Вы находитесь на странице: 1из 47

with particular focus on the molecular regulation of the two main lipases, ATGL and HSL, and

the intracellular and extracellular signals affecting their activity.


Journal of Molecular Endocrinology
(2014) 52, R199R222

T S NIELSEN

and others

Dissecting adipose tissue


lipolysis

Review

52 :3

R199R222

Dissecting adipose tissue lipolysis:


molecular regulation and
implications for metabolic disease
Thomas Svava Nielsen

1,2

, Niels Jessen

Niels Mller and Sten Lund

2,3

, Jens Otto L Jrgensen ,

The Novo Nordisk Foundation Center for Basic Metabolic Research, Section on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b
8000 Aarhus C, Denmark
3
Department of Molecular Medicine, Aarhus University Hospital, Brendstrupga rdsvej 100, 8200 Aarhus N, Denmark

Abstract
Lipolysis is the process by which triglycerides (TGs) are hydrolyzed to free fatty acids (FFAs)
and glycerol. In adipocytes, this is achieved by sequential action of adipose TG lipase
Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Key Words

(ATGL), hormone-sensitive lipase (HSL), and monoglyceride lipase. The activity in the

"

lipolysis

lipolytic pathway is tightly regulated by hormonal and nutritional factors. Under conditions

"

adipose tissue

of negative energy balance such as fasting and exercise, stimulation of lipolysis results in a

"

ATGL

to provide the organism with a sufficient supply of substrate for oxidative metabolism.

"

HSL

However, failure to efficiently suppress lipolysis when FFA demands are low can have

"

free fatty acids

"

type 2 diabetes

profound increase in FFA release from adipose tissue (AT). This response is crucial in order

serious metabolic consequences and is believed to be a key mechanism in the


development of type 2 diabetes in obesity. As the discovery of ATGL in 2004, substantial
progress has been made in the delineation of the remarkable complexity of the
regulatory network controlling adipocyte lipolysis. Notably, regulatory mechanisms have
been identified on multiple levels of the lipolytic pathway, including gene transcription
and translation, post-translational modifications, intracellular localization, proteinprotein
interactions, and protein stability/ degradation. Here, we provide an overview of the
recent advances in the field of AT lipolysis

Introduction
The major energy reserve in mammals consists of fat
stored in adipose tissue (AT). In periods of excess energy
intake, dietary lipids are taken up by fat cells
(adipocytes) in AT and esterified into triglycerides (TGs),
which are stored in cytosolic lipid droplets (LDs). In
conditions like fasting and exercise, when mobilization
of endogenous energy stores is required, TG is
hydrolyzed through the process of lipolysis and released
to the circulation as free fatty acids

http://jme.endocrinology-journals.org
DOI: 10.1530/JME-13-0277

2014 Society for


Endocrinology
Printed in Great

(FFAs). These are delivered to peripheral tissues where


they can serve as substrate for b-oxidation and ATP
production. Only adipocytes have the ability to secrete
FFAs into the circulation (Kolditz & Langin 2010).
Hence, in the post- absorptive state and during
physical exercise, the vast majority of the systemic FFA
originates from AT (Jensen 2003). The unique ability of
AT to balance storage and release of lipids in response
to altered nutrient demands

Published by Bioscientifica Ltd.

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

provides the organism with an FFA-buffering system of


essentially unlimited capacity (Frayn 2002). However, the
metabolic consequences of an excessive expansion of AT
are considerable. In humans, obesity is closely associated
with numerous risk factors that constitute the so-called
metabolic syndrome. This includes abdominal obesity,
hypertension, dyslipidemia, and glucose intolerance,
which are key elements in the pathogenesis of cardiovascular disease and type 2 diabetes (Alberti et al. 2009). The
physiological link between obesity and metabolic disease
in humans is currently not completely understood, and
one of the great enigmas is the remarkable individual
differences in the predisposition to obesity-induced
metabolic disease. This has led to the proposal of the
AT expandability hypothesis (Virtue & Vidal-Puig 2010,
Hardy et al. 2012), which states that the capacity of AT to
expand appropriately when lipid storage is needed is
limited for a given individual. Hence, when the limit is
exceeded, lipids begin to accumulate in ectopic tissues
causing metabolic dysfunction and insulin resistance due
to lipotoxic effects. An emerging view is that this
lipotoxicity is likely not caused by excess TG in itself,
but rather by an excess of lipid intermediates and
metabolites released from hypertrophic adipocytes.
Several recent reviews have addressed these mechanisms
in detail (Boura-Halfon & Zick 2009, Virtue & Vidal-Puig
2010, Copps & White 2012, Hardy et al. 2012, Zechner
et al. 2012, Czech et al. 2013). Notably, it is now clear that
besides serving as energy-dense metabolic substrates,
most, if not all, lipolytic products and intermediates like
diacylglycerol (DG), monoacylglycerol (MG), and FFA
(and metabolites derived from these) play essential roles
in multiple signaling pathways, both at the systemic and
at the intracellular level (Zechner et al. 2012). When
present in excess, several of these lipid intermediates
have been suggested to induce insulin resistance in
ectopic tissues by interfering with insulin signaling at the
level of the insulin receptor substrate (IRS) proteins
(Boura-Halfon
& Zick 2009, Copps & White 2012).
Being the major lipid species released from AT, FFA
is likely one of the key elements in ectopic lipid
accumu- lation and lipotoxicity. Thus, experimental
elevation of plasma FFA levels in human subjects
acutely and dose dependently counteracts peripheral
insulin-stimulated glucose uptake and oxidation (Belfort
et al. 2005, Gormsen et al. 2007, Hoeg et al. 2011).
Furthermore, high FFA levels attenuate the insulinmediated suppression of hepatic glucose production
contributing to the impairment of whole-body glucose

Dissecting adipose tissue


lipolysis

52 :3

R200

tolerance (Roden et al. 2000). Consist- ently, improvements


in whole-body insulin sensitivity

and oral glucose tolerance can be obtained by


pharma- cological reductions of chronically elevated
plasma FFA levels both in type 2 diabetic patients,
obese nondiabetic subjects, and nondiabetic subjects
genetically predisposed to
type
2
diabetes
(Santomauro et al. 1999, Cusi et al. 2007).
Accordingly, excessive FFA mobilization from AT is
widely conceived as playing a pivotal role in insulin
resistance and type 2 diabetes, suggesting that
dysregula- tion of AT lipolysis in the obese state is a
contributing factor to the development of metabolic
disease.

Major signaling pathways in AT lipolysis


Lipolysis is the sequential hydrolysis of one TG
molecule into three FFAs and one glycerol by a class of
hydrolytic enzymes commonly known as lipases. In
mammalian lipolysis, three lipases act in sequence with
the concomi- tant release of one FFA in each step (Fig.
1); adipose TG lipase (ATGL) converts TG to DG and is
the rate-limiting enzyme in the lipolytic pathway
(Zimmermann et al. 2004). DG is hydrolyzed to MG by
hormone-sensitive lipase (HSL; Haemmerle et al. 2002),
and monoglyceride lipase (MGL) cleaves MG into
glycerol and FFA (Fredrikson et al. 1986). The major
positive
regulators
of
human
lipolysis
are

catecholamines and natriuretic peptides (NPs), while


antilipolysis primarily is mediated by insulin and
catecholamines.
Catecholamines
The catecholamines, and specifically the stress hormones
adrenaline and noradrenaline, are the primary mediators
of adrenergic signaling in AT. The manner by which
catecholamines regulate lipolysis is unusual as these
hormones are able to both stimulate and inhibit lipolysis
depending on their relative affinity for different
adrenergic receptors (ARs). Thus, stimulation of lipolysis
requires the activation of b-ARs on the surface of the
adipocyte, while antilipolytic signals are transmitted
by the a2-AR
ATGL
TG

HSL
DG
FFA

MGL
MG
FFA

Glycerol
FFA

Figure 1
Schematic illustration of the lipolytic pathway. To fully hydrolyze TGs,
ATGL, HSL, and MGL act in sequence, with the release of one FFA in
each step. This successively converts TG to DG, then to MG, and finally
to glycerol and a total of tree FFA. TG, triglyceride; ATGL, adipose TG
lipase; DG, diacylglycerol; FFA, free fatty acid; HSL, hormone-sensitive
lipase; MG,
monoacylglycerol; MGL, monoglyceride lipase.

Review

T S NIELSEN

and others

(Robidoux et al. 2004; Fig. 2). Three different b-AR


subtypes exist (b1, b2, and b3), but in humans, only
the b1 and b2 isoforms are involved in lipolysis (Mauriege
et al. 1988, Barbe et al. 1996, Tavernier et al. 1996). Both
a2-AR and b-AR belong to the G-protein-coupled
receptor (GPCR) family: the G-protein associated with
a2-AR contain the inhibitory Gi subunit, while b-ARassociated

G-proteins

contain

the

stimulating

Gs

subunit (Lafontan
& Berlan 1993). The activation of the receptors causes the
G-proteins to interact with adenylyl cyclase (AC), which
is inhibited by interaction with Gi and activated by
interaction with Gs (Lafontan & Berlan 1993). Upon
activation, AC converts ATP to cAMP, resulting in an
increase in intracellular cAMP levels, which activates
protein kinase A (PKA, also known as cAMP-dependent
protein kinase; Langin 2006). Activated PKA phosphorylates the LD-associated protein PLIN1 (Greenberg
Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Garton et al. 1988, Anthonsen et al. 1998). Phosphorylation of PLIN1 promotes the release of comparative gene
identification-58 (CGI-58), which is a potent co-activator
of ATGL (Lass et al. 2006, Granneman et al. 2009). This
facilitates the activation of ATGL, thus initiating the
stimulated lipolytic cascade. Furthermore, PKA-mediated
phosphorylation of HSL causes a rapid activation and
translocation of the lipase from the cytosol to the surface
of the LDs (Egan et al. 1992). Here, it docks on the
phosphorylated PLIN1 and thereby gains access to its DG
substrate, which is being generated by ATGL (Shen et al.
2009, Wang et al. 2009).
Natriuretic peptides
In addition to catecholamines, the cardiac hormones
atrial NP (ANP) and B-type NP (BNP) are important
positive regulators of AT lipolysis in humans. NPs, which

NPR-A

1/2-AR

AC

Gi

GC

Cytosol

Gs

ATP

PKB/Akt

GTP
cAMP

PIP3
PI3K

cGMP

PDE3B

PDE5
PKA

IR

PKG

5-AMP

PIP2

R201

et al. 1991) and cytoplasmic HSL (Stralfors et al. 1984,

2-AR

PDK

52 :3

Dissecting adipose tissue


lipolysis

5-GMP

IRS1/2

HSL

P
P

P P

HSL

Three FFA
+ glycerol
P

Figure 2
Primary signaling pathways in human lipolysis. Black and red lines
indicate pro-lipolytic and anti-lipolytic signaling events, respectively.
Arrows
indicate stimulation and/or translocation and blunt lines indicate inhibition. Stimulation of lipolysis is dependent on PKA- or PKG-mediated
phosphorylation of HSL and PLIN1. PKG is activated by cGMP, which
is
increased in response to activation of the GC-coupled NPR-A. Similarly,
stimulation of the Gs-protein-coupled b1/2-ARs activates AC, which
generates cAMP and activates PKA. Conversely, activation of Gi-protein-

MGL

MG

DG
TG

P
P

Lipid droplet
coupled a2-ARs inhibits AC and thereby reduces cAMP-dependent
signaling to lipolysis. Stimulation of the insulin signaling pathway through
the IR
increases the activity of PDE3B, which converts cAMP to 50 -AMP, thus
decreasing PKA activity and suppressing lipolysis. PKG activity is reduced by

PDE5-mediated conversion of cGMP to 5 0 -GMP, although the upstream


signals regulating this process are currently unknown. The dashed line
indicates a putative Akt-independent insulin pathway acting
selectively on PLIN1. a2-ARs, a2-adrenergic receptors; AC, adenylyl
cyclase; TG, triglyceride; ATGL, adipose TG lipase; b1/2-ARs, b1- and
b2-adrenergic
receptors; CGI-58, comparative gene identification-58; DG,

diacylglycerol; FFA, free fatty acid; GC, guanylyl cyclase; HSL, hormonesensitive lipase; IR, insulin receptor; IRS1/2, IR substrates 1 and 2; MG,
monoacylglycerol; MGL, monoglyceride lipase; NPR-A, type-A natriuretic
peptide receptor; PDE3B, phosphodiesterase 3B; PDK, phosphoinositidedependent kinase; PI3K,
phosphatidylinositol 3-kinase; PKA, protein kinase A; PKB/Akt, protein
kinase B; PLIN1, perilipin 1.

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

are released from the atrial and ventricular walls of the


heart in response to myotube distension (Clerico et al.
2011), stimulate the guanylyl cyclase (GC)-linked type-A
NP receptor on the adipocytes (Sengenes et al. 2000)
(Fig. 2). Accordingly, upon stimulation of the receptor,
GC converts intracellular GTP into cGMP resulting in the
activation of PKG (also known as the cGMP-dependent
protein kinase), and, just like PKA, this kinase activates
the lipolytic cascade by phosphorylation of PLIN1 and
HSL (Sengenes et al. 2003). However, despite the
similarities between PKA- and PKG-mediated lipolysis,
they are distinct pathways and, unlike the cAMPdependent
pathway,
NP-mediated
lipolysis
is
unresponsive
to
the
antilipolytic
effects
of
phosphodiesterase 3B (PDE3B; Sengenes et al. 2000,
Moro et al. 2004a). Instead, counter-regulation of the
NP pathway is believed to occur by hydrolysis of
cGMP by other members of the PDE family of PDEs
(Armani et al. 2011). Indeed, PDE5 expression and
activity has been found in isolated human adipocytes
from both subcutaneous AT (Moro et al. 2007a) and
visceral AT (Aversa et al. 2011); however, this enzyme
appears to be insufcient to control ANP-mediated
lipolysis (Moro et al. 2007a). Hence, at present, the details
of the regulatory pathways counteracting the lipolytic
action of NPs in vivo remains poorly understood (Armani
et al. 2011).
Insulin
Lipolysis is exceptionally sensitive to the action of insulin
(Jensen & Nielsen 2007), which constitutes the major
antilipolytic pathway in human lipolysis (Fig. 2). The IR
possesses intrinsic tyrosine kinase activity. Thus, binding
of insulin induces IR autophosphorylation and subsequent phosphorylation of the IRS1/2 (White 1998).
This promotes the activation of phosphatidylinositol 3kinase (PI3K), which converts phosphatidylinositol- 4,5bisphosphate (PIP2) into phosphatidylinositol-3,4,5triphosphate (PIP3) (Whitman et al. 1988, Carpenter et al.
1990). Generation of PIP3 activates the phosphoinositidedependent kinase causing phosphorylation and
activation of Akt (also known as PKB; Alessi et al. 1997,
Stokoe et al. 1997). Finally, PKB/Akt activates PDE3B,
which degrades cAMP to 50 -AMP (Choi et al. 2006). This
inactivates PKA leading to reduced phosphorylation of
HSL and PLIN1 and suppression of lipolysis.
Interestingly, in a study predating the elucidation of
the canonical insulin signaling pathway, it was demon-

Dissecting adipose tissue


lipolysis

52 :3

R202

strated that besides the inhibitory effect on PKA-mediated


signaling, insulin also acts to reduce lipolysis in primary

rat adipocytes by a cAMP-independent mechanism


(Londos et al. 1985). By carefully measuring PKA
activity ratios in response to increasing concentrations
of iso- prenaline (a b-AR agonist), the authors found
that when insulin was added to the cells at
submaximal
lipolytic stimulation, the insulinmediated reduction in PKA activity was not sufficient
to explain the resulting drop in lipolysis. Conversely,
under conditions of maximal lipolysis, insulin-mediated
changes in PKA activity could fully account for the
resulting change in lipolytic rates. A recent study in
3T3-L1 adipocytes has partially delineated this
bimodal insulin effect by showing that insulinmediated antilipolysis at submaximal b-AR stimulation (but not at maximal stimulation) can proceed
through an alternative PI3K-dependent pathway that
is independent of Akt (Choi et al. 2010). Acting
through as yet unidentified downstream effectors, the
pathway was shown to inhibit lipolysis by selectively
reducing PLIN1 phosphorylation without affecting the
phosphorylation status of HSL (Fig. 2). Considering the
key role of PLIN1 in the regulation of ATGL- and HSLmediated lipolysis such a pathway would indeed be
expected to have a substantial impact on overall
lipolytic rates.
Besides the inhibitory effects on the lipolytic
pathway, insulin also promotes lipid storage by

activating a range of pathways involved in the uptake,


synthesis, and storage of TG in adipocytes. A
comprehensive review of these lipogenic effects of
insulin has been published recently (Czech et al. 2013).

Alternative regulatory pathways


Although catecholamines, insulin, and NPs represent the
major regulators of human lipolysis, several other factors
can modulate lipolysis in AT, either directly by receptormediated signaling or indirectly by remodeling of the
lipolytic cascade. Figure 3 shows an overview of the
different alternative pathways described below.
Agents acting through cAMP-dependent signaling
GPCR pathways affecting the activity of AC are particularly numerous, emphasizing the central role of the
cAMP- dependent pathway in the regulation of TG
hydrolysis.
Thyroid-stimulating hormone (TSH) and the melanocortins (MCs) adrenocorticotrophic hormone (ACTH) and
melanocyte-stimulating hormone stimulate AC by activation of the Gs-coupled TSH receptor (Laugwitz et al.
1996, End o & Kobayashi 2012) and MC receptors
(Cho et al. 2005, Rodrigues et al. 2013) respectively (Fig. 3).

