You are on page 1of 29

REVIEW

Metabolic Flexibility as an Adaptation to Energy


Resources and Requirements in Health and Disease
Reuben L. Smith,1,2 Maarten R. Soeters,2,3 Rob C. I. Wüst,1,4,5 and Riekelt H. Houtkooper1,2,4

1
Laboratory of Genetic Metabolic Diseases, Academic Medical Center, 1105 AZ Amsterdam, Netherlands;
2

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


Amsterdam Gastroenterology and Metabolism, Academic Medical Center, 1105 AZ Amsterdam, Netherlands;
3
Department of Endocrinology and Metabolism, Internal Medicine, Academic Medical Center, 1105 AZ
Amsterdam, Netherlands; 4Amsterdam Cardiovascular Sciences, Academic Medical Center, 1105 AZ Amsterdam,
Netherlands; and 5Amsterdam Movement Sciences, Academic Medical Center, 1105 AZ Amsterdam, Netherlands

ABSTRACT The ability to efficiently adapt metabolism by substrate sensing, trafficking, storage, and utilization, dependent on availability
and requirement, is known as metabolic flexibility. In this review, we discuss the breadth and depth of metabolic flexibility and its impact on
health and disease. Metabolic flexibility is essential to maintain energy homeostasis in times of either caloric excess or caloric restriction, and
in times of either low or high energy demand, such as during exercise. The liver, adipose tissue, and muscle govern systemic metabolic
flexibility and manage nutrient sensing, uptake, transport, storage, and expenditure by communication via endocrine cues. At a molecular
level, metabolic flexibility relies on the configuration of metabolic pathways, which are regulated by key metabolic enzymes and
transcription factors, many of which interact closely with the mitochondria. Disrupted metabolic flexibility, or metabolic inflexibility,
however, is associated with many pathological conditions including metabolic syndrome, type 2 diabetes mellitus, and cancer. Multiple
factors such as dietary composition and feeding frequency, exercise training, and use of pharmacological compounds, influence metabolic
flexibility and will be discussed here. Last, we outline important advances in metabolic flexibility research and discuss medical horizons and
translational aspects. (Endocrine Reviews 39: 489 – 517, 2018)

H uman physiology evolved during times of


dramatic fluctuations in energy supply and
demand. Coping with these changes has entrained the
unprecedented levels of food supply (). This near
continuous intake of calorically dense processed
foods, combined with physical inactivity, reduces a
human body with the ability to manage energy predilection for, and directly impedes, metabolic
metabolism for optimal substrate storage and use flexibility (). This is caused by substrate competition
during states of either food surplus or famine, and and metabolic insensitivity, characterized by dis-
periods of either rest or increased energy demand. The torted nutrient sensing, blunted substrate switching,
ability to efficiently adapt metabolism depending on and impaired energy homeostasis (). Importantly,
demand or supply is known as metabolic flexibility (). this metabolic inflexibility may underlie the epidemic
In general, the human body can aptly use moderate changes in metabolic disease that affect all de- ISSN Print: 0163-769X
amounts of carbohydrates, fatty acids, and amino mographic groups and burden health-care systems ISSN Online: 1945-7189
acids. The modern era, however, is characterized by (, ). Printed: in USA
Copyright © 2018
Endocrine Society
This article has been published
Defining Metabolic Flexibility adaptations of skeletal muscle to exercise (see “Physical under the terms of the
exercise”). The term metabolic flexibility was coined by Creative Commons
Maintaining energy homeostasis requires substrate Kelley et al. () in  when they studied fuel se- Attribution License (CC BY;
https://creativecommons.org/
sensing, trafficking, storage, and utilization, dependent lection in skeletal muscle in lean and obese individuals
licenses/by/4.0/).
on substrate availability (push concept) and energy after an overnight fast. Specifically, they discovered Received: 16 September 2017
requirement (pull concept). Metabolic plasticity (or that skeletal muscle of lean individuals showed a re- Accepted: 19 April 2018
adaptability) was recognized in  by Saltin and markable ability to adapt fuel preference to fasting and First Published Online:
Gollnick () when they reviewed the metabolic insulin infusions and were therefore designated as 24 April 2018

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 489


REVIEW

ESSENTIAL POINTS
· Metabolic flexibility describes efficient switches in metabolism depending on environmental demand
·· Mitochondria play a crucial role in determining metabolic flexibility
Metabolic inflexibility is a hallmark of many age-related metabolic diseases but also plays a central role in, for instance,
cancer and immune metabolism
· Molecular and signaling pathways drive metabolic flexibility and often serve as metabolic sensors
· Metabolic flexibility pathways are therapeutic targets for age-related diseases, similar to caloric restriction or exercise

metabolically flexible (). Insulin-resistant obese pa- diseases such as type  diabetes mellitus (TDM),
tients however manifested a lesser reliance on fatty obesity, cancer, and during conditions of systemic

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


acid oxidation compared with lean individuals and did inflammation such as sepsis. Third, we explore how
not show increased fatty acid oxidation after fasting or metabolic inflexibility can be reversed by lifestyle or
reduced fatty acid oxidation after insulin infusion. pharmacological interventions. Last, we outline future
Because of their inadequate responses to metabolic perspectives for the research field and discuss trans-
challenges, these patients were named “metabolically lational aspects and medical horizons.
inflexible” (). More recent work showed that, upon
consumption of a high-fat diet, lean subjects with
adequate metabolic flexibility were able to increase Physiological Relevance of Metabolic Flexibility
fatty acid oxidation (FAO) at the expense of glucose,
whereas obese individuals were not (). Lean in- Feeding/fasting
dividuals also showed an increased expression of genes In healthy individuals, metabolism is distinctly dif-
involved in fatty acid transport and oxidation com- ferent during a state of fasting compared with a state of
pared with little or no change in their obese coun- caloric availability. In the beginning of the s,
terparts (). Francis Benedict described the adaptation to starvation
The concept of metabolic flexibility was particu- in a single human subject (). To date, this is the most
larly linked to the capacity of mitochondria to select elaborate and detailed human work on what, in ret-
fuel in response to nutritional changes () and placed rospect, can be addressed as metabolic flexibility. Quite
mitochondrial function at its core (). Later, meta- some time later, multiple studies quantified reciprocal
bolic flexibility quickly expanded to encompass the changes in the adaptation to fasting [reviewed in
ability of a given system (whole-body, organ, single Soeters et al. ()] emphasizing the dynamic changes
cell, or organelle) to handle specific nutrients. At a in substrate utilization during feeding and fasting. At
molecular level, metabolic flexibility relies on the the same time, many studies investigated nutrient
configuration of metabolic pathways that manage intake, but it was not until the s that a study
nutrient sensing, uptake, transport, storage, and uti- investigated the postprandial response in relation to
lization. This metabolic organization is mediated by glucose and lipids ().
synthesis, degradation, or activity regulation of key On a systemic level, maintaining metabolic ho-
proteins in metabolic circuits or enzymes with a high meostasis during feeding or fasting relies on multi-
metabolic flux (, ). Metabolic flexibility is not an organ coordinated control of available fuel. During the
“on-off” phenomenon, but involves tightly regulated postprandial period of a carbohydrate-rich meal for
subtle adjustments. Taken together, metabolic flexi- instance, pancreatic b cells respond to the rise in
bility can be understood as an adaptive response of an nutrients by releasing insulin into the bloodstream,
organism’s metabolism to maintain energy homeo- increasing the insulin-glucagon ratio (Fig. ). Under
stasis by matching fuel availability and demand to the influence of insulin, the liver is triggered to absorb
periodic fasting, varying meal composition, physical glucose from the circulation and stop glycogenolysis
activity, and environmental fluctuations (, ). and gluconeogenesis. Skeletal muscle assists in glucose
Here, we review the impact of metabolic flexibility clearance as insulin receptor binding of insulin results
on health and disease. First, we recapitulate how in translocation of glucose transporters (GLUT),
metabolic flexibility is regulated during natural fluc- mainly GLUT, to the plasma membrane, allowing
tuations in dietary fuel availability and during states of glucose to enter the cell. Adipose tissue responds to
increased demand, such as upon physical exertion. We insulin by decreasing the rate of lipolysis and stim-
focus on the mechanistic underpinnings of metabolic ulating fatty acid and triacylglycerol synthesis from
flexibility, denote its regulators, and bring the mito- lipids and glucose (). Collectively, this buffering
chondria forward as central to this process. Second, we capacity ensures that the exposure of tissues to hy-
examine impaired metabolic flexibility as a cause of perglycemia is minimalized and that nutrients are

490 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

stored in adipocytes for release and oxidation in times On the other hand, during fasting, the inhibition of
of scarcity. Fatty acids derived from a meal trigger FAO is released by the action of the energy stress
adipose tissue to reduce nonesterified fatty acid release sensor adenosine monophosphate-activated protein
and prompt hepatocytes to reduce endogenous tri- kinase (AMPK), which inhibits acetyl-CoA carbox-
acylglycerol release, together stimulating the clearance ylase (ACC) by phosphorylation. Inhibition of ACC
of circulating triacylglycerol (). lowers malonyl-CoA concentrations, resulting in an
During fasting, the decrease in circulating dietary increased activity of CPT- and amplified transport of
carbohydrates and lipids and decline in insulin- fatty acids into the mitochondria for b-oxidation.
glucagon ratio induces a switch toward FAO (). When FAO is preferred, acetyl-CoA and reduced
Glucagon stimulates hepatic glycogenolysis and ke- nicotinamide adenine dinucleotide (NADH) levels
togenesis, and the decrease in insulin suppresses he- rise, impeding PDH catalytic activity through allosteric
patocyte malonyl-coenzyme A (CoA) synthesis and inhibition and via activation of PDK. During fasting,
lipogenesis, with concomitant activation of FAO (, PDK gene expression increases through fatty acid–
). After an overnight fast, ketone bodies are in the dependent peroxisome proliferator–activated receptor

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


micromolar range (~ mM for women and ~ mM (PPAR) signaling (). Additionally, an increase in
for men), which is not detected by most assays (). fat availability elevates citrate levels, which inhibit
This is important because it emphasizes that an phosphofructokinase and GLUT, impeding glucose
overnight fast already induces changes in metabolic uptake and use. Inhibition of phosphofructokinase
flexibility in healthy humans. also increases the cytosolic concentration of glucose-
Hydrolytic processes in white adipose tissue (WAT) -phosphate that inhibits hexokinase, making it more
are increased converting stored triacylglycerol to free difficult to metabolize glucose (). This allosteric in-
fatty acids that are used in the periphery as fuel, hibition results in a feed-forward loop, favoring FAO
particularly in the skeletal muscle and heart. Addi- during times of nutrient scarcity to conserve glucose for
tionally, lipolytic processes in the muscle aid in in- biosynthetic processes and brain metabolism. Because
creasing lipid supply (). Because adipose tissue is glucose-derived pyruvate in the liver is no longer
the predominant source of free fatty acids, the ca- converted to acetyl-CoA by PDH, pyruvate can be used
pacity of adipose tissue to store fatty acids during as a gluconeogenic precursor to avoid hypoglycemia.
caloric availability and release fatty acids during In summary, the transition between feed-
caloric restriction is an important determinant of ing and fasting, and vice versa, is regulated by
metabolic flexibility ().

Biochemical transition between feeding White adipose tissue Skeletal muscle


and fasting Fatty acid synthesis Fatty acid oxidation
Triacylglycerol synthesis Glucose uptake & oxidation
Cellular fuel selection depends on the type and Lipolysis
amount of nutrient available. Cellular responses to Glucose
the changes in nutritional state are predominantly
assigned to the mitochondria (see “Mitochondrial
function is essential for metabolic flexibility” and
“Endocrine regulation of metabolic flexibility”).
As proposed in Randle’s glucose-fatty acid cycle
hypothesis, in vitro studies have shown that FAO Pancreas
is suppressed when glucose consumption is in- Insulin/glucagon
creased and vice versa () (Fig. ). In the post-
prandial period of a carbohydrate-rich meal
when glucose and insulin are high, glucose
uptake, glycolysis, and pyruvate oxidation are Heart Brain
favored and FAO is suppressed. These glucose- Fatty acid oxidation Appetite
dependent processes increase the concentration Glucose oxidation
Liver
of malonyl-CoA, which allosterically inhibits Glucose uptake
carnitine palmitoyltransferase- (CPT-), which Lipogenesis
Glycolysis
transports fatty acids into the mitochondria for Gluconeogenesis
b-oxidation. Fatty acids are then alternatively used Glycogenolysis
© 2018 Endocrine Reviews
for triglyceride synthesis and stored. Additionally, ENDOCRINE SOCIETY Ketogenesis
the rise in pyruvate from glycolysis inhibits
Figure 1. Systemic response to insulin. Upon glucose sensing, the pancreas increases the insulin/
pyruvate dehydrogenase kinase (PDK), which glucagon ratio. The rise in insulin stimulates many metabolic processes in the key metabolic organs:
reduces the phosphorylation of pyruvate de- the liver, heart, brain, white adipose tissue, and skeletal muscle. Collectively, these metabolic
hydrogenases (PDHs), resulting in an increased processes switch metabolism from a preference of fatty acid oxidation to glucose uptake and
glucose oxidation (). oxidation.

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 491


REVIEW

communication via endocrine cues and metabolic Prolonged fasting and caloric/dietary restriction
signals that orchestrates fuel oxidation and traf- The importance of energy and nutrient sensing
ficking in tissues throughout the body. The al- transcription factor regulated pathways can be dem-
losteric inhibition of key regulatory enzymes in onstrated during prolonged fasting. It is somewhat
conflicting metabolic pathways ensures a robust surprising that, until , ketone bodies (KBs) were
and typically acute switch between the oxida- thought to have no beneficial physiological role during
tion of glucose (fed state) or fatty acids (fasted prolonged fasting. Currently, we know that ketone
state) (Fig. ). On top of this direct regulation, a bodies are produced by the liver upon prolonged
slower orchestration exists in which metabolic fasting. The central nervous system requires approx-
changes drive the activity of transcription factors imately  g of glucose per day (equivalent to almost
and thereby fine tune the cellular metabolic  kcal), also during fasting. Plasma KB then are an
responses. important source of energy because their blood levels
and oxidation rates increase (). During fasting, KB
can act as an excellent respiratory fuel:  g of D--

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


hydroxybutyrate yields . kg of ATP ( ATP
per molecule D--hydroxybutyrate), whereas  g
of glucose yields only . kg of ATP (). Ketogen-
esis occurs in the liver, where fatty acids undergo
b-oxidation to form acetyl-CoA. This enters the ke-
togenesis pathway, where mitochondrial -hydroxy--
methylglutaryl-CoA synthase is the most important
enzyme involved in ketogenesis. This enzyme is
inhibited by insulin; hence, ketogenesis only occurs
at low insulin (and blood glucose) levels ().
Additionally, insulin inhibits lipolysis by inhibiting
hormone-sensitive lipase in adipose tissue and thereby
prevents the liberation of fatty acids for hepatic
ketogenesis. More recently, ketone bodies have
emerged as an alternative substrate to improve ex-
ercise performance (), and could be used as a
therapeutic target for patients with inborn errors of
metabolism ().
An alternative, and quantitatively less important,
source of fuel during prolonged fasting is the break-
down of amino acids, in particular branched-chain
amino acids (BCAAs). The mitochondrial branched-
chain a-ketoacid dehydrogenase (BCKD) complex is
the rate-limiting step in BCAA catabolism (). BCKD
can be allosterically inhibited when acyl-CoA con-
centrations and NADH levels are sufficient (). This
ensures that under fed conditions and during short
intervals of fasting or light exercise, cellular proteins are
conserved. With nutrient abundance, however, when
BCAA are present in excess such as after a protein-rich
Figure 2. Mechanism of reciprocal inhibition of glucose and fatty acid oxidation. When glucose meal, BCKD also becomes active as BCAA-derived
uptake and consumption increases, fatty acid oxidation is suppressed by malonyl-CoA’s allosteric a-ketoacids allosterically inhibit BCKDs deactivating
inhibition of CPT-1, and increased pyruvate from glycolysis inhibits PDK, which stimulates glucose kinase ().
oxidation (yellow lines). CPT-1 inhibition increases the concentration of LCFA-CoAs, which then
Dietary restriction, more commonly known as
are used for triglyceride synthesis and stored (pink arrow). Vice versa, when fatty acid oxidation is
high, glucose uptake, glycolysis, and pyruvate oxidation are decreased (red lines) because rising caloric restriction (CR), is the prolonged and con-
levels of acetyl-CoA and NADH impede PDH activity. Additionally, increased citrate levels inhibit trolled reduced intake of all dietary constituents while
GLUT4 and PFK-1. PFK-1 inhibition results in increased glucose-6-phosphate concentrations that maintaining appropriate intake of vitamins and
inhibit HK. A decrease in pyruvate oxidation enables pyruvate to be used as either a gluconeogenic minerals. Recycling of cell-intrinsic macromolecules is
precursor or, in energetically demanding tissues, a substrate for PC, which produces oxaloacetate essential to sustain metabolic processes when nutrients
that is used as anaplerotic substrate (purple arrows). During caloric restriction, the rise in AMP/ATP
remain chronically scarce. This intricate salvaging
activates AMPK, which inhibits ACC, stimulating fatty acid uptake by the mitochondria via CPT-1.
ACL, ATP-citrate lyase; CACT, carnitine acylcarnitine translocase; CTP, citrate transport protein;
process is controlled by autophagy (). Autophagy is
CYTO, cytosol; FAS, fatty acid synthase; LCFA, long-chain fatty acid; MITO, mitochondria; MPC, regulated by the deactivation of the nutrient sensors
mitochondrial pyruvate carrier; PC, pyruvate carboxylase. Green arrows indicate stimulatory mechanistic target of rapamycin (mTOR) and v-Akt
reactions. murine thymoma viral oncogene homolog /protein

492 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

kinase B (Akt/PKB), and activation of the cellular caloric excess, elevated levels of acetyl-CoA increase
energy status sensors AMPK and sirtuins (SIRTs) (). protein acetylation events (because they serve as
In mammals, reduced insulin/IGF- signaling acyl donors). A high NADH/NAD+ ratio leads to
through reduced nutrient intake inhibits Akt, which inactivation of mitochondrial and nuclear SIRT
leads to activation of the forkhead box protein O deacetylases, reducing mitochondrial biogenesis and
(FOXO) transcription factors that upregulate the cells’ activity (). Indeed, prolonged high-fat feeding of
maintenance pathways: DNA repair, autophagy, and rodents or continuous fatty acid exposure of myotubes
stress resistance (). mTOR signaling stimulates leads to a reduction in nuclear-encoded mitochondrial
growth and blocks tissue maintenance when nutrients genes (). A reduction in b-oxidation can cause
are plentiful. However, upon CR, reduced intake of accumulation of long-chain fatty acyl-CoA, diacyl-
proteins, particularly of BCAA, downregulates the glycerol, triacylglycerol, and ceramide, which may
mTOR pathway, causing a switch toward salvage increase serine phosphorylation of the insulin receptor
pathways such as autophagy and conserved translation and reduce activation of Akt/PKB, leading to impaired
(). In response to increasing cellular AMP/ATP insulin signaling (). Ultimately, metabolic flexibility is

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


ratios, which rise during CR, AMPK activates cata- impaired during long-term caloric excess, affecting
bolic pathways and represses anabolic pathways many tissues. Among other clinical phenotypes, caloric
(). In parallel, SIRT activity is increased during CR, excess is correlated to metabolic syndrome, obesity,
which depends on oxidized nicotinamide ade- TDM, and cardiovascular disease () (see “Patho-
nine dinucleotide (NAD+) concentrations, and leads to physiology of Metabolic Inflexibility”; Fig. ).
protein deacetylation and improved mitochon-
drial function (see “SIRTs”; Fig. ). Although much Physical exercise
work has been performed on CR and the meta- A good example of cell intrinsic metabolic pro-
bolic adaptations thereof, it is currently unknown gramming upon physiological stimulation occurs in
if this would be a suitable strategy in human skeletal muscle. Skeletal muscle consists of oxidative
interventions. (type I) and glycolytic (type II) fibers, which differ in
their metabolic abilities. Oxidative muscle fibers have a
Caloric excess high mitochondrial density; hence, they prefer oxi-
With caloric excess, the mitochondria are over- dative phosphorylation for ATP production. They also
whelmed by an excess in in substrates derived from contain more lipid droplets and rely on FAO. Gly-
fatty acids, glucose, and amino acids. In this setting, colytic muscle fibers have a low mitochondrial density
metabolic flexibility is altered by a push-mechanism and rely predominantly on the breakdown of stored
(see “Defining Metabolic Flexibility”). Through per- glycogen by glycolysis for their ATP production (,
sistent allosteric inhibition and feed-forward responses ). During low-intensity exercise, oxidative muscle
this surplus leads to mitochondrial metabolic “in- fibers predominantly rely on FAO for their ATP
decision” and ineffective substrate switching, resulting production. During more intense exercise, the rising
in incomplete substrate utilization for energy pro- ATP utilization rate induces a metabolic switch from
duction and subsequent storage of substrates in (ec- FAO to glucose metabolism. When maximal pyruvate
topic) depots (). For instance, high levels of fatty acids production outstrips its mitochondrial import during
can increase expression of PDK through transcrip- severe intensity exercise, pyruvate is converted into
tional activation of PPARa, resulting in inactivation of lactate and NAD+ by lactate dehydrogenase in the
PDH and thus blunting glucose oxidation (). It has cytosol, after which lactate is excreted from the cell.
been suggested that a low ATP utilization in com- NAD+ generation helps maintain cytosolic redox
bination with a high mitochondrial membrane potential and promotes substrate flux through gly-
potential increases reactive oxygen species (ROS) colysis to sustain ATP generation ().
production, causing oxidative damage and triggering As such, regular physical exercise is a classic ex-
signaling events, and importantly affects the activity of ample of how metabolic flexibility is regulated by
redox sensitive metabolic enzymes (). transcription factors. During acute exercise, an in-
With a low electron flux through the electron creased AMP/ATP ratio, sensed by AMPK, in-
transport system, the NADH/NAD+ ratio in the mi- creases transcription, translation, and activity of the
tochondria increases causing redox inhibition of transcriptional coactivator PPAR gamma coactivator
several tricarboxylic acid (TCA) cycle enzymes and -alpha (PGCa) (, ). PGCa is a regulator of
reduces the supply of carbons into the TCA cycle. This exercise-induced adaptations in the capacity of oxi-
results in elevated levels of acetyl-CoA and other acyl- dative phosphorylation (OXPHOS) in skeletal muscle
CoA at crucial sites where catabolism of fatty acids, (, ). In particular, PGCa interacts and coactivates
glucose, and amino acids converge, leading to a state in many transcription factors and nuclear receptors that
which upstream substrate catabolic flux slows down are involved in mitochondrial energy homeostasis
considerably, a phenomenon coined as mitochondrial and metabolic adaptations, such as nuclear respira-
metabolic “gridlock” (). Furthermore, in times of tory factors (NRFs) and PPARs. NRFs regulate the

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 493


REVIEW

Figure 3. Energy- and nutrient-sensing transcription factor–regulated pathways. During caloric excess and obesity, anabolic processes
are stimulated via the insulin/IGF-1 and TOR pathways. Additionally, caloric excess suppresses catabolic processes via a decrease in the
AMP/ATP ratio, leading to reduced activation of AMPK and downstream activity of PGC1a, sirtuins, and FOXO. Simultaneously,
decreased NAD+ levels reduce sirtuin activity, inhibiting PPARg activity and increasing HIF1a activity. Alternatively, during caloric
restriction and fasting, catabolic processes are stimulated by the increase in AMP/ATP ratio and consequential activation of AMPK.
AMPK also reduces anabolic processes through TOR inhibition. AKT, v-Akt murine thymoma viral oncogene homolog.

