relationship between mitochondrial biogenesis, dynamics and turnover. During ageing
activity and circadian rhythm in cells and tissues. a tendency to increase fusion in mitochondria has
In the relationship between circadian rhythm been reported in order to maintain mitochondrial
and mitochondria, sirtuins and AMPK play an activity, however, fat accumulation induces a
important role. A circadian clock of NAD ? higher activity of fission and a block in mitophagy
biosynthesis through modifications of promoting the accumulation of small and damaged
mitochondrial protein acetylation that synchronize mitochondria.
mitochondrial activity with fasting and feeding Probably these two opposite processes end in
cycle has been recently proposed (Peek et al. the accumulation of damaged and ineffective
2013). Levels of NAD? can affect the activity of mitochondria that increase oxidative damage and
complex I by deacetylation (Cela et al. 2016). are related with insulin resistance, metabolic
Interestingly, sirtuins have been associated with dysfunction and the secretion of proinflammatory
the control of the circadian rhythms through their mediators. In a vicious cycle, insulin resistance
activity in the central nervous system (Ramadori and proinflammatory cytokines promotes damage
and Coppari 2010). Neurons mainly located in of mitochondria. Then, the study of the regulation
hypothalamus coordinate long-term energy and of metabolic mediators and the effects of
glucose balance (Elmquist et al. 2005), by sensing- prolongevity factors such as CR, bioactive
mechanisms able to respond to hormonal and compounds or exercise will help us to develop
nutrient levels. Since SIRT1 is known to respond therapeutic strategies to delay ageing and the
to these variations in peripheral tissue, it has been damaging effect of fat accumulation.
proposed that this deacetylase can affect nutrient
response in neurons. In fact, SIRT1 activity in Funding The research group is funded by the Andalusian
Government Grant BIO177 (FEDER funds of European
brain is influenced by changes in nutrient Commission). Research has been funded by the Spanish
availability and affected by obesity (Cohen et al. Ministry of Economy and Competitiveness Grant
2004; Ramadori et al. 2008). Further, AMPK also DEP201239985 and Instituto de Salud Carlos III FIS Grant
PI14/01962. Authors are also members of the CIBERER,
mediates the effects of agents acting on the
Instituto Carlos III, of the Spanish Ministry of Health.
hypothalamus promoting feeding and circadian
rhythms of metabolism (Hardie et al. 2012). It is
then clear that the mechanisms that maintain References
mitochondrial dynamics and turnover are involved
in the regulation of circadian rhythms and that Aagaard-Tillery KM, Grove K, Bishop J, Ke X, Fu Q,
disruption of these mechanisms can be involved in McKnight R, Lane RH (2008) Developmental origins
of disease and determinants of chromatin structure:
the dysfunction of these rhythms during aging and
maternal diet modifies the primate fetal epigenome. J
metabolic disease. At the same time, in a vicious Mol Endocrinol
cycle, dysregulation of circadian rhythms affect 41(2):91–102. https://doi.org/10.1677/JME-08-0025
mitochondrial activity promoting its malfunction. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman
JI, Donato KA, Fruchart JC, James WP, Loria CM,
Smith SC Jr, International Diabetes Federation Task
Force on E, Prevention, Hational Heart L, Blood I,

123
Biogerontology
American Heart A, World Heart F, International Gwinn D, Wang M, Ramaswamy S, Fishbein KW,
Atherosclerosis S, International Association for the Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ,
Study of O (2009) Harmonizing the metabolic Navas P, Puigserver P, Ingram DK, de Cabo R,
syndrome: a joint interim statement of the Sinclair DA (2006) Resveratrol improves health and
International Diabetes Federation Task Force on survival of mice on a high-calorie diet. Nature
Epidemiology and Prevention; National Heart, Lung, 444(7117):337–342.
and Blood Institute; American Heart Association; https://doi.org/10.1038/nature05354
World Heart Federation; International Atherosclerosis BaurJA,ChenD,ChiniEN,ChuaK,CohenHY,deCaboR,
Society; and International Association for the Study of Deng C, Dimmeler S, Gius D, Guarente LP, Helfand
Obesity. Circulation 120(16):1640–1645. SL, Imai S, Itoh H, Kadowaki T, Koya D,
https://doi.org/10.1161/ Leeuwenburgh C, McBurney M, Nabeshima Y, Neri
circulationaha.109.192644 C, Oberdoerffer P, Pestell RG, Rogina B, Sadoshima
Alers S, Loffler AS, Wesselborg S, Stork B (2012) Role of J, Sartorelli V, Serrano M, Sinclair DA, Steegborn C,
AMPK-mTOR-Ulk1/2 in the regulation of autophagy: Tatar M, Tissenbaum HA, Tong Q,
cross talk, shortcuts, and feedbacks. Mol Cell Biol Tsubota K, Vaquero A, Verdin E (2010) Dietary
32(1):2–11. https://doi.org/10.1128/MCB.06159-11 restriction: standing up for sirtuins. Science
Artal-Sanz M, Tavernarakis N (2009) Prohibitin couples 329(5995):1012–1013.
diapause signalling to mitochondrial metabolism https://doi.org/10.1126/science.329.5995.1012
during ageing in C. elegans. Nature 461(7265):793– Bennett CF, Vander Wende H, Simko M, Klum S, Barfield
797. https://doi. org/10.1038/nature08466 S, Choi H, Pineda VV, Kaeberlein M (2014)
Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Activation of the mitochondrial unfolded protein
Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath response does not predict longevity in Caenorhabditis
JR, RabasaLhoret R, Wallberg-Henriksson H, Laville elegans. Nat Commun 5:3483.
M, Palacin M, Vidal H, Rivera F, Brand M, Zorzano https://doi.org/10.1038/ncomms4483
A (2003) Mitofusin-2 determines mitochondrial Beregi E, Regius O (1987) Comparative morphological
network architecture and mitochondrial metabolism. A study of age related mitochondrial changes of the
novel regulatory mechanism altered in obesity. J Biol lymphocytes and skeletal muscle cells. Acta Morphol
Chem 278(19):17190–17197. Hung 35(3–4): 219–224
https://doi.org/10.1074/jbc.M212754200 Beyer RE, Burnett BA, Cartwright KJ, Edington DW,
Bach D, Naon D, Pich S, Soriano FX, Vega N, Rieusset J, Falzon MJ, Kreitman KR, Kuhn TW, Ramp BJ, Rhee
Laville M, Guillet C, Boirie Y, Wallberg-Henriksson SY, Rosenwasser MJ et al (1985) Tissue coenzyme Q
H, Manco M, Calvani M, Castagneto M, Palacin M, (ubiquinone) and protein concentrations over the life
Mingrone span of the laboratory rat. Mech Ageing Dev 32(2–
G, Zierath JR, Vidal H, Zorzano A (2005) Expression 3):267–281
of Mfn2, the Charcot-Marie-Tooth neuropathy type Biel TG, Lee S, Flores-Toro JA, Dean JW, Go KL, Lee
2A gene, in human skeletal muscle: effects of type 2 MH, Law BK, Law ME, Dunn WA Jr, Zendejas I,
diabetes, obesity, weight loss, and the regulatory role Behrns KE, Kim JS (2016) Sirtuin 1 suppresses
of tumor necrosis factor alpha and interleukin-6. mitochondrial dysfunction of ischemic mouse livers in
Diabetes 54(9):2685–2693 a mitofusin 2-dependent manner. Cell Death Differ
Bae SH, Sung SH, Oh SY, Lim JM, Lee SK, Park YN, Lee 23(2):279–290. https://
HE, Kang D, Rhee SG (2013) Sestrins activate Nrf2 doi.org/10.1038/cdd.2015.96
by promoting p62-dependent autophagic degradation Bluher M, Kahn BB, Kahn CR (2003) Extended longevity
of Keap1 and prevent oxidative liver damage. Cell in mice lacking the insulin receptor in adipose tissue.