Review

T S NIELSEN

and others

Dissecting adipose tissue


lipolysis

Growth hormone

A1-R
HM74a GPR81
2-AR

MC
TSH-r

1/2-AR

Gi
TNFR1

AC

Gi

Gs

R203

ANGPTL4

NPY-Y 1

Glucocorticoids

52 :3

AC

ANGPTL4-r

Gs

ATP

ATP
cAMP
PKA

IR

HSL

Three FFA
+ glycerol

MGL

DG
Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

TG
Figure 3
Alternative signaling pathways in lipolysis. Black and red lines indicate
pro- lipolytic and anti-lipolytic signaling events, respectively. Arrows
indicate
stimulation and blunt lines indicate inhibition. Dashed lines illustrate the
indirect lipolytic effects of growth hormone and glucocorticoids by
modulation of receptor sensitivities and ANGPTL4-mediated signaling.
Although the identity of the ANGPTL4 receptor is unknown, the
intracellular signaling has been shown to involve activation of AC.
Stimulation of the Gs-protein-coupled melanocortin (MC) receptor and TSH
receptor (TSH-r) also increases intracellular cAMP levels through activation
of AC. Conversely, the Gi-protein-coupled receptors for NPY/PYY (NPY-Y1),
adenosine (A1-R), b-hydroxybutyrate (HM74a), and lactate (GPR81)

TSH-mediated lipolysis has been found to be particularly


important in neonates and newborns because physiological levels of TSH, as opposed to adrenaline or noradrenaline, potently stimulate human lipolysis at this
developmental stage (Marcus et al. 1988, Janson et al.
1995). By contrast, although a strong lipolytic potential
of the MCs has been observed in several animal species,
including rodents, hamsters, guinea pigs, and rabbits
(Richter & Schwandt 1983, Ng 1990), they seem to have
limited effects on human lipolysis (Bousquet-Melou et al.
1995, Kiwaki & Levine 2003).
Neuropeptide Y (NPY) and peptide YY (PYY) are
released from sympathetic neurons and inhibit AC by
binding to the Gi-protein-coupled NPY-receptor (NPY-Y1;
Fig. 3) on human adipocytes (Serradeil-Le et al. 2000).
Also, in humans, the highest expression of NPY
receptors has been found in subcutaneous AT (Castan et

MG

Lipid droplet
suppress lipolysis by inhibition of AC. Pro-inflammatory signaling through
the TNF-a receptor (TNFR-1) increases lipolysis by suppressing
antilipolytic signaling mediated by the insulin receptor (IR) and a2adrenergic receptors (a2-ARs). For clarity, the intermediate intracellular
steps in the different
signaling pathways have been omitted. AC, adenylyl cyclase;
ANGPTL4, angiopoietin-like protein 4; ATGL, adipose triglyceride
lipase; b1/2-ARs, b1- and b2-adrenergic receptors; CGI-58, comparative
gene
identification-58; DG, diacylglycerol; FFA, free fatty acid; Gi, inhibitory
G-protein; Gs, stimulating G-protein; HSL, hormone-sensitive lipase; MG,
monoacylglycerol; MGL, monoglyceride lipase; PKA, protein kinase A;
PLIN1, perilipin 1; TG, triglyceride; TSH, thyroid-stimulating hormone.

al. 1993), suggesting that the impact of NPY/PYY on


lipolysis is depot specific. The importance of neuronal
regulation of

lipolysis has been underscored by elegant studies in


rodents demonstrating that isolated stimulation of the
hypothalamus with insulin suppresses peripheral AT
lipolysis (Scherer et al. 2011).
The
fasting-induced
circulating
factor
angiopoietin- like protein 4 (ANGPTL4) is a wellestablished negative regulator of lipid uptake in
rodent adipocytes through inhibition of extracellular
lipoprotein lipase activity (Koster et al. 2005,
Sukonina et al. 2006, Lafferty et al. 2013). Recently,

however, ANGPTL4 has also been found to be intimately


involved in the regulation of intracellular cAMPmediated lipolysis in mice (Gray et al. 2012). By an as yet
unidentified mechanism, extracellular ANGPTL4 was
shown to act independently of b-AR activation to
increase intracellular cAMP production via activation of
AC (Gray et al. 2012). The identity of the putative
ANGPTL4 receptor responsible for this effect in
adipocytes is unknown and is currently under
investigation (Koliwad et al. 2012).

Review

T S NIELSEN

and others

In rat and human adipocytes, extracellular adenosine


efciently inhibits lipolysis via the Gi-coupled adenosine
receptor (A1-R; Fig. 3; Lonnroth et al. 1989, Liang et al.
2002). However, in humans, the concentrations required
in the interstitial fluid for a significant reduction of
lipolysis have been found to be at the very high end of
the physiological range and therefore of uncertain
significance (Lonnroth et al. 1989). The ketone body bhydroxybutyrate (b-OHB) has been shown to inhibit
lipolysis in vitro by activating the human Gi-coupled

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

receptor HM74a (Fig. 3; Taggart et al. 2005). HM74a,


which is the ortholog of the mouse PUMA-G receptor, is
also the target of the lipid-lowering drug nicotinic acid
(niacin; Tunaru et al. 2003). Importantly, the observed
inhibition of lipolysis by b-OHB was obtained at concentrations similar to those seen in humans during fasting,
suggesting a feedback mechanism by which b-OHB can
regulate its own production in order to prevent ketoacidosis during starvation (Taggart et al. 2005). Similarly, the
receptor responsible for the antilipolytic effect of lactate
has been identified as GPR81 (Fig. 3; Cai et al. 2008, Liu
et al. 2009a), which is a Gi-coupled receptor highly
homologous to HM74a (Cai et al. 2008). Like b-OHB,
lactate inhibits lipolysis in adipocytes from several
mammalian species including primates, rodents, and
humans at concentrations within the normal physiological range (Liu et al. 2009a). Consequently, it has been
hypothesized that GPR81 could act as a sensor of hypoxia
by suppressing lipolysis in response to increased lactate
production (Cai et al. 2008).
Growth hormone
Growth hormone (GH) potently and dose dependently
stimulates lipolysis in humans (Hansen et al. 2002). In
mice, knockout (KO) of the GH receptor renders the
animals susceptible to obesity, while GH over-expression
results in a lean phenotype (Berryman et al. 2004, 2006).
The nature of the molecular pathway involved in
GH-mediated lipolysis is not entirely clear. However,
results from animal studies indicate that it involves
remodeling of the cAMP-dependent regulatory signaling
pathways such that the responsiveness toward badrenergic signaling is increased (Doris et al. 1994, Yang
et al. 2004) while insulin sensitivity is reduced (Chen et
al. 2001, Johansen et al. 2003; Fig. 3). Although
definitive mechanistic evidence is lacking, this model has
been supported by human studies. Thus, GH administration acutely stimulates lipolysis and causes peripheral

Dissecting adipose tissue


lipolysis

52 :3

R204

insulin resistance (Nellemann et al. 2013). Also, the in vivo

lipolytic effect of GH is counteracted by the AC


inhibitor acipimox (a niacin derivative; Nielsen et al.
2001, 2002). Notably, stimulation with GH does not
increase lipolysis in explants of human AT (Fain et al.
2008) or isolated human adipocytes (Marcus et al.
1994), but the sensitivity toward b-adrenergic agonists is
enhanced by the presence of GH in the culture medium
(Marcus et al. 1994). In line with this, it has recently
been demonstrated that GH administration in human
subjects increases ANGPTL4 levels in plasma (Clasen et
al. 2013). Given the permissive effect of this protein on
cAMP-mediated lipolysis, it seems likely that elevations
in systemic ANGPTL4 levels could be one of the
mechanisms by which GH stimulates AT lipolysis.
Interestingly,
the
responsiveness
toward
GH
stimulation varies among human AT depots, and
visceral AT has been shown to be particularly
sensitive to the lipolytic effects of GH (Nam et al. 2001,
Pasarica et al. 2007, Plockinger & Reuter 2008).
Glucocorticoids
In a manner similar to GH, the lipolytic effects of
glucocorticoids have been attributed to an increased
b-adrenergic responsiveness and a reduction of
insulin- mediated antilipolysis (Fig. 3). Thus, in rat

adipocytes, dexamethasone treatment has been shown


to promote PKA-mediated lipolysis by reducing the
mRNA and protein expression levels of PDE3B (Xu et
al. 2009). Also, dexamethasone potentiates the response
toward b-AR agonists both by inducing an increase in
the number of b-ARs and by increasing the catalytic
response of AC toward receptor-mediated activation
(Lacasa et al. 1988). In agreement with this, elevated
cortisol levels in humans have been found to reduce the
postprandial suppression of FFA release, suggesting a
decreased antilipolytic effect of insulin (Dinneen et al.
1993). As for GH, glucocorticoid- mediated lipolysis in
rodents is partially dependent on an increase in
ANGPTL4 (Koliwad et al. 2009, Gray et al. 2012),
indicating that these hormones share some of the
mechanisms by which they stimulate lipolysis.
However, acute in vivo studies on humans have also
demonstrated additive independent effects of GH and
cortisol on lipolysis, suggesting that alternative, and
distinct, lipolytic pathways exist for these hormones
(Djurhuus et al. 2004).
Tumor necrosis factor a
Multiple effects of the pro-inflammatory cytokine tumor
necrosis factor-a (TNFa) on the lipolytic pathway have

Review

T S NIELSEN

and others

Dissecting adipose tissue


lipolysis

been described. Acting through TNF receptor 1 ( Fig. 3) in


adipocytes from mice (Sethi et al. 2000) and humans
(Ryden et al. 2002), TNFa activates the three MAPKs
p42/44, JNK, and p38 of which p42/44 and JNK are
involved in the induction of lipolysis (Ryden et al. 2002).
In human fat cells, PDE3B protein expression is
decreased dramatically by TNFa (Zhang et al. 2002) and
in rat adipocytes antilipolytic signaling via the a2-AR is
blunted by TNFa by specific proteasomal degradation of
Gi (Gasic et al. 1999, Botion et al. 2001). The combined
effect of these alterations of the insulin and a2-AR
signaling pathway is an increased intracellular cAMP
level and a resulting activation of PKA-mediated
lipolysis. In addition to modulation of antilipolytic
signaling, exposure to TNFa increases ATGL activity
due to remodeling of core components of the lipolytic
machinery. This is discussed in the following section.
Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Physiological
lipolysis

regulation

of

human

AT

As discussed earlier, the lipolytic rate in human AT is


determined by a delicate balance between several regulatory pathways. In healthy subjects, this regulation facilitates a proper lipolytic response to changes in systemic
nutrient demand.
Feeding/fasting
Following the ingestion of a meal, the post-prandial
increase in plasma insulin efciently suppresses lipolysis
to promote the storage of dietary lipids (Roust & Jensen
1993, Jensen 1995). Conversely, in the fasting state, FFA
mobilization is promoted by the combined effects of
reduced plasma insulin and increased release of
adrenaline and noradrenaline (Gjedsted et al. 2007). Also,
it has been demonstrated that lipolysis is further
promoted by a combination of increased b-adrenergic
sensitivity and decreased insulin sensitivity in AT
during fasting (Jensen et al. 1987). Similarly, in obese
individuals subjected to a hypocaloric diet (!3 MJ/day)
for 28 days, lipolytic stimulation by b-adrenergic
agonists as well as by ANP and BNP was enhanced
significantly (Sengenes et al. 2002). These fasting-induced
changes in hormonal sensitivity are mediated, at least
in part, by GH, which is elevated significantly during
prolonged fasting (Norrelund et al. 2001, 2003,
Vendelbo et al. 2010). Likewise, the diurnal fluctuations
in serum FFA levels mirror the pulsatile secretion

52 :3

R205

pattern of GH, and the nocturnal increase in FFA


during sleep is virtually absent in GH-defi ient patients
(Jorgensen et al. 1990).

Exercise
Physical exercise is the other major situation in
which lipolysis is stimulated in humans and this is
believed to involve the concerted action of several
signaling pathways (Frayn 2010). Thus, circulating
levels of adrenaline, noradrenaline, ANP, GH, and
cortisol increase and insulin decreases in proportion to
exercise intensity, and these gradual changes are
reflected in the magnitude of the resulting lipolytic
response (Moro et al. 2007b). Further- more, the
adrenergic responsiveness of subcutaneous AT is
altered with a shift from predominant a-adrenergic
suppression during rest toward predominant badrenergic stimulation during exercise (Arner et al.
1990), and in the post-exercise recovery phase, b-AR
blockade has been shown to dramatically reduce
plasma levels of FFA and glycerol (Wijnen et al. 1993).
Similar to fasting conditions, GH and cortisol are likely
to be some of the hormonal mediators of these
exercise-induced
alterations
in
adrenergic
responsiveness (Kanaley et al. 2004). The primary
adrenergic stimulus of AT during exercise originates
from circulating catecholamines, with only a minor
contri- bution from noradrenaline released from
sympathetic neurons (Stallknecht et al. 2001, de
Glisezinski et al. 2009). Additionally, the NPs have
been found to play a prominent role in exercise-

induced lipolysis in humans (Moro et al. 2004b, de


Glisezinski et al. 2009), and they have been suggested to
account for most of the non- adrenergic lipolytic
signaling in AT during exercise (Moro et al. 2006,
Lafontan et al. 2008).
Depot-specific regulation of lipolysis
AT is not a homogenous organ, and significant regional
differences exist between depots in terms of hormonal
responsiveness and metabolic activity. Also, the
distribution of body fat is gender specific, with men
generally having a more central (upper-body) and women
a more peripheral (lower-body) fat deposition (Demerath
et al. 2007).
Regarding the lipolytic activity of the different
depots, visceral and subcutaneous abdominal ATs are
generally more responsive toward lipolytic stimuli like
catecholamines
or
prolonged
fasting
than
subcutaneous gluteal and femoral fat (Gjedsted et al.
2007, Manolopoulos et al. 2012). The reduced lipolytic
effect of catecholamines in lower-body fat depots is
caused by enhanced a2-AR and reduced b-AR
responsiveness compared with upper-body depots
(Manolopoulos et al. 2012). Additionally, in upper- body
obese women, the antilipolytic effect of insulin is
blunted in the abdominal depots, which enhances

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

lipolysis further (Nellemann et al. 2012). Another important difference between upper- and lower-body subcutaneous AT is the primary way by which adipogenesis
occurs as obesity develops. Thus, AT can expand either
via an increase in the number of fat cells (i.e.
hyperplasia) or by enlargement of the existing
adipocytes (i.e. hypertro- phy), of which the latter has
been found to be an independent marker for increased
metabolic risk (Weyer et al. 2000, Lundgren et al. 2007).
Importantly, irrespective of gender, subcutaneous
abdominal AT is more prone to expansion by
hypertrophy than subcutaneous femoral AT, which
preferentially undergoes hyperplasia (Tchoukalova et al.
2010).
As a consequence of these regional differences in
adipogenesis and lipolytic responsiveness, it has been
found repeatedly that upper-body obesity, but not lowerbody obesity, is associated with elevated systemic FFA
levels and metabolic dysfunction (Nielsen et al. 2004,
Piche et al. 2008, Lapointe et al. 2009, Amati et al. 2012).
In fact, it has been suggested that the preference of
gluteofemoral fat for trapping lipids serves as a
metabolic sink providing metabolic and cardiovascular
protection from the deleterious effects of an excessive
daily influx of dietary lipids (Manolopoulos et al. 2010).
The regulation and implications of these gender- and
depot-specific differences in terms of AT metabolism
and signaling has been covered in great detail in a
recent review (White & Tchoukalova 2014).

The lipolytic pathway: enzymes


and co-regulators
The core enzymatic machinery for TG hydrolysis in AT
consists of ATGL and HSL. Studies of ATGL-KO mice
have revealed that the absence of ATGL reduces the
lipolytic response of adipocytes to b-AR stimulation by
w70%, and by adding a specific HSL inhibitor lipolysis is
reduced by more than 95% (Schweiger et al. 2006).
Furthermore, both the basal and stimulated lipolytic
capacity of human and mouse adipocytes are increased
by ATGL overexpression and deceased by ATGL
silencing (Kershaw et al. 2006, Bezaire et al. 2009). By
contrast, HSL overexpression or silencing does not affect
the basal lipolytic rates in human adipocytes, but the
maximal stimulated lipolytic rate is decreased by
reduced HSL levels (Bezaire et al. 2009).
Adipose TG lipase

Dissecting adipose tissue


lipolysis

52 :3

R206

The important function of ATGL as a TG hydrolase was


discovered simultaneously in 2004 by three different

groups (Jenkins et al. 2004, Villena et al. 2004,


Zimmer- mann et al. 2004). Initially named ATGL
(Zimmermann et al. 2004), phospholipase A2x (Jenkins
et al. 2004), and desnutrin (Villena et al. 2004), the
enzyme is now formally annotated as patatin-like
phospholipase domain- containing protein 2 (Wilson
et al. 2006). Expectedly, the expression of ATGL in
mice has been found to be highest in white AT (WAT)
and brown AT (BAT), but the transcript has been
identified at lower levels in virtually all tissues studied
(Villena et al. 2004, Kershaw et al. 2006).
The transcriptional control of ATGL gene expression
is complex. A peroxisome proliferator-activated
receptor g (PPARg)-responsive element has been
identified in the promoter sequence of the mouse
Atgl gene (Kim et al. 2006), and accordingly
thiazolidinediones
(PPARg
agonists)
like
rosiglitazone increase Atgl expression (Kim et al. 2006,
Liu et al. 2009b). Furthermore, Atgl mRNA
expression in 3T3-L1 adipocytes is negatively
regulated by insulin as well as by TNFa-mediated
p42/44 MAPK activation (Kim et al. 2006). Several
additional studies have addressed the regulation of
ATGL expression, and it has been found that in
humans, ATGL protein is upregulated by fasting
(Nielsen et al. 2011), while in mice, the mRNA is
suppressed by feeding (Kershaw et al. 2006), but

upregulated by glucocorticoids (Villena et al. 2004), and


by SIRT1-mediated activation of the transcrip- tion factor
Foxo1 (Chakrabarti et al. 2011, Shan et al. 2013). However,
numerous studies have found that changes in
mammalian ATGL mRNA and protein levels are often
reciprocal, suggesting that ATGL is subject to extensive
post-transcriptional regulation (Steinberg et al. 2007, Li
et al. 2010, Nielsen et al. 2011, 2012).
ATGL is a specific TG hydrolase, and the activity
toward other lipid substrates like DG, MG, retinylesters
(RE), or cholesterylesters (CE) is very limited
(Zimmermann et al. 2004). Although the 3D structure of
ATGL has not been reported, studies on mutated and
truncated human and murine ATGL have revealed that
the N-terminal half of the enzyme contains the catalytic
patatin domain (Duncan et al. 2010), while the C-terminal
part is believed to be involved in regulation of enzymatic
activity and to mediate the interaction between ATGL
and LDs (Kobayashi et al. 2008, Schweiger et al. 2008).
404

428

The two serine residues Ser


and Ser
in the Cterminal part of the human ATGL sequence
406

430

(corresponding to Ser
and Ser
in murine ATGL)
have been identified as phos- phorylation sites (Bartz
et al. 2007). However, the role of these sites in the
regulation of ATGL activity, and the identity of the
406

upstream kinases is somewhat unclear. Thus, Ser


been suggested to be a consensus site for

has

Review

T S NIELSEN

and others

52 :3

Dissecting adipose tissue


lipolysis

R207

AMP-activated protein kinase (AMPK), and in murine


3T3- L1 adipocytes, it was shown that pharmacological
stimu- lation of lipolysis with the AMPK agonist
406

AICAR was dependent on ATGL Ser phosphorylation


(Ahmadian et al. 2011). However, another study found

A
TG

404

that phos- phorylation of Ser in human ATGL was


increased by b-adrenergic stimulation, while AICAR
treatment had no effect (Pagnon et al. 2012).