Caloric excess/ Caloric restriction/


Obesity Fasting

AMP/ATP

Insulin/

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


IGF-1 AMPK

NAD+

AKT mTOR

sirtuins FOXO

Glucose Protein Mitochondrial Lipid Oxidative


Glycolysis
uptake synthesis function metabolism stress defense
© 2018 Endocrine Reviews ENDOCRINE SOCIETY

expression of nuclear genes encoding OXPHOS cellular glucose handling via GLUT/ expression
proteins, and PPARs regulate the transcription of and insulin sensitivity via nitric oxide (NO) synthase,
genes that encode enzymes involved in lipid transport it is considered a critical player of metabolic flexi-
and catabolism (). The increase of mitochondrial bility during physical activity ().
biogenesis and FAO improves insulin sensitivity. The
role of PGCa in metabolic flexibility is underlined by Hibernation and cold exposure
observations that basal PGCa skeletal muscle ex- Research in migratory birds (), killifish (), and in
pression is reduced in sedentary subjects (). animals that enter states of torpor or hibernation ()
In mouse muscle cells, exercise-dependent calcium has revealed that temperature can influence meta-
influx activates calcineurin that dephosphorylates the bolic reprogramming and metabolic flexibility. Cold-
helix-loop-helix leucine zipper transcription factor EB acclimated birds increase thermogenic capacity
(TFEB) ensuring its localization to the nucleus. Here, through elevated expression of fatty acid transporter
TFEB controls the expression of genes involved in proteins in flight-muscle fibers and mitochondrial
glucose uptake such as GLUT and , hexokinase membranes, and increase mitochondrial density
(HK) I and II, TBC domain family member , and and FAO, OXPHOS, and TCA cycle enzymes ().
glycogen synthase, which collectively lead to glycogen Metabolism in hibernators is also switched to lipids
production to sustain energy generation during later as the major fuel for all organs, although their over-
bouts of exercise (). Additionally, TFEB increases all metabolic rate is severely reduced (). More-
the expression of NRF and mitochondrial tran- over, catabolic processes that consume large
scription factor A (TFAM), which are regulators of amounts of ATP are suppressed, including mito-
mitochondrial biogenesis in muscle. Specifically, sis and cell proliferation, mitochondrial metabolism,
TFAM regulates mitochondrial DNA (mtDNA) transmembrane ion transport, global mRNA tran-
transcription and replication (). Recently, TFEB scription, and protein biosynthesis. Protein bio-
was found to act independently of PGCa to pro- synthesis in particular is linked to decreased Akt and
mote mitochondrial biogenesis and oxidation of mTOR activity (). These studies cautiously sug-
glucose and lipids, and because TFEB directly alters gest that reducing the core body temperature in

494 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

mammals may have a beneficial effect on metabolic feeding/fasting cycles (). The central circadian
health. pacemaker mainly acts through its powerful influence
Until recently the presence of brown adipose tissue over the endocrine system (). The peripheral clocks
(BAT) was thought to be confined to small mammals are synchronized by the central clock and are present
and infants. However, recent studies have shown that in almost all mammalian tissues, where they regulate
in adult humans, BAT activity can be stimulated by tissue specific gene expression ().
mild cold (°C) exposure, suggesting that BAT has Indeed, the circadian clock can have major effects
physiological relevance in humans, too (–). Im- on metabolic flexibility and is even able to coordinate
portantly, brown adipogenesis has been reported in temporal and spatial organization of lipids and cir-
WAT, demonstrating that WAT harbors the potential cadian rhythmicity of mitochondrial function (, ).
to switch to BAT. Brown-like adipocytes within WAT The peripheral circadian clock has several mechanisms
are known as brite (brown-in-white) or beige adipo- to influence metabolism including regulation of
cytes, and transdifferentiation from white to brown is metabolite levels, interaction with nutrient sensors,
commonly referred to as “browning.” control of rate-limiting metabolic enzymes, and

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


Master regulators of browning in humans are modulation of nuclear receptors (). Circadian
PPARg and PGCa (). PPAR agonists in white proteins directly coordinate with key regulators of
adipocytes caused conversion to a brite/beige mo- nutrient homeostasis, including AMPK, cAMP re-
lecular phenotype in vitro, defined by increased sponse element-binding protein, and Akt/PKB, which
mitochondrial oxygen consumption and amplified are themselves driven by daily rhythms of nutrient
expression of the thermogenic gene program, in- supply (, ). Gene expression of key metabolic
cluding uncoupling protein , which is located in the enzymes show diurnal variations (). Expression of
inner mitochondrial membrane and uncouples mi- nuclear receptors such as members of the PPAR family
tochondrial respiration, releasing it as heat (). In and estrogen-related receptor family are also under
mice, transcriptional changes in response to ther- circadian clock control (). Moreover, diurnal vari-
mogenic challenges suggest increased glucose uptake, ations in human skeletal muscle oxidative capacity
glycolysis, glycogen metabolism, pentose phosphate were recently observed and may be linked to the
pathway (PPP) flux, and OXPHOS (), although the circadian clock in muscle ().
exact contribution of such pathways to thermogenesis The circadian clock is under influence from, and
is unclear. In bright/beige adipocytes glucose, utili- can synchronize to, external stimuli such as food
zation is likely switched from OXPHOS to glycer- intake and diet (). For instance, the liver synchro-
oneogenesis favoring triglyceride synthesis. The shift nizes its peripheral circadian rhythm based on the
from glucose to fatty acid use for OXPHOS is enforced availability of circulating metabolites (, ). Pe-
by increased b-oxidation of fatty acids, which restricts ripheral tissues communicate dietary signals to the
complete oxidation of glucose through PDK inhibition brain via endocrine cues such as ghrelin, leptin, and
of the PDH complex. This metabolic pathway rewiring insulin, meaning feeding rhythm strongly contributes
directs pyruvate toward glyceroneogenesis (). to the reciprocal relationship of the circadian clock
Obese subjects showed lower BAT activity than and metabolism (, ). The nutrient sensors AMPK
lean subjects, and low BAT activity is associated with and SIRT that modulate circadian gene expres-
metabolic dysfunctions such as TDM and aging (). sion and circadian oscillating metabolites such as
Although noradrenalin can induce browning, adren- cAMP and NAD+, are known regulators of the cir-
ergic therapy does not activate BAT to the same extent cadian clock (, , ).
as cold exposure, and incurs adverse cardiovascular Although the expression of many genes of both the
effects (). It seems that increasing BAT activity and circadian clock and metabolism fluctuate reciprocally
induction of browning by short-term cold exposure in and in response to environmental cues, enzymatic
humans can increase insulin sensitivity (), but more activity can be modulated by posttranscriptional
work needs to be performed before it can be regarded modification of proteins, which adds an additional
as a suitable treatment of obesity and TDM. layer of rhythmicity to the circadian network ().
Research in this field is very scarce and much is still
Interplay between circadian rhythm and unknown. Not only the timing but also the compo-
metabolic flexibility sition of food intake affect the circadian clock. Mice fed
The circadian clock enables organisms to anticipate on high-fat diets, for instance, showed altered ex-
the diurnal variation in metabolic substrates (). The pression of core clock genes and the genes under their
circadian clock can be divided into the central and control, altered circadian rhythms, and consumed
peripheral clocks. The central, or master clock, is lo- larger amounts of food during their active phase ().
cated in the hypothalamic suprachiasmatic nucleus The extent to which metabolites have control over the
and can function autonomously without any exter- circadian clock is currently unknown. It is likely,
nal input, but can be entrained by temporally rele- however, that an interplay of all metabolites (the
vant external input such as, temperature, light, and metabolome) acts as a cue for the circadian clock

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 495


REVIEW

because many metabolites affect its phase, amplitude, flexibility and prolong healthy aging () (see “Exercise
and/or period of oscillations (). training”).
Disruption of the circadian rhythm, or circadian In summary, on a cellular level, acute metabolic
misalignment, in human subjects can result in insulin flexibility is a universal property of healthy cells ().
resistance (). As a result, (night)shift workers are at a Metabolic flexibility, and therefore substrate flux, is
greater risk to develop obesity, TDM, cardiovascular principally determined by the reciprocity of metabolic
disease, and metabolic syndrome (, ). Currently, circuitries, of which the presence is dependent on the
much more work is required to fully understand the cell’s gene and protein expression, nutrient availability
mechanistic link between a disruption in the circadian and/or demand. On a systemic level, metabolically
rhythm, the loss of metabolic flexibility, and the de- active organs such as the liver, muscle, heart, and
velopment of metabolic disease. adipose tissue, communicate to best organize the
utilization of available fuel. This holistic and vis-à-vis
Aging orchestration of available nutrients to sustain whole-
The underlying multifactorial aspects to aging make it body energy homeostasis has ensured organism sur-

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


difficult to discern specifically which are most caus- vival and is therefore interwoven with both healthy
ative of the aging process. Nonetheless, understanding and diseased states of metabolism.
the fundamental aspects of aging and targeting these
processes using physiological or pharmacological ap-
proaches can limit the progression of many age-related
Regulation of Metabolic Flexibility
diseases ().
The metabolic influence on aging and lifespan has
The intrinsic qualitative and quantitative capacity of
gained increased attention over the past decade; as
cells to oxidize or store energy is dependent on the
such, major regulators of metabolic flexibility play
molecular organization of their metabolic pathways.
dominant roles in aging (, ). Indeed, metabolic
This tissue-specific metabolic programming depends
flexibility is negatively correlated with aging () and
on the coordinated action of various enzymes and
targeting metabolic flexibility as a cause for aging and
transcription factors, which are collectively orches-
related comorbidities may provide cues to delay
trated by intrinsic mitochondrial function), circulating
the onset of age-related diseases and prolong health
endocrine factors, and epigenetic programming.
span. Mitochondrial dysfunction and a cellular shift
toward a glycolytic phenotype is intimately linked to
senescence and the “senescence-associated secretory Mitochondrial function is essential for
phenotype” that entails the secretion of multiple metabolic flexibility
factors such as proinflammatory cytokines, proteases, Mitochondria are equipped to transform metabolism
and growth factors that have potent local and systemic intermediates such as pyruvate, fatty acids, and
effects such as inflammation and metastasis (, ). amino acids, into the reduced energetic equivalents
Perturbation of mitochondrial function and nutrient- NADH and/or flavin adenine dinucleotide through
sensing pathways, particularly related to glucose ho- b-oxidation and the TCA cycle. These are then used in
meostasis, is a hallmark of aging (). As such, the complexes located in the inner mitochondrial mem-
nutrient and energy sensing pathways (insulin/IGF, brane where electrons are transferred from electron
mTOR, AMPK, and SIRTs) are causally involved in donors to acceptors by redox reactions, with oxygen as
fitness and longevity (, ). Consistent with this, CR their ultimate acceptor. During this transfer to a lower
increases health span and/or lifespan in model or- redox potential, the liberated energy is used to extrude
ganisms and improves several markers of health in protons from the mitochondrial matrix into the
humans () (see “Prolonged fasting and caloric/ mitochondrial intermembrane space, generating an
dietary restriction”). electrochemical proton gradient that is used by the
It is currently unclear how metabolic flexibility is FFo-ATP-synthase to generate ATP from ADP and
perturbed in the elderly, because few metabolic flex- inorganic phosphate. As such, mitochondria are the
ibility studies have been conducted in late middle-aged final acceptors for metabolic substrates and are the
and aged populations. One underlying cause to main players for understanding the pull concept of
age-related metabolic pathophysiology is a sedentary metabolic flexibility.
lifestyle that is steadily increasing in prevalence in the Mitochondria are pliable organelles, and they adapt
general population and in particular strongly increases their morphology to nutrient availability and in doing
as people age. A study in middle-aged postmenopausal so regulate OXPHOS activity and substrate prefer-
women showed that endurance training improved ence (, ). Recently, isolated skeletal muscle mito-
work-related ability to mobilize and oxidize free fatty chondria from rats fed high-sugar or high-fat diets
acids, suggesting that in the elderly metabolic flexi- showed reduced metabolic flexibility, indicating
bility can still be trained (). Importantly, exercise that substrate preference is independent of cytosolic-
is a principal preventive strategy to improve metabolic mitochondrial communication and in fact a consequence

496 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

of inherent mitochondrial biochemical network in- Gut endocrine regulation of metabolic flexibility
teractions (, ). The postprandial state is characterized by various,
Mitochondrial bioenergetic function can be con- mainly gut-derived, factors that somehow affect
trolled through both acute changes, aimed to promptly metabolism. The postprandial increase of some of
modify activity, and longer term transcriptional re- these actually modulates metabolic flexibility either
sponses, aimed to regulate mitochondrial volume directly or indirectly via increased insulin secretion.
density. Mitochondrial bioenergetics can be altered For example, glucagonlike peptide- (GLP-) is re-
through calcium activation of mitochondrial enzymes leased by enteral L cells and agonizes pancreatic insulin
(), posttranslational mechanisms such as protein secretion. However, GLP- exerts direct inhibitory
acetylation, and through dynamic adaptations of effects on hepatic glucose production via direct hepatic
morphological architecture by use of mitochondrial or neuronal inhibition (). Also, GLP may con-
fission and fusion components (). For instance, the tribute to reduced intestinal lipoprotein production.
acetylation state of mitochondrial proteins differs Likewise, bile acids also facilitate nutrient trafficking
strongly between the fed and fasting state (). in a hormone-like fashion (). Bile acids induce

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


Mitochondrial morphology is dependent on nu- insulin secretion directly via the transmembrane bile
tritional status of the cell. Cells exposed to a nutrient acid receptor Takeda G protein–coupled receptor 
overload have a fragmented mitochondrial network, (on b cells and indirectly via stimulation of L-cell
whereas upon CR, mitochondria appear more inter- derived GLP- and subsequent insulin release (–).
connected (). Increasing mitochondrial elongation Additionally, the postprandial increase in bile acids
and interconnectivity induces a bioenergetic adapta- also increases insulin sensitivity and energy ex-
tion that increases ATP synthesis capacity and ef- penditure (). In contrast to these two examples,
ficiency (). Conversely, fragmentation of the the peptide ghrelin is produced in stomach X/A cells
mitochondrial network reduces bioenergetic efficiency and actually decreases during food intake. The post-
and might protect against detrimental effects of nu- prandial decrease in ghrelin lowers hepatic glucose
trient overload (, ). production while increasing peripheral glucose-uptake
Many studies support the idea that deregulation of in both skeletal muscle and adipose tissue (). All in
mitochondrial function underlies the onset of meta- all, the combined effects of different factors during food- “Myokines play important
roles in mediating the positive
bolic inflexibility [reviewed in Muoio ()], although a intake and fasting facilitate the organisms’ fuel avail- effects of exercise on whole-
causal link between the two still remains to be fully ability and metabolic flexibility. body metabolism.”
established (, ). Besides regulating glucose
metabolism and fatty acid oxidation in most cell types, Other endocrine factors affecting
mitochondria regulate triglyceride synthesis and glu- metabolic flexibility
coneogenesis in hepatocytes and lipolysis in adipose Many other circulating factors are involved in meta-
tissue (, ). Additionally, insulin secretion from bolic flexibility, including cytokines and other peptides
pancreatic b cells () and synthesis and secretion of that are expressed, produced, and released by adi-
adipokines from WAT is dependent on mitochondrial pocytes (adipokines), muscle (myokines), and liver
function (), suggesting that mitochondria fulfill a (hepatokines). Although the precise function of many
crucial role in determining cellular, tissue, and systemic of these factors remains elusive, some exert autocrine,
metabolic flexibility. Furthermore, a reduction in mito- paracrine, or endocrine effects that are fundamental
chondrial oxidative capacity is linked to the development for organ cross-talk in the regulation of energy ho-
of TDM and obesity (see “Metabolic syndrome and meostasis (). For a comprehensive view on the roles
TDM”), whereas exercise-enhanced mitochondrial of adipokines, myokines, and hepatokines, we refer
performance is related to a better metabolic flexibility and the reader to some excellent reviews on the topic
insulin sensitivity () (see “Cancer”). (–). In this section, we give brief examples to
highlight their role in regulation of metabolic flexi-
Endocrine regulation of metabolic flexibility bility. Figure  summarizes these examples.
The importance of endocrine regulation of metabolic Fully functional adipocytes reduce lipotoxicity in
flexibility, in particular to coordinate complex in- tissues such as the heart and liver and they maintain a
terorgan government of energy storage and oxidation, healthy balance of adipokines, which exert paracrine
is undeniable. As discussed previously, metabolic effects on adipocytes in their direct vicinity and en-
flexibility relies on the action of insulin and glucagon docrine effects on the central nervous system, immune
major regulators of glucose metabolism in response to system, and peripheral tissues. Adiponectin suppresses
dietary stage () (see “Feeding/fasting”, “Biochemical glucose production in the liver and enhances FAO in
transition between feeding and fasting”, and “Pro- skeletal muscle (, ). Indeed, high levels of the
longed fasting and caloric/dietary restriction”). Other insulin-sensitizing, antiapoptotic, and anti-inflammatory
newly discovered paracrine and endocrine factors have hormone adiponectin has been proposed to improve
emerged that also alter metabolism, and some of the metabolic flexibility of adipose tissue, enhancing its
most important will be discussed here. function under metabolic challenges (). Leptin is a

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 497


REVIEW

Figure 4. Circulating factors not produced by specific myostatin knockout mice have significantly im-
endocrine glands are involved in metabolic flexibility. Examples proved insulin sensitivity and glucose uptake, have
include those that are produced by the intestine, adipocytes increased peripheral tissue FAO, and are protected
(adipokines), muscle (myokines), and liver (hepatokines) and
from diet-induced obesity (, ). Another cir-
released into the bloodstream. These endocrine factors act on
metabolism through paracrine and endocrine signaling, and culating factor named meteorin-like has been de-
distal organs include skeletal muscle, adipose tissue, liver, scribed to be released from skeletal muscle after
pancreas, heart, and brain. Much is currently unknown about exercise and in adipose tissue upon cold exposure
these endocrine factors. See “Other endocrine factors affecting (). Meteorin-like is involved in the adaptive
metabolic flexibility” for a brief description of some examples responses to the regulation of energy homeostasis
and their role in regulation of metabolic flexibility.
and tissue inflammation, but the therapeutic po-
tential for metabolic and inflammatory diseases is
currently unknown. Irisin, another myokine, has
been proposed as an important glucoregulatory
candidate. However, contradictory findings con-

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


cerning the role of irisin in humans exist; therefore,
results must be interpreted with caution.
Intestine Hepatokines
Hepatokines can also regulate whole-body meta-
e.g. e.g. bolic flexibility and some are even considered as
• GLP-1 • Fetuin-A
potential targets for the treatment of cardiovascular
• Ghrelin • FGF21
disease [reviewed in Jung et al. ()]. For example,
Circulation fetuin-A has a major role in the regulation of insulin
sensitivity as fetuin-A–deficient mice showed im-
Adipokines Myokines proved insulin sensitivity (). Nucleotide poly-
e.g. e.g. morphisms in human fetuin-A and high levels in
• Adiponectin • Myostatin
• Leptin • Meteorin-like
serum are a predictable marker for the incidence of
• FGF21 • FGF21 TDM (). Additionally, fasting reduces the circu-
lating levels of fetuin-A, whereas high levels of satu-
rated fatty acids and glucose augments the expression
of fetuin-A (). Mice deficient of fetuin-A are re-
© 2018 Endocrine Reviews ENDOCRINE SOCIETY
sistant to high-fat diet–induced obesity and have
improved glucose tolerance ().
Interestingly, some of these paracrine and en-
central feedback indicator for the brain on the docrine factors are secreted by multiple tissues, and
amount of stored energy in the body and rises in their local function and impact on metabolic
concert with the amount of adipose tissue (). flexibility can depend on their origin and local
Leptin also exhibits diurnal expression patterns that plasma concentrations. IL-, for instance, when
are dampened in obese subjects (see “Interplay be- secreted by skeletal muscle, stimulates AMPK ac-
tween circadian rhythm and metabolic flexibility”) tivity and in this way increases glucose uptake and
(). Taken together, adipocytes have an important b-oxidation in muscle and adipose tissue (). IL-
role in systemic neuroendocrine regulation of met- , is, however, also secreted by adipocytes from
abolic flexibility as adipose tissue is both responsive to obese patients and negatively affects metabolic
and responsible for diverse metabolic, inflammatory, flexibility by decreasing insulin signaling and glu-
and hormonal signals [reviewed in Luo and Liu ()]. cose uptake because of its proinflammatory prop-
Myokines provide skeletal muscle with the ability erties (, ). Another example is fibroblast
to mediate whole-body metabolism via endocrine growth factor  (FGF), which, as a myokine,
signaling to adipose tissue, liver, pancreas, heart, and increases GLUT expression in skeletal muscle,
brain (). In particular, myokines play important boosting glucose uptake (). FGF from muscle
roles in mediating the positive effects of exercise on also exerts endocrine-like effects on WAT, in-
whole-body metabolism [reviewed in Oh et al. creasing lipolysis and b-oxidation and inducing
()]. Production of myokines is predominantly browning (). As an adipokine, however, FGF
influenced by skeletal muscle contraction and can stimulates insulin-independent glucose uptake in
alter glucose disposal, FAO, and lipolysis. For peripheral tissues; as a hepatokine, FGF stimu-
instance, the alteration of myostatin expression lates lipolysis in WAT ().
contributes to the proliferation, development, and Although the role of adipokines, myokines, and
metabolism of adipose and skeletal muscle tissue hepatokines in the control of whole-body energy
(). Exercise decreases the expression of myo- homeostasis and metabolic flexibility is only recently
statin in humans and obesity is associated with becoming evident, it is to be reported that they have
increased myostatin expression (). Moreover, clinical relevance and diagnostic potential.