Metab Science 299(5606):572–574. https://doi.org/10.1126/
17(1):73–84. science.1078223
https://doi.org/10.1016/j.cmet.2012.12.002 Boengler K, Kosiol M, Mayr M, Schulz R, Rohrbach S
Barbieri E, Sestili P, Vallorani L, Guescini M, Calcabrini (2017) Mitochondria and ageing: role in heart, skeletal
C, Gioacchini AM, Annibalini G, Lucertini F, Piccoli muscle and adipose tissue. J Cachexia Sarcopenia
G, Stocchi V (2013) Mitohormesis in muscle cells: a Muscle
morphological, molecular, and proteomic approach. 8(3):349–369. https://doi.org/10.1002/jcsm.12178
Muscles Ligaments Tendons J 3(4):254–266 Borengasser SJ, Kang P, Faske J, Gomez-Acevedo H,
Battino M, Gorini A, Villa RF, Genova ML, Bovina C, Blackburn ML, Badger TM, Shankar K (2014) High
Sassi S, Littarru GP, Lenaz G (1995) Coenzyme Q fat diet and in utero exposure to maternal obesity
content in synaptic and non-synaptic mitochondria disrupts circadian rhythm and leads to metabolic
from different brain regions in the ageing rat. Mech programming of liver in rat offspring. PLoS ONE
Ageing Dev 9(1):e84209. https://doi.org/10.
78(3):173–187 1371/journal.pone.0084209
Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Bratic I, Trifunovic A (2010) Mitochondrial energy
Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, metabolism and ageing. Biochim Biophys Acta
Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O,

123
Biogerontology
1797(6–7):961–967. Ciccarone F, Tagliatesta S, Caiafa P, Zampieri M (2017)
https://doi.org/10.1016/j.bbabio.2010.01.004 DNA methylation dynamics in aging: how far are we
Budanov AV, Karin M (2008) p53 target genes sestrin1 and from understanding the mechanisms? Mech Ageing
sestrin2 connect genotoxic stress and mTOR Dev. https://
signaling. Cell 134(3):451–460. doi.org/10.1016/j.mad.2017.12.002
https://doi.org/10.1016/j.cell.2008. 06.028 Clark AT (2015) DNA methylation remodeling in vitro and
Burkewitz K, Zhang Y, Mair WB (2014) AMPK at the in vivo. Curr Opin Genet Dev 34:82–87.
nexus of energetics and aging. Cell Metab 20(1):10– https://doi.org/10. 1016/j.gde.2015.09.002
25. https://doi. org/10.1016/j.cmet.2014.03.002 CohenHY,MillerC,BittermanKJ,WallNR,HekkingB,Kessler
Canto C, Jiang LQ, Deshmukh AS, Mataki C, Coste A, B, Howitz KT, Gorospe M, de Cabo R, Sinclair DA
Lagouge M, Zierath JR, Auwerx J (2010) (2004) Calorie restriction promotes mammalian cell
Interdependence of AMPK and SIRT1 for metabolic survival by inducing the SIRT1 deacetylase. Science
adaptation to fasting and exercise in skeletal muscle. 305(5682): 390–392.
Cell Metab 11(3):213–219. https://doi. https://doi.org/10.1126/science.1099196
org/10.1016/j.cmet.2010.02.006 Conley KE, Jubrias SA, Esselman PC (2000) Oxidative
Cartoni R, Leger B, Hock MB, Praz M, Crettenand A, Pich capacity and ageing in human muscle. J Physiol
S, Ziltener JL, Luthi F, Deriaz O, Zorzano A, Gobelet 526(Pt 1):203–210
C, Kralli A, Russell AP (2005) Mitofusins 1/2 and Cowey S, Hardy RW (2006) The metabolic syndrome: a
ERRalpha expression are increased in human skeletal highrisk state for cancer? Am J Pathol 169(5):1505–
muscle after physical exercise. J Physiol 567(Pt 1522. https://doi.org/10.2353/ajpath.2006.051090
1):349–358. https://doi. Das SK, Balasubramanian P, Weerasekara YK (2017)
org/10.1113/jphysiol.2005.092031 Nutrition modulation of human aging: the calorie
Cela O, Scrima R, Pazienza V, Merla G, Benegiamo G, restriction paradigm. Mol Cell Endocrinol 455:148–
Augello B, Fugetto S, Menga M, Rubino R, Fuhr L, 157. https://doi.org/
Relogio A, Piccoli C, Mazzoccoli G, Capitanio N 10.1016/j.mce.2017.04.011 de Brito OM, Scorrano L
(2016) Clock genesdependent acetylation of complex I (2008) Mitofusin 2 tethers endoplasmic reticulum to
sets rhythmic activity of mitochondrial OxPhos. mitochondria. Nature 456(7222):
Biochim Biophys Acta 605–610. https://doi.org/10.1038/nature07534 de Brito
4:596–606. OM, Scorrano L (2009) Mitofusin-2 regulates
https://doi.org/10.1016/j.bbamcr.2015.12.018 mitochondrial and endoplasmic reticulum morphology and
Chan EY (2009) mTORC1 phosphorylates the ULK1- tethering: the role of Ras. Mitochondrion 9(3):222–226.
mAtg13FIP200 autophagy regulatory complex. Sci https://doi.org/10.1016/j.mito.2009.02.005
Signal de Cabo R, Carmona-Gutierrez D, Bernier M, Hall MN,
2(84):pe51. https://doi.org/10.1126/scisignal.284pe51 Madeo F (2014) The search for antiaging
Chen H, Chan DC (2005) Emerging functions of interventions: from elixirs to fasting regimens. Cell
mammalian mitochondrial fusion and fission. Hum 157(7):1515–1526. https://
Mol Genet doi.org/10.1016/j.cell.2014.05.031
14(2):R283–289. https://doi.org/10.1093/hmg/ddi270 de Goede P, Wefers J, Brombacher EC, Schrauwen P,
Chen G, Liang G, Ou J, Goldstein JL, Brown MS (2004) Kalsbeek A (2018) Circadian rhythms in
Central role for liver X receptor in insulin-mediated mitochondrial respiration. J Mol Endocrinol
activation of Srebp-1c transcription and stimulation of 60(3):R115–R130. https://doi.org/10. 1530/JME-17-
fatty acid synthesis in liver. Proc Natl Acad Sci USA 0196
101(31): Del Pozo-Cruz J, Rodriguez-Bies E, Navas-Enamorado I,
11245–11250. Del Pozo-Cruz B, Navas P, Lopez-Lluch G (2014)
https://doi.org/10.1073/pnas.0404297101 Relationship between functional capacity and body
Chen L, Liu J, Tao X, Wang G, Wang Q, Liu X (2015) The mass index with plasma coenzyme Q10 and oxidative
role of Pin1 protein in aging of human tendon damage in community-dwelling elderly-people. Exp
stem/progenitor cells. Biochem Biophys Res Commun Gerontol 52:46–54.
464(2):487–492. https://doi.org/10.1016/j.exger.2014.01.026
https://doi.org/10.1016/j.bbrc.2015.06.163 Dencher NA, Frenzel M, Reifschneider NH, Sugawa M,
Chistiakov DA, Sobenin IA, Revin VV, Orekhov AN, Krause F (2007) Proteome alterations in rat
Bobryshev YV (2014) Mitochondrial aging and age- mitochondria caused by aging. Ann N Y Acad Sci
related dysfunction of mitochondria. Biomed Res Int 1100:291–298. https://doi.
2014:238463. https://doi.org/10.1155/2014/238463 org/10.1196/annals.1395.030
Choi KM, Lee HL, Kwon YY, Kang MS, Lee SK, Lee CK Denzer I, Munch G, Friedland K (2016) Modulation of
(2013) Enhancement of mitochondrial function mitochondrial dysfunction in neurodegenerative
correlates with the extension of lifespan by caloric diseases via activation of nuclear factor erythroid-2-
restriction and caloric restriction mimetics in yeast. related factor 2 by food-derived compounds.