Lipid droplet

406

Additionally, in mouse AT, Ser phosphorylation was


increased with fasting, exer- cise, and ex vivo stimulation
of the cAMP-dependent pathway in a PKA-dependent
manner, thereby implicating this kinase in the
phosphorylation of ATGL (Pagnon et al. 2012). Notably, in

Acute stimulation

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

PKA

404

human skeletal muscle (SM), Ser


phosphorylation is
also associated with PKA signaling and not with AMPK
signaling, indicating that PKA is indeed the upstream
kinase for this site (Mason et al. 2012). Whether PKA
and/or AMPK are responsible for phosphorylation of

DG

TG

DG

Prolonged stimulation
Degradation

430

Ser is currently unknown, and so far no reports have


been published on the functional role of this site.

ATGL: activation by CGI-58

The primary way by which ATGL activity is increased


under acute lipolytic stimulation is via interaction with
the co-activator CGI-58 (also known as a/b hydrolase
domain-containing protein 5; Fig. 4A and B; Lass et al.
2006). Activation depends on the interaction of CGI-58
with the patatin domain of ATGL (Schweiger et al. 2008,
Cornaciu et al. 2011) and requires direct proteinprotein
interactions between ATGL and CGI-58 (Granneman et al.
2007, Cornaciu et al. 2011). Also, mutational studies have
shown that additional binding of CGI-58 to the LD is
crucial in order to activate ATGL (Gruber et al. 2010). The
molecular mechanism by which CGI-58 activates ATGL
is unclear, but it could potentially involve induction
of conformational changes, presentation of substrate, or
removal of reaction products. Interestingly, in addition to
its role in regulating lipolysis, both mouse and human
CGI-58 has been identified as a CoA-dependent lysophosphatidic acid acyltransferase (LPAAT; Ghosh et al. 2008,
Gruber et al. 2010, Montero-Moran et al. 2010), and it has
been speculated that it promotes lipolysis by channeling
fatty acids released from TG hydrolysis into
phospholipids to reduce end product inhibition of ATGL
and HSL (Montero-Moran et al. 2010). Although the
potential relevance of this LPAAT activity of CGI-58 in
lipolytic regulation remains to be investigated, CGI-58derived phospholipids are essential second messengers

TG

DG

TG
DG

in murine liver and AT when exposed to pro-inflammatory


cytokines like TNFa, interleukin 1b (IL1b), and IL6 (Lord et
al. 2012).

and inflammation, at least in mice.

Figure 4
Regulation of ATGL. (A) In the basal state, CGI-58 is complexed with
PLIN1 and ATGL activity is low. (B) Upon phosphorylation of PLIN1,
CGI-58 is released and associates with ATGL, which increases ATGL
activity. In this
phase, a fraction of the ATGL pool is dominantly inhibited by G0S2.
(C) If the lipolytic stimulation persists, gradual degradation of G0S2
promotes a further increase in ATGL activity. TG, triglyceride; ATGL,
adipose TG lipase; CGI-58, comparative gene identification-58; DG,
diacylglycerol; G0S2, G0/G1 switch gene 2; PKA, protein kinase A;
PLIN1, perilipin 1.

Consequently, as downstream inflammatory stress


kinases can impair insulin signaling, CGI-58 seems to
be involved in cross talk between insulin sensitivity

ATGL: inhibition by G0S2


Recently, the protein product of G0/G1 switch gene 2
(G0S2) was identified as an inhibitor of ATGL (Yang et al.
2010). In mice, G0S2 is primarily expressed in brown
and white adipocytes, but a significant expression has
also been detected in liver, heart, and SM (Zandbergen
et al. 2005). Murine G0S2 mRNA and protein expression
has been shown to be induced by insulin and PPARg
and suppressed by lipolytic agents like TNFa and
the b-AR agonist isoprenaline (Zandbergen et al. 2005,
Yang et al. 2010). Like CGI-58, G0S2 interacts directly with

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

the catalytic patatin domain of ATGL (Yang et al. 2010,


Cornaciu et al. 2011), but the ATGLG0S2 interaction is
independent of the ATGLCGI-58 interaction, and inhibition by G0S2 appears to be dominant to activation by
CGI-58 (Fig. 4B; Yang et al. 2010, 2011, Schweiger et al.
2012). Human G0S2, like the murine ortholog, inhibits
ATGL in a dose-dependent manner and is also involved
in regulating the intracellular localization of ATGL by
recruiting it to LDs (Schweiger et al. 2012).
It has been proposed that G0S2 acts as a long-term
regulator of lipolysis, as G0S2 protein levels are gradually
reduced during prolonged lipolytic stimulation resulting
in increased ATGL activity (Fig. 4C; Yang et al. 2010).
Notably, G0S2 protein and mRNA expression is
dramatically reduced in human AT by prolonged
physiological stimulation of lipolysis with a 72-h fast
(Nielsen et al. 2011) and similar effects have been
observed in birds (Oh et al. 2011) and pigs (Ahn et al.
2013). However, it is currently not known if the
association between ATGL and G0S2 is dynamic and
subject to regulation or if G0S2-mediated ATGL
inhibition primarily depends on the intracellular levels
of G0S2. Recent evidence has suggested the latter
although. Thus, in 3T3-L1 adipocytes, stimulation with
TNFa for up to 16 h reduces G0S2 levels gradually,
while the rate of lipolysis increases nearly proportionally
to the G0S2 reduction (Yang et al. 2011). Conversely,
overexpression of G0S2 signi- ficantly reduces the TNFa
mediated lipolytic response. Murine G0S2 is a shortlived protein with a half-life of
!1 h, and its stability can be greatly improved by
inhibition of the proteasomal pathway (Yang et al.
2011). Hence, it appears that one of the mechanisms by
which TNFa promotes adipocyte lipolysis is by
suppressing G0S2 mRNA expression leading to
cytosolic
depletion
of
G0S2
protein
through
proteasomal degradation and, conse- quently, increased
ATGL activity.
Animal models of ATGL deficiency
Insight into the crucial role of ATGL in whole-body TG
metabolism has been provided from the characterization
of ATGL-deficient mice (Haemmerle et al. 2006). These
animals exhibit massive ectopic lipid accumulation in
virtually all tissues and especially in AT, liver, SM, and
heart (Haemmerle et al. 2006). Accordingly, the animals
become obese even on a low-fat diet, and they suffer from
premature death due to severe cardiac steatosis and
dysfunction (Haemmerle et al. 2006, Schrammel et al.

Dissecting adipose tissue


lipolysis

52 :3

R208

2013). Furthermore, the normal fasting- or exerciseinduced rise in plasma FFA is absent in ATGL-KO animals
indicating a failure to increase lipolysis in WAT (Huijsman
et al. 2009, Schoiswohl et al. 2010). Without sufficient fuel
from lipid substrates, they rely primarily on carbohydrate
metabolism for energy conversion resulting in rapid
depletion of hepatic and SM glycogen stores (Huijsman
et al. 2009, Schoiswohl et al. 2010). Consequently, when
subjected to moderate exercise or short-term fasting, the
mice become hypoglycemic, and if fasting is extended
beyond a modest 812 h, they develop signs of severe
energy starvation like hypothermia, lethargy, reduced
oxygen consumption, and loss of lean mass (Haemmerle
et al. 2006, Schoiswohl et al. 2010, Wu et al. 2012).
Similarly, in spite of massively increased BAT mass,
ATGL- KO mice are unable to increase thermogenesis in
response to cold exposure, indicating that the
mobilization of lipid fuel in BAT is defective
(Haemmerle et al. 2006). In addition to the abnormal
substrate
metabolism,
ATGL deficiency causes
pancreatic steatosis leading to impaired insulin secretion
and hypoinsulinemia (Peyot et al. 2009). Interestingly,
however, despite the massive ectopic lipid accumulation
and b-cell dysfunction, the ATGL-deficient mice have
improved whole-body insulin sensitivity and glucose
tolerance compared with WT animals (Haemmerle et al.
2006, Peyot et al. 2009). Specifically, muscle and WAT
insulin signaling is improved, although in BAT and liver
it is reduced (Kienesberger et al. 2009).
The key role of defective TG catabolism in the
phenotype of ATGL-KO mice has recently been
supported with the generation of transgenic mice with
AT-specific overexpression of G0S2 (Heckmann et al.
2014). Like ATGL-deficient mice, WAT and BAT mass is
increased in these animals due to impaired basal and
stimulated lipolysis, but glucose and insulin tolerance is
improved. Moreover, thermogenesis is attenuated
leading to defec- tive cold adaptation, and the fastinginduced switch from carbohydrate to fatty acid
metabolism is severely impaired (Heckmann et al. 2014).
Conversely, mice with global G0S2 KO are lean and
resistant to high-fat diet-induced obesity and hepatic
steatosis (Zhang et al. 2013). Further- more, hepatic fatty
acid metabolism is enhanced as G0S2 ablation
accelerates ketogenesis and gluconeogenesis while
glycogen breakdown is impaired (Zhang et al. 2013).
Combined with the observations from Atgl-KO mice,
these results support a defining role for ATGL- mediated
lipolysis in whole-body substrate partitioning and

metabolism, at least in mice.


ATGL in human obesity and metabolic disease

The available literature on the expression patterns of


ATGL in human obesity is somewhat conflicting. In a
study

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

among lean and obese women, no difference in ATGL


protein levels were found in subcutaneous abdominal AT
(Ryden et al. 2007). Conversely, in mixed populations of
men and women, ATGL mRNA was increased in subcutaneous abdominal AT in obesity, but the protein levels
were reduced (Steinberg et al. 2007, Yao-Borengasser et al.
2011) and a negative correlation was found between BMI
and ATGL protein expression (Yao-Borengasser et al. 2011).
Furthermore, in paired biopsies, ATGL mRNA expression
was lower in visceral than in subcutaneous abdominal AT
(Yao-Borengasser et al. 2011), and when comparing visceral
AT from obese and lean subjects, ATGL mRNA was
increased in obesity (Steinberg et al. 2007, Tinahones
et al. 2010), while the protein levels were unaffected
(Steinberg et al. 2007). Similarly, in a recent comparison
of lean and obese men, only the mRNA of ATGL was
increased in visceral fat in obesity, but in subcutaneous
abdominal fat, ATGL protein was increased in the obese
subjects (De Naeyer et al. 2011). Furthermore, among obese
males and females with either normal or impaired insulin
sensitivity, insulin resistance has been shown to be
associated with reduced ATGL protein and mRNA in
subcutaneous abdominal AT (Jocken et al. 2007) and
reduced ATGL mRNA in visceral AT (Berndt et al. 2008).
However, whereas the available data on ATGL expression
in obesity is inconclusive, the expression of CGI-58 seems
to be remarkably stable between depots (Yao-Borengasser
et al. 2011) and in obesity (Steinberg et al. 2007).
Gender-specific differences may explain some of the
discrepancies between the studies on ATGL, but in light
of the substantial body of conflicting data, the role of
human ATGL in the pathogenesis of metabolic disease in
obesity is currently unclear.
By contrast, defective ATGL-mediated lipolysis has
been unequivocally identified as the primary defect in the
inherited monogenic disorder neutral lipid storage
disease (NLSD; Lefevre et al. 2001, Lass et al. 2006,
Fischer et al. 2007). Patients with loss-of-function
mutations affecting ATGL are characterized by ectopic
TG accumulation and visceral obesity, skeletal and
cardiac myopathy, and variable degrees of hepatic and
pancreatic steatosis (Schweiger et al. 2009, Laforet et
al. 2013, Natali et al. 2013). However, the metabolic
phenotype of these patients is heterogeneous. Thus,
some are insulin resistant and develop type 2 diabetes
(Laforet et al. 2013), while others have normal insulin
sensitivity but impaired glucose tolerance (Natali et al.
2013). This probably reflects individual differences in the
pattern of ectopic lipid deposition: patients with

Dissecting adipose tissue


lipolysis

52 :3

R209

extensive pancreatic steatosis generally have an impaired


insulin response to an oral

glucose challenge (Natali et al. 2013), while insulin


resistance is more common in patients with severe
muscular involvement and hepatic steatosis (Laforet
et al. 2013). The clinical manifestations of loss-offunction mutations in CGI-58 are similar to those
observed in functional ATGL deficiency except that
these patients do not develop myopathy (Igal et al.
1997). Instead, they suffer from severe ichthyosis
(Chanarin et al. 1975, Lefevre et al. 2001) and,
accordingly, the two types of NLSD are known as
NLSD with myopathy (NLSDM) and NLSD with
ichthyosis (NLSDI, also known as ChanarinDorfman
syndrome) (Fischer et al. 2007). The epidermal
defects observed in NLSDI are not present in NLSDM,
suggesting an ATGL-independent function of CGI-58,
possibly as a LPAAT in the synthesis pathway of
glycerophospholipids and acylceramides required for
the formation and maintenance of the skin
permeability barrier (Igal & Coleman 1996, Radner et
al. 2009). Consistently, KO of CGI-58 in mice results
in a neonatal lethal phenotype caused by leaky skin,
and the pups die from desiccation within hours after
birth (Radner et al. 2009). An overview of studies on
genetic deficiencies affecting ATGL function in humans
and animal models is listed in Table 1.

Hormone-sensitive lipase
HSL was discovered in rat AT in the early 1960s as a
lipolytic enzyme, which was inducible by fasting
and stimulation with ACTH or adrenaline and inhibited
by insulin (Hollenberg et al. 1961, Rizack 1964, Vaughan
et al. 1964).
Like ATGL, HSL is expressed in most tissues examined,
with the highest expression found in WAT and BAT
(Kraemer et al. 1993). The mRNA is generated from a
single gene controlled by a number of alternative
promoters that produce several different tissue-specific
isoforms of the HSL protein that range in size from
w85 kDa and up to 130 kDa (Langin et al. 1993, Mairal
et al. 2002). The HSL isoform found in human AT is a
786 aa protein with an apparent molecular weight of
w88 kDa (Langin et al. 1993).
Efficient lipid hydrolysis by HSL requires the lipase
to form a complex with cytosolic fatty acid-binding
protein 4 (FABP4), which acts as a molecular chaperone
by shuttling the FFA generated by HSL out of the cell
(Fig. 5A; Furuhashi & Hotamisligil 2008). Upon
stimulation of lipolysis, HSL and FABP4 associate in the
cytosol and the complex translocate to LDs (JenkinsKruchten et al. 2003, Smith et al. 2007). Consistently, in
FABP4-KO mice, the lipolytic capacity is reduced, and
the intracellular FFA

Review

Table 1

and others

Affected
protein

Human

NLSDI

CGI-58

Human

NLSDI

CGI-58

Human
Human

NLSDI
NLSDM

CGI-58
ATGL

Human

NLSDM

ATGL

Human

NLSDM

ATGL

Human

NLSDM

ATGL

Mouse
and

KO

ATGL

Mouse
Mouse
Mouse
and

KO
KO
KO

HSL
ATGL
ATGL
ATGL

Mouse
Mouse
Mouse
specific

KO
KO
KO with cardio-

Mouse
Mouse
specific
Mouse
Mouse

Dissecting adipose tissue


lipolysis

52 :3

R210

Overview of genetic studies on ATGL and CGI-58 function


Diagnosis/genetic
model

Specie
s

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

Highlights of the study

Reference

Case report: clinical manifestations and


proposal of diagnostic criteria
Igal et al. (1997)
Identification of defects in CGI-58 as causative for
NLSDI
Lefevre et al. (2001)
Activation of ATGL and rescue of NLSDI by CGI-58
Case report: identification of truncations in
human ATGL
Identification of biochemical defects in truncated
ATGL
Case report: magnetic resonance imaging of
muscles and metabolic characterization
Case report: body composition and glucose/lipid
metabolism

Lass et al. (2006)


Fischer et al. (2007)

Measurement of lipolysis in AT explants from


KO animals

Schweiger et al. (2006)

Phenotyping of KO animals
Insulin sensitivity and glucose/lipid metabolism
Substrate partitioning and metabolism during
rest and exercise

Haemmerle et al. (2006)


Kienesberger et al. (2009)
Huijsman et al. (2009)

HSL
ATGL
CGI-58
ATGL

Evaluation of effects on insulin secretion


Phenotyping of KO animals
Lipid/glucose metabolism during exercise

Peyot et al. (2009)


Radner et al. (2009)
Schoiswohl et al. (2010)

expression
AT-specific KO
KO with cardio-

ATGL
ATGL

Phenotyping and fed/fasting metabolism


Cardiac metabolism

Wu et al. (2012)
Schrammel et al. (2013)

KOexpression
AT-specific overexpression

G0S2
G0S2

Phenotyping of transgenic animals


Phenotyping of transgenic animals

Zhang et al. (2013)


Heckmann et al. (2014)

levels in adipocytes are increased (Coe et al. 1999).


Activation of HSL requires phosphorylation by PKA or
PKG on specific regulatory serine residues. In rat HSL,
563

659

660

these sites are Ser , Ser , and Ser (Stralfors et al. 1984,
Garton et al. 1988, Anthonsen et al. 1998) corresponding
552

649

650

to the human residues Ser , Ser , and Ser (Contreras


et al. 1998, Watt et al. 2006). The functional role of these
563

sites is different; phosphorylation of Ser is thought to


promote the translocation of HSL from the cytosol to LDs
659

(Daval et al. 2005), while phosphorylation of Ser

and

660

Ser is critical for activation of the intrinsic enzymatic


activity (Anthonsen et al. 1998). Conversely, phosphoryl565

554

ation of rat HSL on Ser


(human Ser ) by AMPK
inhibits HSL activation, most likely by steric
563

hindrance of phosphorylation of the adjacent Ser ,


thus preventing the translocation of HSL to the LDs
(Daval et al. 2005) (Fig. 5B). In terms of specificity,

Kobayashi et al. (2008)


Laforet et al. (2013)
Natali et al. (2013)

HSL is a much more promiscuous lipase than ATGL


and readily hydrolyze several lipid substrates, including
TG, DG, MG, RE, and CE in vitro (Fredrikson et al. 1981,
Wei et al. 1997). Among the substrates and intermediates
in TG lipolysis, the affinity for DG is approximately
tenfold higher than for TG and MG (Fredrikson et al.
1981, 1986).

Animal models of HSL deficiency

(Osuga et al. 2000, Harada et al. 2003, Park et al. 2005). This

The key role of HSL as a DG hydrolase in vivo was

unexpected observation was found to be caused by a


compensatory reduction in fatty acid esterification and

revealed with the generation of HSL-KO mice, which


were found to accumulate intracellular DG in AT, SM,
cardiac muscle, and testis (Haemmerle et al. 2002).
However, in contrast to ATGL-KO mice, HSL-deficient
mice do not suffer from severe systemic lipid
accumulation, although they tend to have enlargement
of internal organs like liver, heart, pancreas, and spleen
(Harada et al. 2003). Surprisingly, the WAT mass is
slightly reduced, and they are resistant to high- fat dietinduced obesity and peripheral insulin resistance

de novo lipogenesis to counteract the reduced release of


FFA to the circulation (Zimmermann et
al. 2003).
However, similar to ATGL-KO mice, HSL-KO mice have
increased BAT mass and enlargement of brown
adipocytes (Harada et al. 2003), but this is not
associated with impaired thermo- genesis, as they retain
a normal sensitivity to cold exposure (Osuga et al. 2000).
An overview of animal studies on genetic deficiencies
affecting HSL is listed in Table 2.