498 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

Epigenetic regulation of metabolic flexibility an altered epigenetic landscape associated with


Metabolic flexibility in response to environmental disrupted lipid oxidative metabolism and mito-
stimuli, such as diet and exercise, are dramatically chondrial function in adipose tissue, skeletal muscle,
influenced by epigenetic factors as they influence gene and liver ().
expression by regulating access of transcriptional
machinery to DNA. Evidence that epigenetic changes
drive metabolic inflexibility in humans is emerging Pathophysiology of Metabolic Inflexibility
().
Metabolic networks, in particular those in the Although inborn errors of metabolism are clear ex-
mitochondria, directly transmit information about amples of metabolic inflexibility, here we focus on
the cells metabolic state to epigenetic programming acquired metabolic inflexibility. For specific in-
enzymes that, for instance, add or remove epigenetic formation on inborn errors of metabolism we refer the
markers onto chromatin (, ). In this way, reader to a comprehensive book on the subject ().
histones can act as metabolic sensors, converting Here, we discuss the pathophysiology of metabolic

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


changes in metabolism into stable patterns of gene flexibility in the context of obesity, metabolic syn-
expression, a concept named “metabolic memory” drome and TDM, as well as systemic inflammation,
(, ). Both global fluctuations in me- cardiovascular disease, and cancer.
tabolite levels caused by nutritional inputs, circa-
dian rhythm, and oxygenation, or local changes Obesity
depending on intracellular metabolite distribution, An estimated % of the adult US population is obese
can translate into epigenetic changes (, ). The or overweight (). At the heart of obesity lies the
abundance of cofactors and the metabolic enzymes inability to regulate lipolytic and antilipolytic processes
that generate them not only alter epigenetic enzyme in adipose tissue during starvation and feeding, re-
histone modification, but also affect DNA methyl- spectively. Obesity is predominantly associated with
ation and posttranslational modification of the elevated levels of plasma free fatty acids (). High
epigenetic enzymes themselves, resulting in a com- circulating levels of free fatty acids inhibit glycogen
plex feedback network (, ). Moreover, the synthase activity and PDH activity, which leads to
amplitude and duration of the metabolic stimulus reduced disposal and oxidation of glucose. Besides
required to alter the epigenome is dependent on the adipocyte metabolic dysfunction, skeletal muscle mi-
vastly different kinetics of epigenetic modifier en- tochondrial capacity and b-oxidation are reduced.
zymes (). Metabolic sensory epigenetic pro- Specifically, upregulation of PPARa and its down-
gramming enzymes include: histone acetyltransferases stream targets in response to high-fat feeding are
that acetylate histones using acetyl-CoA (), SIRTs defective (). Excess calories are then stored in
that deacetylase histones using NAD+ as a cofactor (), peripheral fat depots as triglyceride; when these depots
histone methyl-transferases that methylate or remove reach their maximum capacity and fail to expand, fat
methyl marks using S-adenosylmethionine as a methyl accumulates in ectopic depots, including skeletal
donor, or lysine-specific demethylases that use flavin muscle and the liver. Ectopic fat deposition is related to
adenine dinucleotide as a cofactor (). How these metabolic abnormalities and defects in insulin sensi-
epigenetic regulators are targeted to specific sites, such tivity, TDM, cardiovascular disease, and cancer ().
as promotor regions, how transient their epigenetic Finally, obesity is associated with a state of chronic
markers are, and how these changes are inherited, is still low-grade inflammation because ectopic fat depots
under active investigation (). release more inflammatory mediators than peripheral
People with a family history for TDM have an fat depots and infiltration of macrophages ().
increased risk for developing metabolic inflexibility; Metabolic inflexibility and fat deposition therefore
the lower HK II activity and PGC expression play a likely reinforce one another in a vicious cycle.
role in this (). For instance, skeletal muscle from
families with a history of TDM has altered meth- Metabolic syndrome and T2DM
ylation status of genes involved in muscle function Metabolic syndrome is defined by visceral obesity and
and insulin and calcium signaling (). Tissue- at least two of the following factors: elevated blood
specific epigenetic regulation may be of particular pressure, raised fasting plasma glucose concentration,
importance for metabolic flexibility because over- elevated triglyceride concentrations, and/or low high-
weight patients with TDM also have hyper- density lipoprotein cholesterol. Together with excess
methylated promoter regions of PGCa and an body fat and physical inactivity, metabolic syndrome
OXPHOS complex I subunit in skeletal muscle is a major risk factor for developing TDM and re-
(). Promoters of many genes that are important lated complications include cardiovascular disease; in-
for pancreatic b-cell survival and function are dif- creased rates of specific cancers, physical, and
ferentially methylated in TDM patients compared cognitive disability (); and is associated with in-
with controls (). Moreover, obese patients have creased risk for TDM and cardiovascular disease,

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 499


REVIEW

among cancer (). Consequently, individuals with can also trigger systemic inflammation. How this is
metabolic syndrome have increased mortality and a regulated at the cellular and molecular level is cur-
shortened lifespan (). rently unknown, but hyperglycemia-induced mito-
The best example of compromised metabolic chondrial ROS production can stimulate inflammation
flexibility in metabolic syndrome is a deteriorated by signaling factors (), such as protein kinase C,
insulin-mediated substrate switching. As such, meta- p MAPK, and c-Jun-N-terminal kinase ().
bolic inflexibility is at the core of the pathophysiology Systemic low-grade inflammation as a trigger of
of insulin resistance (). After a high-fat meal, pa- metabolic inflexibility is best described in the context
tients with metabolic syndrome have higher levels of of obesity and lipid toxicity (). As a result of excess
glycaemia and lower skeletal muscle free fatty acid fatty acid intake, organs that reach the maximum of
uptake compared with healthy individuals. In response their storage capacity and ectopic tissues that accu-
to fasting, skeletal muscle from patients with insulin mulate fatty acids upon overspill can become infil-
resistance are less able to switch to FAO compared trated by immune cells resulting in inflammatory
with healthy individuals (). An increased de- processes. Dysregulated release and storage of fatty

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


pendency on glucose oxidation and decreased reliance acids can lead to an increased release of inflammatory
on FAO in offspring from patients with TDM sug- cytokines such as TNFa and monocyte chemo-
gests that impaired FAO may precede insulin re- attractant protein- and decreased secretion of anti-
sistance (–). inflammatory adipokines such as adiponectin. This
Moreover, studies strongly imply that impaired can result in recruitment of M type macrophages and
mitochondrial function precedes insulin resistance (, T cells. Additionally, B lymphocytes, neutrophils,
). The importance of OXPHOS and its mainte- eosinophils, mast cells, and natural killer cells have all
nance in relation to insulin resistance is underscored been implicated in adipose tissue dysfunction. This
by observations that skeletal muscle mitochondria lipid toxicity can therefore generate signaling in-
from patients with TDM or obesity are unable to termediates that can interfere with local and systemic
increase replication of mtDNA, which encodes es- immune responses, causing a vicious cycle of immune-
sential OXPHOS components, in response to exercise metabolic degradation ().
combined with CR (). Moreover, skeletal muscle Although the mechanism and specific mediators in
mitochondria from insulin-resistant patients have lipid-induced inflammation are not completely un-
lower expression of PGCa and its downstream derstood, the endoplasmic reticulum (ER) is central to
targets, and differ in mass, morphology, and function these responses because this is where both lipid bio-
(). In particular, muscle mitochondria from synthesis and esterification processes as well as
patients with TDM show reduced expression of inflammatory pathways converge. Disrupted lipid
mitofusin-, which regulates mitochondrial outer synthesis in the ER can change ER membrane com-
membrane fusion, and thus mitochondrial dynamics position, leading to ER stress, dysfunction, and ulti-
and quality control (). They also have a lower mately cell death, triggering inflammation ().
maximal oxidative capacity, smaller mitochondria and Lipids are also able to instigate inflammatory processes
reduced NADH oxidase (complex I) activity () through interaction with cell-surface receptors, such as
Interestingly, studies have demonstrated that Toll-like receptor-, and stress kinases in the cyto-
BCAA and associated metabolites are strongly asso- plasm, such as protein kinase R that through down-
ciated with insulin resistance and TDM (). Based stream signaling can induce the expression of genes
on the theory of mitochondrial metabolic gridlock and that mediate inflammation and apoptosis, and pro-
anaplerosis, excessive BCAA metabolites are proposed mote inflammasome activity. Moreover, there is
to clog the b-oxidation machinery, particularly in emerging evidence that lipids engage intracellular
skeletal muscle and liver, and thus contribute to ac- signaling pathways via protein kinase C isoforms that
cumulation of incompletely oxidized intermediates of are related to T-cell activation and LPS responses
fatty acids, particularly in the presence of a high-fat (). It is unlikely, however, that one of such re-
diet. Collectively, under these conditions, such sponses underlies lipotoxicity, but that a combina-
byproducts render glucose superfluous as a substrate tion of factors mediate lipid-associated inflammation
and, combined with the upsurge in ROS, can lead to (, ).
insulin resistance ().
Metabolic flexibility in immune responses
Systemic low-grade inflammation Metabolic flexibility and the accompanied rerouting of
One of the hallmarks of metabolic syndrome is low- metabolic flux are essential for immune function.
grade chronic systemic inflammation (, ). In the Following immune stimulation, naive lymphocytes
case of obesity and insulin resistance, systemic in- that rely on b-oxidation of fatty acids and pyruvate
flammation can trigger and propagate metabolic inflex- oxidation via the TCA cycle become active and en-
ibility. Systemic inflammation and metabolic inflexibility gage in glycolysis and glutaminolysis (). During
can cause a vicious circle because metabolic inflexibility glycolysis, glucose is metabolized to lactate in the

500 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

presence of oxygen, which allows for augmented rates Adipose tissue macrophages that have been activated
of glycolytic flux and regeneration of NAD+. Addi- and rely on glucose are proinflammatory (type M) and
tionally, the switch to glycolysis enables glycolysis and contribute to adipose inflammation and insulin resis-
TCA cycle intermediates to be used as key sources of tance. Conversely, macrophages that rely on fatty
carbon molecules for biosynthesis of nucleotides, acid metabolism secrete anti-inflammatory cytokines
amino acids, and lipids. In this way, glycolysis facili- and thus preserve insulin sensitivity of liver and adi-
tates robust growth, rapid cellular proliferation, and pose tissue (type M) (, ). Proinflammatory
the production of large quantities of effector mole- activation can be achieved by overexpression of GLUT,
cules, ultimately to mount a sufficient immune re- even in the absence of other conventional stimuli, or
sponse. The exact molecular regulation and thus the by decreasing expression of lipid trafficking proteins,
dependency on this metabolic switch differs between such as fatty acid transport protein  (FATP). FATP
specific lymphocyte subsets (). knockout mice fed high-fat diets showed an increased
Reminiscent of the characteristic metabolic net- proinflammatory phenotype and worsened meta-
work adaptation of tumor cells (see “Cancer”) mye- bolic syndrome than mice with normal FATP

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


locytomatosis oncogene (Myc)–regulated glutamine expression. Alternatively, overexpression of FATP
dependency is important for activated lymphocytes decreased substrate switching to glucose and re-
because glutamine fulfills a vital role as anaplerotic duced inflammation (). Thus, macrophage inflam-
substrate via a-ketoglutarate to refill the TCA cycle, matory status is mediated by rerouting metabolic
supporting the robust metabolic switch to glycoly- pathways.
sis and assisting in ATP production and bio- The metabolic switch of glucose metabolism
genesis of citrate and pyruvate (). Therefore, generates ROS that drive the production of in-
activated lymphocytes sustain OXPHOS for ATP flammatory enzymes, cytokines, and chemokines such
production, which enhances cell survival and lifespan as IL-, monocyte chemoattractant protein-, TNF-a,
of lymphocytes and is essential for immune memory and inducible NO synthase (iNOS). iNOS is an im-
(). Memory T cells also use glucose and other portant metabolic regulator of the immune response
fuels to synthesize triglycerides, which are then used in because NO inhibits OXPHOS and oxidative meta-
FAO (). bolism, thus promoting the glycolytic and proin- “Metabolic inflexibility is
correlated to an increased risk
Contrary to the dogma that innate immunity is flammatory phenotype (, ). In this way, of certain types of cancers.”
nonspecific and lacks memory, classic innate immune low-grade systemic inflammation (defined as a two-
cells such as macrophages, natural killer cells, and fold to threefold increase of circulating inflammatory
monocytes can become epigenetically reprogrammed mediators) including the infiltration of immune cells,
by infection or vaccination, which confers nonspecific particularly in metabolic tissues that have reached
protection from secondary infection, a phenomenon their capacity limits, can be driven by metabolic in-
called trained immunity (). Similar to lymphocytes, flexibility (). Recently, inhibition of iNOS in mouse
there is a metabolic basis to “training” these cells. macrophages was shown to dampen the M pheno-
Specifically, in monocytes stimulated with lipopoly- type through reduction of NO-induced OXPHOS
saccharide, a switch from OXPHOS to glycolysis inhibition and assist in the phenotypic and metabolic
underlies these changes, resulting in increased glucose M to M repolarization, suggesting that editing
consumption, lactate production, and NAD+/NADH macrophage (re)polarization is a promising target to
ratio (). The increase in glycolytic metabolism reduce inflammation and promote tissue repair ().
enables a more robust and swift response to intruding An example of metabolic inflexibility and disrupted
pathogens (). Training of immune cells is de- inflammatory assuagement is sepsis. During sepsis, a
pendent on Akt, mTOR, hypoxia-inducible factor a profound change in acute leukocyte metabolism oc-
(HIFa), and, to a lesser extent, SIRTs. Their crucial curs. Metabolic inflexibility drives sepsis-related innate
roles were affirmed by inhibition of Akt by wort- immunoparalysis as the metabolism through glycol-
mannin, mTOR by rapamycin, HIFa by ascorbate, ysis, b-oxidation, and OXPHOS pathways in leuko-
and activation of SIRT by resveratrol, because these cytes is downregulated, resulting in their inability to
compounds blunt trained immunity (). Recently, mount any response whatsoever (). A sudden
however, the notion that a shift from OXPHOS to mitochondrial complex I dysfunction in sepsis (),
glycolysis underlies activation of all immune cells possibly linked to the overproduction of NO and ROS,
upon microbial stimulation was challenged because may be one of the causes of an upstream mito-
pathogen-specific metabolic rewiring was observed in chondrial gridlock, and has been observed to relate to
human monocytes. This pathogen specificity was organ dysfunction (). Moreover, the impaired
proposed to derive from signaling strength, rather than metabolic rate has been associated with reduced levels
qualitative signaling differences between microbial of mtDNA and mRNA expression of OXPHOS
stimuli, and consequently mediates different func- components (, ). In summary, metabolic flex-
tional outputs such as phagocytic capacity (). ibility is not only necessary to mount an adequate

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 501


REVIEW

immune response but also for mitigation of the in- malonyl-CoA concentration, which inhibits CPT-
flammatory process.  and thus FAO ().

Cardiovascular disease Cancer


Cardiac performance is sustained by fatty acid and Metabolic inflexibility is correlated to an increased risk
glucose oxidation, although fatty acids are the pre- of certain types of cancers () and metabolic syn-
ferred substrate in the heart because of the higher drome is associated with a % elevated total cancer
energy yield compared with glucose. This flux is mortality (). Epidemiological evidence shows that
mediated by a high expression of PPARa-regulated through their relation to insulin resistance, excess body
genes encoding key proteins in fatty acid uptake, es- weight, and TDM are associated with an increased
terification, and oxidation (). Under energetically risk of pancreatic, liver, and endometrial cancers,
demanding conditions such as exercise, the heart among others, and of colon cancer in males ().
switches to the oxidation of glucose and lactate (). Excess body weight increases the risk of cancer via
An increase in heart rate increases mitochondrial augmented circulating levels of leptin and decreased

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


calcium concentration (), allowing higher mito- circulating levels of adiponectin (). Physical exer-
chondrial ATP production rates to sustain the cise is inversely related to certain types of cancers
increased energetic load of the heart. Upon exercise- and may reduce the risk of cancer by changing in-
induced sympathetic nervous system stimulation, sulin and IGF and/or sex hormone levels, decreasing
b-adrenergic signaling increases glycolytic flux via body weight and positively modulating immune
cAMP activation of cAMP-dependent protein ki- function ().
nase A, increasing pyruvate production and glu- Diet composition is also correlated to development
cose metabolism. Protein kinase A also activates of certain cancers [reviewed in Potter et al. )].
phosphofructokinase- and PDH, stimulating the High-fat diets for instance have particularly been re-
heart to rapidly oxidize glucose even in the presence of lated to increased risk of colorectal (), pancreatic
fatty acids (). As a consequence, triglyceride ac- (), breast (), lung (), and prostate cancer
cumulation in cardiomyocytes likely leads to abnormal (). Recently, tumor cell metastasis in mice was
lipid signaling, increased ROS production, ER stress, found to be under strong control of the tumor cells’
and mitochondrial dysfunction (). Glucose meta- potential to express the fatty acid receptor CD and
bolism is enhanced in a similar manner through in- metabolize fatty acids. Additionally, dietary fatty acid
sulin and nutrient stress signaling via Akt and AMPK, exposure increased tumor cell expression of CD and
respectively (). increased metastasis in mice (). Besides diets with a
A dependency on glucose (and ketone body) high fatty acid content, diets with a high amount of
metabolism is also observed in myocardial ischemia, animal-derived amino acids also increase the risk of
ventricular hypertrophy, and systemic hypertension cancer in the middle-aged human population ().
(, ), as is mitochondrial dysfunction (, ). Caloric restriction, however, is known to reduce cancer
A recent study in mice demonstrated that mildly occurrence () (see “Prolonged fasting and caloric/
increasing PPARa expression in the progressive phase dietary restriction”). Rodent studies have shown that
of heart failure, when FAO is decreased, maintains CR alters insulin and IGF- levels via a reduction in
myocardial function and energetics, suggesting that phosphatidylinositol -kinase (PIK)/Akt signaling.
modulating substrate utilization may be a promising Moreover, inhibition of mTOR, which is downstream
therapeutic strategy for heart failure (). of PIK/Akt, by prolonged rapamycin treatment,
Obesity can cause metabolic inflexibility of the delays cancer formation in aged mice (). Reducing
heart and alter substrate selection (). High-fat carbohydrate intake reduced tumor growth in mice
diet feeding and consequent insulin insensitivity, (). In rhesus monkeys, CR reduced the incidence of
for instance, are known to cause cardiac metabolic neoplasia by % (). Currently, clinical studies are
inflexibility and reliance on fatty acids for energy under way, but various human studies point toward a
production through PDK inhibition of PDH. reduced incidence of cancer after caloric restriction
Similar to TDM, increased circulating fatty acids ().
only exacerbates the feed-forward dependency
on fatty acid substrates for energy production Tumor growth and metabolic flexibility
through the allosteric inhibition of enzymes in- Our understanding of cancer metabolism has rapidly
volved in glycolysis (). Conversely, the failing advanced in recent years. Most cancer cells show a
heart becomes metabolically inflexible with a remarkable metabolic flexibility, which allows a
decreased capacity to use fatty acids and an in- survival advantage in the face of their energetic
creased dependence on glucose metabolism (). demand and the environmental supply of nutrients.
The switch from fatty acid preference to glucose Mitochondrial-mediated flexibility is central in this
is maintained by increased acetyl-CoA produc- process [reviewed in Vyas et al. ()]. Metabolic
tion from pyruvate and subsequent increases in adaptations that underlie clonal evolution of tumor

502 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

cells to a metastatic phenotype suggest that tumor dampens glucose metabolism through negative feed-
cells do not become metabolically hardwired but back on PDH (, ). A glycolytic switch involving
remain able to reroute metabolism to adapt to differential expression of pyruvate kinase isoforms
their phenotype and the newly acquired envi- and stabilization of HIFa upregulates rate-limiting
ronment (). Indeed, reducing metabolic flex- enzymes within branching pathways of glycolysis,
ibility in cancer cells may lead to potential ensuring that glycolytic intermediates are free to take
treatment options, because metabolic interfer- part in diverse biosynthetic reactions that are essential
ence can come at a substantial cost to oncogenic for increased proliferation (). These alternative
potential (). pathways include the PPP using glucose--phosphate,
Tumors and their environment can be very diverse hexosamine biosynthesis using fructose--phosphate,
and, as such, their metabolism and substrate prefer- phospholipid biosynthesis using dihydroxyacetone
ence is also diverse (). Common traits, however, phosphate, and glycine and serine biosynthesis using
include increased glucose consumption via glycolysis -phosphoglycerate. The PPP is chiefly used for
and enhanced glutamine metabolism to support the NADPH and ribose synthesis to produce nucleotides

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


energetic and anabolic demands of proliferation. (). Of particular significance is serine production
Notwithstanding the diversity of cancers, malignant because it is a major substrate for the folate/one-
cells share a common metabolic trait, namely that they carbon cycle whose metabolites contribute to vari-
can acquire and use nutrients from a predominantly ous cellular biosynthetic and regulatory processes (,
nutrient-poor environment, a modus operandi that ). Lactate, produced from glycolysis, is excreted
emerged as a promising target to battle tumors from the cell or used in biosynthetic reactions such as
(–). Most tumors display one, if not several, aspartate synthesis (). Aspartate is used to support
cancer associated metabolic hallmarks: () deregulated protein and nucleotide synthesis in proliferating cells
uptake of glucose and amino acids, () use of op- and sustains proliferation in the face of OXPHOS
portunistic modes of nutrient acquisition, () use of impairment (). Even TCA cycle intermediates are
glycolysis/TCA cycle intermediates for biosynthe- not solely used to produce NADH for mitochondrial
sis and reduced nicotinamide adenine dinucleotide respiration, but intermediates can also be used to form “Some tumors and embryonic
phosphate (NADPH) production, () increased de- nonessential amino acids and fatty acids, which fa- stem cells are capable of
mand for nitrogen, () alterations in metabolite-driven cilitate protein synthesis, membrane construction and proliferation without an
gene regulation, and () specialized metabolic in- cholesterol synthesis (). exogenous supply of
teractions with the microenvironment (). As a Glutamine is an essential nutrient for cancer glutamine.”
prominent feature of cell activation and proliferation, proliferation and its dependency is also referred to as
tumor cells chiefly require increased amounts of “glutamine addiction” (). Glutamine can either be
glucose and glutamine to survive (). used as an important anaplerotic substrate in the TCA
The metabolic reprogramming that underlies in- cycle, a carbon and nitrogen donor, or for production
creased glucose consumption for use in glycolysis, as of purine and pyrimidine nucleotides that are nec-
opposed to OXPHOS, is known as the Warburg effect. essary for DNA replication (). Intracellular gluta-
In , Otto Warburg discovered that cancer cells mine can also be used as a substrate for the large
metabolize glucose differently than cells of normal neutral amino acid antiporter (LAT). LAT can
tissues: that even in conditions of sufficient oxygen couple glutamine export with import of essential
availability, cancer cells convert glucose into lactate amino acids. Compared with glucose, glutamine
instead of using glucose for OXPHOS (). Warburg tumorigenesis-associated metabolic reprogramming is
hypothesized that cancer cells have mitochondrial only recently becoming clear. In proliferating cells, the
defects and impaired aerobic respiration that forces transcription factor Myc is a major driver of glutamine
them to rely on glycolysis. Today, we understand that utilization and is frequently targeted for upregulation
mitochondrial respiration is not impaired but that in various tumors, despite the abundance of glucose.
cancer cells place emphasis on acquisition and gen- This glutamine addiction is beneficial for the cancer
eration of building blocks necessary for cell division. cell because it maintains mitochondrial TCA cycle
They do so by enhancing biosynthetic metabolism integrity and provides the cell with large quantities of
using glycolytic intermediates (). Increased glucose NADPH needed to meet the demands of cell pro-
uptake and metabolism is very often mediated by the liferation (). Besides Myc-regulated glutamine
oncogenic signaling protein Ras, which activates the addiction, the activity of the Rb tumor suppressor
PIK/Akt pathway. PIK/Akt signaling increases protein family, which negatively regulates glutamine
the expression of GLUT and its translocation to the uptake, is reduced, facilitating increased uptake of
cell surface, and enhances HK and phosphofructoki- glutamine. However, not all tumors are glutamine
nase activity, thus capturing glucose and increasing dependent because some tumors and embryonic
glycolytic flux (). Interestingly, glucose catabolism stem cells are capable of proliferation without an
in cancer cells is partially uncoupled from the TCA exogenous supply of glutamine, because they can
cycle and OXPHOS because increased activity of PDK synthesize it ().