Biochem Biophys Res Commun 441(1):236–242. Pharmacol Res 103:80–94.
https://doi.org/10.1016/j.bbrc. 2013.10.049 https://doi.org/10.1016/j.phrs.2015.11.019

123
Biogerontology
Dolinsky VW, Rogan KJ, Sung MM, Zordoky BN, monozygotic twins. Proc Natl Acad Sci USA
Haykowsky MJ, Young ME, Jones LW, Dyck JR 102(30):10604–10609. https://doi.org/10.1073/
(2013) Both aerobic exercise and resveratrol pnas.0500398102
supplementation attenuate doxorubicin-induced Garatachea N, Pareja-Galeano H, Sanchis-Gomar F,
cardiac injury in mice. Am J Physiol Endocrinol SantosLozano A, Fiuza-Luces C, Moran M, Emanuele
Metab 305(2):E243–253. https://doi.org/10. E, Joyner MJ, Lucia A (2015) Exercise attenuates the
1152/ajpendo.00044.2013 major hallmarks of aging. Rejuvenation Res 18(1):57–
Elmquist JK, Coppari R, Balthasar N, Ichinose M, Lowell 89. https://doi. org/10.1089/rej.2014.1623
BB (2005) Identifying hypothalamic pathways Genova ML, Lenaz G (2015) The interplay between
controlling food intake, body weight, and glucose respiratory supercomplexes and ROS in aging.
homeostasis. J Comp Neurol 493(1):63–71. Antioxid Redox Signal 23:208–238.
https://doi.org/10.1002/cne.20786 https://doi.org/10.1089/ars.2014.6214
Fabbri E, Chia CW, Spencer RG, Fishbein KW, Reiter DA, Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH,
Cameron D, Zane AC, Moore ZA, Gonzalez-Freire M, Mostoslavsky R, Alt FW, Wu Z, Puigserver P (2007)
Zoli M, Studenski SA, Kalyani RR, Egan JM, Ferrucci Metabolic control of muscle mitochondrial function
L (2017) Insulin resistance is associated with reduced and fatty acid oxidation through SIRT1/PGC-1alpha.
mitochondrial oxidative capacity measured by 31P- EMBO J 26(7): 1913–1923.
magnetic resonance spectroscopy in participants https://doi.org/10.1038/sj.emboj.7601633
without diabetes from the baltimore longitudinal study Gomes LC, Scorrano L (2008) High levels of Fis1, a pro-
of aging. Diabetes fission mitochondrial protein, trigger autophagy.
66(1):170–176. https://doi.org/10.2337/db16-0754 Biochim Biophys Acta 1777(7–8):860–866.
Fang EF, Waltz TB, Kassahun H, Lu Q, Kerr JS, Morevati https://doi.org/10.1016/j. bbabio.2008.05.442
M, Fivenson EM, Wollman BN, Marosi K, Iser WB, Gomes LC, Scorrano L (2011) Mitochondrial elongation
Eckley DM, Zhang Y, Lehrmann E, Goldberg IG, during autophagy: a stereotypical response to survive
Scheibye-Knudsen M, Mattson MP, Nilsen H, Bohr in difficult times. Autophagy 7(10):1251–1253.
VA, Becker KG (2017) Tomatidine enhances lifespan https://doi.org/10. 4161/auto.7.10.16771
and healthspan in C. elegans through mitophagy Gomez LA, Hagen TM (2012) Age-related decline in
induction via the SKN-1/Nrf2 pathway. Sci Rep mitochondrial bioenergetics: does supercomplex
7:46208. https://doi.org/10.1038/srep46208 destabilization determine lower oxidative capacity and
Feige JN, Lagouge M, Auwerx J (2008) Dietary higher superoxide production? Semin Cell Dev Biol
manipulation of mouse metabolism. Curr Protoc Mol 23(7):758–767.
Biol 84(1):29B-5. https://doi.org/10.1016/j.semcdb.2012.04. 002
https://doi.org/10.1002/0471142727.mb29b05s84 Goncalves RL, Quinlan CL, Perevoshchikova IV, Hey-
Feil R, Fraga MF (2012) Epigenetics and the environment: Mogensen M, Brand MD (2015) Sites of superoxide
emerging patterns and implications. Nat Rev Genet and hydrogen peroxide production by muscle
13(2):97–109. https://doi.org/10.1038/nrg3142 mitochondria assessed ex vivo under conditions
Feng S, Jacobsen SE, Reik W (2010) Epigenetic mimicking rest and exercise. J Biol Chem 290(1):209–
reprogramming in plant and animal development. 227. https://doi.org/10. 1074/jbc.M114.619072
Science 330(6004): 622–627. Gong C, Li C, Qi X, Song Z, Wu J, Hughes ME, Li X
https://doi.org/10.1126/science.1190614 (2015) The daily rhythms of mitochondrial gene
Fernandez-Ayala DJ, Chen S, Kemppainen E, O’Dell KM, expression and oxidative stress regulation are altered
Jacobs HT (2010) Gene expression in a Drosophila by aging in the mouse liver. Chronobiol Int
model 32(9):1254–1263. https://doi.
of mitochondrial disease. PLoS ONE 5(1):e8549. org/10.3109/07420528.2015.1085388
https:// doi.org/10.1371/journal.pone.0008549 Gonzalez-Freire M, Cabo Rd, Bernier M, Sollott SJ, Fabbri
Fernandez-Ayala DJ, Guerra I, Jimenez-Gancedo S, E, Navas P, Ferrucci L (2015) Reconsidering the role
Cascajo MV, Gavilan A, Dimauro S, Hirano M, of mitochondria in aging. J Gerontol Ser A.
Briones P, Artuch R, De Cabo R, Salviati L, Navas P https://doi.org/10. 1093/gerona/glv070
(2013) Survival transcriptome in the coenzyme Q10 Goto T, Takano M (2009) Transcriptional role of FOXO1
deficiency syndrome is acquired by epigenetic in drug resistance through antioxidant defense
modifications: a modelling study for human coenzyme systems. Adv Exp Med Biol 665:171–179
Q10 deficiencies. BMJ Open 3(3): Gouspillou G, Sgarioto N, Norris B, Barbat-Artigas S,
e002524. https://doi.org/10.1136/bmjopen-2012- AubertinLeheudreM,MoraisJA,BurelleY,TaivassaloT,
002524 HeppleRT (2014) The relationship between muscle
Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, fiber type-specific PGC-1alpha content and
Ballestar ML, Heine-Suner D, Cigudosa JC, Urioste mitochondrial content varies between rodent models
M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, and humans. PLoS ONE 9(8):
Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, e103044. https://doi.org/10.1371/journal.pone.0103044
Spector TD, Wu YZ, Plass C, Esteller M (2005) Grazioli E, Dimauro I, Mercatelli N, Wang G, Pitsiladis Y,
Epigenetic differences arise during the lifetime of Di Luigi L, Caporossi D (2017) Physical activity in

123
Biogerontology
the prevention of human diseases: role of epigenetic coactivators, energy homeostasis, and metabolism.
modifications. BMC Genomics 18(Suppl 8):802. Endocr Rev
https://doi.org/10. 1186/s12864-017-4193-5 27(7):728–735. https://doi.org/10.1210/er.2006-0037
Grefhorst A, Elzinga BM, Voshol PJ, Plosch T, Kok T, Hardie DG (2011) Sensing of energy and nutrients by
Bloks VW, van der Sluijs FH, Havekes LM, Romijn AMPactivated protein kinase. Am J Clin Nutr
JA, Verkade HJ, Kuipers F (2002) Stimulation of 93(4):891S–896.
lipogenesis by pharmacological activation of the liver https://doi.org/10.3945/ajcn.110.001925
X receptor leads to production of large, triglyceride- Hardie DG, Ross FA, Hawley SA (2012) AMPK: a nutrient
rich very low density lipoprotein particles. J Biol and energy sensor that maintains energy homeostasis.