Review

T S NIELSEN

and others

52 :3

Dissecting adipose tissue


lipolysis

R211

FFA

B
FFA- FABP4

cAMP

PDE3B

PKA
P

HSL

5-AMP

FABP4

HSL

P
P

P
P

P
P

DG

MG

Lipid droplet
Figure 5

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

AMPK

Regulation of HSL. (A) Phosphorylation of Ser552, Ser649, and Ser650 on


human HSL promotes lipase activation and association with FABP4 in
the cytosol. Subsequent translocation of this complex to the LD surface
is
dependent on both HSL and PLIN1 phosphorylation and results in full
activation of HSL activity. Acting as a molecular chaperone, FABP4
shuttles the FFAs released by HSL from the LD to the plasma membrane
of the
adipocyte where they are secreted. (B) LD-targeting of cytoplasmic HSL

HSL in human obesity and metabolic disease


While changes in ATGL expression patterns in obesity
are of uncertain significance, the importance of HSL
expression is somewhat more well established, although
discrepancies certainly exist. Thus, the expression of HSL
mRNA in subcutaneous abdominal AT in obesity has
been reported to be increased (Ray et al. 2009), reduced
(Large et al. 1999, Mairal et al. 2006), or not affected
(Steinberg et al. 2007, De Naeyer et al. 2011). However,
irrespective of gender, the majority of studies have found
the correspond- ing HSL protein levels to be reduced in
obesity (Large et al. 1999, Langin et al. 2005, Ryden et al.
2007, Ray et al. 2009). Similarly, in the obese state, insulin
resistance is associated with a reduction in HSL mRNA
and protein in subcu- taneous abdominal AT (Jocken et
al. 2007). In visceral AT, HSL mRNA levels have
consistently been found to be upregulated in obesity
(Mairal et al. 2006, Steinberg et al. 2007, Ray et al. 2009,
De Naeyer et al. 2011), but the protein levels seem to be
unaffected (De Naeyer et al. 2011) or possibly reduced
(Ray et al. 2009).
So far, no human examples of loss-of-function
mutations affecting HSL have been reported, and in light

DG

HSL

HSL

PKA

MG

Lipid droplet
requires cAMP-dependent phosphorylation on Ser552 by PKA. Conversely,
AMPK is activated by 5 0 -AMP and phosphorylate HSL on the adjacent
Ser554. As phosphorylation on Ser552 and Ser554 is mutually exclusive, AMPK
reduces the LD association of HSL. AMPK, AMP-activated protein kinase;
DG, diacylglycerol; FABP4, fatty acid-binding protein 4; FFA, free fatty acid;
HSL, hormone-sensitive lipase; MG, monoacylglycerol; PDE3B, phosphodiesterase 3B; PKA, protein kinase A.

of the relatively mild phenotype of HSL-KO mice, it seems


unlikely that HSL deficiency per se is associated with severe
metabolic defects in humans.

Monoglyceride lipase
MGL was identified in rats as an MG-specific lipase
with no affinity toward TG or DG (Tornqvist &
Belfrage 1976). The first MGL-KO mouse model has
recently been generated (Table 2), and these animals
accumulate MG in WAT, brain, and liver (Taschler
et al. 2011). Also, it was found that HSL partially
compensated for the absence of MGL in AT, as the
stimulated glycerol release was reduced by a modest
43% compared with WT mice. However, upon specifi
inhibition of HSL in cultured fat pads, the MGhydrolase activity was almost completely abolished
(Taschler et al. 2011). To date, no reports have been

published indicating that MGL expression and enzymatic


activity in AT is regulated by hormonal signals or
nutritional status, suggesting that the enzyme is constitutively active in the lipolytic cascade.
LD-associated proteins: the CIDE family
The PLIN proteins and the CIDE proteins constitute the
two major families of LD-associated proteins in adipocytes. The pro-apoptotic CIDE proteins (cell-death inducing DNA fragmentation factor-a-like effector) comprise
three members: CIDEA, CIDEB, and CIDEC (also known
as fat-specific protein of 27 kDa, Fsp27) (Yonezawa et al.
2011).

Review

T S NIELSEN

and others

Table 2 Overview of genetic studies on HSL, MGL, and


FABP4 function

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Specie
s

Geneti
c
model

Affecte
d
protein

Mouse

KO

FABP4

Mouse

KO

HSL

Mouse

KO

HSL

Mouse

KO

HSL

Mouse

KO

HSL

Mouse

KO

HSL

Mouse

KO

MGL

Coe et al.
(1999)
Osuga et al. (2000)

Highlights
of the study
Reference

Phenotyping of
KO animals
Phenotyping of
KO animals
Involvement of
HSL in
whole-body
DG catabolism
Lipid metabolism in
diet-induced
obesity
AT adaptations
to HSL
deficiency
Insulin sensitivity and
glucose/lipid
metabolism
Phenotyping of
KO animals

Haemmerle
et al. (2002)

Harada et al.
(2003)

Zimmermann
et al. (2003)
Park et al.
(2005)

Taschler et al.
(2011)

Dissecting adipose tissue


lipolysis

52 :3

R212

functional relationship, they are now annotated as PLIN1


(perilipin A), PLIN2 (adipophilin), PLIN3 (TIP-47),
PLIN4 (S3-12), and PLIN5 (MLDP/OXPAT) (Kimmel et
al. 2010).
Of the PLIN proteins, PLIN1 is particularly important in
the regulation of AT lipolysis, while the other family
members are involved in adipogenesis and LD formation
(PLIN2), LD biosynthesis and stabilization (PLIN3),
LD maturation (PLIN4) and regulation of lipolysis in
oxidative tissues not expressing PLIN1 (PLIN5) (Bickel
et al. 2009).
Multiple regulatory roles of murine PLIN1 have been
described in the lipolytic cascade, and it has been
suggested that PLIN1 is the master regulator of PKAstimulated lipolysis in mice (Miyoshi et al. 2007). As such,
PLIN1 either directly or indirectly regulates the activity
of ATGL and HSL as well as their access to lipid
substrates in the LDs. The LD targeting of HSL is partly
governed by PLIN1; in the basal state, as much as
half of the total cellular HSL content is located in the
cytoplasm, but PKA- mediated PLIN1 and HSL
phosphorylation has been shown to significantly
enhance the co-localization and association of the two
proteins on LDs (Miyoshi et al.

CIDEA and CIDEC from mice and humans have recently


been shown to be negative regulators of lipolysis (Nord-

the family also includes the proteins S3-12 and myocyte


LD protein (MLDP, also known as OXPAT) (Bickel et al.

strom et al. 2005, Puri et al. 2008, Christianson et al. 2010),


although their specific role is not well characterized yet.

2009). However, due to their evolutionary, structural, and


2006). However, studies on mutant PLIN1 lacking all
phosphosites have revealed that in the absence of
phosphorylation of PLIN1, the PKA-mediated increase in
HSL activity is blunted, although the lipase is phosphorylated and translocated (Miyoshi et al. 2006). In other
words, the lipolytic action of LD-associated and phosphorylated HSL are critically dependent on PLIN1 phosphorylation in mouse adipocytes.
Similarly, the activation of murine ATGL depends
on phosphorylation of PLIN1, although by a different
mechanism. In the basal state, unphosphorylated PLIN1
has been shown to negatively regulate ATGL by
efciently sequestering CGI-58, thereby preventing
activation of ATGL (Granneman et al. 2007, 2009). Upon
stimulation of lipolysis, phosphorylation of PLIN1 causes
the dissoci- ation of CGI-58, which can then bind and
activate ATGL (Granneman et al. 2009; Fig. 4).
Interestingly, a single amino acid in PLIN1 has been
identified as the crucial residue for regulation of HSL-

However, it appears that they promote lipid storage


through their involvement in LD formation, fusion, and
stabilization (Puri et al. 2008, Christianson et al. 2010, Ito
et al. 2010). Accordingly, human CIDEC has been found
to interact with PLIN1 (see below) and the interaction
between these two proteins is critical for the formation of
large LDs and unilocular adipocytes (i.e. cells containing
a single big LD) (Grahn et al. 2013, Sun et al. 2013).
The mechanism by which the CIDE proteins inhibit
lipolysis is incompletely understood but it seems to
involve shielding of the LDs from the action of lipases by
providing a physical barrier around the lipid core
(Christianson et al. 2010, Yang et al. 2013).
LD-associated proteins: the PLIN family
The other family of LD-associated proteins has been
studied in much more detail. The PLIN proteins were
originally called the PAT family after perilipin A, adipophilin, and tail-interacting protein of 47 kDa (TIP-47), and

517

and ATGL-mediated lipolysis, as mutation of Ser in


the mouse sequence almost completely abolishes PKA-

stimulated FFA and glycerol release (Miyoshi et al.


2007).
Animal models of deficiencies in LD-associated
proteins

In mice, the CIDE proteins exhibit a distinct expression


pattern. Thus, while CIDEA is predominantly expressed
in BAT (Zhou et al. 2003), CIDEB is almost
exclusively

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

expressed in the liver (Li et al. 2007), and CIDEC is


primarily found in WAT (Nishino et al. 2008), suggesting
a tissue-specific role of these proteins. Consistently,
CIDEA- deficient mice are characterized by elevations
in body temperature and overall metabolic rate due to
accelerated BAT lipolysis and thermogenesis (Zhou et al.
2003). Consequently, these mice are lean and resistant
to diet- induce obesity and glucose intolerance. A liverspecific function of CIDEA has also been demonstrated,
as hepatic CIDEA knockdown in genetically obese ob/ob
mice reduces hepatic TG accumulation and LD size and
accelerates overall energy expenditure (Zhou et al. 2012).
Similarly, KO of CIDEB results in lean mice with
improved glucose tolerance, insulin sensitivity, and
resistance to hepatic steatosis, but BAT metabolism is
normal in these animals (Li et al. 2007). Instead,
ketogenesis and overall metabolic rate is accelerated,
suggesting an overall shift in substrate utilization toward
lipid metabolism (Li et al. 2007). In terms of metabolic
rate, susceptibility to obesity, hepatic steatosis, and
disturbances in glucose homeostasis and metabolism,
the overall phenotype of CIDEC-KO mice is very similar
to the phenotype of CIDEA- and CIDEB- deficient
animals (Nishino et al. 2008, Toh et al. 2008). Notably,
however, while the metabolic rate in BAT is reduced,
mitochondrial biogenesis, oxygen consumption, and
basal lipolytic rates are increased in WAT (Nishino et al.
2008). Consistently, WAT expression of specific factors
inhibiting BAT differentiation is reduced and a
concomitant increase in the expression of BAT-specific
genes (e.g. UCP1) causes a shift toward a more brownlike phenotype (Toh et al. 2008). Thus, in spite of tissuespecific differences in the expression and regulation of
the CIDE proteins, they seem to share a crucial role in
the regulation of overall substrate partitioning and
metabolism.
In agreement with the role of PLIN1 as a negative
regulator of ATGL activity, PLIN1-deficient mice have
been found to have dramatically decreased fat mass and
increased basal lipolysis (Martinez-Botas et al. 2000,
Tansey et al. 2001). This observation has been supported
by in vitro studies; stimulation of 3T3-L1 cells with TNFa
was found to increase basal lipolysis due to a reduction in
PLIN1 protein levels, and this effect could be reversed by
simultaneous overexpression of PLIN1 (Souza et al. 1998).
Furthermore, PLIN1-KO animals are lean and resistant to
diet-induced obesity, but nevertheless they are prone
to develop glucose intolerance and peripheral insulin
resistance (Martinez-Botas et al. 2000, Tansey et al. 2001).

52 :3

Dissecting adipose tissue


lipolysis

R213

Interestingly, in the absence of PLIN1, the expression of


PLIN2 is increased and, consequently, PLIN2 is the major
LD-associated protein in these animals (Tansey et al.
2001). In cultured cells, overexpression of PLIN2 has
been shown to promote lipid storage by negatively
regulating the access of ATGL to LDs (Listenberger et al.
2007), but the underlying mechanism is currently
unknown, and it remains to be determined whether
PLIN1 defi iency is associated with increased PLIN2
expression in humans in vivo.
LD-associated proteins in human obesity and
metabolic disease
As for ATGL and HSL, the available data on the
association between obesity and the expression of LDassociated proteins are rather inconclusive. In the
obese state, PLIN1 mRNA expression has been reported
to be increased in visceral AT (Ray et al. 2009, Tinahones
et al. 2010) but unaffected in subcutaneous abdominal
AT (Ray et al. 2009). However, others have found a
negative correlation between BMI and visceral PLIN1
mRNA levels, suggesting that the expression is reduced
in obesity (Moreno- Navarrete et al. 2013). In obesity,
PLIN1 protein expression is reduced in both visceral and
subcutaneous abdominal AT (Ray et al. 2009), and
among obese subjects, the protein levels in subcutaneous
abdominal AT are lower in insulin-resistant subjects than
in insulin-sensitive subjects (Moreno-Navarrete et al.
2013).
Recently, two different loss-of-function mutations in
PLIN1 have been identified in three patients diagnosed
with a rare autosomal dominant partial lipodystrophy
(Gandotra et al. 2011a). Both mutations introduced a
frameshift causing the loss of three regulatory PKA sites,
517

including the crucial Ser . Consequently, these mutants


fail to sequester CGI-58 leading to permanently elevated
basal lipolysis due to constitutive activation of ATGL
(Gandotra et al. 2011b). The clinical manifestations of
these mutations included dyslipidemia, hypertension,
lipoatrophy, hepatic steatosis, severe insulin resistance,
and type 2 diabetes. Remarkably, a very similar
phenotype has been found in a patient carrying a lossof-function mutation in the LD-targeting domain of
CIDEC, suggesting that the integrity of the interaction
between CIDEC and PLIN1 is crucial for proper LD
dynamics and lipolytic control in humans in vivo
(Rubio-Cabezas et al. 2009). Additionally, the CIDECdefi
AT had an

unusually high occurrence of multilocular adipocytes


supporting a role for CIDEC in the formation and
stabilization of large LDs (Rubio-Cabezas et al. 2009).

An overview of studies on genetic defi iencies affecting


LD-associated proteins in humans and animal models is
listed in Table 3.

Review

T S NIELSEN

and others

52 :3

Dissecting adipose tissue


lipolysis

R214

Table 3 Overview of genetic studies on LD-associated proteins

Species
model

Diagnosis/genetic

Affected
protein

Human

Partial lipodystrophy

CIDEC

Human
truncations

Partial lipodystrophy

PLIN1

Human

Highlights of the study

Reference

Identification of CIDEC truncation as


the causative mutation
Identification of two PLIN1

Rubio-Cabezas et al.
(2009) Gandotra et al.

Partial lipodystrophy

PLIN1

Mouse
(2000) Mouse
Mouse
Mouse
Mouse
Mouse

KO
KO
KO
KO
KO
KO

PLIN1
PLIN1
CIDEA
CIDEB
CIDEC
CIDEC

Mouse

KO

CIDEA

as causative mutations
Characterization of PLIN1 mutants in
regulation of lipolysis in cell
culture
Phenotyping of KO animals
Phenotyping of KO animals
Phenotyping of KO animals
Phenotyping of KO animals
Phenotyping of KO animals
Analysis of WAT browning in KO
animals
Characterization of the role of
CIDEA in hepatic metabolism

(2011a) Gandotra et al.


(2011b)
Martinez-Botas et al.
Tansey et al. (2001)
Zhou et al. (2003)
Li et al. (2007)
Nishino et al. (2008)
Toh et al. (2008)
Zhou et al.
(2012)

Intracellular lipolysis in non-ATs


In this review, we have focused on lipolysis in AT.
However, the ability to store and re-hydrolyze TG is not
unique for adipocytes, and most cell types are able to
form LDs by taking up and esterifying FFA into TG
(Greenberg et al. 2011). In fact, the importance of tight
lipolytic control in other tissues has become apparent
with the characterization of the phenotypes associated
with defi lipase activity, particu- larly with respect to
ATGL. Like the myocardial defects observed in Atgl KO
mice, muscle-specific CGI-58 KO mice also suffer from
cardiomyopathy and cardiac steatosis caused by a severe
impairment of TG catabolism (Zierler et al. 2013).
Conversely, myocardial specific ATGL overexpression in
type 1 diabetic mice confers protection against diabetesinduced cardiomyopathy (Pulinilkunnil et al. 2013), supporting a key role of ATGL in the regulation of metabolism
and lipid homeostasis in the heart. Similar results have
been obtained in the liver, where ATGL deficiency causes
hepatic steatosis and reduced b-oxidation (Ong et al. 2011,
Wu et al. 2011) while liver-specific overexpression of
ATGL or HSL reduces obesity-induced steatosis and
increases b-oxidation (Reid et al. 2008, Ong et al. 2011).
Surprisingly although, in spite of these effects of altered
lipase expression on hepatic TG content and substrate
utilization, liver-specifi KO or
overexpression of ATGL only has minor effects on hepatic
insulin sensitivity and whole-body metabolic parameters

DG to TG hydrolase activity (Jocken et al. 2010). Finally, in


obese type 2 diabetic patients, basal SM lipolysis is
elevated and the anti-lipolytic action of insulin is
blunted (Jocken et al. 2013). Whether this impairment is a
result of peripheral insulin resistance or a cause of it is
unclear although, and at present, the mechanistic
connection between intramyocel- lular lipolysis and
insulin resistance is a matter of intense debate and
research.
In summary, since the discovery of ATGL in 2004
tremendous progress has been made in the characterization of AT lipolysis. However, with the increasing
number of newly identified enzymes and regulatory
proteins, the remarkable complexity of the hormonal
and intracellular signaling network regulating the lipolytic pathway has also become clear. Considering the
severe phenotypes associated with defective lipolysis in
AT, pancreas, liver, heart, and SM, it is evident that the
balance between lipid mobilization, utilization, and
storage is crucial in most tissues. Consequently, by
delineating the processes regulating lipid metabolism in
adipose and non-ATs alike, we can also advance our
understanding of glucose metabolism and identify new
pathways to target in the treatment of metabolic disease
like type 2 diabetes.

(Turpin et al. 2011, Wu et al. 2011). In human SM, ATGL

protein is increased significantly by endurance training,


indicating that mobilization of intramyocellular TG stores
involves ATGL (Alsted et al. 2009). Obesity is also
associated with increased SM content of ATGL, whereas
HSL is decreased, resulting in a substantial decrease in the
ratio of

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Declaration of interest
The authors declare that there is no conflict of interest that could be
perceived as prejudicing the impartiality of the review.