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 503


REVIEW

Interventions to Improve Metabolic Flexibility part from increased volume of the mitochondrial
reticulum and elevated levels of cardiolipin, a lipid that
Lifestyle is necessary for the assembly of OXPHOS complexes
Lifestyle interventions are pertinent for patients with (, , ).
metabolic syndrome. Most patients with TDM are Regular physical exercise positively influences
overweight or obese and do not exercise frequently. insulin-stimulated glucose uptake and mitochondrial
Interventions that reduce body weight by as little as function in skeletal muscle, and, importantly, in pa-
%, however, can reduce obesity-related metabolic tients with TDM (). FAO in skeletal muscle in-
disorders (). In particular, loss of visceral adipose creases during physical exertion independent of body
tissue is related to improved metabolic control of mass index, although regular exercise is likely needed
fasting glucose, cholesterol/high-density lipoprotein to sustain a long-lasting impact on metabolic flexibility
ratio, triglycerides, and diastolic blood pressure (). (). Particularly combined with weight loss, exercise
Lifestyle interventions to reduce body weight pre- training improves insulin sensitivity, mitochondrial
dominantly include exercise training and controlled content, and fasting FAO (). Intriguingly, type II,

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


reduced caloric intake, but their efficacy depends on glycolytic, muscle fiber density is higher in obese and
age, sex, ethnicity, and body weight upon inclusion insulin-resistant patients, although it is unknown
(). As such, caution must be taken when inter- whether these are due to inactivity or impaired glucose
preting results when assessing metabolic flexibility metabolism (). Observations of reduced PGCa,
using suboptimal methods, because individual vari- AMPK (), and mitofusin- () expression in
ability and experimental setup can considerably in- insulin-resistant individuals after exercise might pro-
fluence results. vide mechanistic information as to why mitochondrial
function improves more in healthy volunteers com-
Exercise training pared with patients with TDM and obesity.
Physical inactivity is likely one of the primary causes of Although skeletal muscle is considered the main
metabolic inflexibility (, ); regular habitual site at which the adaptive responses of exercise on
physical exercise has long been known to increase metabolic flexibility occur, it is becoming increasingly
metabolic flexibility (). As such, exercise training evident that other tissues are also involved (see
regimens can be used as an intervention to improve “Physical exercise”). Regular exercise can, for instance,
metabolic flexibility. Generally, exercise can be divided reduce adipose cell size and enhance adipose glucose
into two extremes: aerobic (or endurance-based) and metabolism, resulting in improved insulin sensitivity
anaerobic/glycolytic (resistance-based) activities. Both in both adipose and muscle tissue. Moreover, habitual
promote considerable health benefits such as increased physical exercise remodels subcutaneous adipose tis-
mitochondrial content and improvements in glycemic sue by stimulating browning in mice (). In rats,
control (). For example, a -day endurance ex- chronic endurance exercise induces browning in
ercise training regimen increases FAO in the absence subcutaneous WAT concomitant with increased mo-
of increased mitochondrial content. A high-intensity bilization of energy stores, which were attenuated in
exercise training program, however, showed elevated animals fed high-fat diets. The browning program
citrate synthase and b-hydroxyacyl CoA dehydroge- initiated by exercise training promoted expression of
nase activity after  days and increased levels of mi- PPARa and PPARg, AMPK, PGCa, and adipose
tochondrial complexes after  days (). triglyceride lipase (). Although the exact mech-
AMPK is an important regulator of exercise- anisms underlying this beneficial effect are still under
induced effects on metabolic flexibility (). Acute investigation, exercise training in humans reduced
AMPK activation reduces glycogen and protein syn- intrahepatic lipid content (). In mice, PGCa is
thesis while promoting glucose transport and FAO required for an exercise-induced increase in mito-
(). A higher mitochondrial volume density and chondrial volume density and reduction in intra-
improved mitochondrial quality can be seen as con- hepatic lipid content (). Also, in the human heart,
sequences of chronic activation of AMPK and ex- exercise reduces cardiometabolic risk factors by in-
pression induction of PGCa, myocyte-specific creasing insulin sensitivity, decreasing cardiac lipid
enhancer factor  (MEF), NRF- and NRF-, and content, and improving glucose tolerance ().
nuclear expulsion of histone deacetylase  and  (, In mice, exercise increased cardiac PGCa, NRF,
, ). Contraction-induced calcium uptake acutely and TFAM expression and augmented mitochondrial
increases OXPHOS (, ) and augments glucose volume and number, which were all dependent on
transport and stimulates lipid uptake and oxida- endothelial nitric oxide synthase ().
tion through MEF-induced expression of GLUT Interestingly, exercise training also drives meta-
and PGCa, respectively. Additionally, PGCa is bolic adaptations through epigenetic mechanisms.
expressed upon muscle contraction-induced activation Short-term, high-intensity exercise decreases muscle
of p MAPK (). Endurance exercise increases promoter methylation of genes involved in mito-
the activity of muscle oxidative enzymes and FAO, in chondrial function such as PGCa, TFAM, MEFA,

504 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

and PDK, whereas in patients with TDM, these Recently, attention has arisen for food intake re-
regions usually have higher methylation levels (). stricted to the active time phase (). Alterations in
High-fat feeding in mice induced PGCa hyper- feeding patterns outside the active phase can disrupt
methylation that was transferable to the offspring. the synchrony between the central and peripheral
Maternal exercise, on the other hand, prevented high- clocks and disturb metabolic flexibility (see “Interplay
fat feeding hypermethylation of PGCa and mitigated between circadian rhythm and metabolic flexibility”).
epigenetic associated metabolic dysfunction in the In rodents, food intake outside the active phase causes
offspring (). Indeed, moderate exercise during obesity, whereas time-restricted feeding protects
pregnancy in humans is advised because it reduces the against obesity and insulin resistance (). Time-
risk for obesity during the progeny’s childhood and restricted feeding restores both cycling of metabolic
preadolescence (). Although more research is regulators such as cAMP response element-binding
necessary, it is clear that regular exercise and exercise protein, mTOR and AMPK, and circadian clock gene
training may aid in reversing the pandemic of met- expression ().
abolic disease. Besides dietary interventions to reduce overall

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


energy intake or restrict energy intake to restricted
Dietary interventions periods, specific dietary constituents can induce
Weight loss is an important step in restoring metabolic changes in metabolic flexibility. For instance, carnitine
flexibility and is the most common intervention for is closely associated with the mechanism of metabolic
obesity and obesity-related metabolic comorbidities. flexibility (). Carnitine plays a role in the import of
Generally, energy-restricting diets are aimed at in- long-chain fatty acids into the mitochondria for use in
ducing a state of negative energy balance so that stored b-oxidation and in the mitochondrial efflux of excess
lipids inside adipocytes are used as alternative sub- carbons in the form of acyl-carnitines (). Mecha-
strates (). Energy-restricting dietary regimens have nistically, during substrate opulence or a deficiency of
proven effective in augmenting metabolic flexibility in carnitine or carnitine acetyltransferase, accumulation
animal studies and hold promise for application in of acetyl-CoA in skeletal muscle allosterically inhibits
humans (). During intermittent fasting, subjects go PDH resulting in impaired glucose utilization and
for extended periods with little or no energy intake, whole-body glucose tolerance (). In obese rats, free “Exercise training regimens can
with intervening periods of normal energy intake. carnitine in skeletal muscle is decreased and supple- be used as an intervention to
Intermittent fasting in rodents improved insulin and mentation of L-carnitine restored metabolic flexibility improve metabolic flexibility.”
leptin sensitivity increases ketone body levels and (). In patients with TDM, carnitine acetyl-
reduced adiposity and inflammation (). CR with- transferase expression is severely perturbed and free
out hunger- or disease-related malnutrition in animals carnitine concentrations in diabetic mice are decreased
and humans results in healthier aging through im- compared with controls (, ). Although not yet
proved metabolic health, reduced obesity, and the risk of in clinical practice, L-carnitine supplementation im-
TDM, cancer, and cardiovascular disease (). Ad- proves metabolic flexibility by decreasing plasma
ditionally, CR is considered the most robust interven- glucose and insulin levels and increasing PDH activity
tion to increase longevity in animal model systems and in muscle of insulin-resistant subjects (). Carnitine
ameliorate age-associated disease in primates and metabolism may also be involved in the regulation of
humans (see “Prolonged fasting and caloric/dietary mitochondrial protein acetylation, because acetyl-CoA
restriction” and “Aging”) (). serve as acetyl donors and protein hyperacetylation is
Maintaining an energy-restricting diet, however, is observed in high-fat feeding of mice ().
challenging, because most people have difficulties main-
taining compliance over long periods. Moreover, a recent Pharmaceuticals
study highlighted the paucity of clinical evidence sup- Pharmaceutical approaches to improve metabolic
porting energy-restricting diets in humans (). Although flexibility have been studied in great detail. Most
weight loss is generally achieved in overweight and obese pharmaceutical therapeutics target major players or
subjects, potential adverse effects exist for leaner subjects key nodes in metabolic circuits, many acting on mi-
(). Recommendations such as interruption of sedentary tochondrial function (). The examples that follow
behavior with repeated low-intensity exercise, as well as provide strong support for continuing the search for
“exercise snacking” regimens wherein brief vigorous ex- future pharmacological principles that enhance met-
ercise is followed by meal consumption, may prove more abolic flexibility (Fig. ). For more details on the
effective than the standard recommendation to “eat less mechanisms underlying the beneficial effects of these
and move more” (). Because compliance to energy- treatments on metabolic flexibility, we refer the reader
restricting diets is challenging, interventions that alter to some excellent reviews described in each topic.
meal timing without reducing total caloric intake are ac-
tively pursued. Popular concepts such as increasing or AMPK
decreasing meal frequency, however, lack concrete scientific AMPK is activated by a high intracellular AMP/ATP
evidence supporting their efficacy (). ratio and activates transcription factors, such as the

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 505


REVIEW

FOXO proteins, to increase mitochondrial energy studies in humans suggest that resveratrol may im-
production. Metformin is a biguanide and reduces prove insulin sensitivity and reduce plasma levels
hepatic glucose production and increases insulin of glucose and insulin in patients with TDM and
sensitivity by activating AMPK, although several mimic CR in obese subjects (, ). As such,
AMPK-independent mechanisms have been proposed resveratrol use by humans is particularly beneficial in
[reviewed in Pryor and Cabreiro ()]. Metformin is reversing the early stages of metabolic disorders. Full
one of the first-line treatments of patients with TDM, confirmation of these beneficial effects in humans
but it has also been used to treat patients at risk for by placebo-controlled clinical trials remains rela-
TDM, such as those with metabolic syndrome (). tively limited. Variation in duration and dose of
Resveratrol (,,9-trihydroxystilbene) is a plant- resveratrol may explain the diverse outcomes of these
derived polyphenol that activates AMPK and, via studies ().
SIRT, increases PGCa, PGCa deacetylation, mi- The AMPK agonist -aminoimidazole--carboxamide
tochondrial size and density, and mtDNA content in riboside (AICAR) improves skeletal muscle glucose
skeletal muscle [reviewed in de Ligt et al. ()]. uptake and transport, fatty acid uptake, mitochondrial

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


Resveratrol treatment also increased physical endur- protein content, and insulin sensitivity in mice ().
ance and protected from high-fat diet-induced muscle AICAR also rescued mitochondrial function in mice
accumulation of diacylglyceride and ceramide, and deficient in cytochrome c oxidase and augmented
related mitochondrial dysfunction (). Because exercise endurance in healthy animals in a PGCa-
resveratrol activates mitochondrial biogenesis through dependent manner, even if they were untrained (,
the AMPK-SIRT-PGCa axis, it prompts mito- ). Chronic exposure of AICAR reduced white
chondrial biogenesis, the unfolded protein response, adiposity and increased OXPHOS in rat hearts by
and autophagy machinery that are known to extend increasing PGCa expression and FAO (), as well
longevity in animals (, ). Animal studies have as glucose uptake (). Figure  summarizes the
also demonstrated that resveratrol could stimulate potential role of AMPK activators for metabolic
energy expenditure and protect against a high-fat diet- flexibility.
induced weight gain (), via induction of FAO and
reduced lipogenesis, mediated by activation of the SIRTs
AMPK-SIRT axis (, ). In the context of Through SIRTss, NAD+ provides a direct link be-
insulin-controlled metabolic flexibility, rodent studies tween the cellular energy state of the cell and control
largely show improved insulin sensitivity and glucose of signaling and transcriptional events [reviewed
tolerance in models of obesity, diabetes, and metabolic in Houtkooper et al. ()]. The SIRT family of
dysfunction [reviewed in de Ligt et al. ()]. Clinical NAD+-dependent deacylases, comprising seven mem-
bers that vary in tissue specificity, subcellular localiza-
tion, enzymatic activity, and targets, mediates metabolic
AICAR flexibility in animal models (). SIRT and SIRT have
particularly received attention in this respect. SIRT is
Resveratrol AMPK predominantly found in the nucleus, although it can
also be found in the cytosol. SIRT controls the activity
of transcription factors and cofactors such as p,
MEF, FOXO, and PGCa, which govern mitochon-
NR/NMN/NA/PARPi NAD+ drial biogenesis and activity and lipid and glucose
metabolism (). SIRT, which is localized in the mi-
tochondrial matrix, targets many proteins involved in
SIRT1 metabolic homeostasis, including OXPHOS subunits
(). Acetylation of mitochondrial proteins propagates
Mitochondrial biogenesis and metabolic inflexibility and deacetylation promotes
Ac p improved mitochondrial function metabolic flexibility (, ).
TF targets NAD+ levels can be increased by supplying bio-
synthetic precursors, intermediates, or by inhibiting
NAD+ consuming enzymes [reviewed in Carafa et al.
)]. Nicotinamide riboside, nicotinamide mono-
© 2018 Endocrine Reviews ENDOCRINE SOCIETY
nucleotide, and nicotinic acid are pursued as strategies
to boost metabolic flexibility through increased NAD+
Figure 5. A selection of pharmaceutical compounds that target major players or key nodes in and consequent activation of SIRTs (, ) (Fig. ).
metabolic circuits, such as AMPK and sirtuins. Via altered transcription factors, these compounds
act on mitochondrial function and positively affect metabolic flexibility. See “Pharmaceuticals” for
Boosting NAD+ levels has also been investigated as a
a brief description of the examples mentioned here. Ac, acetyl; NA, nicotinic acid; NMN, strategy to increase lifespan as NAD+ levels decline
nicotinamide mononucleotide; NR, nicotinamide riboside; P, phosphate; PARPi, poly (ADP-ribose) during aging in model systems and humans (). For
polymerase inhibitor; TF, transcription factor. instance, nicotinamide riboside, which is converted to

506 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

nicotinamide mononucleotide via the NAD+ salvage rats by reducing hepatic glucose output, restoring
pathway in eukaryote cells, can boost NAD+ synthesis skeletal muscle glucose uptake, and suppressing free
and can enhance oxidative metabolism and protect fatty acid release by adipocytes (). Finally, thia-
mouse tissues from high-fat diet-induced metabolic zolidinediones, such as rosiglitazone, potently activate
abnormalities () by means of PPARg and anti- PPARg and lower blood glucose levels in patients with
oxidant gene upregulation (). Additionally, nico- TDM. Thiazolidinediones improve skeletal muscle
tinamide riboside treatment in aging mice increased glucose disposal via upregulation of GLUT, decrease
skeletal muscle function by preventing stem cell se- liver glucose output, and, as a primary target, improve
nescence, improved mitochondrial function, and a the lipid-buffering capacity of WAT and as such
higher expression of genes involved in the TCA cycle improve hepatic steatosis (, ). However, thia-
and OXPHOS (). Nicotinamide mononucleotide zolidinediones were rapidly sidelined as therapeutics
conversion to NAD+ activates SIRT and improves because of their potential adverse effects such as an
glucose homeostasis in mice (). Nicotinamide increased risk of myocardial infarction ().
mononucleotide administration to mice also enhances

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


energy metabolism, promotes physical activity, im- mTOR
proves lipid profiles, and ameliorates age-related mTOR is a central regulator of growth and meta-
pathophysiology (). Alternatively, pharmacologi- bolism in all eukaryotes and its activity depends on the
cal inhibition of the NAD+ consuming enzyme poly cells energy and nutrient levels [reviewed in Laplante
(ADP-ribose) polymerase in mice activates SIRT and and Sabatini ()]. In general, anabolic processes such
increases mitochondrial respiratory capacity leading to as protein and lipid synthesis, and protein turnover are
augmented fitness and protection from diet-induced controlled by mTOR complex  (mTORC). As such,
obesity (). Moreover, poly (ADP-ribose) poly- mTORC controls the balance between anabolism and
merase inhibition rescued mitochondrial respiration catabolism in response to changing environments.
defects and increased FAO in myotubes from obese mTORC facilitates cellular growth by a shift in
patients by augmenting mitochondrial function (). glucose metabolism from OXPHOS to glycolysis,
likely through an mTORC-mediated increase in the
PPARs translation of the transcription factor HIFa and an
PPARs are lipid sensors that transcriptionally mod- increased flux through the PPP, which uses carbons
ulate metabolic programs in response to nutrition and from glucose to generate NADPH and other in-
are interesting drug targets to improve metabolic termediary metabolites needed for proliferation and
flexibility [reviewed in Bugge and Holst ()]. There growth (). A reduction in cellular energy charge,
are three PPAR isotypes (PARa, PPARb/d, and such as during glucose deprivation or fasting, inhibits
PPARg), which differ in ligand affinity and tissue mTORC by AMPK-dependent as well as AMPK-
distribution [reviewed in Gross et al. ()]. Fibrates independent pathways, and is required for the gen-
activate PPARa and are commonly used for treatment eration of ketone bodies in the liver (). Similarly,
of hyperlipidemia. Of the fibrate drug class, fenofibrate low levels of amino acids (particularly arginine and
and bezafibrate have recently gained interest as in- leucine) and interaction with AMPK inhibit mTORC
terventions to improve metabolic flexibility, in par- activity. In these ways, energy-consuming processes
ticular for the treatment of insulin resistance. such as mRNA translation are inhibited during pe-
Fenofibrate improves FAO in primary human skeletal riods of low energy.
muscle cell cultures from obese and insulin resistant mTORC instead mainly controls cellular pro-
subjects. In vitro and in animal models, PPARa ac- liferation and survival, and has more limited effects on
tivation increased expression of PDK and CPT (). metabolic flexibility per se. What should be noted,
In insulin-deficient mice, bezafibrate improves im- however, is that mTORC directly activates Akt
paired glucose metabolism by augmenting hepatic downstream of insulin/PIK signaling; as such,
mitochondrial performance [reviewed in Komen and mTORC inhibition disrupts the physiological re-
Thorburn ()], suppressing hepatic inflammatory sponse to insulin.
pathways, and improving insulin sensitivity (). Tissue-specific mTOR signaling can have profound
Similarly, synthetic PPARb/d agonists such as effects on whole-body metabolism. For instance, in-
GW or L- reduce adiposity improve creased mTOR signaling in either adipose tissue,
insulin sensitivity and hyperlipidemia in obese pa- skeletal muscle, or the liver, negatively affects systemic
tients, through their ability to augment mitochondrial glucose and insulin homeostasis (). Indeed, mTOR
biogenesis and activity (). PPARb/d agonists also signaling is dysregulated in cancer, TDM and obesity,
enhance energy expenditure by increasing fatty acid and is therefore actively perused as a promising drug
catabolism in adipose tissue and skeletal muscle (). target [reviewed in Laplante and Sabatini ( and
Another PPAR agonist is tesaglitazar, which binds and Albert and Hall ()].
activates PPARa and PPARg. Tesaglitazar increases One compound that particularly received ample
whole-body glucose metabolic insulin action in obese attention as an inhibitor of mTOR is rapamycin.