Chem 277(37):34182–34190. Nat Rev Mol Cell Biol 13(4):251–262.
https://doi.org/10.1074/jbc.M204887200 https://doi.org/10.1038/
Griffin TM, Humphries KM, Kinter M, Lim HY, Szweda nrm3311
LI Harman D (1956) Aging: a theory based on free radical and
(2015) Nutrient sensing and utilization: getting to the radiation chemistry. J Gerontol 11(3):298–300
heart of metabolic flexibility. Biochimie. Harman D (1972) The biologic clock: the mitochondria? J
https://doi.org/10. 1016/j.biochi.2015.10.013 Am Geriatr Soc 20(4):145–147
Guaras A, Perales-Clemente E, Calvo E, Acin-Perez R, Harper JW, Ordureau A, Heo JM (2018) Building and
Loureiro-Lopez M, Pujol C, Martinez-Carrascoso I, decoding ubiquitin chains for mitophagy. Nat Rev Mol
Nunez E, Garcia-Marques F, Rodriguez-Hernandez Cell Biol 19(2):93–108.
MA, Cortes A, Diaz F, Perez-Martos A, Moraes CT, https://doi.org/10.1038/nrm.2017.129
Fernandez-Silva P, Trifunovic A, Navas P, Vazquez J, Hernandez-Camacho JD, Bernier M, Lopez-Lluch G, Navas
Enriquez JA (2016) The CoQH2/CoQ ratio serves as a P (2018) Coenzyme Q10 supplementation in aging and
sensor of respiratory chain efficiency. Cell Rep disease. Front Physiol 9:44.
15(1):197–209. https://doi.org/10. https://doi.org/10.3389/fphys. 2018.00044
1016/j.celrep.2016.03.009 Holmstrom MH, Iglesias-Gutierrez E, Zierath JR, Garcia-
Guarente L (2011) Sirtuins, aging, and metabolism. Cold Roves PM (2012) Tissue-specific control of
Spring Harb Symp Quant Biol 76:81–90. mitochondrial respiration in obesity-related insulin
https://doi.org/10.1101/ sqb.2011.76.010629 resistance and diabetes. Am J Physiol Endocrinol
Guarente L (2013) Calorie restriction and sirtuins revisited. Metab 302(6):E731–739. https://
Genes Dev 27(19):2072–2085. doi.org/10.1152/ajpendo.00159.2011
https://doi.org/10.1101/ gad.227439.113 Horvath S (2013) DNA methylation age of human tissues
Gvozdjakova A, Kucharska J, Tkacov M, Singh RB, and cell types. Genome Biol 14(10):R115.
Hlavata A (2012) Ratio of lipid parameters to https://doi.org/10. 1186/gb-2013-14-10-r115
coenzyme Q10 could be used as biomarker of the Huffman DM, Barzilai N (2009) Role of visceral adipose
development of early complications of obesity in tissue in aging. Biochim Biophys Acta
children. Bratisl Lek Listy 113(1):21–25 1790(10):1117–1123.
Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, https://doi.org/10.1016/j.bbagen.2009.01.008
Mery A, Vasquez DS, Turk BE, Shaw RJ (2008) Imai S, Guarente L (2010) Ten years of NAD-dependent
AMPK phosphorylation of raptor mediates a SIR2 family deacetylases: implications for metabolic
metabolic checkpoint. Mol Cell 30(2):214–226. diseases. Trends Pharmacol Sci 31(5):212–220.
https://doi.org/10.1016/j.molcel. 2008.03.003 https://doi.org/10. 1016/j.tips.2010.02.003
Hafizi Abu Bakar M, Kian Kai C, Wan Hassan WN, Inoki K, Zhu T, Guan KL (2003) TSC2 mediates cellular
Sarmidi MR, Yaakob H, Zaman Huri H (2015) energy response to control cell growth and survival.
Mitochondrial dysfunction as a central event for Cell 115(5):577–590
mechanisms underlying insulin resistance: the roles of Ishihara M, Urushido M, Hamada K, Matsumoto T,
long chain fatty acids. Diabetes Metab Res Rev Shimamura Y, Ogata K, Inoue K, Taniguchi Y,
31(5):453–475. https://doi.org/10. 1002/dmrr.2601 Horino T, Fujieda M, Fujimoto S, Terada Y (2013)
Halling JF, Pilegaard H (2017) Autophagy-dependent Sestrin-2 and BNIP3 regulate autophagy and
beneficial effects of exercise. Cold Spring Harb mitophagy in renal tubular cells in acute kidney injury.
Perspect Med 7(8): a029777. Am J Physiol Renal Physiol 305(4):
https://doi.org/10.1101/cshperspect.a029777 F495–509.
Halling JF, Ringholm S, Olesen J, Prats C, Pilegaard H https://doi.org/10.1152/ajprenal.00642.2012
(2017) Exercise training protects against aging- Jager S, Handschin C, St-Pierre J, Spiegelman BM (2007)
induced mitochondrial fragmentation in mouse AMPactivated protein kinase (AMPK) action in
skeletal muscle in a PGC-1alpha dependent manner. skeletal muscle via direct phosphorylation of PGC-
Exp Gerontol 96:1–6. 1alpha. Proc Natl Acad Sci USA 104(29):12017–
https://doi.org/10.1016/j.exger.2017.05.020 12022. https://doi.org/10.1073/ pnas.0705070104
Handschin C, Spiegelman BM (2006) Peroxisome Jagota A, Kalyani D (2010) Effect of melatonin on age
proliferatoractivated receptor gamma coactivator 1 induced changes in daily serotonin rhythms in
suprachiasmatic nucleus of male Wistar rat.

123
Biogerontology
Biogerontology 11(3):299–308. mitochondrial toxin 3-nitropropionic acid. ASN Neuro
https://doi.org/10.1007/s10522-009-9248-9 6(1):e00133. https://doi.org/ 10.1042/AN20130042
Jheng HF, Tsai PJ, Guo SM, Kuo LH, Chang CS, Su IJ, Kume S, Uzu T, Horiike K, Chin-Kanasaki M, Isshiki K,
Chang CR, Tsai YS (2012) Mitochondrial fission Araki S, Sugimoto T, Haneda M, Kashiwagi A, Koya
contributes to mitochondrial dysfunction and insulin D (2010) Calorie restriction enhances cell adaptation
resistance in skeletal muscle. Mol Cell Biol to hypoxia through Sirt1-dependent mitochondrial
32(2):309–319. https://doi. org/10.1128/MCB.05603- autophagy in mouse aged kidney. J Clin Invest
11 120(4):1043–1055. https://doi.org/10.1172/JCI41376
Jin SH, Yang JH, Shin BY, Seo K, Shin SM, Cho IJ, Ki SH Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H,
(2013) Resveratrol inhibits LXRalpha-dependent Lerin C, Daussin F, Messadeq N, Milne J, Lambert P,
hepatic lipogenesis through novel antioxidant Sestrin2 Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J
gene induction. Toxicol Appl Pharmacol 271(1):95– (2006)
105. https:// Resveratrol improves mitochondrial function and
doi.org/10.1016/j.taap.2013.04.023 protects against metabolic disease by activating SIRT1
Joseph SB, Laffitte BA, Patel PH, Watson MA, Matsukuma and PGC1alpha. Cell 127(6):1109–1122.
KE, Walczak R, Collins JL, Osborne TF, Tontonoz P https://doi.org/10.1016/j. cell.2006.11.013
(2002) Direct and indirect mechanisms for regulation Lambert AJ, Brand MD (2009) Reactive oxygen species
of fatty acid synthase gene expression by liver X production by mitochondria. Methods Mol Biol
receptors. J Biol Chem 277(13):11019–11025. 554:165–181. https://doi.org/10.1007/978-1-59745-
https://doi.org/10.1074/jbc. M111041200 521-3_11
Kahn BB, Flier JS (2000) Obesity and insulin resistance. J Lane RK, Hilsabeck T, Rea SL (2015) The role of
Clin mitochondrial dysfunction in age-related diseases.