Funding
This work was supported by grants from: i) The FOOD Study
Group/Ministry of Food, Agriculture and Fisheries & Ministry of Family
and Consumer

Review

T S NIELSEN

and others

Affairs, Denmark, ii) The Lundbeck Foundation, Denmark, iii) The Novo
Nordisk Foundation, Denmark, iv) Augustinus Fonden, Denmark, and
v) Aase og Ejnar Danielsens Fond, Denmark.

Acknowledgements
The Novo Nordisk Foundation Center for Basic Metabolic Research is an
independent Research Center at the University of Copenhagen partially
funded by an unrestricted donation from the Novo Nordisk Foundation
(www.metabol.ku.dk).

References

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

Ahmadian M, Abbott MJ, Tang T, Hudak CS, Kim Y, Bruss M, Hellerstein


MK, Lee HY, Samuel VT, Shulman GI et al. 2011 Desnutrin/ATGL is
regulated by AMPK and is required for a brown adipose phenotype.
Cell Metabolism 13 739748. (doi:10.1016/j.cmet.2011.05.002)
Ahn J, Oh SA, Suh Y, Moeller SJ & Lee K 2013 Porcine G(0)/G(1) switch
gene 2 (G0S2) expression is regulated during adipogenesis and shortterm in-vivo nutritional interventions. Lipids 48 209218.
(doi:10.1007/ s11745-013-3756-8)
Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA,
Fruchart JC, James WP, Loria CM & Smith SC Jr 2009 Harmonizing the
metabolic syndrome: a joint interim statement of the International
Diabetes Federation Task Force on Epidemiology and Prevention;
National Heart, Lung, and Blood Institute; American Heart
Association; World Heart Federation; International Atherosclerosis
Society; and
International Association for the Study of Obesity. Circulation 120
16401645. (doi:10.1161/CIRCULATIONAHA.109.192644)
Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB &
Cohen P 1997 Characterization of a 3-phosphoinositide-dependent
protein kinase which phosphorylates and activates protein kinase Ba.
Current Biology 7 261269. (doi:10.1016/S0960-9822(06)00122-9)
Alsted TJ, Nybo L, Schweiger M, Fledelius C, Jacobsen P, Zimmermann R,
Zechner R & Kiens B 2009 Adipose triglyceride lipase in human
skeletal muscle is upregulated by exercise training. American Journal of
Physiology. Endocrinology and Metabolism 296 E445E453.
(doi:10.1152/ ajpendo.90912.2008)
Amati F, Pennant M, Azuma K, Dube JJ, Toledo FG, Rossi AP, Kelley DE &
Goodpaster BH 2012 Lower thigh subcutaneous and higher visceral
abdominal adipose tissue content both contribute to insulin resistance.
Obesity 20 11151117. (doi:10.1038/oby.2011.401)
Anthonsen MW, Ronnstrand L, Wernstedt C, Degerman E & Holm C 1998
Identification of novel phosphorylation sites in hormone-sensitive
lipase that are phosphorylated in response to isoproterenol and
govern activation properties in vitro. Journal of Biological Chemistry 273
215221. (doi:10.1074/jbc.273.1.215)
Armani A, Marzolla V, Rosano GM, Fabbri A & Caprio M 2011
Phosphodiesterase type 5 (PDE5) in the adipocyte: a novel player in
fat metabolism? Trends in Endocrinology and Metabolism 22 404411.
(doi:10.1016/j.tem.2011.05.004)
Arner P, Kriegholm E, Engfeldt P & Bolinder J 1990 Adrenergic regulation
of lipolysis in situ at rest and during exercise. Journal of Clinical
Investigation 85 893898. (doi:10.1172/JCI114516)
Aversa A, Caprio M, Antelmi A, Armani A, Brama M, Greco EA,
Francomano D, Calanchini M, Spera G, Di Luigi L et al. 2011 Exposure
to phosphodiesterase type 5 inhibitors stimulates aromatase
expression in human adipocytes in vitro. Journal of Sexual Medicine 8
696704.
(doi:10.1111/j.1743-6109.2010.02152.x)

Dissecting adipose tissue


lipolysis

52 :3

R215

Barbe P, Millet L, Galitzky J, Lafontan M & Berlan M 1996 In situ assessment


of the role of the b1-, b2- and b3-adrenoceptors in the control of
lipolysis and nutritive blood flow in human subcutaneous adipose
tissue. British Journal of Pharmacology 117 907913. (doi:10.1111/
j.1476-5381.1996.tb15279.x)

Bartz R, Zehmer JK, Zhu M, Chen Y, Serrero G, Zhao Y & Liu P 2007
Dynamic activity of lipid droplets: protein phosphorylation and
GTP-mediated protein translocation. Journal of Proteome Research 6
32563265. (doi:10.1021/pr070158j)
Belfort R, Mandarino L, Kashyap S, Wirfel K, Pratipanawatr T, Berria
R, DeFronzo RA & Cusi K 2005 Doseresponse effect of elevated
plasma free fatty acid on insulin signaling. Diabetes 54 1640
1648. (doi:10.2337/diabetes.54.6.1640)
Berndt J, Kralisch S, Kloting N, Ruschke K, Kern M, Fasshauer M, Schon
MR, Stumvoll M & Bluher M 2008 Adipose triglyceride lipase gene
expression in human visceral obesity. Experimental and Clinical
Endocrinology & Diabetes 116 203210. (doi:10.1055/s-2007993148)
Berryman DE, List EO, Coschigano KT, Behar K, Kim JK & Kopchick JJ
2004 Comparing adiposity profiles in three mouse models with
altered GH signaling. Growth Hormone & IGF Research 14 309318.
(doi:10.1016/
j.ghir.2004.02.005)
Berryman DE, List EO, Kohn DT, Coschigano KT, Seeley RJ & Kopchick
JJ 2006 Effect of growth hormone on susceptibility to diet-induced
obesity. Endocrinology 147 28012808. (doi:10.1210/en.2006-0086)
Bezaire V, Mairal A, Ribet C, Lefort C, Girousse A, Jocken J, Laurencikiene
J,
Anesia R, Rodriguez AM, Ryden M et al. 2009 Contribution of
adipose triglyceride lipase and hormone-sensitive lipase to lipolysis
in hMADS adipocytes. Journal of Biological Chemistry 284 18282
18291. (doi:10.1074/jbc.M109.008631)
Bickel PE, Tansey JT & Welte MA 2009 PAT proteins, an ancient family of
lipid droplet proteins that regulate cellular lipid stores. Biochimica et
Biophysica Acta 1791 419440. (doi:10.1016/j.bbalip.2009.04.002)
Botion LM, Brasier AR, Tian B, Udupi V & Green A 2001 Inhibition of
proteasome activity blocks the ability of TNFa to down-regulate G(i)
proteins and stimulate lipolysis. Endocrinology 142 50695075.
(doi:10.1210/endo.142.12.8518)
Boura-Halfon S & Zick Y 2009 Phosphorylation of IRS proteins, insulin

action, and insulin resistance. American Journal of Physiology.


Endocrinology and Metabolism 296 E581E591.
(doi:10.1152/ajpendo.90437.2008)
Bousquet-Melou A, Galitzky J, Lafontan M & Berlan M 1995 Control of
lipolysis in intra-abdominal fat cells of nonhuman primates:
comparison with humans. Journal of Lipid Research 36 451461.
Cai TQ, Ren N, Jin L, Cheng K, Kash S, Chen R, Wright SD, Taggart AK &
Waters MG 2008 Role of GPR81 in lactate-mediated reduction of
adipose lipolysis. Biochemical and Biophysical Research Communications
377 987991. (doi:10.1016/j.bbrc.2008.10.088)
Carpenter CL, Duckworth BC, Auger KR, Cohen B, Schaffhausen BS &
Cantley LC 1990 Purification and characterization of phosphoinositide
3-kinase from rat liver. Journal of Biological Chemistry 265 1970419711.
Castan I, Valet P, Larrouy D, Voisin T, Remaury A, Daviaud D, Laburthe
M
& Lafontan M 1993 Distribution of PYY receptors in human fat cells:
an antilipolytic system alongside the a2-adrenergic system. American
Journal of Physiology 265 E74E80.
Chakrabarti P, English T, Karki S, Qiang L, Tao R, Kim J, Luo Z, Farmer SR
& Kandror KV 2011 SIRT1 controls lipolysis in adipocytes via
FOXO1-mediated expression of ATGL. Journal of Lipid Research 52
16931701. (doi:10.1194/jlr.M014647)
Chanarin I, Patel A, Slavin G, Wills EJ, Andrews TM & Stewart G 1975
Neutral-lipid storage disease: a new disorder of lipid metabolism.
BMJ 1 553555. (doi:10.1136/bmj.1.5957.553)
Chen XL, Lee K, Hartzell DL, Dean RG, Hausman GJ, McGraw RA,
Della-Fera MA & Baile CA 2001 Adipocyte insensitivity to insulin
in growth hormone-transgenic mice. Biochemical and Biophysical
Research Communications 283 933937. (doi:10.1006/bbrc.2001.4882)
Cho KJ, Shim JH, Cho MC, Choe YK, Hong JT, Moon DC, Kim JW &
Yoon DY 2005 Signaling pathways implicated in a-melanocyte
stimulating hormone-induced lipolysis in 3T3-L1 adipocytes. Journal
of Cellular Biochemistry 96 869878. (doi:10.1002/jcb.20561)
Choi YH, Park S, Hockman S, Zmuda-Trzebiatowska E, Svennelid F,
Haluzik M, Gavrilova O, Ahmad F, Pepin L, Napolitano M et al. 2006
Alterations in regulation of energy homeostasis in cyclic nucleotide

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

phosphodiesterase 3B-null mice. Journal of Clinical Investigation 116


32403251. (doi:10.1172/JCI24867)
Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS,
Monks BR & Birnbaum MJ 2010 Insulin regulates adipocyte lipolysis
via an Akt-independent signaling pathway. Molecular and Cellular
Biology 30 50095020. (doi:10.1128/MCB.00797-10)
Christianson JL, Boutet E, Puri V, Chawla A & Czech MP 2010
Identification of the lipid droplet targeting domain of the Cidea
protein. Journal of Lipid Research 51 34553462.
(doi:10.1194/jlr.M009498)
Clasen BF, Krusenstjerna-Hafstrom T, Vendelbo MH, Thorsen K, Escande
C, Moller N, Pedersen SB, Jorgensen JO & Jessen N 2013 Gene
expression in skeletal muscle after an acute intravenous GH bolus in
human subjects: identification of a mechanism regulating ANGPTL4.
Journal of Lipid Research 54 19881997. (doi:10.1194/jlr.P034520)
Clerico A, Giannoni A, Vittorini S & Passino C 2011 Thirty years of the
heart as an endocrine organ: physiological role and clinical utility
of cardiac natriuretic hormones. American Journal of Physiology. Heart
and Circulatory Physiology 301 H12H20.
(doi:10.1152/ajpheart.00226.2011)
Coe NR, Simpson MA & Bernlohr DA 1999 Targeted disruption of the
adipocyte lipid-binding protein (aP2 protein) gene impairs fat cell
lipolysis and increases cellular fatty acid levels. Journal of Lipid
Research 40 967972.
Contreras JA, Danielsson B, Johansson C, Osterlund T, Langin D & Holm
C 1998 Human hormone-sensitive lipase: expression and large-scale
purification from a baculovirus/insect cell system. Protein Expression
and Purification 12 9399. (doi:10.1006/prep.1997.0821)
Copps KD & White MF 2012 Regulation of insulin sensitivity by
serine/threonine phosphorylation of insulin receptor substrate
proteins IRS1 and IRS2. Diabetologia 55 25652582.
(doi:10.1007/ s00125-012-2644-8)
Cornaciu I, Boeszoermenyi A, Lindermuth H, Nagy HM, Cerk IK, Ebner
C, Salzburger B, Gruber A, Schweiger M, Zechner R et al. 2011 The
minimal domain of adipose triglyceride lipase (ATGL) ranges until
leucine 254 and can be activated and inhibited by CGI-58 and
G0S2,
respectively.
PLoS
ONE
6
e26349.
(doi:10.1371/journal.pone.0026349)
Cusi K, Kashyap S, Gastaldelli A, Bajaj M & Cersosimo E 2007 Effects on
insulin secretion and insulin action of a 48-h reduction of plasma
free fatty acids with acipimox in nondiabetic subjects genetically
predisposed to type 2 diabetes. American Journal of Physiology.
Endocrinology and Metabolism 292 E1775E1781.
(doi:10.1152/ajpendo.00624.2006)
Czech MP, Tencerova M, Pedersen DJ & Aouadi M 2013 Insulin signalling
mechanisms for triacylglycerol storage. Diabetologia 56 949964.
(doi:10.1007/s00125-013-2869-1)
Daval M, Diot-Dupuy F, Bazin R, Hainault I, Viollet B, Vaulont S, Hajduch
E, Ferre P & Foufelle F 2005 Anti-lipolytic action of AMP-activated
protein kinase in rodent adipocytes. Journal of Biological Chemistry 280
2525025257. (doi:10.1074/jbc.M414222200)
Demerath EW, Sun SS, Rogers N, Lee M, Reed D, Choh AC, Couch W,
Czerwinski SA, Chumlea WC, Siervogel RM et al. 2007 Anatomical
patterning of visceral adipose tissue: race, sex, and age variation.
Obesity 15 29842993. (doi:10.1038/oby.2007.356)
De Naeyer H, Ouwens DM, Van Nieuwenhove Y, Pattyn P, t Hart LM,
Kaufman JM, Sell H, Eckel J, Cuvelier C, Taes YE et al. 2011 Combined
gene and protein expression of hormone-sensitive lipase and adipose
triglyceride lipase, mitochondrial content, and adipocyte size in
subcutaneous and visceral adipose tissue of morbidly obese men.
Obesity Facts 4 407416. (doi:10.1159/000333445)
Dinneen S, Alzaid A, Miles J & Rizza R 1993 Metabolic effects of the
nocturnal rise in cortisol on carbohydrate metabolism in normal
humans. Journal of Clinical Investigation 92 22832290. (doi:10.1172/
JCI116832)

Dissecting adipose tissue


lipolysis

52 :3

R216

Djurhuus CB, Gravholt CH, Nielsen S, Pedersen SB, Moller N & Schmitz O
2004 Additive effects of cortisol and growth hormone on regional
and systemic lipolysis in humans. American Journal of Physiology.
Endocrinology and Metabolism 286 E488E494. (doi:10.1152/ajpendo.
00199.2003)

Doris R, Vernon RG, Houslay MD & Kilgour E 1994 Growth hormone


decreases the response to anti-lipolytic agonists and decreases the
levels of Gi2 in rat adipocytes. Biochemical Journal 297 4145.
Duncan RE, Wang Y, Ahmadian M, Lu J, Sarkadi-Nagy E & Sul HS
2010 Characterization of desnutrin functional domains: critical
residues for triacylglycerol hydrolysis in cultured cells. Journal of
Lipid Research 51 309317. (doi:10.1194/jlr.M000729)
Egan JJ, Greenberg AS, Chang MK, Wek SA, Moos MC Jr & Londos C
1992 Mechanism of hormone-stimulated lipolysis in adipocytes:
transloca- tion of hormone-sensitive lipase to the lipid storage
droplet. PNAS 89 85378541. (doi:10.1073/pnas.89.18.8537)
Endo T & Kobayashi T 2012 Expression of functional TSH receptor in
white adipose tissues of hyt/hyt mice induces lipolysis in vivo.
American Journal of Physiology. Endocrinology and Metabolism 302
E1569E1575. (doi:10.1152/ajpendo.00572.2011)
Fain JN, Cheema P, Tichansky DS & Madan AK 2008 Stimulation of
human omental adipose tissue lipolysis by growth hormone plus
dexametha- sone. Molecular and Cellular Endocrinology 295 101105.
(doi:10.1016/
j.mce.2008.05.014)
Fischer J, Lefevre C, Morava E, Mussini JM, Laforet P, Negre-Salvayre A,
Lathrop M & Salvayre R 2007 The gene encoding adipose triglyceride
lipase (PNPLA2) is mutated in neutral lipid storage disease with
myopathy. Nature Genetics 39 2830. (doi:10.1038/ng1951)
Frayn KN 2002 Adipose tissue as a buffer for daily lipid flux. Diabetologia
45
12011210. (doi:10.1007/s00125-002-0873-y)
Frayn KN 2010 Fat as a fuel: emerging understanding of the adipose
tissueskeletal muscle axis. Acta Physiologica 199 509518.
(doi:10.1111/j.1748-1716.2010.02128.x)
Fredrikson G, Stralfors P, Nilsson NO & Belfrage P 1981 Hormonesensitive lipase of rat adipose tissue. Purification and some
properties. Journal of Biological Chemistry 256 63116320.
Fredrikson G, Tornqvist H & Belfrage P 1986 Hormone-sensitive lipase
and monoacylglycerol lipase are both required for complete
degradation of adipocyte triacylglycerol. Biochimica et Biophysica

Acta 876 288293.