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 507


REVIEW

Rapamycin is a natural bacterial product that acutely with citrate synthase rates, and pyruvate carboxyl-
inhibits mTORC and, after prolonged treatment, also ase rates showed the highest fold change in response
inhibits mTORC in some cell types. As such, rapa- to glucose stimulation, confirming these enzymes
mycin has both positive and negative effects on as important nodes in glucose metabolism ().
metabolism depending on dose and duration. Short- Labeling with [,-C]-glucose and [U-C]-glutamine
term rapamycin treatment in mice instigates glucose revealed considerable differences in metabolic pathway
intolerance, insulin resistance, and immunodeficiency. fluxes between the exponential growth and stationary
Prolonged treatment improves metabolic profiles, phase of Chinese hamster ovary cells. Specifically, gly-
increases oxygen consumption and ketogenesis, and colytic flux and lactate production was high and PPP
markedly enhances insulin sensitivity [reviewed in Li flux low in the exponential growth phase, which was
et al. ()]. Additionally, rapamycin has earned great collectively reversed in the stationary phase concomitant
interest over the years to prolong lifespan (, ), with lactate consumption and reduced TCA cycle flux.
even when administered to aged animals (). Interestingly fatty acid biosynthesis remained high in
Rapamycin, however, has poor solubility and phar- both growth conditions (). Recent advances in

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


macokinetics, which led to the production of rapa- metabolite extraction from isolated mitochondria
mycin analogs (rapalogs), which are currently used in demonstrate that metabolic flux models can be built
some cancer therapies (). with increasing precision (). Translation of the
Manipulating mTOR signaling is, however, very collected data, however, will likely be the most chal-
complex, because many positive and negative feedback lenging task in the future (). Finally, metabolic
loops exist, reducing efficacy of mTOR targeting flexibility modeling can give us insight into the evo-
compounds. Moreover, because mTOR controls many lution of metabolic pathways (), demonstrating that
vital cellular processes, its complete inhibition by high genes encoding enzymes with high connectivity and
doses of rapamycin may negatively affect the main- high metabolic flux have a greater chance of conser-
tenance of tissue functions and provoke adverse events vation () and are the most likely sites of metabolic
(). Our molecular understanding of mTOR sig- regulation ().
naling is not yet comprehensive, and future research
into rapalogs with fewer pleiotropic and adverse effects How to understand metabolic flexibility from
is necessary and will likely lead to therapeutics to ease whole body/animal physiology
metabolic disease. The continued use of model organisms to understand
metabolic flexibility regulation is vital. Over the past
few decades, model organisms such as primates, ro-
Future Perspectives dents, flies, nematodes, and yeast have proven their
effectiveness in elucidating pathways and regulatory
Metabolic flexibility can be studied at various levels, components involved in metabolic flexibility. In
from cell to whole-body studies. However, metabolic humans, mixed meal tests, glucose clamping, isotope
flexibility is likely best understood as a system of administration, and indirect calorimetry are regularly
interacting components (). As such, metabolic used to assess metabolic flexibility in intervention
flexibility should be measured by the ability to adapt studies. Scientists have limited their research on a
to conditions of temporary stress, such as physical small number of animal models to further understand
exercise, infections, or mental stress, in a healthy metabolic flexibility and its regulation. Some examples
manner (). are discussed in the following sections.
The round worm Caenorhabditis elegans, for in-
How to understand metabolic flexibility from stance, is used to investigate metabolic pathways and
cellular studies has been instrumental in elucidating key genetic
To understand metabolism at the system level, cellular regulators of metabolism in relation to lifespan (,
metabolic programming, such as biochemical path- , ). Inbred lines have been generated for
ways and networks thereof, should be well annotated C. elegans () and, recently, metabolic network
first. Metabolic flux analysis, flux balance analysis, and models became available (, ). With relative ease,
metabolic pathway analysis are among the most dietary and genetic influences can be simultaneously
popular tools in stoichiometric network analysis and investigated in C. elegans (). Moreover, important
are used extensively to study (cancer) metabolism advances have been made in quantifying the metabolic
(, ). In general, intracellular fluxes are measured spectrum in C. elegans ().
using isotope labeling of substrates, after which iso- Work in mice and rats has particularly forwarded
topomer distribution in metabolites can be quantified the field. For example, mice have been the driv-
(). For instance, mass-isotopomeric flux analysis of ing force behind research into CR (), and the use
glycolysis and the TCA cycle in a rat-derived b-cell of recombinant inbred lines such as the BXD
line using [U-C]-D-glucose has demonstrated that mouse strains now offer insights into the role of ge-
insulin secretion and oxygen consumption correlate netic background effects and can help translate the

508 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

differences between gene variants into meaningful data released in  and included  cell type–specific
for human populations and disease. The natural models (). In , Recon was updated as Recon
variation in gene expression that is present in the . and includes a total of  metabolites, 
human population is also recapitulated in these reactions, and  associated genes. Among other
models and therefore more honestly reflect genotype- topics, Recon is used to predict biomarkers for inborn
phenotype associations than artificial loss-of-function errors of metabolism, to identify drug targets and
situations, such as gene knockouts or knock-down possible side effects, and to study cancer metabolism
experiments (). For instance, % of the BXD and the interactions between microorganisms and
strains, which were generated by crossing CBL/J their host (). Although the models are not yet all-
and DBA/J, have a hypersecretion of insulin in re- inclusive, they can yield meaningful insights into
sponse to glucose (). Moreover, OXPHOS gene the regulation of metabolic flexibility. For instance,
expression and supercomplex assembly, and PPAR genome-scale models of cancer metabolism can help
and cholesterol biosynthesis pathways, which are us comprehend the complexity of human cancer
known to influence metabolic diseases, are highly heterogeneity in relation to substrate preference and

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


variable in the BXD population (). therefore expedite the discovery of putative anticancer
Although rats and mice share a high metabolic targets (). Genome-scale metabolic models can also
homogeneity with humans, they differ in many as- be applied to investigate genetic variation in genotype-
pects. For instance, vitamin C and bile acid synthesis phenotype interactions and as such can be used to
pathways are different in rats and humans (). identify metabolic biomarkers and discover treatments
Additionally, the discrepancies between sex, or strain based on genetic markers ().
variations in sensing and handling of substrates, may Recently, a compartmentalized mathematical model
convolute the interpretation of data derived from of human metabolism, which was not based on Recon ,
animal model systems (). Differences in experi- was implemented to study the effects of insulin re-
mental approaches and the techniques used to mea- sistance on individual tissues (). The model describes
sure metabolic flexibility may influence outcomes the transport, storage, and utilization of glucose and fats
(). Methodological differences can severely impact in the human body and suggested that insulin resistance
interpretation of study data (), advocating for a in one tissue created a knock-on effect in other tissues “The continued use of model
organisms to understand
gold-standard application of available methods and that tries to compensate for the reduced flexibility. The metabolic flexibility regulation
technology. For instance, rodent models are sus- model also simulated that insulin resistance causes fatty is vital.”
ceptible to temperature and as such can show di- liver regardless of the site of insulin resistance in the
vergent metabolism depending on their environment. body, and that slightly reduced skeletal muscle meta-
Moreover, cage-housing density of mice can strongly bolic flexibility is caused by insulin resistance of indi-
influence glucose metabolism (). To standardize vidual tissues, although the strongest effect on muscle
and validate phenotyping in mice, standard operating metabolic flexibility is observed when the whole body is
procedures were developed by the Eumorphia pro- insulin resistant (). At the organ level, flux balance
gram to ensure that test outcomes are comparable computational modeling of muscle metabolism during
between different laboratories (). Finally, many the fasted to fed state transition showed that key
different mechanisms influence metabolism, making metabolic phenotypes characteristic of human insulin
the identification of causal mechanisms of meta- resistance can be recapitulated by decreasing flux
bolic inflexibility challenging. Therefore, high-quality through the PDH complex (). Notably, these ob-
measurements of multiple complementing layers servations corroborate knowledge that competition
of data including genomics, transcriptomics, pro- between fatty acids and glucose occurs at the level of the
teomics, metabolomics, and phenomics are needed. pyruvate dehydrogenase complex (). Additionally,
Such a multilayered cross-omics approach can help maximum glycogen accumulation and homeostatic
elucidate complex traits associated with metabolic energy demand were found among the most important
flexibility (). parameters regulating muscle cell metabolism (),
which supports previous findings that improving gly-
Systems biology, mathematical modeling, and cogen synthesis strongly influences exercise-induced
stoichiometric network analysis skeletal muscle insulin sensitivity (). Collectively,
Through development of mathematical and compu- these advances enable us to understand how bio-
tational models, systems biology can identify mean- chemical networks behave under different conditions
ingful components and interactions, especially at and help us identify important actors that can be tar-
critical nodes in the network. As such, systems bi- geted to eliminate the undesired effects of environ-
ological approaches can represent, analyze, and predict mental or inherent factors ().
the behavior of the whole system (). A good ex-
ample of this is the global human metabolic net- Medical horizons and translational aspects
work, Recon , which was released in  (). The wealth of available preclinical data suggests
A community-driven reconstruction, Recon , was that metabolic flexibility is the cornerstone of many

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 509


REVIEW

(patho)biological processes. It is this wealth, however, underpinnings (). However, because of the
that hampers translation to applicable human in- complexity of metabolic flexibility, many ends
terventions. Metabolic flexibility occurs at the sub- remain untied and solid translational steps are
strate, cell, tissue, organ, and organism level and is often missing. The use of isotope labeling to
affected by disease, diet, body composition, and many determine metabolic flux and substrate prefer-
other (epigenetic) factors. ence is a common practice (). Flux mea-
To gain further insights into our understanding of surements can aptly portray the dynamics of
metabolic flexibility, a coherent, multidisciplinary metabolism, whereas protein and gene expression
approach is warranted. Such an approach includes analyses are typically static snapshots in time. A
specialists in phenomics, metabolomics, proteomics, biochemical revival of flux metabolism research
genomics, enzymology, molecular biology, bio- will surely reveal insights into metabolic
informatics, systems biology, clinical genetics, nutri- flexibility.
tion, whole-cell systems, model systems, and in vivo Unveiling the key denominators of metabolic
human studies. The latter are particularly required to flexibility as targets for intervention strategies is par-

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


study the metabolically flexible processes to prevent amount to stop the rise of metabolic disease. Besides
merely superficial and associative data. However, abating metabolic inflexibility such strategies may even
recent studies show that personalized analysis and prolong health span and lifespan. To advance trans-
tailoring of metabolism open up therapeutic possi- lational research from bench to bedside, clinical
bilities (–). Supporting this collaboration, trials need to be performed that are well-controlled
current diagnosis and research tools need to be and use state-of-the-art methods to measure metabolic
improved, for instance in the field of metabolomics, flexibility.
in which faster and more accurate analyses are With this review, we have summarized the
needed. Major advances can be made by combining current status of metabolic flexibility research and
standalone analyses into combined methods and realize that, because of the vast amount of literature
applying targeted metabolite platforms that cover and the complexity of this research field, it is not
emerging groups of metabolites, with a primary comprehensive. Nonetheless, it is evident that
focus on human translation. metabolic flexibility can be placed in the broad
context of health and disease and a deeper un-
derstanding of its intricacies will significantly affect
Conclusion health care. As a final note, the situation portrayed
here does not necessarily reflect that of each in-
Metabolic flexibility is likely best understood as a dividual. Because of the genetic and epigenetic
system of interacting components to manage energy disparity of humans and the enormous varieties in
resources and requirements in health and disease lifestyle, it is not unthinkable that each person
(). Together with the endocrine system, mito- fashions a unique way to maintain energy ho-
chondria orchestrate the metabolic reprogramming meostasis. In light of the rapid developments in the
necessary to sustain cell function in both healthy and field of nutrigenomics and personalized medicine,
pathological conditions. future research will likely focus on the union be-
Metabolic flexibility has a broad scope that can tween metabolic flexibility and personalized
be daunting; nevertheless great progress has been medicine. Clearly, there is still much work to be
made over the past few decades to understand its done, yet interesting times lie ahead.

References
1. Olson KA, Schell JC, Rutter J. Pyruvate and metabolic publications/grd-2016/en/. Accessed 24 April 8. Kelley DE, Goodpaster B, Wing RR, Simoneau JA.
flexibility: illuminating a path toward selective cancer 2018.doi:978 92 4 156525 7. Skeletal muscle fatty acid metabolism in association
therapies. Trends Biochem Sci. 2016;41(3):219–230. 6. International Food Policy Research Institute. with insulin resistance, obesity, and weight loss. Am
2. Speakman JR. Evolutionary perspectives on the Global Nutrition report 2016: from promise to J Physiol. 1999;277(6 Pt 1):E1130–E1141.
obesity epidemic: adaptive, maladaptive, and neu- impact ending malnutrition by 2030. Available at: 9. Kelley DE, Mandarino LJ. Fuel selection in human
tral viewpoints. Annu Rev Nutr. 2013;33(1):289–317. www.ifpri.org/publication/global-nutrition-report- skeletal muscle in insulin resistance: a reexamina-
3. López-Otı́n C, Galluzzi L, Freije JMP, Madeo F, 2016-promise-impact-ending-malnutrition-2030. tion. Diabetes. 2000;49(5):677–683.
Kroemer G. Metabolic control of longevity. Cell. Accessed 24 April 2018. doi:10.2499/9780896295841. 10. Battaglia GM, Zheng D, Hickner RC, Houmard JA.
2016;166(4):802–821. 7. Saltin B, Gollnick PD. Skeletal muscle adaptability: Effect of exercise training on metabolic flexibility
4. Muoio DM. Metabolic inflexibility: when mito- significance for metabolism and performance. In: in response to a high-fat diet in obese individuals.
chondrial indecision leads to metabolic gridlock. Peachy L, Adrian R, Geiger S, eds. Handbook of Am J Physiol Endocrinol Metab. 2012;303(12):
Cell. 2014;159(6):1253–1262. Physiology: section 10: Skeletal Muscle. Bethesda, E1440–E1445.
5. World Health Organization. Global report on MD: American Physiological Society; 1983: 11. Obre E, Rossignol R. Emerging concepts in bio-
diabetes. Available at: www.who.int/diabetes/ 555–631. energetics and cancer research: metabolic flexibility,

510 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

coupling, symbiosis, switch, oxidative tumors, 31. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, 49. Lira VA, Benton CR, Yan Z, Bonen A. PGC-1alpha
metabolic remodeling, signaling and bio- Smith A, Murray AJ, Stubbs B, West J, McLure SW, regulation by exercise training and its influences on
energetic therapy. Int J Biochem Cell Biol. 2015; King MT, Dodd MS, Holloway C, Neubauer S, muscle function and insulin sensitivity. Am J Physiol
59:167–181. Drawer S, Veech RL, Griffin JL, Clarke K. Nutritional Endocrinol Metab. 2010;299(2):E145–E161.
12. Vitkup D, Kharchenko P, Wagner A. Influence of ketosis alters fuel preference and thereby endur- 50. Rowe GC, Patten IS, Zsengeller ZK, El-Khoury R,
metabolic network structure and function on en- ance performance in athletes. Cell Metab. 2016; Okutsu M, Bampoh S, Koulisis N, Farrell C,
zyme evolution. Genome Biol. 2006;7(5):R39. 24(2):256–268. Hirshman MF, Yan Z, Goodyear LJ, Rustin P, Arany
13. Carstens MT, Goedecke JH, Dugas L, Evans J, Kroff J, 32. Wüst RCI, Visser G, Wanders RJA, Houtkooper RH. Z. Disconnecting mitochondrial content from re-
Levitt NS, Lambert EV. Fasting substrate oxidation Ketones and inborn errors of metabolism: old spiratory chain capacity in PGC-1-deficient skeletal
in relation to habitual dietary fat intake and insulin friends revisited. J Inherit Metab Dis. 2017;40(1):3–4. muscle. Cell Reports. 2013;3(5):1449–1456.
resistance in non-diabetic women: a case for 33. Shimomura Y, Honda T, Shiraki M, Murakami T, 51. Perry CGR, Lally J, Holloway GP, Heigenhauser GJF,
metabolic flexibility? Nutr Metab (Lond). 2013;10(1): Sato J, Kobayashi H, Mawatari K, Obayashi M, Harris Bonen A, Spriet LL. Repeated transient mRNA
8. RA. Branched-chain amino acid catabolism in ex- bursts precede increases in transcriptional and
14. van Ommen B, Keijer J, Heil SG, Kaput J. Challenging ercise and liver disease. J Nutr. 2006;136(1, Suppl): mitochondrial proteins during training in hu-
homeostasis to define biomarkers for nutrition 250S–253S. man skeletal muscle. J Physiol. 2010;588(Pt 23):
related health. Mol Nutr Food Res. 2009;53(7): 34. Harris RA, Joshi M, Jeoung NH, Obayashi M. 4795–4810.
795–804. Overview of the molecular and biochemical basis of 52. Mansueto G, Armani A, Viscomi C, D’Orsi L, De
branched-chain amino acid catabolism. J Nutr. Cegli R, Polishchuk EV, Lamperti C, Di Meo I,

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


15. Benedict FG. An experiment on a fasting man.
Science. 1912;35(909):865. 2005;135(6 suppl):1527S–1530S. Romanello V, Marchet S, Saha PK, Zong H, Blaauw B,
16. Soeters MR, Soeters PB, Schooneman MG, Houten 35. Brosnan JT, Brosnan ME. Branched-chain amino Solagna F, Tezze C, Grumati P, Bonaldo P, Pessin JE,
SM, Romijn JA. Adaptive reciprocity of lipid and acids: enzyme and substrate regulation. J Nutr. Zeviani M, Sandri M, Ballabio A. Transcription
glucose metabolism in human short-term starva- 2006;136(1, Suppl):207S–211S. factor EB controls metabolic flexibility during ex-
tion. Am J Physiol Endocrinol Metab. 2012;303(12): 36. Kapahi P, Kaeberlein M, Hansen M. Dietary re- ercise. Cell Metab. 2017;25(1):182–196.
E1397–E1407. striction and lifespan: lessons from invertebrate 53. Stager M, Swanson DL, Cheviron ZA. Regulatory
17. Ahmed M, Gannon MC, Nuttall FQ. Postprandial models. Ageing Res Rev. 2017;39:3–14. mechanisms of metabolic flexibility in the dark-
plasma glucose, insulin, glucagon and triglyceride 37. Madeo F, Zimmermann A, Maiuri MC, Kroemer G. eyed junco (Junco hyemalis). J Exp Biol. 2015;218(Pt
responses to a standard diet in normal subjects. Essential role for autophagy in life span extension. 5):767–777.
Diabetologia. 1976;12(1):61–67. J Clin Invest. 2015;125(1):85–93. 54. Anderson SN, Podrabsky JE. The effects of hypoxia
18. Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, 38. Fontana L, Partridge L. Promoting health and and temperature on metabolic aspects of embry-
longevity through diet: from model organisms to onic development in the annual killifish Austro-
Raptis SA. Insulin effects in muscle and adipose
humans. Cell. 2015;161(1):106–118. fundulus limnaeus. J Comp Physiol B. 2014;184(3):
tissue. Diabetes Res Clin Pract. 2011;93(Suppl 1):
39. Kenyon CJ. The genetics of ageing [published 355–370.
S52–S59.
correction appears in Nature. 2010;467:622]. Nature. 55. Wu C-W, Storey KB. Life in the cold: links between
19. Frayn KN. Adipose tissue as a buffer for daily lipid
2010;464(7288):504–512. mammalian hibernation and longevity. Biomol
flux. Diabetologia. 2002;45(9):1201–1210.
40. Cantó C, Jiang LQ, Deshmukh AS, Mataki C, Coste Concepts. 2016;7(1):41–52.
20. Longo VD, Mattson MP. Fasting: molecular
A, Lagouge M, Zierath JR, Auwerx J. In- 56. van Marken Lichtenbelt WD, Vanhommerig JW,
mechanisms and clinical applications. Cell Metab.
terdependence of AMPK and SIRT1 for metabolic Smulders NM, Drossaerts JMAFL, Kemerink GJ,
2014;19(2):181–192.
adaptation to fasting and exercise in skeletal Bouvy ND, Schrauwen P, Teule GJJ. Cold-activated
21. Ramnanan CJ, Edgerton DS, Kraft G, Cherrington
muscle. Cell Metab. 2010;11(3):213–219. brown adipose tissue in healthy men. N Engl J Med.
AD. Physiologic action of glucagon on liver glucose
41. Schrauwen-Hinderling VB, Kooi ME, Schrauwen P. 2009;360(15):1500–1508.
metabolism. Diabetes Obes Metab. 2011;13(Suppl
Mitochondrial function and diabetes: conse- 57. Cypess AM, Lehman S, Williams G, Tal I, Rodman D,
1):118–125.
quences for skeletal and cardiac muscle meta- Goldfine AB, Kuo FC, Palmer EL, Tseng Y-H, Doria A,
22. McGarry JD, Foster DW. Regulation of hepatic fatty
bolism. Antioxid Redox Signal. 2016;24(1):39–51. Kolodny GM, Kahn CR. Identification and impor-
acid oxidation and ketone body production. Annu 42. Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as tance of brown adipose tissue in adult humans.
Rev Biochem. 1980;49(1):395–420. regulators of metabolism and healthspan. Nat Rev N Engl J Med. 2009;360(15):1509–1517.
23. Marinou K, Adiels M, Hodson L, Frayn KN, Karpe F, Mol Cell Biol. 2012;13(4):225–238. 58. Virtanen KA, Lidell ME, Orava J, Heglind M,
Fielding BA. Young women partition fatty acids 43. Devarshi PP, McNabney SM, Henagan TM. Skeletal Westergren R, Niemi T, Taittonen M, Laine J, Savisto
towards ketone body production rather than muscle nucleo-mitochondrial crosstalk in obesity N-J, Enerbäck S, Nuutila P. Functional brown adi-
VLDL-TAG synthesis, compared with young men. and type 2 diabetes. Int J Mol Sci. 2017;18(4):831. pose tissue in healthy adults. N Engl J Med. 2009;
Br J Nutr. 2011;105(6):857–865. 44. Brøns C, Grunnet LG. Mechanisms in endocrinol- 360(15):1518–1525.
24. Paniagua JA. Nutrition, insulin resistance and dys- ogy: skeletal muscle lipotoxicity in insulin resistance 59. Lee P, Swarbrick MM, Ho KKY. Brown adipose tissue
functional adipose tissue determine the different and type 2 diabetes: a causal mechanism or an in adult humans: a metabolic renaissance. Endocr
components of metabolic syndrome. World J Di- innocent bystander? Eur J Endocrinol. 2017;176(2): Rev. 2013;34(3):413–438.
abetes. 2016;7(19):483–514. R67–R78. 60. Barquissau V, Beuzelin D, Pisani DF, Beranger GE,
25. Hue L, Taegtmeyer H. The Randle cycle revisited: 45. Picard M, Hepple RT, Burelle Y. Mitochondrial Mairal A, Montagner A, Roussel B, Tavernier G,
a new head for an old hat. Am J Physiol Endocrinol functional specialization in glycolytic and oxidative Marques MA, Moro C, Guillou H, Amri EZ, Langin
Metab. 2009;297(3):E578–E591. muscle fibers: tailoring the organelle for optimal D. White-to-brite conversion in human adipocytes
26. Sugden MC, Holness MJ. Mechanisms underlying function. Am J Physiol Cell Physiol. 2012;302(4): promotes metabolic reprogramming towards fatty
regulation of the expression and activities of the C629–C641. acid anabolic and catabolic pathways. Mol Metab.
mammalian pyruvate dehydrogenase kinases. Arch 46. Wüst RCI, Jaspers RT, van Heijst AF, Hopman MTE, 2016;5(5):352–365.
Physiol Biochem. 2006;112(3):139–149. Hoofd LJC, van der Laarse WJ, Degens H. Region- 61. Hao Q, Yadav R, Basse AL, Petersen S, Sonne SB,
27. Spriet LL. New insights into the interaction of specific adaptations in determinants of rat skeletal Rasmussen S, Zhu Q, Lu Z, Wang J, Audouze K,
carbohydrate and fat metabolism during exercise. muscle oxygenation to chronic hypoxia. Am J Gupta R, Madsen L, Kristiansen K, Hansen JB.
Sports Med. 2014;44(suppl 1):S87–S96. Physiol Heart Circ Physiol. 2009;297(1):H364–H374. Transcriptome profiling of brown adipose tissue
28. Owen OE, Morgan AP, Kemp HG, Sullivan JM, 47. Adeva-Andany M, López-Ojén M, Funcasta- during cold exposure reveals extensive regulation of
Herrera MG, Cahill GF Jr. Brain metabolism during Calderón R, Ameneiros-Rodrı́guez E, Donapetry- glucose metabolism. Am J Physiol Endocrinol Metab.
fasting. J Clin Invest. 1967;46(10):1589–1595. Garcı́a C, Vila-Altesor M, Rodrı́guez-Seijas J. 2015;308(5):E380–E392.
29. Manninen AH. Metabolic effects of the very-low- Comprehensive review on lactate metabolism in 62. Mottillo EP, Desjardins EM, Crane JD, Smith BK,
carbohydrate diets: misunderstood “villains” of human health. Mitochondrion. 2014;17:76–100. Green AE, Ducommun S, Henriksen TI, Rebalka IA,
human metabolism. J Int Soc Sports Nutr. 2004;1(2): 48. Jager S, Handschin C, St-Pierre J, Spiegelman BM. Razi A, Sakamoto K, Scheele C, Kemp BE, Hawke TJ,
7–11. AMP-activated protein kinase (AMPK) action in Ortega J, Granneman JG, Steinberg GR. Lack of
30. Akram M. A focused review of the role of ketone skeletal muscle via direct phosphorylation of PGC- adipocyte AMPK exacerbates insulin resistance and
bodies in health and disease. J Med Food. 2013; 1alpha. Proc Natl Acad Sci USA. 2007;104(29): hepatic steatosis through brown and beige adipose
16(11):965–967. 12017–12022. tissue function. Cell Metab. 2016;24(1):118–129.