Invest 106(4):473–481. Biochim Biophys Acta 11:1387–1400.
https://doi.org/10.1172/JCI10842 https://doi.org/10.1016/j.bbabio.2015.05. 021
Katic M, Kennedy AR, Leykin I, Norris A, McGettrick A, Lapinska K, Faria G, McGonagle S, Macumber KM,
Gesta S, Russell SJ, Bluher M, Maratos-Flier E, Kahn Heerboth S, Sarkar S (2018) Cancer progenitor cells:
CR (2007) Mitochondrial gene expression and the result of an epigenetic event? Anticancer Res
increased oxidative metabolism: role in increased 38(1):1–6. https://doi.
lifespan of fat-specific insulin receptor knock-out org/10.21873/anticanres.12184
mice. Aging Cell 6(6):827–839. Laplante M, Sabatini DM (2009) mTOR signaling at a
https://doi.org/10.1111/j.1474-9726.2007.00346.x glance. J Cell Sci 122(Pt 20):3589–3594.
Kendrick AA, Choudhury M, Rahman SM, McCurdy CE, https://doi.org/10.1242/ jcs.051011
Friederich M, Van Hove JL, Watson PA, Birdsey N, Lee S, Jeong SY, Lim WC, Kim S, Park YY, Sun X, Youle
Bao J, Gius D, Sack MN, Jing E, Kahn CR, Friedman RJ, Cho H (2007) Mitochondrial fission and fusion
JE, Jonscher KR (2011) Fatty liver is associated with mediators, hFis1 and OPA1, modulate cellular
reduced SIRT3 activity and mitochondrial protein senescence. J Biol Chem 282(31):22977–22983.
hyperacetylation. Biochem J 433(3):505–514. https://doi.org/10.1074/jbc. M700679200
https://doi.org/10.1042/ BJ20100791 Lee JH, Budanov AV, Park EJ, Birse R, Kim TE, Perkins
Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo GA, Ocorr K, Ellisman MH, Bodmer R, Bier E, Karin
AM, Epel ES, Franceschi C, Lithgow GJ, Morimoto M (2010) Sestrin as a feedback inhibitor of TOR that
RI, Pessin JE, Rando TA, Richardson A, Schadt EE, prevents agerelated pathologies. Science
Wyss-Coray T, Sierra F (2014) Geroscience: linking 327(5970):1223–1228.
aging to chronic disease. Cell 159(4):709–713. https://doi.org/10.1126/science.1182228
https://doi.org/10.1016/j. cell.2014.10.039 Lee TH, Pastorino L, Lu KP (2011) Peptidyl-prolyl cis-
Khraiwesh H, Lopez-Dominguez JA, Lopez-Lluch G, trans isomerase Pin1 in ageing, cancer and Alzheimer
Navas P, de Cabo R, Ramsey JJ, Villalba JM, disease. Expert Rev Mol Med 13:e21.
Gonzalez-Reyes JA (2013) Alterations of https://doi.org/10.1017/ S1462399411001906
ultrastructural and fission/fusion markers in Lee JH, Budanov AV, Talukdar S, Park EJ, Park HL, Park
hepatocyte mitochondria from mice following calorie HW, Bandyopadhyay G, Li N, Aghajan M, Jang I,
restriction with different dietary fats. J Gerontol A Wolfe AM, Perkins GA, Ellisman MH, Bier E,
Biol Sci Med Sci 68(9):1023–1034. https://doi.org/10. Scadeng M, Foretz M, Viollet B, Olefsky J, Karin M
1093/gerona/glt006 (2012) Maintenance of metabolic homeostasis by
Kim J, Kundu M, Viollet B, Guan KL (2011) AMPK and Sestrin2 and Sestrin3. Cell Metab 16(3):311–321.
mTOR regulate autophagy through direct https://doi.org/10.1016/j.cmet. 2012.08.004
phosphorylation of Ulk1. Nat Cell Biol 13(2):132– Lee JH, Budanov AV, Karin M (2013) Sestrins orchestrate
141. https://doi.org/10. cellular metabolism to attenuate aging. Cell Metab
1038/ncb2152 18(6):792–801. https://doi.org/10.1016/j.cmet.2013.08.018
Kudo T, Loh DH, Tahara Y, Truong D, Hernandez- Leenen FA, Muller CP, Turner JD (2016) DNA
Echeagaray E, Colwell CS (2014) Circadian methylation: conducting the orchestra from exposure
dysfunction in response to in vivo treatment with the

123
Biogerontology
to phenotype? Clin Epigenetics 8:92. Lowell BB, Shulman GI (2005) Mitochondrial dysfunction
https://doi.org/10.1186/s13148-0160256-8 and type 2 diabetes. Science 307(5708):384–387.
Lenhare L, Crisol BM, Silva VRR, Katashima CK, https://doi. org/10.1126/science.1104343
Cordeiro AV, Pereira KD, Luchessi AD, da Silva Manikonda PK, Jagota A (2012) Melatonin administration
ASR, Cintra DE, Moura LP, Pauli JR, Ropelle ER differentially affects age-induced alterations in daily
(2017) Physical exercise increases Sestrin 2 protein rhythms of lipid peroxidation and antioxidant enzymes
levels and induces autophagy in the skeletal muscle of in male rat liver. Biogerontology 13(5):511–524.
old mice. Exp Gerontol 97:17–21. https://doi.org/10. 1007/s10522-012-9396-1
https://doi.org/10.1016/j.exger.2017.07.009 Martin-Montalvo A, de Cabo R (2013) Mitochondrial
Liesa M, Borda-d’Agua B, Medina-Gomez G, Lelliott CJ, metabolic reprogramming induced by calorie
Paz JC, Rojo M, Palacin M, Vidal-Puig A, Zorzano A restriction. Antioxid Redox Signal 19(3):310–320.
(2008) Mitochondrial fusion is increased by the https://doi.org/10.1089/ars. 2012.4866
nuclear coactivator PGC-1beta. PLoS ONE Matsumoto AM (2002) Andropause: clinical implications
3(10):e3613. https://doi.org/ of the decline in serum testosterone levels with aging
10.1371/journal.pone.0003613 in men. J Gerontol A Biol Sci Med Sci 57(2):M76–99
Liesa M, Palacin M, Zorzano A (2009) Mitochondrial Mattson MP, Moehl K, Ghena N, Schmaedick M, Cheng A
dynamics in mammalian health and disease. Physiol (2018) Intermittent metabolic switching,
Rev 89(3): 799–845. neuroplasticity and brain health. Nat Rev Neurosci
https://doi.org/10.1152/physrev.00030.2008 19(2):63–80. https://
Lin YF, Haynes CM (2016) Metabolism and the UPR(mt). doi.org/10.1038/nrn.2017.156
Mol Cell 61(5):677–682. McKendry J, Breen L, Shad BJ, Greig CA (2018) Muscle
https://doi.org/10.1016/j.molcel. 2016.02.004 morphology and performance in master athletes: a
Lopez-Lluch G (2017) Mitochondrial activity and dynamics systematic review and meta-analyses. Ageing Res Rev
changes regarding metabolism in ageing and obesity. 45:62–82. https://doi.org/10.1016/j.arr.2018.04.007
Mech Ageing Dev 162:108–121. Meister P, Schott S, Bedet C, Xiao Y, Rohner S, Bodennec
https://doi.org/10.1016/j.mad. 2016.12.005 S, Hudry B, Molin L, Solari F, Gasser SM, Palladino
Lopez-Lluch G, Navas P (2016) Calorie restriction as an F (2011) Caenorhabditis elegans Heterochromatin
intervention in ageing. J Physiol 594(8):2043–2060. protein 1 (HPL2) links developmental plasticity,
https://doi. org/10.1113/JP270543 longevity and lipid metabolism. Genome Biol
Lopez-Lluch G, Hunt N, Jones B, Zhu M, Jamieson H, 12(12):R123. https://doi.org/10. 1186/gb-2011-12-12-
Hilmer S, Cascajo MV, Allard J, Ingram DK, Navas P, r123
de Cabo R Menshikova EV, Ritov VB, Fairfull L, Ferrell RE, Kelley
(2006) Calorie restriction induces mitochondrial DE, Goodpaster BH (2006) Effects of exercise on
biogenesis mitochondrial content and function in aging human
andbioenergeticefficiency.ProcNatlAcadSciUSA103(6 skeletal muscle. J Gerontol A Biol Sci Med Sci
): 1768–1773. 61(6):534–540
https://doi.org/10.1073/pnas.0510452103 Merry BJ (2002) Molecularmechanisms
Lopez-Lluch G, Irusta PM, Navas P, de Cabo R (2008) linking calorie restriction and
Mitochondrial biogenesis and healthy aging. Exp longevity. Int J Biochem Cell
Gerontol 43(9):813–819. Biol 34(11):1340–1354
https://doi.org/10.1016/j.exger.2008.06. 014 Moehle EA, Shen K, Dillin A (2018) Mitochondrial
Lopez-Lluch G, Rodriguez-Aguilera JC, Santos-Ocana C, proteostasis in the context of cellular and organismal
Navas P (2010) Is coenzyme Q a key factor in aging? health and aging. J Biol Chem.