(doi:10.1016/0005-2760(86)90286-9)
Furuhashi M & Hotamisligil GS 2008 Fatty acid-binding proteins: role in
metabolic diseases and potential as drug targets. Nature Reviews.
Drug Discovery 7 489503. (doi:10.1038/nrd2589)
Gandotra S, Le Dour C, Bottomley W, Cervera P, Giral P, Reznik Y,
Charpentier G, Auclair M, Delepine M, Barroso I et al. 2011a Perilipin
deficiency and autosomal dominant partial lipodystrophy. New
England Journal of Medicine 364 740748.
(doi:10.1056/NEJMoa1007487)
Gandotra S, Lim K, Girousse A, Saudek V, ORahilly S & Savage DB 2011b
Human frame shift mutations affecting the carboxyl terminus of
perilipin increase lipolysis by failing to sequester the adipose
triglyceride lipase (ATGL) coactivator AB-hydrolase-containing 5
(ABHD5). Journal of Biological Chemistry 286 3499835006.
(doi:10.1074/jbc.M111.278853)
Garton AJ, Campbell DG, Cohen P & Yeaman SJ 1988 Primary structure of
the site on bovine hormone-sensitive lipase phosphorylated by cyclic
AMP-dependent protein kinase. FEBS Letters 229 6872. (doi:10.1016/
0014-5793(88)80799-3)
Gasic S, Tian B & Green A 1999 Tumor necrosis factor a stimulates
lipolysis in adipocytes by decreasing Gi protein concentrations.
Journal of Biological Chemistry 274 67706775.
(doi:10.1074/jbc.274.10.6770)
Ghosh AK, Ramakrishnan G, Chandramohan C & Rajasekharan R 2008
CGI-58, the causative gene for ChanarinDorfman syndrome,
mediates acylation of lysophosphatidic acid. Journal of Biological
Chemistry 283 2452524533. (doi:10.1074/jbc.M801783200)
Gjedsted J, Gormsen LC, Nielsen S, Schmitz O, Djurhuus CB, Keiding S,
ORskov H, Tonnesen E & Moller N 2007 Effects of a 3-day fast on
regional lipid and glucose metabolism in human skeletal muscle and
adipose tissue. Acta Physiologica 191 205216. (doi:10.1111/j.17481716.2007.01740.x)
de Glisezinski I, Larrouy D, Bajzova M, Koppo K, Polak J, Berlan M, Bulow J,
Langin D, Marques MA, Crampes F et al. 2009 Adrenaline but not
noradrenaline is a determinant of exercise-induced lipid mobilization

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

in human subcutaneous adipose tissue. Journal of Physiology 587


33933404. (doi:10.1113/jphysiol.2009.168906)
Gormsen LC, Jessen N, Gjedsted J, Gjedde S, Norrelund H, Lund S,
Christiansen JS, Nielsen S, Schmitz O & Moller N 2007 Doseresponse
effects of free fatty acids on glucose and lipid metabolism during
somatostatin blockade of growth hormone and insulin in humans.
Journal of Clinical Endocrinology and Metabolism 92 1834
1842. (doi:10.1210/jc.2006-2659)
Grahn TH, Zhang Y, Lee MJ, Sommer AG, Mostoslavsky G, Fried SK,
Greenberg AS & Puri V 2013 FSP27 and PLIN1 interaction promotes
the formation of large lipid droplets in human adipocytes. Biochemical
and
Biophysical
Research
Communications
432
296301.
(doi:10.1016/j.bbrc.
2013.01.113)
Granneman JG, Moore HP, Granneman RL, Greenberg AS, Obin MS &
Zhu Z 2007 Analysis of lipolytic protein trafcking and interactions in
adipocytes. Journal of Biological Chemistry 282 57265735.
(doi:10.1074/ jbc.M610580200)
Granneman JG, Moore HP, Krishnamoorthy R & Rathod M 2009 Perilipin
controls lipolysis by regulating the interactions of AB-hydrolase
containing 5 (Abhd5) and adipose triglyceride lipase (Atgl). Journal of
Biological Chemistry 284 3453834544.
(doi:10.1074/jbc.M109.068478)
Gray NE, Lam LN, Yang K, Zhou AY, Koliwad S & Wang JC 2012
Angiopoietin-like 4 (Angptl4) protein is a physiological mediator of
intracellular lipolysis in murine adipocytes. Journal of Biological
Chemistry 287 84448456. (doi:10.1074/jbc.M111.294124)
Greenberg AS, Egan JJ, Wek SA, Garty NB, Blanchette-Mackie EJ &
Londos C 1991 Perilipin, a major hormonally regulated adipocytespecific phosphoprotein associated with the periphery of lipid storage
droplets. Journal of Biological Chemistry 266 1134111346.
Greenberg AS, Coleman RA, Kraemer FB, McManaman JL, Obin MS, Puri
V, Yan QW, Miyoshi H & Mashek DG 2011 The role of lipid
droplets in metabolic disease in rodents and humans. Journal of
Clinical Investi- gation 121 21022110. (doi:10.1172/JCI46069)
Gruber A, Cornaciu I, Lass A, Schweiger M, Poeschl M, Eder C, Kumari M,
Schoiswohl G, Wolinski H, Kohlwein SD et al. 2010 The N-terminal
region of comparative gene identification-58 (CGI-58) is important for
lipid droplet binding and activation of adipose triglyceride lipase.
Journal of Biological Chemistry 285 1228912298. (doi:10.1074/jbc.
M109.064469)
Haemmerle G, Zimmermann R, Hayn M, Theussl C, Waeg G, Wagner E,
Sattler W, Magin TM, Wagner EF & Zechner R 2002 Hormonesensitive lipase deficiency in mice causes diglyceride accumulation in
adipose tissue, muscle, and testis. Journal of Biological Chemistry 277
48064815. (doi:10.1074/jbc.M110355200)
Haemmerle G, Lass A, Zimmermann R, Gorkiewicz G, Meyer C, Rozman
J, Heldmaier G, Maier R, Theussl C, Eder S et al. 2006 Defective
lipolysis and altered energy metabolism in mice lacking adipose
triglyceride lipase. Science 312 734737.
(doi:10.1126/science.1123965)
Hansen TK, Gravholt CH, ORskov H, Rasmussen MH, Christiansen JS &
Jorgensen JO 2002 Dose dependency of the pharmacokinetics and
acute lipolytic actions of growth hormone. Journal of Clinical
Endocrinology and Metabolism 87 46914698. (doi:10.1210/jc.2002020563)
Harada K, Shen WJ, Patel S, Natu V, Wang J, Osuga J, Ishibashi S &
Kraemer FB 2003 Resistance to high-fat diet-induced obesity and
altered expression of adipose-specific genes in HSL-deficient mice.
American Journal of Physiology. Endocrinology and Metabolism 285
E1182E1195. (doi:10.1152/ajpendo.00259.2003)
Hardy OT, Czech MP & Corvera S 2012 What causes the insulin resistance
underlying obesity? Current Opinion in Endocrinology, Diabetes, and
Obesity 19 8187. (doi:10.1097/MED.0b013e3283514e13)
Heckmann BL, Zhang X, Xie X, Saarinen A, Lu X, Yang X & Liu J 2014
Defective adipose lipolysis and altered global energy metabolism in

Dissecting adipose tissue


lipolysis

52 :3

R217

mice with adipose overexpression of the lipolytic inhibitor G0/G1


switch gene 2 (G0S2). Journal of Biological Chemistry 289 19051916.
(doi:10.1074/jbc.M113.522011)

Hoeg LD, Sjoberg KA, Jeppesen J, Jensen TE, Frosig C, Birk JB, Bisiani B,
Hiscock N, Pilegaard H, Wojtaszewski JF et al. 2011 Lipid-induced
insulin resistance affects women less than men and is not
accompanied by inflammation or impaired proximal insulin
signaling. Diabetes 60
6473. (doi:10.2337/db10-0698)
Hollenberg CH, Raben MS & Astwood EB 1961 The lipolytic response to
corticotropin. Endocrinology 68 589598. (doi:10.1210/endo-68-4589) Huijsman E, van de Par C, Economou C, van der Poel C, Lynch GS,
Schoiswohl G, Haemmerle G, Zechner R & Watt MJ 2009 Adipose
triacylglycerol lipase deletion alters whole body energy metabolism
and impairs exercise performance in mice. American Journal of
Physiology. Endocrinology and Metabolism 297 E505E513.
(doi:10.1152/ ajpendo.00190.2009)
Igal RA & Coleman RA 1996 Acylglycerol recycling from triacylglycerol
to phospholipid, not lipase activity, is defective in neutral lipid
storage disease fibroblasts. Journal of Biological Chemistry 271
1664416651.
(doi:10.1074/jbc.271.28.16644)
Igal RA, Rhoads JM & Coleman RA 1997 Neutral lipid storage disease
with fatty liver and cholestasis. Journal of Pediatric Gastroenterology
and Nutrition 25 541547. (doi:10.1097/00005176-19971100000011)
Ito M, Nagasawa M, Hara T, Ide T & Murakami K 2010 Differential roles
of CIDEA and CIDEC in insulin-induced anti-apoptosis and lipid
droplet formation in human adipocytes. Journal of Lipid Research 51
16761684. (doi:10.1194/jlr.M002147)
Janson A, Karlsson FA, Micha-Johansson G, Bolme P, Bronnegard M &
Marcus C 1995 Effects of stimulatory and inhibitory thyrotropin
receptor antibodies on lipolysis in infant adipocytes. Journal of
Clinical Endocrinology and Metabolism 80 17121716.
(doi:10.1210/jcem.80.5.
7745024)
Jenkins CM, Mancuso DJ, Yan W, Sims HF, Gibson B & Gross RW 2004
Identification, cloning, expression, and purification of three novel
human calcium-independent phospholipase A2 family members
possessing triacylglycerol lipase and acylglycerol transacylase

activities.
Journal of Biological Chemistry 279 4896848975. (doi:10.1074/jbc.
M407841200)
Jenkins-Kruchten AE, Bennaars-Eiden A, Ross JR, Shen WJ, Kraemer FB &
Bernlohr DA 2003 Fatty acid-binding proteinhormone-sensitive
lipase interaction. Fatty acid dependence on binding. Journal of
Biological Chemistry 278 4763647643. (doi:10.1074/jbc.M307680200)
Jensen MD 1995 Gender differences in regional fatty acid
metabolism before and after meal ingestion. Journal of Clinical
Investigation 96 22972303. (doi:10.1172/JCI118285)
Jensen MD 2003 Fate of fatty acids at rest and during exercise: regulatory
mechanisms. Acta Physiologica Scandinavica 178 385390.
(doi:10.1046/ j.1365-201X.2003.01167.x)
Jensen MD & Nielsen S 2007 Insulin dose response analysis of free fatty
acid kinetics. Metabolism 56 6876.
(doi:10.1016/j.metabol.2006.08.022)
Jensen MD, Haymond MW, Gerich JE, Cryer PE & Miles JM 1987 Lipolysis
during fasting. Decreased suppression by insulin and increased
stimulation by epinephrine. Journal of Clinical Investigation 79 207213.
(doi:10.1172/JCI112785)
Jocken JW, Langin D, Smit E, Saris WH, Valle C, Hul GB, Holm C, Arner P
& Blaak EE 2007 Adipose triglyceride lipase and hormone-sensitive
lipase protein expression is decreased in the obese insulin-resistant
state.
Journal of Clinical Endocrinology and Metabolism 92 2292
2299. (doi:10.1210/jc.2006-1318)
Jocken JW, Moro C, Goossens GH, Hansen D, Mairal A, Hesselink MK,
Langin D, van Loon LJ & Blaak EE 2010 Skeletal muscle lipase content
and activity in obesity and type 2 diabetes. Journal of Clinical
Endocrinology and Metabolism 95 54495453. (doi:10.1210/jc.20100776)
Jocken JW, Goossens GH, Boon H, Mason RR, Essers Y, Havekes B, Watt
MJ, van Loon LJ & Blaak EE 2013 Insulin-mediated suppression of
lipolysis in adipose tissue and skeletal muscle of obese type 2 diabetic
men and men with normal glucose tolerance. Diabetologia 56 2255
2265.
(doi:10.1007/s00125-013-2995-9)

Review

M
ol
ec
ul
ar
E
n
d
oc
ri
n
ol
o
g
y

T S NIELSEN

and others

Johansen T, Richelsen B, Hansen HS, Din N & Malmlof K 2003 Growth


hormone-mediated breakdown of body fat: effects of GH on lipases in
adipose tissue and skeletal muscle of old rats fed different diets.
Hormone and Metabolic Research 35 243250. (doi:10.1055/s-200339481)
Jorgensen JO, Moller N, Lauritzen T, Alberti KG, ORskov H &
Christiansen JS 1990 Evening versus morning injections of growth
hormone (GH) in GH-deficient patients: effects on 24-hour patterns
of circulating hormones and metabolites. Journal of Clinical
Endocrinology and Metabolism 70 207214. (doi:10.1210/jcem-70-1207)
Kanaley JA, Dall R, Moller N, Nielsen SC, Christiansen JS, Jensen MD &
Jorgensen JO 2004 Acute exposure to GH during exercise stimulates
the turnover of free fatty acids in GH-deficient men. Journal of Applied
Physiology 96 747753. (doi:10.1152/japplphysiol.00711.2003)
Kershaw EE, Hamm JK, Verhagen LA, Peroni O, Katic M & Flier JS 2006
Adipose triglyceride lipase: function, regulation by insulin, and
comparison with adiponutrin. Diabetes 55 148157. (doi:10.2337/
diabetes.55.01.06.db05-0982)
Kienesberger PC, Lee D, Pulinilkunnil T, Brenner DS, Cai L, Magnes C,
Koefeler HC, Streith IE, Rechberger GN, Haemmerle G et al. 2009
Adipose triglyceride lipase deficiency causes tissue-specific changes in
insulin signaling. Journal of Biological Chemistry 284 3021830229.
(doi:10.1074/jbc.M109.047787)
Kim JY, Tillison K, Lee JH, Rearick DA & Smas CM 2006 The adipose tissue
triglyceride lipase ATGL/PNPLA2 is downregulated by insulin and
TNF-a in 3T3-L1 adipocytes and is a target for transactivation by
PPARgamma. American Journal of Physiology. Endocrinology and Metabolism 291 E115E127. (doi:10.1152/ajpendo.00317.2005)
Kimmel AR, Brasaemle DL, McAndrews-Hill M, Sztalryd C & Londos C
2010 Adoption of PERILIPIN as a unifying nomenclature for the
mammalian PAT-family of intracellular lipid storage droplet proteins.
Journal of Lipid Research 51 468471. (doi:10.1194/jlr.R000034)
Kiwaki K & Levine JA 2003 Differential effects of adrenocorticotropic
hormone on human and mouse adipose tissue. Journal of Comparative
Physiology. B, Biochemical, Systemic, and Environmental Physiology 173
675678. (doi:10.1007/s00360-003-0377-1)
Kobayashi K, Inoguchi T, Maeda Y, Nakashima N, Kuwano A, Eto E, Ueno N
, Sasaki S, Sawada F, Fujii M et al. 2008 The lack of the C-terminal
domain of adipose triglyceride lipase causes neutral lipid storage
disease through impaired interactions with lipid droplets. Journal of
Clinical Endocrinology and Metabolism 93 28772884.
(doi:10.1210/jc. 2007-2247)
Kolditz CI & Langin D 2010 Adipose tissue lipolysis. Current Opinion in
Clinical Nutrition and Metabolic Care 13 377381. (doi:10.1097/MCO.
0b013e32833bed6a)
Koliwad SK, Kuo T, Shipp LE, Gray NE, Backhed F, So AY, Farese RV Jr &
Wang JC 2009 Angiopoietin-like 4 (ANGPTL4, fasting-induced
adipose factor) is a direct glucocorticoid receptor target and
participates in glucocorticoid-regulated triglyceride metabolism.
Journal of Biological Chemistry 284 2559325601.
(doi:10.1074/jbc.M109.025452)
Koliwad SK, Gray NE & Wang JC 2012 Angiopoietin-like 4 (Angptl4): a
glucocorticoid-dependent gatekeeper of fatty acid flux during fasting.
Adipocytes 1 182187. (doi:10.4161/adip.20787)
Koster A, Chao YB, Mosior M, Ford A, Gonzalez-DeWhitt PA, Hale JE, Li
D, Qiu Y, Fraser CC, Yang DD et al. 2005 Transgenic angiopoietin-like
(angptl)4 overexpression and targeted disruption of angptl4 and
angptl3: regulation of triglyceride metabolism. Endocrinology 146
49434950. (doi:10.1210/en.2005-0476)
Kraemer FB, Patel S, Saedi MS & Sztalryd C 1993 Detection of hormonesensitive lipase in various tissues. I. Expression of an HSL/bacterial
fusion protein and generation of anti-HSL antibodies. Journal of Lipid
Research 34 663671.
Lacasa D, Agli B & Giudicelli Y 1988 Permissive action of glucocorticoids
on catecholamine-induced lipolysis: direct in vitro effects on the fat

Dissecting adipose tissue


lipolysis

52 :3

R218

cell b-adrenoreceptor-coupled-adenylate cyclase system. Biochemical


and Biophysical Research Communications 153 489497. (doi:10.1016/
S0006-291X(88)81121-5)

Lafferty MJ, Bradford KC, Erie DA & Neher SB 2013 Angiopoietin-like


Protein 4 Inhibition of Lipoprotein Lipase: evidence for reversible
complex formation. Journal of Biological Chemistry 288 28524
28534. (doi:10.1074/jbc.M113.497602)
Lafontan M & Berlan M 1993 Fat cell adrenergic receptors and the control
of white and brown fat cell function. Journal of Lipid Research 34
10571091.
Lafontan M, Moro C, Berlan M, Crampes F, Sengenes C & Galitzky J
2008 Control of lipolysis by natriuretic peptides and cyclic GMP.
Trends in Endocrinology and Metabolism 19 130137.
(doi:10.1016/j.tem.2007.
11.006)
Laforet P, Stojkovic T, Bassez G, Carlier PG, Clement K, Wahbi K, Petit FM,
Eymard B & Carlier RY 2013 Neutral lipid storage disease with
myopathy: a whole-body nuclear MRI and metabolic study.
Molecular Genetics and Metabolism 108 125131.
(doi:10.1016/j.ymgme.2012.
12.004)
Langin D 2006 Control of fatty acid and glycerol release in adipose
tissue lipolysis. Comptes Rendus Biologies 329 598607.
(doi:10.1016/j.crvi.
2005.10.008)
Langin D, Laurell H, Holst LS, Belfrage P & Holm C 1993 Gene
organization and primary structure of human hormone-sensitive
lipase: possible
significance of a sequence homology with a lipase of Moraxella
TA144,
an
antarctic
bacterium.
PNAS
90
48974901.
(doi:10.1073/pnas.90.11. 4897)
Langin D, Dicker A, Tavernier G, Hoffstedt J, Mairal A, Ryden M, Arner E,
Sicard A, Jenkins CM, Viguerie N et al. 2005 Adipocyte lipases and
defect of lipolysis in human obesity. Diabetes 54 31903197.
(doi:10.2337/diabetes.54.11.3190)
Lapointe A, Piche ME, Weisnagel SJ, Bergeron J & Lemieux S 2009
Associations between circulating free fatty acids, visceral adipose
tissue accumulation, and insulin sensitivity in postmenopausal
women.
Metabolism 58 180185. (doi:10.1016/j.metabol.2008.09.011)
Large V, Reynisdottir S, Langin D, Fredby K, Klannemark M, Holm C