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 511


REVIEW

63. Hanssen MJW, Hoeks J, Brans B, van der Lans AAJJ, 81. Lee D, Hwang W, Artan M, Jeong D-E, Lee S-J. Effects 100. Gao AW, Cantó C, Houtkooper RH. Mitochondrial
Schaart G, van den Driessche JJ, Jörgensen JA, of nutritional components on aging. Aging Cell. response to nutrient availability and its role in
Boekschoten MV, Hesselink MK, Havekes B, Kersten 2015;14(1):8–16. metabolic disease. EMBO Mol Med. 2014;6(5):
S, Mottaghy FM, van Marken Lichtenbelt WD, 82. Houtkooper RH, Williams RW, Auwerx J. Metabolic 580–589.
Schrauwen P. Short-term cold acclimation im- networks of longevity. Cell. 2010;142(1):9–14. 101. van de Weijer T, Sparks LM, Phielix E, Meex RC, van
proves insulin sensitivity in patients with type 2 83. Stull AJ, Galgani JE, Johnson WD, Cefalu WT. The Herpen NA, Hesselink MKC, Schrauwen P,
diabetes mellitus. Nat Med. 2015;21(8):863–865. contribution of race and diabetes status to met- Schrauwen-Hinderling VB. Relationships between
64. Milev NB, Reddy AB. Circadian redox oscillations abolic flexibility in humans. Metabolism. 2010;59(9): mitochondrial function and metabolic flexibility in
and metabolism. Trends Endocrinol Metab. 2015; 1358–1364. type 2 diabetes mellitus. PLoS One. 2013;8(2):e51648.
26(8):430–437. 84. Wiley CDD, Campisi J. From ancient pathways to 102. Caton PW, Nayuni NK, Khan NQ, Wood EG, Corder
65. Sahar S, Sassone-Corsi P. Regulation of metabolism: aging cells—connecting metabolism and cellular R. Fructose induces gluconeogenesis and lipo-
the circadian clock dictates the time. Trends senescence. Cell Metab. 2016;23(6):1013–1021. genesis through a SIRT1-dependent mechanism.
Endocrinol Metab. 2012;23(1):1–8. 85. Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, J Endocrinol. 2011;208(3):273–283.
66. Chatham JC, Young ME. Regulation of myocardial Freund A, Shirakawa K, Lim HW, Davis SS, 103. Saltiel AR, Kahn CR. Insulin signalling and the
metabolism by the cardiomyocyte circadian clock. Ramanathan A, Gerencser AA, Verdin E, Campisi J. regulation of glucose and lipid metabolism. Nature.
Mitochondrial dysfunction induces senescence 2001;414(6865):799–806.
J Mol Cell Cardiol. 2013;55(1):139–146.
with a distinct secretory phenotype. Cell Metab. 104. Wiederkehr A, Wollheim CB. Mitochondrial signals
67. Aviram R, Manella G, Kopelman N, Neufeld-Cohen
2016;23(2):303–314. drive insulin secretion in the pancreatic b-cell. Mol

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


A, Zwighaft Z, Elimelech M, Adamovich Y, Golik M,
86. López-Otı́n C, Blasco MA, Partridge L, Serrano M, Cell Endocrinol. 2012;353(1-2):128–137.
Wang C, Han X, Asher G. Lipidomics analyses reveal
Kroemer G. The hallmarks of aging. Cell. 2013; 105. Koh EH, Park J-Y, Park H-S, Jeon MJ, Ryu JW, Kim M,
temporal and spatial lipid organization and uncover
153(6):1194–1217. Kim SY, Kim M-S, Kim S-W, Park IS, Youn JH, Lee K-
daily oscillations in intracellular organelles. Mol Cell. 87. Fadini GP, Ceolotto G, Pagnin E, de Kreutzenberg S, U. Essential role of mitochondrial function in adi-
2016;62(4):636–648. Avogaro A. At the crossroads of longevity and ponectin synthesis in adipocytes. Diabetes. 2007;
68. de Goede P, Wefers J, Brombacher EC, Schrauwen P, metabolism: the metabolic syndrome and lifespan 56(12):2973–2981.
Kalsbeek A. Circadian rhythms in mitochondrial determinant pathways. Aging Cell. 2011;10(1): 106. Dubé JJ, Coen PM, DiStefano G, Chacon AC,
respiration. J Mol Endocrinol. 2018;60(3):R115–R130. 10–17. Helbling NL, Desimone ME, Stafanovic-Racic M,
69. Lee Y, Kim E-K. AMP-activated protein kinase as a 88. DiPietro L. Exercise training and fat metabolism Hames KC, Despines AA, Toledo FGS, Goodpaster
key molecular link between metabolism and after menopause: implications for improved met- BH. Effects of acute lipid overload on skeletal
clockwork. Exp Mol Med. 2013;45(7):e33. abolic flexibility in aging. J Appl Physiol (1985). 2010; muscle insulin resistance, metabolic flexibility, and
70. Asher G, Schibler U. Crosstalk between compo- 109(6):1569–1570. mitochondrial performance. Am J Physiol Endocrinol
nents of circadian and metabolic cycles in mam- 89. Cartee GD, Hepple RT, Bamman MM, Zierath JR. Metab. 2014;307(12):E1117–E1124.
mals. Cell Metab. 2011;13(2):125–137. Exercise promotes healthy aging of skeletal muscle. 107. Goodpaster BH, Sparks LM. Metabolic flexibility in
71. Sahar S, Sassone-Corsi P. Metabolism and cancer: Cell Metab. 2016;23(6):1034–1047. health and disease. Cell Metab. 2017;25(5):
the circadian clock connection. Nat Rev Cancer. 90. Cogliati S, Frezza C, Soriano ME, Varanita T, 1027–1036.
2009;9(12):886–896. Quintana-Cabrera R, Corrado M, Cipolat S, Costa V, 108. Jin T, Weng J. Hepatic functions of GLP-1 and its
72. van Moorsel D, Hansen J, Havekes B, Scheer FAJL, Casarin A, Gomes LC, Perales-Clemente E, Salviati L, based drugs: current disputes and perspectives. Am
Jörgensen JA, Hoeks J, Schrauwen-Hinderling VB, Fernandez-Silva P, Enriquez JA, Scorrano L. Mito- J Physiol Endocrinol Metab. 2016;311(3):E620–E627.
Duez H, Lefebvre P, Schaper NC, Hesselink MKC, chondrial cristae shape determines respiratory 109. van Nierop FS, Scheltema MJ, Eggink HM, Pols TW,
Staels B, Schrauwen P. Demonstration of a day- chain supercomplexes assembly and respiratory Sonne DP, Knop FK, Soeters MR. Clinical relevance
night rhythm in human skeletal muscle oxidative efficiency. Cell. 2013;155(1):160–171. of the bile acid receptor TGR5 in metabolism.
capacity. Mol Metab. 2016;5(8):635–645. 91. Cogliati S, Enriquez JA, Scorrano L. Mitochondrial Lancet Diabetes Endocrinol. 2017;5(3):224–233.
73. Reinke H, Asher G. Circadian clock control of liver cristae: where beauty meets functionality. Trends 110. Kumar DP, Rajagopal S, Mahavadi S, Mirshahi F,
metabolic functions. Gastroenterology. 2016;150(3): Biochem Sci. 2016;41(3):261–273. Grider JR, Murthy KS, Sanyal AJ. Activation of
574–580. 92. Jørgensen W, Rud KA, Mortensen OH, Frandsen L, transmembrane bile acid receptor TGR5 stimulates
74. Damiola F, Le Minh N, Preitner N, Kornmann B, Grunnet N, Quistorff B. Your mitochondria are insulin secretion in pancreatic b cells. Biochem
Fleury-Olela F, Schibler U. Restricted feeding un- what you eat: a high-fat or a high-sucrose diet Biophys Res Commun. 2012;427(3):600–605.
couples circadian oscillators in peripheral tissues eliminates metabolic flexibility in isolated mito- 111. Vettorazzi JF, Ribeiro RA, Borck PC, Branco RCS,
from the central pacemaker in the suprachiasmatic chondria from rat skeletal muscle. Physiol Rep. 2017; Soriano S, Merino B, Boschero AC, Nadal A,
nucleus. Genes Dev. 2000;14(23):2950–2961. 5(6):e13207. Quesada I, Carneiro EM. The bile acid TUDCA
75. Eckel-Mahan K, Sassone-Corsi P. Metabolism and 93. Kuzmiak-Glancy S, Willis WT. Skeletal muscle fuel increases glucose-induced insulin secretion via the
the circadian clock converge. Physiol Rev. 2013; selection occurs at the mitochondrial level. J Exp cAMP/PKA pathway in pancreatic beta cells.
93(1):107–135. Biol. 2014;217(Pt 11):1993–2003. Metabolism. 2016;65(3):54–63.
76. Masri S. Sirtuin-dependent clock control: new ad- 94. Wüst RCI, van der Laarse WJ, Rossiter HB. On-off 112. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J,
asymmetries in oxygen consumption kinetics of Rizzo G, Macchiarulo A, Yamamoto H, Mataki C,
vances in metabolism, aging and cancer. Curr Opin
single Xenopus laevis skeletal muscle fibres suggest Pruzanski M, Pellicciari R, Auwerx J, Schoonjans K.
Clin Nutr Metab Care. 2015;18(6):521–527.
higher-order control. J Physiol. 2013;591(3):731–744. TGR5-mediated bile acid sensing controls glucose
77. Eckel-Mahan KL, Patel VR, de Mateo S, Orozco-Solis
95. Wai T, Langer T. Mitochondrial dynamics and homeostasis. Cell Metab. 2009;10(3):167–177.
R, Ceglia NJ, Sahar S, Dilag-Penilla SA, Dyar KA, Baldi
metabolic regulation. Trends Endocrinol Metab. 113. Alamri BN, Shin K, Chappe V, Anini Y. The role of
P, Sassone-Corsi P. Reprogramming of the circadian
2016;27(2):105–117. ghrelin in the regulation of glucose homeostasis.
clock by nutritional challenge. Cell. 2013;155(7): 96. Jing E, O’Neill BT, Rardin MJ, Kleinridders A, Ilkeyeva Horm Mol Biol Clin Investig. 2016;26(1):3–11.
1464–1478. OR, Ussar S, Bain JR, Lee KY, Verdin EM, Newgard 114. Oh KJ, Lee DS, Kim WK, Han BS, Lee SC, Bae KH.
78. Scheer FAJL, Hilton MF, Mantzoros CS, Shea SA. CB, Gibson BW, Kahn CR. Sirt3 regulates metabolic Metabolic adaptation in obesity and type ii di-
Adverse metabolic and cardiovascular conse- flexibility of skeletal muscle through reversible abetes: myokines, adipokines and hepatokines. Int J
quences of circadian misalignment. Proc Natl Acad enzymatic deacetylation. Diabetes. 2013;62(10): Mol Sci. 2016;18(1):1–31.
Sci USA. 2009;106(11):4453–4458. 3404–3417. 115. Luo L, Liu M. Adipose tissue in control of meta-
79. Karlsson B, Knutsson A, Lindahl B. Is there an as- 97. Glancy B, Hartnell LM, Malide D, Yu Z-X, Combs bolism. J Endocrinol. 2016;231(3):R77–R99.
sociation between shift work and having a meta- CA, Connelly PS, Subramaniam S, Balaban RS. Mi- 116. Jung TW, Yoo HJ, Choi KM. Implication of hep-
bolic syndrome? Results from a population based tochondrial reticulum for cellular energy distribu- atokines in metabolic disorders and cardiovascular
study of 27,485 people. Occup Environ Med. 2001; tion in muscle. Nature. 2015;523(7562):617–620. diseases. BBA Clin. 2016;5:108–113.
58(11):747–752. 98. Liesa M, Shirihai OS. Mitochondrial dynamics in the 117. Coles CA. Adipokines in healthy skeletal muscle and
80. Pan A, Schernhammer ES, Sun Q, Hu FB. Rotating regulation of nutrient utilization and energy ex- metabolic disease. Adv Exp Med Biol. 2016;900:
night shift work and risk of type 2 diabetes: two penditure. Cell Metab. 2013;17(4):491–506. 133–160.
prospective cohort studies in women. PLoS Med. 99. Speijer D. Being right on Q: shaping eukaryotic 118. Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H,
2011;8(12):e1001141. evolution. Biochem J. 2016;473(22):4103–4127. Uchida S, Yamashita S, Noda M, Kita S, Ueki K, Eto

512 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

K, Akanuma Y, Froguel P, Foufelle F, Ferre P, Carling 136. Ozanne SE, Constância M. Mechanisms of disease: 155. Hotamisligil GS, Erbay E. Nutrient sensing and in-
D, Kimura S, Nagai R, Kahn BB, Kadowaki T. Adi- the developmental origins of disease and the role of flammation in metabolic diseases. Nat Rev Immunol.
ponectin stimulates glucose utilization and fatty- the epigenotype. Nat Clin Pract Endocrinol Metab. 2008;8(12):923–934.
acid oxidation by activating AMP-activated protein 2007;3(7):539–546. 156. Hotamisligil GS. Inflammation and metabolic dis-
kinase. Nat Med. 2002;8(11):1288–1295. 137. Kirchner H, Osler ME, Krook A, Zierath JR. Epige- orders. Nature. 2006;444(7121):860–867.
119. Asterholm IW, Scherer PE. Enhanced metabolic netic flexibility in metabolic regulation: disease 157. Du X, Matsumura T, Edelstein D, Rossetti L,
flexibility associated with elevated adiponectin cause and prevention? Trends Cell Biol. 2013;23(5): Zsengellér Z, Szabó C, Brownlee M. Inhibition of
levels. Am J Pathol. 2010;176(3):1364–1376. 203–209. GAPDH activity by poly(ADP-ribose) polymerase
120. Considine RV, Sinha MK, Heiman ML, Kriauciunas 138. Barrès R, Zierath JR. The role of diet and exercise in activates three major pathways of hyperglycemic
A, Stephens TW, Nyce MR, Ohannesian JP, the transgenerational epigenetic landscape of damage in endothelial cells. J Clin Invest. 2003;
Marco CC, McKee LJ, Bauer TL, et al. Serum T2DM. Nat Rev Endocrinol. 2016;12(8):441–451. 112(7):1049–1057.
immunoreactive-leptin concentrations in normal- 139. Saudubray J-M, van den Berghe G, Walter JH, eds. 158. Calçada D, Vianello D, Giampieri E, Sala C, Castellani
weight and obese humans. N Engl J Med. 1996; Inborn Metabolic Diseases, 5th ed. Berlin: Springer; G, de Graaf A, Kremer B, van Ommen B, Feskens E,
334(5):292–295. 2012. Santoro A, Franceschi C, Bouwman J. The role of
121. Bray MS, Young ME. Circadian rhythms in the 140. Flegal KM, Kruszon-Moran D, Carroll MD, Fryar CD, low-grade inflammation and metabolic flexibility in
development of obesity: potential role for the Ogden CL. Trends in obesity among adults in the aging and nutritional modulation thereof: a systems
circadian clock within the adipocyte. Obes Rev. United States, 2005 to 2014. JAMA. 2016;315(21): biology approach. Mech Ageing Dev. 2014;136-137:
2284–2291. 138–147.

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


2007;8(2):169–181.
122. Pedersen BK, Febbraio MA. Muscles, exercise and 141. Aucouturier J, Duché P, Timmons BW. Metabolic 159. Gregor MF, Hotamisligil GS. Inflammatory mech-
obesity: skeletal muscle as a secretory organ. Nat Rev flexibility and obesity in children and youth. Obes anisms in obesity. Annu Rev Immunol. 2011;29(1):
Endocrinol. 2012;8(8):457–465. Rev. 2011;12(5):e44–e53. 415–445.
123. Li F, Li Y, Duan Y, Hu CAA, Tang Y, Yin Y. Myokines 142. Boyle KE, Friedman JE, Janssen RC, Underkofler C, 160. Ertunc ME, Hotamisligil GS. Lipid signaling and
and adipokines: Involvement in the crosstalk be- Houmard JA, Rasouli N. Metabolic inflexibility with lipotoxicity in metaflammation: indications for
tween skeletal muscle and adipose tissue. Cytokine obesity and the effects of fenofibrate on skeletal metabolic disease pathogenesis and treatment.
Growth Factor Rev. 2017;33:73–82. muscle fatty acid oxidation. Horm Metab Res. 2017; J Lipid Res. 2016;57(12):2099–2114.
124. Kostrominova TY. Role of myokines in the main- 49(1):50–57. 161. Donnelly RP, Finlay DK. Glucose, glycolysis and lym-
tenance of whole-body metabolic homeostasis. 143. Tahergorabi Z, Khazaei M, Moodi M, Chamani E. phocyte responses. Mol Immunol. 2015;68(2, 2 Pt C):
Minerva Endocrinol. 2016;41(4):403–420. From obesity to cancer: a review on proposed 513–519.
125. Rao RR, Long JZ, White JP, Svensson KJ, Lou J, mechanisms. Cell Biochem Funct. 2016;34(8): 162. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R,
Lokurkar I, Jedrychowski MP, Ruas JL, Wrann CD, Lo 533–545. Finkelstein D, McCormick LL, Fitzgerald P, Chi H,
144. Storlien L, Oakes ND, Kelley DE. Metabolic flexibility. Munger J, Green DR. The transcription factor
JC, Camera DM, Lachey J, Gygi S, Seehra J, Hawley JA,
Proc Nutr Soc. 2004;63(2):363–368. Myc controls metabolic reprogramming upon
Spiegelman BM. Meteorin-like is a hormone that
145. Guize L, Thomas F, Pannier B, Bean K, Jego B, T lymphocyte activation. Immunity. 2011;35(6):
regulates immune-adipose interactions to in-
Benetos A. All-cause mortality associated with 871–882.
crease beige fat thermogenesis. Cell. 2014;157(6):
specific combinations of the metabolic syndrome 163. Assmann N, Finlay DK. Metabolic regulation of
1279–1291.
according to recent definitions. Diabetes Care. 2007; immune responses: therapeutic opportunities. J Clin
126. Rodrı́guez A, Becerril S, Ezquerro S, Méndez-
30(9):2381–2387. Invest. 2016;126(6):2031–2039.
Giménez L, Frühbeck G. Crosstalk between adi-
146. Galgani JE, Moro C, Ravussin E. Metabolic flexibility 164. Netea MG, Joosten LAB, Latz E, Mills KHG, Natoli G,
pokines and myokines in fat browning. Acta Physiol
and insulin resistance. Am J Physiol Endocrinol Stunnenberg HG, O’Neill LAJ, Xavier RJ. Trained
(Oxf). 2017;219(2):362–381.
Metab. 2008;295(5):E1009–E1017. immunity: a program of innate immune memory in
127. Drong AW, Lindgren CM, McCarthy MI. The ge-
147. Kelley DE. Skeletal muscle triglycerides: an aspect of health and disease. Science. 2016;352(6284):aaf1098.
netic and epigenetic basis of type 2 diabetes and
regional adiposity and insulin resistance. Ann N Y 165. Lachmandas E, Boutens L, Ratter JM, Hijmans A,
obesity. Clin Pharmacol Ther. 2012;92(6):707–715.
Acad Sci. 2002;967(1):135–145. Hooiveld GJ, Joosten LAB, Rodenburg RJ, Fransen
128. Sen P, Shah PP, Nativio R, Berger SL. Epigenetic
148. Schrauwen P, Schrauwen-Hinderling V, Hoeks J, JAM, Houtkooper RH, van Crevel R, Netea MG,
mechanisms of longevity and aging. Cell. 2016; Hesselink MKC. Mitochondrial dysfunction and Stienstra R. Microbial stimulation of different Toll-
166(4):822–839. lipotoxicity. Biochim Biophys Acta. 2010;1801(3): like receptor signalling pathways induces diverse
129. Aon MA, Cortassa S, Juhaszova M, Sollott SJ. Mi- 266–271. metabolic programmes in human monocytes. Nat
tochondrial health, the epigenome and healthspan. 149. Phielix E, Mensink M. Type 2 diabetes mellitus and Microbiol. 2016;2(December):16246.
Clin Sci (Lond). 2016;130(15):1285–1305. skeletal muscle metabolic function. Physiol Behav. 166. Cheng SC, Quintin J, Cramer RA, Shepardson KM,
130. Katada S, Imhof A, Sassone-Corsi P. Connecting 2008;94(2):252–258. Saeed S, Kumar V, Giamarellos-Bourboulis EJ,
threads: epigenetics and metabolism. Cell. 2012; 150. Ritov VB, Menshikova EV, Azuma K, Wood R, Martens JH, Rao NA, Aghajanirefah A, Manjeri GR, Li
148(1-2):24–28. Toledo FGS, Goodpaster BH, Ruderman NB, Kelley Y, Ifrim DC, Arts RJ, van der Veer BM, Deen PM,
131. Reddy MA, Zhang E, Natarajan R. Epigenetic DE. Deficiency of electron transport chain in human Logie C, O’Neill LA, Willems P, van de Veerdonk FL,
mechanisms in diabetic complications and meta- skeletal muscle mitochondria in type 2 diabetes van der Meer JW, Ng A, Joosten LA, Wijmenga C,
bolic memory. Diabetologia. 2015;58(3):443–455. mellitus and obesity. Am J Physiol Endocrinol Metab. Stunnenberg HG, Xavier RJ, Netea MG. mTOR- and
132. Taylor EM, Jones AD, Henagan TM. A review of 2010;298(1):E49–E58. HIF-1a-mediated aerobic glycolysis as metabolic
mitochondrial-derived fatty acids in epigenetic 151. Hesselink MKC, Schrauwen-Hinderling V, Schrauwen P. basis for trained immunity [published correction
regulation of obesity and type 2 diabetes. J Nutrit Skeletal muscle mitochondria as a target to pre- appears in Science. 2014;346(6210):aaa1503]. Science.
Health Food Sci. 2014;2(3):1–4. vent or treat type 2 diabetes mellitus. Nat Rev 2014;345(6204):1250684.
133. Menzies KJ, Zhang H, Katsyuba E, Auwerx J. Protein Endocrinol. 2016;12(11):633–645. 167. Johnson AR, Milner JJ, Makowski L. The in-
acetylation in metabolism - metabolites and co- 152. Kelley DE, He J, Menshikova EV, Ritov VB. Dys- flammation highway: metabolism accelerates in-
factors. Nat Rev Endocrinol. 2016;12(1):43–60. function of mitochondria in human skeletal muscle flammatory traffic in obesity. Immunol Rev. 2012;
134. Teperino R, Schoonjans K, Auwerx J. Histone methyl in type 2 diabetes. Diabetes. 2002;51(10):2944–2950. 249(1):218–238.
transferases and demethylases; can they link 153. Jang C, Oh SF, Wada S, Rowe GC, Liu L, Chan MC, 168. Kang K, Reilly SM, Karabacak V, Gangl MR,
metabolism and transcription? Cell Metab. 2010; Rhee J, Hoshino A, Kim B, Ibrahim A, Baca LG, Kim E, Fitzgerald K, Hatano B, Lee C-H. Adipocyte-derived
12(4):321–327. Ghosh CC, Parikh SM, Jiang A, Chu Q, Forman DE, Th2 cytokines and myeloid PPARdelta regulate
135. Chen Z, Miao F, Paterson AD, Lachin JM, Zhang L, Lecker SH, Krishnaiah S, Rabinowitz JD, Weljie AM, macrophage polarization and insulin sensitivity. Cell
Schones DE, Wu X, Wang J, Tompkins JD, Genuth S, Baur JA, Kasper DL, Arany Z. A branched-chain Metab. 2008;7(6):485–495.
Braffett BH, Riggs AD, Natarajan R, Natarajan R; amino acid metabolite drives vascular fatty acid 169. Johnson AR, Qin Y, Cozzo AJ, Freemerman AJ,
DCCT/EDIC Research Group. Epigenomic profiling transport and causes insulin resistance. Nat Med. Huang MJ, Zhao L, Sampey BP, Milner JJ, Beck MA,
reveals an association between persistence of DNA 2016;22(4):421–426. Damania B, Rashid N, Galanko JA, Lee DP, Edin ML,
methylation and metabolic memory in the DCCT/ 154. Newgard CB. Interplay between lipids and Zeldin DC, Fueger PT, Dietz B, Stahl A, Wu Y,
EDIC type 1 diabetes cohort. Proc Natl Acad Sci USA. branched-chain amino acids in development of Mohlke KL, Makowski L. Metabolic reprogramming
2016;113(21):E3002–E3011. insulin resistance. Cell Metab. 2012;15(5):606–614. through fatty acid transport protein 1 (FATP1)