Mech Ageing Dev 131(4):225–235. https://doi.org/10.1074/jbc.TM117.000893
https://doi.org/10.1016/j. mad.2010.02.003 Moreno-Loshuertos R, Enriquez JA (2016) Respiratory
Lopez-Lluch G, Santos-Ocana C, Sanchez-Alcazar JA, supercomplexes and the functional segmentation of
Fernandez-Ayala DJ, Asencio-Salcedo C, Rodriguez- the CoQ pool. Free Radic Biol Med.
Aguilera JC, Navas P (2015) Mitochondrial responsibility https://doi.org/10.1016/j. freeradbiomed.2016.04.018
in ageing process: innocent, suspect or guilty. Mottillo S, Filion KB, Genest J, Joseph L, Pilote L, Poirier
Biogerontology 16(5): 599–620. P, Rinfret S, Schiffrin EL, Eisenberg MJ (2010) The
https://doi.org/10.1007/s10522-015-9585-9 Lopez-Otin C, metabolic syndrome and cardiovascular risk a
Blasco MA, Partridge L, Serrano M, Kroemer G (2013) systematic review and meta-analysis. J Am Coll
The hallmarks of aging. Cell 153(6):1194–1217. Cardiol 56(14):
https://doi.org/10.1016/j.cell.2013.05.039 1113–1132. https://doi.org/10.1016/j.jacc.2010.05.034
Lourenco AB, Munoz-Jimenez C, Venegas-Caleron M, Mourier A, Motori E, Brandt T, Lagouge M, Atanassov I,
ArtalSanz M (2015) Analysis of the effect of the Galinier A, Rappl G, Brodesser S, Hultenby K,
mitochondrial prohibitin complex, a context- Dieterich C, Larsson NG (2015) Mitofusin 2 is
dependent modulator of longevity, on the C. elegans required to maintain mitochondrial coenzyme Q
metabolome. Biochim Biophys Acta 11:1457–1468. levels. J Cell Biol
https://doi.org/10.1016/j.bbabio. 2015.06.003

123
Biogerontology
208(4):429–442. Park YY, Lee S, Karbowski M, Neutzner A, Youle RJ, Cho
https://doi.org/10.1083/jcb.201411100 H (2010) Loss of MARCH5 mitochondrial E3
Nakatsu Y, Sakoda H, Kushiyama A, Zhang J, Ono H, ubiquitin ligase induces cellular senescence through
Fujishiro M, Kikuchi T, Fukushima T, Yoneda M, dynamin-related protein 1 and mitofusin 1. J Cell Sci
Ohno H, Horike N, Kanna M, Tsuchiya Y, Kamata H, 123(Pt
Nishimura F, Isobe T, Ogihara T, Katagiri H, Oka Y, 4):619–626. https://doi.org/10.1242/jcs.061481
Takahashi S, Kurihara H, Uchida T, Asano T (2011) Pastore S, Hood DA (2013) Endurance training ameliorates
Peptidyl-prolyl cis/trans isomerase NIMA-interacting the metabolic and performance characteristics of
1 associates with insulin receptor substrate-1 and circadian Clock mutant mice. J Appl Physiol
enhances insulin actions and adipogenesis. J Biol 114(8):1076–1084.
Chem 286(23):20812–20822. https:// https://doi.org/10.1152/japplphysiol.01505.2012
doi.org/10.1074/jbc.M110.206904 Patti ME, Corvera S (2010) The role of mitochondria in the
Nakatsu Y, Iwashita M, Sakoda H, Ono H, Nagata K, pathogenesis of type 2 diabetes. Endocr Rev
Matsunaga Y, Fukushima T, Fujishiro M, Kushiyama 31(3):364–395. https://doi.org/10.1210/er.2009-0027
A, Kamata H, Takahashi S, Katagiri H, Honda H, Payne BA, Chinnery PF (2015) Mitochondrial dysfunction
Kiyonari H, Uchida T, Asano T (2015) Prolyl in aging: much progress but many unresolved
isomerase Pin1 negatively regulates AMP-activated questions. Biochim Biophys Acta 11:1347–1353.
protein kinase (AMPK) by associating with the CBS https://doi.org/10. 1016/j.bbabio.2015.05.022
domain in the gamma subunit. J Biol Chem Peek CB, Affinati AH, Ramsey KM, Kuo HY, Yu W, Sena
290(40):24255–24266. LA, Ilkayeva O, Marcheva B, Kobayashi Y, Omura C,
https://doi.org/10.1074/jbc.M115. 658559 Levine DC, Bacsik DJ, Gius D, Newgard CB,
Naon D, Zaninello M, Giacomello M, Varanita T, Grespi F, Goetzman E, Chandel NS, Denu JM, Mrksich M, Bass
Lakshminaranayan S, Serafini A, Semenzato M, J (2013) Circadian clock NAD ? cycle drives
Herkenne S, Hernandez-Alvarez MI, Zorzano A, De mitochondrial oxidative metabolism in mice. Science
Stefani D, Dorn GW 2nd, Scorrano L (2016) Critical 342(6158):1243417. https://
reappraisal confirms that Mitofusin 2 is an doi.org/10.1126/science.1243417
endoplasmic reticulum-mitochondria tether. Proc Natl Pich S, Bach D, Briones P, Liesa M, Camps M, Testar X,
Acad Sci USA 113(40):11249–11254. Palacin M, Zorzano A (2005) The Charcot-Marie-
https://doi.org/10.1073/pnas.1606786113 Tooth type 2A gene product, Mfn2, up-regulates fuel
Nemoto S, Fergusson MM, Finkel T (2005) SIRT1 oxidation through expression of OXPHOS system.
functionally interacts with the metabolic regulator and Hum Mol Genet
transcriptional coactivator PGC-1{alpha}. J Biol 14(11):1405–1415.
Chem 280(16): https://doi.org/10.1093/hmg/ddi149
16456–16460. Qiu X, Brown K, Hirschey MD, Verdin E, Chen D (2010)
https://doi.org/10.1074/jbc.M501485200 Calorie restriction reduces oxidative stress by SIRT3-
Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R, mediated SOD2 activation. Cell Metab 12(6):662–
Tedesco L, Falcone S, Valerio A, Cantoni O, Clementi 667. https://doi.org/10.1016/j.cmet.2010.11.015
E, Moncada S, Carruba MO (2005) Calorie restriction Rai N, Venugopalan G, Pradhan R, Ambastha A, Upadhyay
promotes mitochondrial biogenesis by inducing the AD, Dwivedi S, Dey AB, Dey S (2018) Exploration of
expression of eNOS. Science 310(5746):314–317. novel anti-oxidant protein sestrin in frailty syndrome
https://doi.org/10.1126/ science.1117728 in elderly. Aging Dis 9(2):220–227.