& Arner P 1999 Decreased expression and function of adipocyte


hormone-sensitive lipase in subcutaneous fat cells of obese subjects.
Journal of Lipid Research 40 20592066.
Lass A, Zimmermann R, Haemmerle G, Riederer M, Schoiswohl G,
Schweiger M, Kienesberger P, Strauss JG, Gorkiewicz G & Zechner R
2006 Adipose triglyceride lipase-mediated lipolysis of cellular fat
stores is activated by CGI-58 and defective in ChanarinDorfman
Syndrome. Cell Metabolism 3 309319.
(doi:10.1016/j.cmet.2006.03.005)
Laugwitz KL, Allgeier A, Offermanns S, Spicher K, Van Sande J, Dumont JE
& Schultz G 1996 The human thyrotropin receptor: a heptahelical
receptor capable of stimulating members of all four G protein
families. PNAS 93 116120. (doi:10.1073/pnas.93.1.116)
Lefevre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, Lakhdar
H, Wollenberg A, Verret JL, Weissenbach J et al. 2001 Mutations in
CGI-58,
the
gene
encoding
a
new
protein
of
the
esterase/lipase/thioesterase
subfamily, in ChanarinDorfman syndrome. American Journal of
Human Genetics 69 10021012. (doi:10.1086/324121)
Li JZ, Ye J, Xue B, Qi J, Zhang J, Zhou Z, Li Q, Wen Z & Li P 2007 Cideb
regulates diet-induced obesity, liver steatosis, and insulin sensitivity
by controlling lipogenesis and fatty acid oxidation. Diabetes 56
25232532. (doi:10.2337/db07-0040)
Li YC, Zheng XL, Liu BT & Yang GS 2010 Regulation of ATGL expression
mediated by leptin in vitro in porcine adipocyte lipolysis. Molecular
and Cellular Biochemistry 333 121128. (doi:10.1007/s11010-0090212-4)
Liang HX, Belardinelli L, Ozeck MJ & Shryock JC 2002 Tonic activity of
the rat adipocyte A1-adenosine receptor. British Journal of
Pharmacology 135 14571466. (doi:10.1038/sj.bjp.0704586)
Listenberger LL, Ostermeyer-Fay AG, Goldberg EB, Brown WJ & Brown
DA 2007 Adipocyte differentiation-related protein reduces the lipid
droplet association of adipose triglyceride lipase and slows
triacylglycerol turnover. Journal of Lipid Research 48 27512761.
(doi:10.1194/jlr.
M700359-JLR200)
Liu C, Wu J, Zhu J, Kuei C, Yu J, Shelton J, Sutton SW, Li X, Yun SJ,
Mirzadegan T et al. 2009a Lactate inhibits lipolysis in fat cells through

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

activation of an orphan G-protein-coupled receptor, GPR81. Journal


of Biological Chemistry 284 28112822.
(doi:10.1074/jbc.M806409200)
Liu LF, Purushotham A, Wendel AA, Koba K, Deiuliis J, Lee K & Belury
MA 2009b Regulation of adipose triglyceride lipase by rosiglitazone.
Diabetes, Obesity & Metabolism 11 131142. (doi:10.1111/j.14631326. 2008.00916.x)
Londos C, Honnor RC & Dhillon GS 1985 cAMP-dependent protein
kinase and lipolysis in rat adipocytes. III. Multiple modes of insulin
regulation of lipolysis and regulation of insulin responses by
adenylate cyclase regulators. Journal of Biological Chemistry 260
1513915145.
Lonnroth P, Jansson PA, Fredholm BB & Smith U 1989 Microdialysis of
intercellular adenosine concentration in subcutaneous tissue in
humans. American Journal of Physiology 256 E250E255.
Lord CC, Betters JL, Ivanova PT, Milne SB, Myers DS, Madenspacher J,
Thomas G, Chung S, Liu M, Davis MA et al. 2012 CGI-58/ABHD5derived signaling lipids regulate systemic inflammation and insulin
action. Diabetes 61 355363. (doi:10.2337/db11-0994)
Lundgren M, Svensson M, Lindmark S, Renstrom F, Ruge T & Eriksson
JW 2007 Fat cell enlargement is an independent marker of insulin
resistance and hyperleptinaemia. Diabetologia 50 625633.
(doi:10.1007/s00125-006-0572-1)
Mairal A, Melaine N, Laurell H, Grober J, Holst LS, Guillaudeux T, Holm
C, Jegou B & Langin D 2002 Characterization of a novel testicular
form of human hormone-sensitive lipase. Biochemical and Biophysical
Research Communications 291 286290.
(doi:10.1006/bbrc.2002.6427)
Mairal A, Langin D, Arner P & Hoffstedt J 2006 Human adipose triglyceride
lipase (PNPLA2) is not regulated by obesity and exhibits low in vitro
triglyceride hydrolase activity. Diabetologia 49 16291636.
(doi:10.1007/s00125-006-0272-x)
Manolopoulos KN, Karpe F & Frayn KN 2010 Gluteofemoral body fat as a
determinant of metabolic health. International Journal of Obesity 34
949959. (doi:10.1038/ijo.2009.286)
Manolopoulos KN, Karpe F & Frayn KN 2012 Marked resistance of
femoral adipose tissue blood flow and lipolysis to adrenaline in vivo.
Diabetologia 55 30293037. (doi:10.1007/s00125-012-2676-0)
Marcus C, Bolme P, Micha-Johansson G, Margery V & Bronnegard M 1994
Growth hormone increases the lipolytic sensitivity for catecholamines
in adipocytes from healthy adults. Life Sciences 54 13351341.
(doi:10.1016/0024-3205(94)00512-5)
Marcus C, Ehren H, Bolme P & Arner P 1988 Regulation of lipolysis
during the neonatal period. Importance of thyrotropin. Journal of
Clinical Investigation 82 17931797. (doi:10.1172/JCI113793)
Martinez-Botas J, Anderson JB, Tessier D, Lapillonne A, Chang BH,
Quast MJ, Gorenstein D, Chen KH & Chan L 2000 Absence of perilipin
results in leanness and reverses obesity in Lepr(db/db) mice. Nature
Genetics 26 474479. (doi:10.1038/82630)
Mason RR, Meex RC, Lee-Young R, Canny BJ & Watt MJ 2012
Phosphorylation of adipose triglyceride lipase Ser(404) is not related
to 50 -AMPK activation during moderate-intensity exercise in humans.
American Journal of Physiology. Endocrinology and Metabolism 303
E534E541. (doi:10.1152/ajpendo.00082.2012)
Mauriege P, De PG, Berlan M & Lafontan M 1988 Human fat cell
b-adrenergic receptors: b-agonist-dependent lipolytic responses and
characterization of b-adrenergic binding sites on human fat cell
membranes with highly selective b1-antagonists. Journal of Lipid
Research 29 587601.
Miyoshi H, Souza SC, Zhang HH, Strissel KJ, Christoffolete MA, Kovsan J,
Rudich A, Kraemer FB, Bianco AC, Obin MS et al. 2006 Perilipin
promotes hormone-sensitive lipase-mediated adipocyte lipolysis via
phosphorylation-dependent and -independent mechanisms. Journal of
Biological Chemistry 281 1583715844. (doi:10.1074/jbc.M601097200)
Miyoshi H, Perfield JW, Souza SC, Shen WJ, Zhang HH, Stancheva ZS,
Kraemer FB, Obin MS & Greenberg AS 2007 Control of adipose

Dissecting adipose tissue


lipolysis

52 :3

R219

triglyceride lipase action by serine 517 of perilipin A globally regulates


protein kinase A-stimulated lipolysis in adipocytes. Journal of Biological
Chemistry 282 9961002. (doi:10.1074/jbc.M605770200)

Montero-Moran G, Caviglia JM, McMahon D, Rothenberg A,


Subramanian V, Xu Z, Lara-Gonzalez S, Storch J, Carman GM &
Brasaemle DL 2010 CGI-58/ABHD5 is a coenzyme A-dependent
lysophosphatidic acid acyltransferase. Journal of Lipid Research 51
709719. (doi:10.1194/jlr.M001917)
Moreno-Navarrete JM, Ortega F, Serrano M, Rodriguez-Hermosa JI,
Ricart W, Mingrone G & Fernandez-Real JM 2013 CIDEC/FSP27 and
PLIN1 gene expression run in parallel to mitochondrial genes in
human adipose tissue, both increasing after weight loss.
International
Journal
of
Obesity
[in
press].
(doi:10.1038/ijo.2013.171)
Moro C, Galitzky J, Sengenes C, Crampes F, Lafontan M & Berlan M
2004a Functional and pharmacological characterization of the
natriuretic peptide-dependent lipolytic pathway in human fat cells.
Journal of Pharmacological and Experimental Therapeutics 308 984992.
(doi:10.1124/jpet.103.060913)
Moro C, Crampes F, Sengenes C, de GI, Galitzky J, Thalamas C, Lafontan
M
& Berlan M 2004b Atrial natriuretic peptide contributes to
physiologi- cal control of lipid mobilization in humans. FASEB
Journal 18 908910. (doi:10.1096/fj.03-1086fje)
Moro C, Polak J, Hejnova J, Klimcakova E, Crampes F, Stich V, Lafontan M
& Berlan M 2006 Atrial natriuretic peptide stimulates lipid
mobilization during repeated bouts of endurance exercise. American
Journal of Physiology. Endocrinology and Metabolism 290 E864E869.
(doi:10.1152/ ajpendo.00348.2005)
Moro C, Klimcakova E, Lafontan M, Berlan M & Galitzky J 2007a
Phosphodiesterase-5A and neutral endopeptidase activities in human
adipocytes do not control atrial natriuretic peptide-mediated
lipolysis. British Journal of Pharmacology 152 11021110.
(doi:10.1038/sj.bjp.
0707485)
Moro C, Pillard F, de GI, Crampes F, Thalamas C, Harant I, Marques
MA, Lafontan M & Berlan M 2007b Sex differences in lipolysisregulating mechanisms in overweight subjects: effect of exercise
intensity. Obesity 15 22452255. (doi:10.1038/oby.2007.267)
Nam SY, Kim KR, Cha BS, Song YD, Lim SK, Lee HC & Huh KB 2001

Low-dose growth hormone treatment combined with diet restriction


decreases insulin resistance by reducing visceral fat and increasing
muscle mass in obese type 2 diabetic patients. International Journal of
Obesity and Related Metabolic Disorders 25 11011107.
(doi:10.1038/sj.ijo.0801636)
Natali A, Gastaldelli A, Camastra S, Baldi S, Quagliarini F, Minicocci I,
Bruno C, Pennisi E & Arca M 2013 Metabolic consequences of adipose
triglyceride lipase deficiency in humans: an in vivo study in patients
with neutral lipid storage disease with myopathy. Journal of Clinical
Endocrinology and Metabolism 98 E1540E1548. (doi:10.1210/jc.20131444)
Nellemann B, Gormsen LC, Sorensen LP, Christiansen JS & Nielsen S 2012
Impaired insulin-mediated antilipolysis and lactate release in adipose
tissue of upper-body obese women. Obesity 20 5764.
(doi:10.1038/oby. 2011.290)
Nellemann B, Vendelbo MH, Nielsen TS, Bak AM, Hogild M, Pedersen
SB, Bienso RS, Pilegaard H, Moller N, Jessen N et al. 2004 Growth
hormone- induced insulin resistance in human subjects involves
reduced
pyruvate dehydrogenase activity. Acta Physiologica 210 392402.
(doi:10.1111/apha.12183)
Ng TB 1990 Studies on hormonal regulation of lipolysis and
lipogenesis in fat cells of various mammalian species. Comparative
Biochemistry and Physiology. B, Comparative Biochemistry 97 441446.
(doi:10.1016/0305- 0491(90)90141-F)
Nielsen S, Moller N, Christiansen JS & Jorgensen JO 2001 Pharmacological
antilipolysis restores insulin sensitivity during growth hormone
exposure. Diabetes 50 23012308. (doi:10.2337/diabetes.50.10.2301)
Nielsen S, Jorgensen JO, Hartmund T, Norrelund H, Nair KS, Christiansen
JS & Moller N 2002 Effects of lowering circulating free fatty acid levels
on protein metabolism in adult growth hormone deficient patients.
Growth Hormone & IGF Research 12 425433. (doi:10.1016/S10966374(02)00119-3)
Nielsen S, Guo Z, Johnson CM, Hensrud DD & Jensen MD 2004
Splanchnic lipolysis in human obesity. Journal of Clinical Investigation
113
15821588. (doi:10.1172/JCI21047)

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

Nielsen TS, Vend elbo MH, Jessen N, Pedersen SB, Jorgensen JO, Lund S &
Moller N 2011 Fasting, but not exercise, increases adipose triglyceride
lipase (ATGL) protein and reduces G(0)/G(1) switch gene 2 (G0S2)
protein and mRNA content in human adipose tissue. Journal of Clinical
Endocrinology and Metabolism 96 E1293E1297. (doi:10.1210/jc.20110149)
Nielsen TS, Kampmann U, Nielsen RR, Jessen N, Orskov L, Pedersen SB,
Jorgensen JO, Lund S & Moller N 2012 Reduced mRNA and protein
expression of perilipin A and G0/G1 switch gene 2 (G0S2) in human
adipose tissue in poorly controlled type 2 diabetes. Journal of Clinical
Endocrinology and Metabolism 97 E1348E1352. (doi:10.1210/jc.20121159)
Nishino N, Tamori Y, Tateya S, Kawaguchi T, Shibakusa T, Mizunoya W,
Inoue K, Kitazawa R, Kitazawa S, Matsuki Y et al. 2008 FSP27
contributes to efcient energy storage in murine white adipocytes by
promoting the formation of unilocular lipid droplets. Journal of
Clinical Investigation 118 28082821. (doi:10.1172/JCI34090)
Nordstrom EA, Ryden M, Backlund EC, Dahlman I, Kaaman M, Blomqvist L,
Cannon B, Nedergaard J & Arner P 2005 A human-specific role of cell
death-inducing DFFA (DNA fragmentation factor-a)-like effector A
(CIDEA) in adipocyte lipolysis and obesity. Diabetes 54 17261734.
(doi:10.2337/diabetes.54.6.1726)
Norrelund H, Moller N, Nair KS, Christiansen JS & Jorgensen JO 2001
Continuation of growth hormone (GH) substitution during fasting in
GH-deficient patients decreases urea excretion and conserves protein
synthesis. Journal of Clinical Endocrinology and Metabolism 86 3120
3129. (doi:10.1210/jcem.86.7.7618)
Norrelund H, Nair KS, Nielsen S, Frystyk J, Ivarsen P, Jorgensen JO,
Christiansen JS & Moller N 2003 The decisive role of free fatty acids
for protein conservation during fasting in humans with and without
growth hormone. Journal of Clinical Endocrinology and Metabolism 88
43714378. (doi:10.1210/jc.2003-030267)
Oh SA, Suh Y, Pang MG & Lee K 2011 Cloning of avian G(0)/G(1) switch
gene 2 genes and developmental and nutritional regulation of
G(0)/G(1) switch gene 2 in chicken adipose tissue. Journal of Animal
Science 89 367375. (doi:10.2527/jas.2010-3339)
Ong KT, Mashek MT, Bu SY, Greenberg AS & Mashek DG 2011 Adipose
triglyceride lipase is a major hepatic lipase that regulates
triacylglycerol turnover and fatty acid signaling and partitioning.
Hepatology 53
116126. (doi:10.1002/hep.24006)
Osuga J, Ishibashi S, Oka T, Yagyu H, Tozawa R, Fujimoto A, Shionoiri F,
Yahagi N, Kraemer FB, Tsutsumi O et al. 2000 Targeted disruption of
hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity. PNAS 97 787792.
(doi:10.1073/pnas.97.2.787)
Pagnon J, Matzaris M, Stark R, Meex RC, Macaulay SL, Brown W, OBrien
PE, Tiganis T & Watt MJ 2012 Identification and functional
characterization of protein kinase A phosphorylation sites in the
major lipolytic protein, adipose triglyceride lipase. Endocrinology 153
42784289. (doi:10.1210/ en.2012-1127)
Park SY, Kim HJ, Wang S, Higashimori T, Dong J, Kim YJ, Cline G, Li H,
Prentki M, Shulman GI et al. 2005 Hormone-sensitive lipase knockout
mice have increased hepatic insulin sensitivity and are protected from
short-term diet-induced insulin resistance in skeletal muscle and
heart. American Journal of Physiology. Endocrinology and Metabolism 289
E30E39. (doi:10.1152/ajpendo.00251.2004)
Pasarica M, Zachwieja JJ, Dejonge L, Redman S & Smith SR 2007 Effect of
growth hormone on body composition and visceral adiposity in
middle-aged men with visceral obesity. Journal of Clinical
Endocrinology and Metabolism 92 42654270. (doi:10.1210/jc.20070786)
Peyot ML, Guay C, Latour MG, Lamontagne J, Lussier R, Pineda M,
Ruderman NB, Haemmerle G, Zechner R, Joly E et al. 2009 Adipose
triglyceride lipase is implicated in fuel- and non-fuel-stimulated
insulin secretion. Journal of Biological Chemistry 284 1684816859.

Dissecting adipose tissue


lipolysis

52 :3

R220

(doi:10.1074/jbc.M109.006650)
Piche ME, Lapointe A, Weisnagel SJ, Corneau L, Nadeau A, Bergeron J &
Lemieux S 2008 Regional body fat distribution and metabolic profile in
postmenopausal women. Metabolism 57 11011107. (doi:10.1016/
j.metabol.2008.03.015)

Plockinger U & Reuter T 2008 Pegvisomant increases intra-abdominal


fat in patients with acromegaly: a pilot study. European Journal of
Endocrinology 158 467471. (doi:10.1530/EJE-07-0637)
Pulinilkunnil T, Kienesberger PC, Nagendran J, Waller TJ, Young ME,
Kershaw EE, Korbutt G, Haemmerle G, Zechner R & Dyck JR 2013
Myocardial adipose triglyceride lipase overexpression protects
diabetic mice from the development of lipotoxic cardiomyopathy.
Diabetes 62 14641477. (doi:10.2337/db12-0927)
Puri V, Ranjit S, Konda S, Nicoloro SM, Straubhaar J, Chawla A,
Chouinard M, Lin C, Burkart A, Corvera S et al. 2008 Cidea is
associated with lipid droplets and insulin sensitivity in humans.
PNAS 105
78337838. (doi:10.1073/pnas.0802063105)
Radner FP, Streith IE, Schoiswohl G, Schweiger M, Kumari M, Eichmann
TO, Rechberger G, Koefeler HC, Eder S, Schauer S et al. 2009 Growth
retardation, impaired triacylglycerol catabolism, hepatic steatosis,
and lethal skin barrier defect in mice lacking comparative gene
identification-58 (CGI-58). Journal of Biological Chemistry 285 73007311.
(doi:10.1074/jbc.M109.081877)
Ray H, Pinteur C, Frering V, Beylot M & Large V 2009 Depot-specific
differences in perilipin and hormone-sensitive lipase expression in
lean and obese. Lipids in Health and Disease 8 58.
(doi:10.1186/1476-511X-8-58)
Reid BN, Ables GP, Otlivanchik OA, Schoiswohl G, Zechner R, Blaner
WS, Goldberg IJ, Schwabe RF, Chua SC Jr & Huang LS 2008
Hepatic overexpression of hormone-sensitive lipase and adipose
triglyceride lipase promotes fatty acid oxidation, stimulates direct
release of free fatty acids, and ameliorates steatosis. Journal of
Biological Chemistry 283 1308713099.
(doi:10.1074/jbc.M800533200)
Richter WO & Schwandt P 1983 In vitro lipolysis of proopiocortin
peptides.
Life Sciences 33 (Suppl 1) 747750. (doi:10.1016/00243205(83)90610-0)
Rizack MA 1964 Activation of an epinephrine-sensitive lipolytic activity
from adipose tissue by adenosine 3 0 ,50 -phosphate. Journal of
Biological
Chemistry 239 392395.