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 513


REVIEW

regulates macrophage inflammatory potential mortality in the Third National Health and Nu- 201. Strickland M, Stoll EA. Metabolic reprogramming in
and adipose inflammation. Mol Metab. 2016;5(7): trition Examination Survey. Cancer Causes Control. glioma. Front Cell Dev Biol. 2017;5(April):43.
506–526. 2017;28(2):127–136. 202. Zhao S, Torres A, Henry RA, Trefely S, Wallace M,
170. Van den Bossche J, Baardman J, Otto NA, van der 185. Scappaticcio L, Maiorino MI, Bellastella G, Giugliano Lee JV, Carrer A, Sengupta A, Campbell SL, Kuo Y-M,
Velden S, Neele AE, van den Berg SM, Luque-Martin D, Esposito K. Insights into the relationships be- Frey AJ, Meurs N, Viola JM, Blair IA, Weljie AM,
R, Chen HJ, Boshuizen MCS, Ahmed M, Hoeksema tween diabetes, prediabetes, and cancer. Endocrine. Metallo CM, Snyder NW, Andrews AJ, Wellen KE.
MA, de Vos AF, de Winther MPJ. Mitochondrial 2017;56(2):231–239. ATP-citrate lyase controls a glucose-to-acetate meta-
dysfunction prevents repolarization of inflamma- 186. Inoue M, Tsugane S. Insulin resistance and cancer: bolic switch. Cell Reports. 2016;17(4):1037–1052.
tory macrophages. Cell Reports. 2016;17(3):684–696. epidemiological evidence. Endocr Relat Cancer. 2012; 203. Zhao X, Jiang P, Deng X, Li Z, Tian F, Guo F, Li X,
171. Cheng S-C, Scicluna BP, Arts RJW, Gresnigt MS, 19(5):F1–F8. Wang S. Inhibition of mTORC1 signaling sensitizes
Lachmandas E, Giamarellos-Bourboulis EJ, Kox M, 187. Potter J, Brown L, Williams RL, Byles J, Collins CE. hepatocellular carcinoma cells to glycolytic stress.
Manjeri GR, Wagenaars JAL, Cremer OL, Leentjens J, Diet quality and cancer outcomes in adults: a sys- Am J Cancer Res. 2016;6(10):2289–2298.
van der Meer AJ, van de Veerdonk FL, Bonten MJ, tematic review of epidemiological studies. Int J Mol 204. Martinez-Outschoorn UE, Peiris-Pagés M, Pestell
Schultz MJ, Willems PHGM, Pickkers P, Joosten LAB, Sci. 2016;17(7):1–30. RG, Sotgia F, Lisanti MP. Cancer metabolism:
van der Poll T, Netea MG. Broad defects in the 188. Ridlon JM, Wolf PG, Gaskins HR. Taurocholic acid a therapeutic perspective [published correction
energy metabolism of leukocytes underlie immu- metabolism by gut microbes and colon cancer. Gut appears in Nat Rev Clin Oncol. 2017;14:113]. Nat Rev
noparalysis in sepsis. Nat Immunol. 2016;17(4): Microbes. 2016;7(3):201–215. Clin Oncol. 2017;14(1):11–31.
406–413.

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


189. Gordon-Dseagu VLZ, Thompson FE, Subar AF, 205. Chaube B, Malvi P, Singh SV, Mohammad N, Meena
172. Brealey D, Brand M, Hargreaves I, Heales S, Land J, Ruder EH, Thiébaut ACM, Potischman N, Stolzenberg- AS, Bhat MK. Targeting metabolic flexibility by si-
Smolenski R, Davies NA, Cooper CE, Singer M. Solomon R. A cohort study of adolescent and multaneously inhibiting respiratory complex I and
Association between mitochondrial dysfunction midlife diet and pancreatic cancer risk in the lactate generation retards melanoma progression.
and severity and outcome of septic shock. Lancet. NIH-AARP Diet and Health Study. Am J Epidemiol. Oncotarget. 2015;6(35):37281–37299.
2002;360(9328):219–223. 2017;186(3):305–317. 206. Pavlova NN, Thompson CB. The emerging hall-
173. Quoilin C, Mouithys-Mickalad A, Lécart S, Fontaine- 190. Magaki M, Ishii H, Yamasaki A, Kitai Y, Kametani S, marks of cancer metabolism. Cell Metab. 2016;23(1):
Aupart M-P, Hoebeke M. Evidence of oxidative Nakai R, Dabid A, Tsuda H, Ohnishi T. A high-fat 27–47.
stress and mitochondrial respiratory chain dys- diet increases the incidence of mammary cancer 207. Vazquez A, Kamphorst JJ, Markert EK, Schug ZT,
function in an in vitro model of sepsis-induced inc-Ha-ras proto-oncogene transgenic rats. J Toxicol Tardito S, Gottlieb E. Cancer metabolism at a
kidney injury. Biochim Biophys Acta. 2014;1837(10): Pathol. 2017;30(2):145–152. glance. J Cell Sci. 2016;129(18):3367–3373.
1790–1800. 191. Sundaram S, Yan L. Dietary energy restriction re- 208. Vander Heiden MG, Cantley LC, Thompson CB.
174. Lewis AJ, Billiar TR, Rosengart MR. Biology and duces high-fat diet-enhanced metastasis of Lewis Understanding the Warburg effect: the metabolic
metabolism of sepsis: innate immunity, bio- lung carcinoma in mice. Oncotarget. 2016;7(40): requirements of cell proliferation. Science. 2009;
energetics, and autophagy. Surg Infect (Larchmt).
65669–65675. 324(5930):1029–1033.
2016;17(3):286–293.
192. Ferro M, Terracciano D, Buonerba C, Lucarelli G, 209. Lunt SY, Vander Heiden MG. Aerobic glycolysis:
175. Kaimoto S, Hoshino A, Ariyoshi M, Okawa Y,
Bottero D, Perdonà S, Autorino R, Serino A, meeting the metabolic requirements of cell pro-
Tateishi S, Ono K, Uchihashi M, Fukai K, Iwai-Kanai
Cantiello F, Damiano R, Andras I, De Placido S, Di liferation. Annu Rev Cell Dev Biol. 2011;27(1):
E, Matoba S. Activation of PPAR-a in the early stage
Lorenzo G, Battaglia M, Jereczek-Fossa BA, Mirone 441–464.
of heart failure maintained myocardial function and
V, De Cobelli O. The emerging role of obesity, diet 210. Holness MJ, Sugden MC. Regulation of pyruvate
energetics in pressure-overload heart failure. Am J
and lipid metabolism in prostate cancer. Future dehydrogenase complex activity by reversible
Physiol Heart Circ Physiol. 2017;312(2):H305–H313.
Oncol. 2017;13(3):285–293. phosphorylation. Biochem Soc Trans. 2003;31(6):
176. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial
193. Pascual G, Avgustinova A, Mejetta S, Martı́n M, 1143–1151.
substrate metabolism in the normal and failing
Castellanos A, Attolini CS-O, Berenguer A, Prats N, 211. Kaplon J, Zheng L, Meissl K, Chaneton B, Selivanov
heart. Physiol Rev. 2005;85(3):1093–1129.
Toll A, Hueto JA, Bescós C, Di Croce L, Benitah SA. VA, Mackay G, van der Burg SH, Verdegaal EME,
177. Wüst RCI, Helmes M, Martin JL, van der Wardt TJT,
Targeting metastasis-initiating cells through the Cascante M, Shlomi T, Gottlieb E, Peeper DS. A key
Musters RJP, van der Velden J, Stienen GJM. Rapid
fatty acid receptor CD36. Nature. 2017;541(7635): role for mitochondrial gatekeeper pyruvate de-
frequency-dependent changes in free mitochon-
drial calcium concentration in rat cardiac myocytes. 41–45. hydrogenase in oncogene-induced senescence.
J Physiol. 2017;595(6):2001–2019. 194. Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Nature. 2013;498(7452):109–112.
178. Griffin TM, Humphries KM, Kinter M, Lim H-YY, Cheng CW, Madia F, Fontana L, Mirisola MG, 212. Maddocks ODK, Berkers CR, Mason SM, Zheng L,
Szweda LI. Nutrient sensing and utilization: getting Guevara-Aguirre J, Wan J, Passarino G, Kennedy BK, Blyth K, Gottlieb E, Vousden KH. Serine starvation
to the heart of metabolic flexibility. Biochimie. 2016; Wei M, Cohen P, Crimmins EM, Longo VD. Low induces stress and p53-dependent metabolic
124:74–83. protein intake is associated with a major reduction remodelling in cancer cells. Nature. 2013;493(7433):
179. Pascual F, Coleman RA. Fuel availability and fate in in IGF-1, cancer, and overall mortality in the 65 and 542–546.
cardiac metabolism: a tale of two substrates. Bio- younger but not older population. Cell Metab. 213. Meiser J, Tumanov S, Maddocks O, Labuschagne CF,
chim Biophys Acta. 2016;1861(10):1425–1433. 2014;19(3):407–417. Athineos D, Van Den Broek N, Mackay GM,
180. Lopaschuk GD, Ussher JR, Folmes CDL, Jaswal JS, 195. Riera CE, Dillin A. Tipping the metabolic scales Gottlieb E, Blyth K, Vousden K, Kamphorst JJ,
Stanley WC. Myocardial fatty acid metabolism in towards increased longevity in mammals. Nat Cell Vazquez A. Serine one-carbon catabolism with
health and disease. Physiol Rev. 2010;90(1):207–258. Biol. 2015;17(3):196–203. formate overflow. Sci Adv. 2016;2(10):e1601273.
181. Schwarzer M, Osterholt M, Lunkenbein A, 196. Muñoz-Pinedo C, El Mjiyad N, Ricci J-E. Cancer 214. Sullivan LB, Gui DY, Hosios AM, Bush LN, Freinkman
Schrepper A, Amorim P, Doenst T. Mitochondrial metabolism: current perspectives and future di- E, Vander Heiden MG. Supporting aspartate bio-
reactive oxygen species production and respiratory rections. Cell Death Dis. 2012;3(1):e248. synthesis is an essential function of respiration in
complex activity in rats with pressure overload- 197. Colman RJ, Anderson RM, Johnson SC, Kastman EK, proliferating cells. Cell. 2015;162(3):552–563.
induced heart failure. J Physiol. 2014;592(17): Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, 215. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N,
3767–3782. Simmons HA, Kemnitz JW, Weindruch R. Caloric Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff
182. Wüst RCI, de Vries HJ, Wintjes LT, Rodenburg RJ, restriction delays disease onset and mortality in M, McMahon SB, Thompson CB. Myc regulates a
Niessen HWM, Stienen GJM. Mitochondrial com- rhesus monkeys. Science. 2009;325(5937):201–204. transcriptional program that stimulates mito-
plex I dysfunction and altered NAD(P)H kinetics in 198. Most J, Tosti V, Redman LM, Fontana L. Calorie chondrial glutaminolysis and leads to glutamine
rat myocardium in cardiac right ventricular hy- restriction in humans: an update. Ageing Res Rev. addiction. Proc Natl Acad Sci USA. 2008;105(48):
pertrophy and failure. Cardiovasc Res. 2016;111(4): 2017;39:36–45. 18782–18787.
362–372. 199. Vyas S, Zaganjor E, Haigis MC. Mitochondria and 216. Tardito S, Oudin A, Ahmed SU, Fack F, Keunen O,
183. Tsugane S, Inoue M. Insulin resistance and cancer: cancer. Cell. 2016;166(3):555–566. Zheng L, Miletic H, Sakariassen PØ, Weinstock A,
epidemiological evidence. Cancer Sci. 2010;101(5): 200. Herst PM, Berridge MV. Cell hierarchy, metabolic Wagner A, Lindsay SL, Hock AK, Barnett SC, Ruppin
1073–1079. flexibility and systems approaches to cancer E, Mørkve SH, Lund-Johansen M, Chalmers AJ,
184. Gathirua-Mwangi WG, Monahan PO, Murage MJ, treatment. Curr Pharm Biotechnol. 2013;14(3): Bjerkvig R, Niclou SP, Gottlieb E. Glutamine syn-
Zhang J. Metabolic syndrome and total cancer 289–299. thetase activity fuels nucleotide biosynthesis and

514 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

supports growth of glutamine-restricted glioblas- exercise and fasting-induced adaptations in mouse 247. de Ligt M, Timmers S, Schrauwen P. Resveratrol and
toma. Nat Cell Biol. 2015;17(12):1556–1568. liver. Am J Physiol Regul Integr Comp Physiol. 2011; obesity: can resveratrol relieve metabolic distur-
217. DeLany JP, Kelley DE, Hames KC, Jakicic JM, 301(5):R1501–R1509. bances? Biochim Biophys Acta. 2015;1852(6):
Goodpaster BH. Effect of physical activity on weight 232. Vettor R, Valerio A, Ragni M, Trevellin E, Granzotto 1137–1144.
loss, energy expenditure, and energy intake during M, Olivieri M, Tedesco L, Ruocco C, Fossati A, Fabris 248. Um J-H, Park S-J, Kang H, Yang S, Foretz M,
diet induced weight loss. Obesity (Silver Spring). R, Serra R, Carruba MO, Nisoli E. Exercise training McBurney MW, Kim MK, Viollet B, Chung JH. AMP-
2014;22(2):363–370. boosts eNOS-dependent mitochondrial biogenesis activated protein kinase-deficient mice are resistant
218. Gallagher D, Heshka S, Kelley DE, Thornton J, Boxt L, in mouse heart: role in adaptation of glucose to the metabolic effects of resveratrol. Diabetes.
Pi-Sunyer FX, Patricio J, Mancino J, Clark JM; MRI metabolism. Am J Physiol Endocrinol Metab. 2014; 2010;59(3):554–563.
Ancillary Study Group of Look AHEAD Research 306(5):E519–E528. 249. Andreux PA, Houtkooper RH, Auwerx J. Pharma-
Group. Changes in adipose tissue depots and 233. de la Iglesia R, Loria-Kohen V, Zulet MA, Martinez cological approaches to restore mitochondrial
metabolic markers following a 1-year diet and ex- JA, Reglero G, Ramirez de Molina A. Dietary function. Nat Rev Drug Discov. 2013;12(6):465–483.
ercise intervention in overweight and obese pa- strategies implicated in the prevention and treat- 250. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane
tients with type 2 diabetes. Diabetes Care. 2014; ment of metabolic syndrome. Int J Mol Sci. 2016; H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert
37(12):3325–3332. 17(11):1–21. P, Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J.
219. Pownall HJ, Bray GA, Wagenknecht LE, Walkup MP, 234. Tay J, Thompson CH, Luscombe-Marsh ND, Resveratrol improves mitochondrial function and
Heshka S, Hubbard VS, Hill J, Kahn SE, Nathan DM, Wycherley TP, Noakes M, Buckley JD, Wittert GA, protects against metabolic disease by activating
Schwartz AV, Johnson KC, Group LAR; Look SIRT1 and PGC-1alpha. Cell. 2006;127(6):1109–1122.