Olichon A, Emorine LJ, Descoins E, Pelloquin L, Brichese https://doi.org/10.14336/AD. 2017.0423
L, Gas N, Guillou E, Delettre C, Valette A, Hamel CP, Ramadori G, Coppari R (2010) Pharmacological
Ducommun B, Lenaers G, Belenguer P (2002) The manipulations of CNS sirtuins: potential effects on
human dynamin-related protein OPA1 is anchored to metabolic homeostasis. Pharmacol Res 62(1):48–54.
the mitochondrial inner membrane facing the inter- https://doi.org/10.1016/j.
membrane space. FEBS Lett 523(1–3):171–176 phrs.2010.02.002
Page MM, Robb EL, Salway KD, Stuart JA (2010) Ramadori G, Lee CE, Bookout AL, Lee S, Williams KW,
Mitochondrial redox metabolism: aging, longevity and Anderson J, Elmquist JK, Coppari R (2008) Brain
dietary effects. Mech Ageing Dev 131(4):242–252. SIRT1: anatomical distribution and regulation by
https://doi.org/ 10.1016/j.mad.2010.02.005 energy availability. J Neurosci 28(40):9989–9996.
Palacios OM, Carmona JJ, Michan S, Chen KY, Manabe Y, https://doi.org/10. 1523/JNEUROSCI.3257-08.2008
Ward JL 3rd, Goodyear LJ, Tong Q (2009) Diet and Rauthan M, Ranji P, Aguilera Pradenas N, Pitot C, Pilon M
exercise signals regulate SIRT3 and activate AMPK (2013) The mitochondrial unfolded protein response
and PGC-1alpha in skeletal muscle. Aging (Albany activator ATFS-1 protects cells from inhibition of the
NY) 1(9):771–783 mevalonate pathway. Proc Natl Acad Sci USA
Palikaras K, Lionaki E, Tavernarakis N (2015) Coupling 110(15):
mitogenesis and mitophagy for longevity. Autophagy 5981–5986. https://doi.org/10.1073/pnas.1218778110
11(8):1428–1430. Ravussin E, Redman LM, Rochon J, Das SK, Fontana L,
https://doi.org/10.1080/15548627.2015. 1061172 Kraus WE, Romashkan S, Williamson DA, Meydani

123
Biogerontology
SN, Villareal DT, Smith SR, Stein RI, Scott TM, in the aging process. In: Clark KB (ed) Bioenergetics.
Stewart TM, Saltzman E, Klein S, Bhapkar M, Martin InTech, Rijeka,
CK, Gilhooly CH, Holloszy JO, Hadley EC, Roberts pp 95–120
SB, Group CS (2015) A 2-year randomized controlled Santel A, Frank S (2008) Shaping mitochondria: the
trial of human caloric restriction: feasibility and complex posttranslational regulation of the
effects on predictors of health span and longevity. J mitochondrial fission protein DRP1. IUBMB Life
Gerontol A Biol Sci Med Sci 60(7):448–455. https://doi. org/10.1002/iub.71
70(9):1097–1104. Schottl T, Kappler L, Fromme T, Klingenspor M (2015)
https://doi.org/10.1093/gerona/glv057 Limited OXPHOS capacity in white adipocytes is a
Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, hallmark of obesity in laboratory mice irrespective of
Shimomura I, Shan B, Brown MS, Goldstein JL, the glucose tolerance status. Mol Metab 4(9):631–642.
Mangelsdorf DJ (2000) Regulation of mouse sterol https://doi.org/10. 1016/j.molmet.2015.07.001
regulatory elementbinding protein-1c gene (SREBP- Schubeler D (2015) Function and information content of
1c) by oxysterol receptors. LXRalpha and LXRbeta. DNA methylation. Nature 517(7534):321–326.
Genes Dev 14(22): 2819–2830 https://doi.org/ 10.1038/nature14192
Riera CE, Merkwirth C, De Magalhaes Filho CD, Dillin A Scialo F, Sriram A, Fernandez-Ayala D, Gubina N, Lohmus
(2016) Signaling networks determining life span. M, Nelson G, Logan A, Cooper HM, Navas P,
Annu Rev Biochem 85:35–64. Enriquez JA, Murphy MP, Sanz A (2016)
https://doi.org/10.1146/annurevbiochem-060815- Mitochondrial ROS produced via reverse electron
014451 transport extend animal lifespan. Cell Metab
Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte 23(4):725–734. https://doi.org/10.1016/j.cmet.
GR, Tran D, Perez G, Gutierrez-Casado E, Koike S, 2016.03.009
Knotts TA, Imai DM, Griffey SM, Kim K, Hagopian Scrima R, Cela O, Merla G, Augello B, Rubino R, Quarato
K, McMackin MZ, Haj FG, Baar K, Cortopassi GA, G, Fugetto S, Menga M, Fuhr L, Relogio A, Piccoli C,
Ramsey JJ, LopezDominguez JA (2018) A ketogenic Mazzoccoli G, Capitanio N (2016) Clock-genes and
diet extends longevity and healthspan in adult mice. mitochondrial respiratory activity: evidence of a
Cell Metab 27(5):1156. reciprocal interplay. Biochim Biophys Acta 8:1344–
https://doi.org/10.1016/j.cmet.2018.04.005 1351. https://doi. org/10.1016/j.bbabio.2016.03.035
Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Sebastian D, Palacin M, Zorzano A (2017) Mitochondrial
Puigserver P (2005) Nutrient control of glucose dynamics: coupling mitochondrial fitness with healthy
homeostasis through a complex of PGC-1alpha and aging. Trends Mol Med 23(3):201–215.
SIRT1. Nature 434(7029):113–118. https://doi.org/10. 1016/j.molmed.2017.01.003
https://doi.org/10.1038/ Shaw JE, Zimmet PZ, George K, Alberti MM (2005)
nature03354 Metabolic syndrome-do we really need a new
Rodriguez-Bies E, Navas P, Lopez-Lluch G (2015) Age- definition? Metab Syndr Relat Disord 3(3):191–193.
dependent effect of every-other-day feeding and https://doi.org/10.1089/ met.2005.3.191
aerobic exercise in ubiquinone levels and related Short KR, Bigelow ML, Kahl J, Singh R, Coenen-Schimke
antioxidant activities in mice muscle. J Gerontol A J, Raghavakaimal S, Nair KS (2005) Decline in
Biol Sci Med Sci 70(1):33–43. skeletal muscle mitochondrial function with aging in
https://doi.org/10.1093/gerona/glu002 humans. Proc Natl Acad Sci USA 102(15):5618–5623.
Romanello V, Sandri M (2015) Mitochondrial quality https://doi.org/10. 1073/pnas.0501559102
control and muscle mass maintenance. Front Physiol Smirnova E, Griparic L, Shurland DL, van der Bliek AM
6:422. https:// doi.org/10.3389/fphys.2015.00422 (2001) Dynamin-related protein Drp1 is required for
Ruiz R, Perez-Villegas EM, Manuel Carrion A (2016) mitochondrial division in mammalian cells. Mol Biol
AMPK function in aging process. Curr Drug Targets Cell 12(8):2245–2256
17(8):932–941 Son C, Hosoda K, Ishihara K, Bevilacqua L, Masuzaki H,
Ryan BJ, Hoek S, Fon EA, Wade-Martins R (2015) Fushiki T, Harper ME, Nakao K (2004) Reduction of
Mitochondrial dysfunction and mitophagy in dietinduced obesity in transgenic mice overexpressing
Parkinson’s: from familial to sporadic disease. Trends uncoupling protein 3 in skeletal muscle. Diabetologia
Biochem Sci 47(1):47–54. https://doi.org/10.1007/s00125-003-
40(4):200–210. 1272-8
https://doi.org/10.1016/j.tibs.2015.02.003 Soriano FX, Liesa M, Bach D, Chan DC, Palacin M,
Samant SA, Zhang HJ, Hong Z, Pillai VB, Sundaresan NR, Zorzano A (2006) Evidence for a mitochondrial
Wolfgeher D, Archer SL, Chan DC, Gupta MP (2014) regulatory pathway defined by peroxisome
SIRT3 deacetylates and activates OPA1 to regulate proliferator-activated receptorgamma coactivator-1
mitochondrial dynamics during stress. Mol Cell Biol alpha, estrogen-related receptor-alpha, and mitofusin
34(5):807–819. https://doi.org/10.1128/MCB.01483- 2. Diabetes 55(6):1783–1791. https://
13 Santa-Cruz Calvo S, Navas P, Lo´pez-Lluch G doi.org/10.2337/db05-0509
(2012) Sirtuindependent meabolic control and its role Sousa JS, D’Imprima E, Vonck J (2018) Mitochondrial
respiratory chain complexes. Subcell Biochem

123
Biogerontology
87:167–227. https://doi.org/10.1007/978-981-10- murine hearts. Mech Ageing Dev 167:30–45.
7757-9_7 https://doi.org/10.1016/j.mad.2017. 09.002
Steinberg GR, O’Neill HM, Dzamko NL, Galic S, Naim T, Viana-Huete V, Guillen C, Garcia G, Fernandez S,
Koopman R, Jorgensen SB, Honeyman J, Hewitt K, GarciaAguilar A, Kahn CR, Benito M (2018) Male
Chen brown fatspecific double knockout of IGFIR/IR:
ZP, Schertzer JD, Scott JW, Koentgen F, Lynch GS, atrophy, mitochondrial fission failure, impaired
Watt MJ, van Denderen BJ, Campbell DJ, Kemp BE thermogenesis, and obesity. Endocrinology
(2010) Whole body deletion of AMP-activated protein 159(1):323–340. https://doi.org/ 10.1210/en.2017-
kinase {beta}2 reduces muscle AMPK activity and 00738
exercise capacity. J Biol Chem 285(48):37198–37209. Vina J, Gomez-Cabrera MC, Borras C, Froio T, Sanchis-
https://doi. Gomar F, Martinez-Bello VE, Pallardo FV (2009)
org/10.1074/jbc.M110.102434 Mitochondrial biogenesis in exercise and in ageing.
Storlien L, Oakes ND, Kelley DE (2004) Metabolic Adv Drug Deliv Rev 61(14):1369–1374.
flexibility. Proc Nutr Soc 63(2):363–368. https://doi.org/10.1016/j.addr.2009.06. 006
https://doi.org/10.1079/ PNS2004349 Wallace DC (2012) Mitochondria and cancer. Nat Rev
Sundstrom J, Riserus U, Byberg L, Zethelius B, Lithell H, Cancer 12(10):685–698.
Lind L (2006) Clinical value of the metabolic https://doi.org/10.1038/nrc3365
syndrome for long term prediction of total and Wang L, Ishihara T, Ibayashi Y, Tatsushima K, Setoyama
cardiovascular mortality: prospective, population D,
based cohort study. BMJ 332(7546):878–882. Hanada Y, Takeichi Y, Sakamoto S, Yokota S, Mihara
https://doi.org/10.1136/bmj.38766. 624097.1F K, Kang D, Ishihara N, Takayanagi R, Nomura M
Szafranski K, Mekhail K (2014) The fine line between (2015) Disruption of mitochondrial fission in the liver
lifespan extension and shortening in response to protects mice from diet-induced obesity and metabolic
caloric restriction. Nucleus 5(1):56–65. deterioration. Diabetologia 58(10):2371–2380.
https://doi.org/10.4161/nucl.27929 https://doi.org/10. 1007/s00125-015-3704-7
Tao R, Xiong X, Liangpunsakul S, Dong XC (2015) Sestrin Wang M, Xu Y, Liu J, Ye J, Yuan W, Jiang H, Wang Z,
3 protein enhances hepatic insulin sensitivity by direct Jiang H, Wan J (2017) Recent insights into the
activation of the mTORC2-Akt signaling. Diabetes biological functions of sestrins in health and disease.
64(4):1211–1223. https://doi.org/10.2337/db14-0539 Cell Physiol Biochem 43(5):1731–1741.
Tatar M, Sedivy JM (2016) Mitochondria: masters of https://doi.org/10.1159/000484060
epigenetics. Cell 165(5):1052–1054. Weber TA, Reichert AS (2010) Impaired quality control of
https://doi.org/10.1016/j. cell.2016.05.021 mitochondria: aging from a new perspective. Exp
Theiss AL, Vijay-Kumar M, Obertone TS, Jones DP, Gerontol 45(7–8):503–511.
Hansen JM, Gewirtz AT, Merlin D, Sitaraman SV https://doi.org/10.1016/j.exger.2010.03. 018
(2009) Prohibitin is a novel regulator of antioxidant Weinert D (2010) Circadian temperature variation and
response that attenuates colonic inflammation in mice. ageing. Ageing Res Rev 9(1):51–60.
Gastroenterology 137(1):199–208. https://doi.org/10.1016/j.arr. 2009.07.003
https://doi.org/10.1053/j.gastro.2009.03. 033 Willcox BJ, Willcox DC (2014) Caloric restriction, caloric
Tian Y, Garcia G, Bian Q, Steffen KK, Joe L, Wolff S, restriction mimetics, and healthy aging in Okinawa:
Meyer BJ, Dillin A (2016) Mitochondrial stress controversies and clinical implications. Curr Opin Clin
induces chromatin reorganization to promote Nutr Metab Care 17(1):51–58.
longevity and UPR(mt). Cell 165(5):1197–1208. https://doi.org/10.1097/MCO. 0000000000000019
https://doi.org/10.1016/j.cell.2016.04. 011 Wroblewski AP, Amati F, Smiley MA, Goodpaster B,
Toledo FG, Goodpaster BH (2013) The role of weight loss Wright V (2011) Chronic exercise preserves lean
and exercise in correcting skeletal muscle muscle mass in masters athletes. Phys Sportsmed
mitochondrial abnormalities in obesity, diabetes and 39(3):172–178. https://
aging. Mol Cell Endocrinol 379(1–2):30–34. doi.org/10.3810/psm.2011.09.1933
https://doi.org/10.1016/j. mce.2013.06.018 Yang YL, Loh KS, Liou BY, Chu IH, Kuo CJ, Chen HD,
Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Chen CS (2013) SESN-1 is a positive regulator of
Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, lifespan in Caenorhabditis elegans. Exp Gerontol
Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, 48(3):371–379.
Shirihai OS (2008) Fission and selective fusion govern https://doi.org/10.1016/j.exger.2012.12.011
mitochondrial segregation and elimination by Yoon Y, Krueger EW, Oswald BJ, McNiven MA (2003)
autophagy. EMBO J The mitochondrial protein hFis1 regulates
27(2):433–446. https://doi.org/10.1038/sj.emboj.7601963 mitochondrial fission in mammalian cells through an
Valli H, Ahmad S, Chadda KR, Al-Hadithi A, Grace AA, interaction with the dynamin-like protein DLP1. Mol
Jeevaratnam K, Huang CL (2017) Age-dependent Cell Biol
atrial arrhythmic phenotype secondary to 23(15):5409–5420
mitochondrial dysfunction in Pgc-1beta deficient Yoon YS, Yoon DS, Lim IK, Yoon SH, Chung HY, Rojo
M, Malka F, Jou MJ, Martinou JC, Yoon G (2006)

123
Biogerontology
Formation of elongated giant mitochondria in DFO- radical theory of aging. Aging Cell 14(1):1–7.
induced cellular senescence: involvement of enhanced https://doi.org/10. 1111/acel.12287
fusion process through modulation of Fis1. J Cell
Physiol
209(2):468–480. https://doi.org/10.1002/jcp.20753
Zampieri M, Ciccarone F, Calabrese R, Franceschi C,
Burkle A, Caiafa P (2015) Reconfiguration of DNA
methylation in aging. Mech Ageing Dev 151:60–70.
https://doi.org/10. 1016/j.mad.2015.02.002
Zeng N, D’Souza RF, Mitchell CJ, Cameron-Smith D
(2018)
Sestrins are differentially expressed with age in the
skeletal muscle of men: a cross-sectional analysis. Exp
Gerontol. https://doi.org/10.1016/j.exger.2018.05.006
Ziegler DV, Wiley CD, Velarde MC (2015) Mitochondrial
effectors of cellular senescence: beyond the free

123

You might also like