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


Yancy WS Jr, Brinkworth GD. Effects of an energy-
AHEAD Research Group. Changes in body com- restricted low-carbohydrate, high unsaturated fat/ 251. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C,
position over 8 years in a randomized trial of a low saturated fat diet versus a high-carbohydrate, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis
lifestyle intervention: the look AHEAD study. low-fat diet in type 2 diabetes: a 2-year randomized K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D,
Obesity (Silver Spring). 2015;23(3):565–572. clinical trial. Diabetes Obes Metab. 2018;20(4): Wang M, Ramaswamy S, Fishbein KW, Spencer RG,
220. Bergouignan A, Antoun E, Momken I, Schoeller DA, 858–871. Lakatta EG, Le Couteur D, Shaw RJ, Navas P,
Gauquelin-Koch G, Simon C, Blanc S. Effect of 235. Mattson MP, Longo VD, Harvie M. Impact of in- Puigserver P, Ingram DK, de Cabo R, Sinclair DA.
contrasted levels of habitual physical activity on termittent fasting on health and disease processes. Resveratrol improves health and survival of mice
metabolic flexibility. J Appl Physiol (1985). 2013; Ageing Res Rev. 2017;39:46–58. on a high-calorie diet. Nature. 2006;444(7117):
114(3):371–379. 236. Mattison JA, Colman RJ, Beasley TM, Allison DB, 337–342.
221. Rynders CA, Blanc S, DeJong N, Bessesen DH, Kemnitz JW, Roth GS, Ingram DK, Weindruch R, de 252. Shang J, Chen LL, Xiao FX, Sun H, Ding HC, Xiao H.
Bergouignan A. Sedentary behaviour is a key de- Cabo R, Anderson RM. Caloric restriction improves Resveratrol improves non-alcoholic fatty liver dis-
terminant of metabolic inflexibility. J Physiol. 2018; ease by activating AMP-activated protein kinase.
health and survival of rhesus monkeys. Nat Com-
596(8):1319–1330. Acta Pharmacol Sin. 2008;29(6):698–706.
mun. 2017;8:14063.
222. Egan B, Zierath JR. Exercise metabolism and the 253. Timmers S, Konings E, Bilet L, Houtkooper RH, van
237. Harvie M, Howell A. Potential benefits and harms of
molecular regulation of skeletal muscle adaptation. de Weijer T, Goossens GH, Hoeks J, van der Krieken
intermittent energy restriction and intermittent
Cell Metab. 2013;17(2):162–184. S, Ryu D, Kersten S, Moonen-Kornips E, Hesselink
fasting amongst obese, overweight and normal
223. Kjøbsted R, Munk-Hansen N, Birk JB, Foretz M, MKC, Kunz I, Schrauwen-Hinderling VB, Blaak E,
weight subjects-a narrative review of human and
Viollet B, Björnholm M, Zierath JR, Treebak JT, Auwerx J, Schrauwen P. Calorie restriction-like ef-
animal evidence. Behav Sci (Basel). 2017;7(1):4.
Wojtaszewski JFP. Enhanced muscle insulin sensi- fects of 30 days of resveratrol supplementation on
238. Hutchison AT, Heilbronn LK. Metabolic impacts of
tivity after contraction/exercise is mediated by energy metabolism and metabolic profile in obese
altering meal frequency and timing - Does when we
AMPK. Diabetes. 2017;66(3):598–612. humans. Cell Metab. 2011;14(5):612–622.
eat matter? Biochimie. 2016;124:187–197.
224. McGee SL, Fairlie E, Garnham AP, Hargreaves M. 254. Bhatt JK, Thomas S, Nanjan MJ. Resveratrol sup-
239. Asher G, Sassone-Corsi P. Time for food: the in-
Exercise-induced histone modifications in human plementation improves glycemic control in type 2
timate interplay between nutrition, metabolism,
skeletal muscle. J Physiol. 2009;587(24):5951–5958. diabetes mellitus. Nutr Res. 2012;32(7):537–541.
225. Greggio C, Jha P, Kulkarni SS, Lagarrigue S, Broskey and the circadian clock. Cell. 2015;161(1):84–92. 255. Komen JC, Thorburn DR. Turn up the power -
NT, Boutant M, Wang X, Conde Alonso S, Ofori E, 240. Zarrinpar A, Chaix A, Panda S. Daily eating patterns
pharmacological activation of mitochondrial bio-
Auwerx J, Cantó C, Amati F. Enhanced respiratory and their impact on health and disease. Trends genesis in mouse models. Br J Pharmacol. 2014;
chain supercomplex formation in response to ex- Endocrinol Metab. 2016;27(2):69–83. 171(8):1818–1836.
ercise in human skeletal muscle. Cell Metab. 2017; 241. Miyata Y, Shimomura I. Metabolic flexibility and 256. Narkar VA, Downes M, Yu RT, Embler E, Wang YX,
25(2):301–311. carnitine flux: The role of carnitine acyltransferase in Banayo E, Mihaylova MM, Nelson MC, Zou Y,
226. da Silva Aragão R, Guzmán-Quevedo O, Pérez- glucose homeostasis. J Diabetes Investig. 2013;4(3): Juguilon H, Kang H, Shaw RJ, Evans RM. AMPK and
Garcı́a G, Manhães-de-Castro R, Bolaños-Jiménez 247–249. PPARdelta agonists are exercise mimetics. Cell. 2008;
F. Maternal protein restriction impairs the tran- 242. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, 134(3):405–415.
scriptional metabolic flexibility of skeletal muscle in Ilkayeva O, Stevens RD, Hegardt FG, Muoio DM. 257. Russell RR III, Bergeron R, Shulman GI, Young LH.
adult rat offspring. Br J Nutr. 2014;112(3):328–337. Carnitine insufficiency caused by aging and over- Translocation of myocardial GLUT-4 and increased
227. Malin SK, Haus JM, Solomon TPJ, Blaszczak A, nutrition compromises mitochondrial performance glucose uptake through activation of AMPK by
Kashyap SR, Kirwan JP. Insulin sensitivity and and metabolic control. J Biol Chem. 2009;284(34): AICAR. Am J Physiol. 1999;277(2 Pt 2):H643–H649.
metabolic flexibility following exercise training 22840–22852. 258. Houtkooper RH, Cantó C, Wanders RJ, Auwerx J.
among different obese insulin-resistant phenotypes. 243. Muoio DM, Noland RC, Kovalik J-PP, Seiler SE, The secret life of NAD+: an old metabolite con-
Am J Physiol Endocrinol Metab. 2013;305(10): Davies MN, DeBalsi KL, Ilkayeva OR, Stevens RD, trolling new metabolic signaling pathways. Endocr
E1292–E1298. Kheterpal I, Zhang J, Covington JD, Bajpeyi S, Rev. 2010;31(2):194–223.
228. Wallberg-Henriksson H, Zierath JR. Metabolism. Ravussin E, Kraus W, Koves TR, Mynatt RL. Muscle- 259. Bleeker JC, Houtkooper RH. Sirtuin activation as a
Exercise remodels subcutaneous fat tissue and specific deletion of carnitine acetyltransferase therapeutic approach against inborn errors of
improves metabolism. Nat Rev Endocrinol. 2015; compromises glucose tolerance and metabolic metabolism. J Inherit Metab Dis. 2016;39(4):565–572.
11(4):198–200. flexibility. Cell Metab. 2012;15(5):764–777. 260. Carafa V, Rotili D, Forgione M, Cuomo F, Serretiello
229. Sepa-Kishi DM, Ceddia RB. Exercise-mediated ef- 244. Xia Y, Li Q, Zhong W, Dong J, Wang Z, Wang C. L- E, Hailu GS, Jarho E, Lahtela-Kakkonen M, Mai A,
fects on white and brown adipose tissue plasticity carnitine ameliorated fatty liver in high-calorie diet/ Altucci L. Sirtuin functions and modulation: from
and metabolism. Exerc Sport Sci Rev. 2016;44(1): STZ-induced type 2 diabetic mice by improving chemistry to the clinic. Clin Epigenetics. 2016;8(1):61.
37–44. mitochondrial function. Diabetol Metab Syndr. 2011; 261. Cantó C, Menzies KJ, Auwerx J. NAD(+) metabolism
230. Brouwers B, Hesselink MKC, Schrauwen P, 3(1):31. and the control of energy homeostasis: a balancing
Schrauwen-Hinderling VB. Effects of exercise 245. Handschin C. Caloric restriction and exercise “mi- act between mitochondria and the nucleus. Cell
training on intrahepatic lipid content in humans. metics”: ready for prime time? Pharmacol Res. 2016; Metab. 2015;22(1):31–53.
Diabetologia. 2016;59(10):2068–2079. 103:158–166. 262. Mouchiroud L, Houtkooper RH, Auwerx J. NAD+
231. Haase TN, Ringholm S, Leick L, Biensø RS, Kiilerich K, 246. Pryor R, Cabreiro F. Repurposing metformin: an old metabolism: a therapeutic target for age-related
Johansen S, Nielsen MM, Wojtaszewski JF, Hidalgo J, drug with new tricks in its binding pockets. Biochem metabolic disease. Crit Rev Biochem Mol Biol.
Pedersen PA, Pilegaard H. Role of PGC-1a in J. 2015;471(3):307–322. 2013;48(4):397–408.

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 515


REVIEW

263. Finkel T. The metabolic regulation of aging. Nat Clish CB, Murphy LO, Manning BD. Activation 293. Maglioni S, Ventura N. C. elegans as a model or-
Med. 2015;21(12):1416–1423. of a metabolic gene regulatory network down- ganism for human mitochondrial associated dis-
264. Cantó C, Houtkooper RH, Pirinen E, Youn DY, stream of mTOR complex 1. Mol Cell. 2010;39(2): orders. Mitochondrion. 2016;30:117–125.
Oosterveer MH, Cen Y, Fernandez-Marcos PJ, 171–183. 294. Lapierre LR, Hansen M. Lessons from C. elegans:
Yamamoto H, Andreux PA, Cettour-Rose P, 277. Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini signaling pathways for longevity. Trends Endocrinol
Gademann K, Rinsch C, Schoonjans K, Sauve AA, DM. mTORC1 controls fasting-induced ketogenesis Metab. 2012;23(12):637–644.
Auwerx J. The NAD(+) precursor nicotinamide and its modulation by ageing. Nature. 2010; 295. Doroszuk A, Snoek LB, Fradin E, Riksen J, Kammenga
riboside enhances oxidative metabolism and pro- 468(7327):1100–1104. J. A genome-wide library of CB4856/N2 in-
tects against high-fat diet-induced obesity. Cell 278. Albert V, Hall MN. mTOR signaling in cellular and trogression lines of Caenorhabditis elegans. Nucleic
Metab. 2012;15(6):838–847. organismal energetics. Curr Opin Cell Biol. 2015; Acids Res. 2009;37(16):e110.
265. Shi W, Hegeman MA, van Dartel DAM, Tang J, 33(1):55–66. 296. Yilmaz LS, Walhout AJ. A Caenorhabditis elegans
Suarez M, Swarts H, van der Hee B, Arola L, Keijer J. 279. Li J, Kim SG, Blenis J. Rapamycin: one drug, many genome-scale metabolic network model. Cell Syst.
Effects of a wide range of dietary nicotinamide effects. Cell Metab. 2014;19(3):373–379. 2016;2(5):297–311.
riboside (NR) concentrations on metabolic flexi- 280. Kennedy BKK, Lamming DWW. The mechanistic 297. Gebauer J, Gentsch C, Mansfeld J, Schmeißer K,
bility and white adipose tissue (WAT) of mice fed a target of rapamycin: the grand conducTOR of Waschina S, Brandes S, Klimmasch L, Zamboni N,
mildly obesogenic diet. Mol Nutr Food Res. 2017; metabolism and aging. Cell Metab. 2016;23(6): Zarse K, Schuster S, Ristow M, Schäuble S, Kaleta C.
61(8):1600878. A genome-scale database and reconstruction of
990–1003.
266. Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, Caenorhabditis elegans metabolism. Cell Syst. 2016;

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


281. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle
D’Amico D, Ropelle ER, Lutolf MP, Aebersold R, 2(5):312–322.
CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K,
Schoonjans K, Menzies KJ, Auwerx J. NAD+ re- 298. Watson E, MacNeil LT, Arda HE, Zhu LJ, Walhout
Carter CS, Pahor M, Javors MA, Fernandez E, Miller
pletion improves mitochondrial and stem cell AJM. Integration of metabolic and gene regula-
RA. Rapamycin fed late in life extends lifespan in
function and enhances life span in mice. Science. tory networks modulates the C. elegans die-
genetically heterogeneous mice. Nature. 2009;
2016;352(6292):1436–1443. tary response [published correction appears in
267. Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, 460(7253):392–395. Cell. 2013;153(6):1406–1407]. Cell. 2013;153(1):
Sasaki Y, Redpath P, Migaud ME, Apte RS, Uchida K, 282. van Ommen B, van der Greef J, Ordovas JM, Daniel 253–266.
Yoshino J, Imai SI. Long-term administration of H. Phenotypic flexibility as key factor in the human 299. Gao AW, Chatzispyrou IA, Kamble R, Liu YJ, Herzog
nicotinamide mononucleotide mitigates age- nutrition and health relationship. Genes Nutr. 2014; K, Smith RL, van Lenthe H, Vervaart MAT, van
associated physiological decline in mice. Cell 9(5):423. Cruchten A, Luyf AC, van Kampen A, Pras-Raves
Metab. 2016;24(6):795–806. 283. Stroeve JHM, van Wietmarschen H, Kremer BHA, ML, Vaz FM, Houtkooper RH. A sensitive mass
268. Pirinen E, Cantó C, Jo YS, Morato L, Zhang H, van Ommen B, Wopereis S. Phenotypic flexibility spectrometry platform identifies metabolic changes
Menzies KJ, Williams EG, Mouchiroud L, Moullan N, as a measure of health: the optimal nutritional of life history traits in C. elegans. Sci Rep. 2017;7(1):
Hagberg C, Li W, Timmers S, Imhof R, Verbeek J, stress response test. Genes Nutr. 2015;10(3):13. 2408.
Pujol A, van Loon B, Viscomi C, Zeviani M, 284. Orman MA, Berthiaume F, Androulakis IP, 300. Demetrius L. Of mice and men. When it comes to
Schrauwen P, Sauve AA, Schoonjans K, Auwerx J. Ierapetritou MG. Advanced stoichiometric analysis studying ageing and the means to slow it down,
Pharmacological Inhibition of poly(ADP-ribose) of metabolic networks of mammalian systems. Crit mice are not just small humans. EMBO Rep. 2005;
polymerases improves fitness and mitochondrial Rev Biomed Eng. 2011;39(6):511–534. 6(Spec No):S39–S44.
function in skeletal muscle. Cell Metab. 2014;19(6): 285. Lewis NE, Abdel-Haleem AM. The evolution of 301. Wong N, Morahan G, Stathopoulos M, Proietto J,
1034–1041. genome-scale models of cancer metabolism. Front Andrikopoulos S. A novel mechanism regulating
269. Bugge A, Holst D. PPAR agonists, - could tissue Physiol. 2013;4:237. insulin secretion involving Herpud1 in mice. Dia-
targeting pave the way? Biochimie. 2017;136: 286. Bequette BJ, Sunny NE, El-Kadi SW, Owens SL. betologia. 2013;56(7):1569–1576.
100–104. Application of stable isotopes and mass isotopomer 302. Williams EG, Wu Y, Jha P, Dubuis S, Blattmann P,
270. Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in distribution analysis to the study of intermediary Argmann CA, Houten SM, Amariuta T, Wolski W,
obesity-induced T2DM, dyslipidaemia and NAFLD. metabolism of nutrients. J Anim Sci. 2006;84(suppl): Zamboni N, Aebersold R, Auwerx J. Systems pro-
Nat Rev Endocrinol. 2017;13(1):36–49. E50–E59. teomics of liver mitochondria function. Science.
271. Franko A, Huypens P, Neschen S, Irmler M, Rozman 287. Alves TC, Pongratz RL, Zhao X, Yarborough O, 2016;352(6291):aad0189.
J, Rathkolb B, Neff F, Prehn C, Dubois G, Baumann Sereda S, Shirihai O, Cline GW, Mason G, Kibbey RG. 303. Blais EM, Rawls KD, Dougherty BV, Li ZI, Kolling GL,
M, Massinger R, Gradinger D, Przemeck GKH, Repp Integrated, step-wise, mass-isotopomeric flux Ye P, Wallqvist A, Papin JA. Reconciled rat and
B, Aichler M, Feuchtinger A, Schommers P, Stöhr O, analysis of the TCA cycle. Cell Metab. 2015;22(5): human metabolic networks for comparative tox-
Sanchez-Lasheras C, Adamski J, Peter A, Prokisch H, 936–947. icogenomics and biomarker predictions. Nat
Beckers J, Walch AK, Fuchs H, Wolf E, Schubert M, 288. Ahn WS, Antoniewicz MR. Parallel labeling exper- Commun. 2017;8:14250.
Wiesner RJ, Hrabě de Angelis M. Bezafibrate im- iments with [1,2-(13)C]glucose and [U-(13)C]glu- 304. Bardova K, Horakova O, Janovska P, Hansikova J,
proves insulin sensitivity and metabolic flexibility in tamine provide new insights into CHO cell Kus V, van Schothorst EM, Hoevenaars FPM, Uil
STZ-induced diabetic mice. Diabetes. 2016;65(9): M, Hensler M, Keijer J, Kopecky J. Early differences
metabolism. Metab Eng. 2013;15(1):34–47.
2540–2552. in metabolic flexibility between obesity-resistant
289. Chen WW, Freinkman E, Wang T, Birsoy K, Sabatini
272. Wallenius K, Kjellstedt A, Thalén P, Löfgren L, Oakes and obesity-prone mice. Biochimie. 2016;124:
DM. Absolute quantification of matrix metabolites
ND. The PPAR a/ghagonist, tesaglitazar, improves 163–170.
reveals the dynamics of mitochondrial metabolism.
insulin mediated switching of tissue glucose and 305. Yen CA, Curran SP. Gene-diet interactions and
Cell. 2016;166(5):1324–1337.
free fatty acid utilization in vivo in the obese Zucker aging in C. elegans. Exp Gerontol. 2016;86:
290. Previs SF, Kelley DE. Tracer-based assessments of
rat. PPAR Res. 2013;2013:305347. 106–112.
hepatic anaplerotic and TCA cycle flux: practi-
273. Sugii S, Olson P, Sears DD, Saberi M, Atkins AR, 306. Hylander BL, Repasky EA. Thermoneutrality, mice,
Barish GD, Hong S-H, Castro GL, Yin Y-Q, Nelson cality, stoichiometry, and hidden assumptions. and cancer: a heated opinion. Trends Cancer. 2016;
MC, Hsiao G, Greaves DR, Downes M, Yu RT, Am J Physiol Endocrinol Metab. 2015;309(8): 2(4):166–175.
Olefsky JM, Evans RM. PPARgamma activation in E727–E735. 307. Argmann CA, Chambon P, Auwerx J. Mouse phe-
adipocytes is sufficient for systemic insulin sensi- 291. Abedpour N, Kollmann M. Resource constrained nogenomics: the fast track to “systems metabolism.”
tization. Proc Natl Acad Sci USA. 2009;106(52): flux balance analysis predicts selective pressure on Cell Metab. 2005;2(6):349–360.
22504–22509. the global structure of metabolic networks. BMC 308. Bordbar A, Palsson BO. Using the reconstructed
274. Kung J, Henry RR. Thiazolidinedione safety. Expert Syst Biol. 2015;9(1):88. genome-scale human metabolic network to study
Opin Drug Saf. 2012;11(4):565–579. 292. Kuehne A, Mayr U, Sévin DC, Claassen M, physiology and pathology. J Intern Med. 2012;271(2):
275. Laplante M, Sabatini DM. mTOR signaling in Zamboni N. Metabolic network segmentation: 131–141.
growth control and disease. Cell. 2012;149(2): a probabilistic graphical modeling approach to 309. Thiele I, Swainston N, Fleming RMT, Hoppe A,
274–293. identify the sites and sequential order of meta- Sahoo S, Aurich MK, Haraldsdottir H, Mo ML,
276. Düvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, bolic regulation from non-targeted metab- Rolfsson O, Stobbe MD, Thorleifsson SG, Agren R,
Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver olomics data. PLOS Comput Biol. 2017;13(6): Bölling C, Bordel S, Chavali AK, Dobson P, Dunn
S, Vander Heiden MG, MacKeigan JP, Finan PM, e1005577. WB, Endler L, Hala D, Hucka M, Hull D, Jameson D,

516 Smith et al Metabolic Flexibility in Health and Disease Endocrine Reviews, August 2018, 39(4):489–517
REVIEW

Jamshidi N, Jonsson JJ, Juty N, Keating S, Nookaew I, Le 315. Taghipoor M, van Milgen J, Gondret F. A systemic Disclosure Summary: The authors have nothing to
Novère N, Malys N, Mazein A, Papin JA, Price approach to explore the flexibility of energy stores disclose.
ND, Selkov E Sr, Sigurdsson MI, Simeonidis E, at the cellular scale: examples from muscle cells.
Sonnenschein N, Smallbone K, Sorokin A, van Beek J Theor Biol. 2016;404:331–341.
JHGM, Weichart D, Goryanin I, Nielsen J, Westerhoff 316. Vernieri C, Casola S, Foiani M, Pietrantonio F, de Abbreviations
HV, Kell DB, Mendes P, Palsson BØ. A community- Braud F, Longo V. Targeting cancer metabolism: ACC, acetyl-CoA carboxylase; AICAR, 5-aminoimidazole-4-
driven global reconstruction of human metabolism. dietary and pharmacologic interventions. Cancer carboxamide riboside; Akt/PKB, v-Akt murine thymoma viral
Nat Biotechnol. 2013;31(5):419–425. Discov. 2016;6(12):1315–1333. oncogene homolog 1/protein kinase B; AMPK, adenosine
310. Swainston N, Smallbone K, Hefzi H, Dobson PD, 317. Riscuta G. Nutrigenomics at the interface of aging, monophosphate-activated protein kinase; BAT, brown adi-
Brewer J, Hanscho M, Zielinski DC, Ang KS, Gardiner lifespan, and disease pevention. J Nutr. 2013;146(10): pose tissue; BCAA, branched chain amino acids; BCKD,
NJ, Gutierrez JM, Kyriakopoulos S, Lakshmanan M, Li 1931–1939. branched-chain a-ketoacid dehydrogenase; CoA, coenzyme
S, Liu JK, Martı́nez VS, Orellana CA, Quek L-E, 318. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, A; CPT-1, carnitine palmitoyltranferase-1; CR, caloric re-
Weinberger A, Ben-Yacov O, Lador D, Avnit-Sagi T, striction; ER, endoplasm reticulum; FAO, fatty acid oxidation;
Thomas A, Zanghellini J, Borth N, Lee D-Y, Nielsen LK,
FATP1, fatty acid transport protein 1; FGF21, fibroblast
Kell DB, Lewis NE, Mendes P. Recon 2.2: from re- Lotan-Pompan M, Suez J, Mahdi JA, Matot E, Malka
growth factor 21; FOXO, forkhead box protein O; GLP-1,
construction to model of human metabolism. G, Kosower N, Rein M, Zilberman-Schapira G,
glucagonlike peptide-1; GLUT, glucose transporter; HIF1a,
Metabolomics. 2016;12(7):109. Dohnalová L, Pevsner-Fischer M, Bikovsky R,
hypoxia-inducible factor 1 alpha; HK, hexokinase; iNOS, in-
311. McGarrity S, Halldórsson H, Palsson S, Johansson PI, Halpern Z, Elinav E, Segal E. Personalized nutrition

Downloaded from https://academic.oup.com/edrv/article/39/4/489/4982126 by guest on 23 August 2021


ducible nitric oxide synthase; KB, ketone body; LAT1, large
Rolfsson Ó. Understanding the causes and im- by prediction of glycemic responses. Cell. 2015;
neutral amino acid antiporter; MEF2, myocyte-specific en-
plications of endothelial metabolic variation in 163(5):1079–1094. hancer factor 2; mtDNA, mitochondrial DNA; mTOR,
cardiovascular disease through genome-scale 319. Amoedo NDD, Obre E, Rossignol R. Drug discovery mechanistic target of rapamycin; mTORC1/2, mechanistic target of
metabolic modeling. Front Cardiovasc Med. 2016;3: strategies in the field of tumor energy metabolism: rapamycin complex 1/2; Myc, myelocytomatosis oncogene; NAD+,
10. Limitations by metabolic flexibility and metabolic oxidized nicotinamide adenine dinucleotide; NADH, reduced
312. Pearson T, Wattis JAD, King JR, MacDonald IA, resistance to chemotherapy. Biochim Biophys Acta. nicotinamide adenine dinucleotide; NADPH, reduced nico-
Mazzatti DJ. The effects of insulin resistance on 2017;1858(8):674–685. tinamide adenine dinucleotide phosphate; NO, nitric oxide;
individual tissues: an application of a mathematical NRF, nuclear respiratory factor; OXPHOS, oxidative phos-
model of metabolism in humans. Bull Math Biol. phorylation; PDH, pyruvate dehydrogenase; PDK, pyruvate
2016;78(6):1189–1217. Acknowledgments dehydrogenase kinase; PGC1a, peroxisome proliferator–
313. Nogiec C, Burkart A, Dreyfuss JM, Lerin C, Kasif S, Patti Financial Support: This work was supported by European activated receptor g coactivator 1-a; PI3K, phosphatidyli-
M-EE. Metabolic modeling of muscle metabolism Research Council Starting Grant 638290 and ZonMw-VIDI nositol 3-kinase; PPAR, peroxisome proliferator–activated
identifies key reactions linked to insulin resistance Grant 917.15.305 to (R.H.H.). receptor; PPP, pentose phosphate pathway; ROS, reactive
phenotypes. Mol Metab. 2015;4(3):151–163. Correspondence and Reprint Requests: Riekelt H. oxygen species; SIRT, sirtuin; T2DM, type 2 diabetes mellitus;
314. Zhang S, Hulver MW, McMillan RP, Cline MA, Gilbert ER. Houtkooper, PhD, Laboratory of Genetic Metabolic Diseases, TCA, tricarboxylic acid; TFAM, mitochondrial transcription
The pivotal role of pyruvate dehydrogenase kinases in Academic Medical Center, Meibergdreef 9, 1105 AZ factor A; TFEB, helix-loop-helix leucine zipper transcription
metabolic flexibility. Nutr Metab (Lond). 2014;11(1):10. Amsterdam, Netherlands. E-mail: r.h.houtkooper@amc.nl. factor EB; WAT, white adipose tissue.

doi: 10.1210/er.2017-00211 https://academic.oup.com/edrv 517

You might also like