Robidoux J, Martin TL & Collins S 2004 b-Adrenergic receptors and


regulation of energy expenditure: a family affair. Annual Review of
Pharmacology and Toxicology 44 297323. (doi:10.1146/annurev.
pharmtox.44.101802.121659)
Roden M, Stingl H, Chandramouli V, Schumann WC, Hofer A, Landau
BR, Nowotny P, Waldhausl W & Shulman GI 2000 Effects of free fatty
acid elevation on postabsorptive endogenous glucose production and
gluconeogenesis in humans. Diabetes 49 701707. (doi:10.2337/
diabetes.49.5.701)
Rodrigues AR, Almeida H & Gouveia AM 2013 a-MSH signalling
via melanocortin 5 receptor promotes lipolysis and impairs
re-esterification in adipocytes. Biochimica et Biophysica Acta 1831
12671275. (doi:10.1016/j.bbalip.2013.04.008)
Roust LR & Jensen MD 1993 Postprandial free fatty acid kinetics are
abnormal in upper body obesity. Diabetes 42 15671573.
(doi:10.2337/diab.42.11.1567)
Rubio-Cabezas O, Puri V, Murano I, Saudek V, Semple RK, Dash S,
Hyden CS, Bottomley W, Vigouroux C, Magre J et al. 2009 Partial
lipodystrophy and insulin resistant diabetes in a patient with a
homozygous nonsense mutation in CIDEC. EMBO Molecular Medicine 1
280287. (doi:10.1002/emmm.200900037)
Ryden M, Dicker A, van HV, Hauner H, Brunnberg M, Perbeck L, Lonnqvist
F
& Arner P 2002 Mapping of early signaling events in tumor
necrosis factor-a-mediated lipolysis in human fat cells. Journal of
Biological Chemistry 277 10851091.
(doi:10.1074/jbc.M109498200)
Ryden M, Jocken J, van HV, Dicker A, Hoffstedt J, Wiren M, Blomqvist
L, Mairal A, Langin D, Blaak E et al. 2007 Comparative studies of
the role of hormone-sensitive lipase and adipose triglyceride lipase in
human fat cell lipolysis. American Journal of Physiology. Endocrinology
and Metabolism 292 E1847E1855.
(doi:10.1152/ajpendo.00040.2007)
Santomauro AT, Boden G, Silva ME, Rocha DM, Santos RF, Ursich MJ,
Strassmann PG & Wajchenberg BL 1999 Overnight lowering of free
fatty acids with Acipimox improves insulin resistance and glucose
tolerance in obese diabetic and nondiabetic subjects. Diabetes 48
18361841. (doi:10.2337/diabetes.48.9.1836)

Review

M
ol
ec
ul
ar
E
n
d
oc
ri
n
ol
o
g
y

T S NIELSEN

and others

Scherer T, OHare J, Diggs-Andrews K, Schweiger M, Cheng B, Lindtner


C, Zielinski E, Vempati P, Su K, Dighe S et al. 2011 Brain insulin
controls adipose tissue lipolysis and lipogenesis. Cell Metabolism 13
183194. (doi:10.1016/j.cmet.2011.01.008)
Schoiswohl G, Schweiger M, Schreiber R, Gorkiewicz G, Preiss-Landl K,
Taschler U, Zierler KA, Radner FP, Eichmann TO, Kienesberger PC et
al. 2010 Adipose triglyceride lipase plays a key role in the supply of
the working muscle with fattyacids. Journal of Lipid Research 51 490
499. (doi:10.1194/jlr.M001073)
Schrammel A, Mussbacher M, Winkler S, Haemmerle G, Stessel H,
Wolkart G, Zechner R & Mayer B 2013 Cardiac oxidative stress in a
mouse model of neutral lipid storage disease. Biochimica et
Biophysica Acta 1831 16001608. (doi:10.1016/j.bbalip.2013.07.004)
Schweiger M, Schreiber R, Haemmerle G, Lass A, Fledelius C, Jacobsen P,
Tornqvist H, Zechner R & Zimmermann R 2006 Adipose triglyceride
lipase and hormone-sensitive lipase are the major enzymes in adipose
tissue triacylglycerol catabolism. Journal of Biological Chemistry 281
4023640241. (doi:10.1074/jbc.M608048200)
Schweiger M, Schoiswohl G, Lass A, Radner FP, Haemmerle G, Malli R,
Graier W, Cornaciu I, Oberer M, Salvayre R et al. 2008 The Cterminal region of human adipose triglyceride lipase affects enzyme
activity and lipid droplet binding. Journal of Biological Chemistry
283 1721117220. (doi:10.1074/jbc.M710566200)
Schweiger M, Lass A, Zimmermann R, Eichmann TO & Zechner R 2009
Neutral lipid storage disease: genetic disorders caused by mutations
in adipose triglyceride lipase/PNPLA2 or CGI-58/ABHD5. American
Journal of Physiology. Endocrinology and Metabolism 297 E289E296.
(doi:10.1152/ajpendo.00099.2009)
Schweiger M, Paar M, Eder C, Brandis J, Moser E, Gorkiewicz G, Grond S,
Radner FP, Cerk I, Cornaciu I et al. 2012 G0/G1 switch gene-2
regulates human adipocyte lipolysis by affecting activity and
localization of adipose triglyceride lipase. Journal of Lipid Research 53
23072317.
(doi:10.1194/jlr.M027409)
Sengenes C, Berlan M, de GI, Lafontan M & Galitzky J 2000 Natriuretic
peptides: a new lipolytic pathway in human adipocytes. FASEB
Journal 14 13451351. (doi:10.1096/fj.14.10.1345)
Sengenes C, Stich V, Berlan M, Hejnova J, Lafontan M, Pariskova Z &
Galitzky J 2002 Increased lipolysis in adipose tissue and lipid
mobilization to natriuretic peptides during low-calorie diet in obese
women. International Journal of Obesity and Related Metabolic Disorders
26 2432. (doi:10.1038/sj.ijo.0801845)
Sengenes C, Bouloumie A, Hauner H, Berlan M, Busse R, Lafontan M &
Galitzky J 2003 Involvement of a cGMP-dependent pathway in the
natriuretic peptide-mediated hormone-sensitive lipase phosphorylation in human adipocytes. Journal of Biological Chemistry 278
4861748626. (doi:10.1074/jbc.M303713200)
Serradeil-Le GC, Lafontan M, Raufaste D, Marchand J, Pouzet B, Casellas
P, Pascal M, Maffrand JP & Le FG 2000 Characterization of NPY
receptors controlling lipolysis and leptin secretion in human
adipocytes. FEBS Letters 475 150156. (doi:10.1016/S00145793(00)01649-5)
Sethi JK, Xu H, Uysal KT, Wiesbrock SM, Scheja L & Hotamisligil GS 2000
Characterisation of receptor-specific TNFa functions in adipocyte cell
lines lacking type 1 and 2 TNF receptors. FEBS Letters 469 7782.
(doi:10.1016/S0014-5793(00)01250-3)
Shan T, Ren Y & Wang Y 2013 Sirtuin 1 affects the transcriptional
expression of adipose triglyceride lipase in porcine adipocytes.
Journal of Animal Science 91 12471254. (doi:10.2527/jas.20115030)
Shen WJ, Patel S, Miyoshi H, Greenberg AS & Kraemer FB 2009 Functional
interaction of hormone-sensitive lipase and perilipin in lipolysis.
Journal of Lipid Research 50 23062313. (doi:10.1194/jlr.M900176JLR200)
Smith AJ, Thompson BR, Sanders MA & Bernlohr DA 2007 Interaction of

Dissecting adipose tissue


lipolysis

52 :3

R221

the adipocyte fatty acid-binding protein with the hormone-sensitive


lipase: regulation by fatty acids and phosphorylation. Journal of
Biological Chemistry 282 3242432432. (doi:10.1074/jbc.M703730200)
Souza SC, de Vargas LM, Yamamoto MT, Lien P, Franciosa MD, Moss LG &
Greenberg AS 1998 Overexpression of perilipin A and B blocks the

ability of tumor necrosis factor a to increase lipolysis in 3T3-L1


adipocytes. Journal of Biological Chemistry 273 2466524669.
(doi:10.1074/jbc.273.38.24665)
Stallknecht B, Lorentsen J, Enevoldsen LH, Bulow J, Biering-Sorensen F,
Galbo H & Kjaer M 2001 Role of the sympathoadrenergic system in
adipose tissue metabolism during exercise in humans. Journal of
Physiology 536 283294. (doi:10.1111/j.1469-7793.2001.00283.x)
Steinberg GR, Kemp BE & Watt MJ 2007 Adipocyte triglyceride lipase
expression in human obesity. American Journal of Physiology.
Endocrinology and Metabolism 293 E958E964.
(doi:10.1152/ajpendo.00235.2007)
Stokoe D, Stephens LR, Copeland T, Gaffney PR, Reese CB, Painter GF,
Holmes AB, McCormick F & Hawkins PT 1997 Dual role of
phosphatidylinositol-3,4,5-trisphosphate in the activation of
protein kinase B. Science 277 567570.
(doi:10.1126/science.277.5325.567)
Stralfors P, Bjorgell P & Belfrage P 1984 Hormonal regulation of
hormone- sensitive lipase in intact adipocytes: identification of
phosphorylated
sites and effects on the phosphorylation by lipolytic hormones
and insulin. PNAS 81 33173321. (doi:10.1073/pnas.81.11.3317)
Sukonina V, Lookene A, Olivecrona T & Olivecrona G 2006
Angiopoietin- like protein 4 converts lipoprotein lipase to inactive
monomers and modulates lipase activity in adipose tissue. PNAS
103 1745017455.
(doi:10.1073/pnas.0604026103)
Sun Z, Gong J, Wu H, Xu W, Wu L, Xu D, Gao J, Wu JW, Yang H, Yang M
et al. 2013 Perilipin1 promotes unilocular lipid droplet formation
through the activation of Fsp27 in adipocytes. Nature Communications
4
1594. (doi:10.1038/ncomms2581)
Taggart AK, Kero J, Gan X, Cai TQ, Cheng K, Ippolito M, Ren N, Kaplan
R, Wu K, Wu TJ et al. 2005 (D)-b-hydroxybutyrate inhibits adipocyte
lipolysis via the nicotinic acid receptor PUMA-G. Journal of Biological
Chemistry 280 2664926652. (doi:10.1074/jbc.C500213200)
Tansey JT, Sztalryd C, Gruia-Gray J, Roush DL, Zee JV, Gavrilova O,
Reitman ML, Deng CX, Li C, Kimmel AR et al. 2001 Perilipin
ablation results in a lean mouse with aberrant adipocyte lipolysis,
enhanced leptin production, and resistance to diet-induced obesity.

PNAS 98
64946499. (doi:10.1073/pnas.101042998)
Taschler U, Radner FP, Heier C, Schreiber R, Schweiger M, Schoiswohl G,
Preiss-Landl K, Jaeger D, Reiter B, Koefeler HC et al. 2011
Monoglyceride lipase deficiency in mice impairs lipolysis and
attenuates diet-induced insulin resistance. Journal of Biological
Chemistry 286 1746717477.
(doi:10.1074/jbc.M110.215434)
Tavernier G, Barbe P, Galitzky J, Berlan M, Caput D, Lafontan M & Langin
D 1996 Expression of b3-adrenoceptors with low lipolytic action in
human subcutaneous white adipocytes. Journal of Lipid Research 37
8797.
Tchoukalova YD, Votruba SB, Tchkonia T, Giorgadze N, Kirkland JL &
Jensen MD 2010 Regional differences in cellular mechanisms of
adipose tissue gain with overfeeding. PNAS 107 1822618231.
(doi:10.1073/
pnas.1005259107)
Tinahones FJ, Garrido-Sanchez L, Miranda M, Garcia-Almeida JM,
Macias-Gonzalez M, Ceperuelo V, Gluckmann E, Rivas-Marin J,
Vendrell J & Garcia-Fuentes E 2010 Obesity and insulin resistancerelated changes in the expression of lipogenic and lipolytic genes in
morbidly obese subjects. Obesity Surgery 20 15591567.
(doi:10.1007/s11695-010-0194-z)
Toh SY, Gong J, Du G, Li JZ, Yang S, Ye J, Yao H, Zhang Y, Xue B, Li Q et al.
2008 Up-regulation of mitochondrial activity and acquirement of
brown adipose tissue-like property in the white adipose tissue of
fsp27 deficient mice. PLoS ONE 3 e2890.
(doi:10.1371/journal.pone.0002890)
Tornqvist H & Belfrage P 1976 Purification and some properties of a
monoacylglycerol-hydrolyzing enzyme of rat adipose tissue. Journal of
Biological Chemistry 251 813819.
Tunaru S, Kero J, Schaub A, Wufka C, Blaukat A, Pfeffer K & Offermanns S
2003 PUMA-G and HM74 are receptors for nicotinic acid and
mediate its anti-lipolytic effect. Nature Medicine 9 352355.
(doi:10.1038/nm824)
Turpin SM, Hoy AJ, Brown RD, Rudaz CG, Honeyman J, Matzaris M
& Watt MJ 2011 Adipose triacylglycerol lipase is a major regulator
of hepatic lipid metabolism but not insulin sensitivity in mice.
Diabetologia 54 146156. (doi:10.1007/s00125-010-1895-5)

Review

Jo
ur
n
al
of
M
ol
ec
ul
ar
E
n
d
oc
ri
n

T S NIELSEN

and others

Vaughan M, Berger JE & Steinberg D 1964 Hormone-sensitive lipase and


monoglyceride lipase activities in adipose tissue. Journal of Biological
Chemistry 239 401409.
Vendelbo MH, Jorgensen JO, Pedersen SB, Gormsen LC, Lund S, Schmitz
O,
Jessen N & Moller N 2010 Exercise and fasting activate growth
hormone-dependent myocellular signal transducer and activator of
transcription-5b phosphorylation and insulin-like growth factor-I
messenger ribonucleic acid expression in humans. Journal of
Clinical Endocrinology and Metabolism 95 E64E68.
(doi:10.1210/jc.2010-0689)
Villena JA, Roy S, Sarkadi-Nagy E, Kim KH & Sul HS 2004 Desnutrin, an
adipocyte gene encoding a novel patatin domain-containing protein,
is induced by fasting and glucocorticoids: ectopic expression of
desnutrin increases triglyceride hydrolysis. Journal of Biological
Chemistry 279
4706647075. (doi:10.1074/jbc.M403855200)
Virtue S & Vidal-Puig A 2010 Adipose tissue expandability, lipotoxicity
and the metabolic syndrome an allostatic perspective. Biochimica et
Biophysica Acta 1801 338349. (doi:10.1016/j.bbalip.2009.12.006)
Wang H, Hu L, Dalen K, Dorward H, Marcinkiewicz A, Russell D, Gong
D, Londos C, Yamaguchi T, Holm C et al. 2009 Activation of
hormone- sensitive lipase requires two steps, protein phosphorylation
and
binding to the PAT-1 domain of lipid droplet coat proteins. Journal
of Biological Chemistry 284 3211632125.
(doi:10.1074/jbc.M109.006726)
Watt MJ, Holmes AG, Pinnamaneni SK, Garnham AP, Steinberg GR,
Kemp BE & Febbraio MA 2006 Regulation of HSL serine phosphorylation in skeletal muscle and adipose tissue. American Journal of
Physiology. Endocrinology and Metabolism 290 E500E508.
(doi:10.1152/ ajpendo.00361.2005)
Wei S, Lai K, Patel S, Piantedosi R, Shen H, Colantuoni V, Kraemer FB &
Blaner WS 1997 Retinyl ester hydrolysis and retinol efflux from BFC1b adipocytes. Journal of Biological Chemistry 272 1415914165.
(doi:10.1074/jbc.272.22.14159)
Weyer C, Foley JE, Bogardus C, Tataranni PA & Pratley RE 2000 Enlarged
subcutaneous abdominal adipocyte size, but not obesity itself,
predicts type II diabetes independent of insulin resistance.
Diabetologia 43
14981506. (doi:10.1007/s001250051560)
White MF 1998 The IRS-signalling system: a network of docking proteins
that mediate insulin action. Molecular and Cellular Biochemistry 182
311. (doi:10.1023/A:1006806722619)
White UA & Tchoukalova YD 2014 Sex dimorphism and depot
differences in adipose tissue function. Biochimica et Biophysica Acta
1842 377392. (doi:10.1016/j.bbadis.2013.05.006)
Whitman M, Downes CP, Keeler M, Keller T & Cantley L 1988 Type I
phosphatidylinositol kinase makes a novel inositol phospholipid,
phosphatidylinositol-3-phosphate. Nature 332 644646. (doi:10.1038/
332644a0)
Wijnen JA, van Baak MA, de HC, Boudier HA, Tan FS & Van Bortel
LM 1993 b-Blockade and lipolysis during endurance exercise.
European
Journal
of
Clinical
Pharmacology
45
101105.
(doi:10.1007/BF00315488)
Wilson PA, Gardner SD, Lambie NM, Commans SA & Crowther DJ 2006
Characterization of the human patatin-like phospholipase family.
Journal of Lipid Research 47 19401949. (doi:10.1194/jlr.M600185JLR200) Wu JW, Wang SP, Alvarez F, Casavant S, Gauthier N, Abed L,
Soni KG,
Yang G & Mitchell GA 2011 Deficiency of liver adipose triglyceride
lipase in mice causes progressive hepatic steatosis. Hepatology 54
122132. (doi:10.1002/hep.24338)
Wu JW, Wang SP, Casavant S, Moreau A, Yang GS & Mitchell GA 2012
Fasting energy homeostasis in mice with adipose deficiency of
desnutrin/adipose triglyceride lipase. Endocrinology 153 2198

Dissecting adipose tissue


lipolysis

52 :3

R222

2207. (doi:10.1210/en.2011-1518)
Xu C, He J, Jiang H, Zu L, Zhai W, Pu S & Xu G 2009 Direct effect of
glucocorticoids on lipolysis in adipocytes. Molecular Endocrinology 23
11611170. (doi:10.1210/me.2008-0464)

Yang S, Mulder H, Holm C & Eden S 2004 Effects of growth hormone


on the function of b-adrenoceptor subtypes in rat adipocytes.
Obesity Research 12 330339. (doi:10.1038/oby.2004.41)
Yang X, Lu X, Lombes M, Rha GB, Chi YI, Guerin TM, Smart EJ & Liu J
2010 The G(0)/G(1) switch gene 2 regulates adipose lipolysis
through association with adipose triglyceride lipase. Cell Metabolism
11
194205. (doi:10.1016/j.cmet.2010.02.003)
Yang X, Zhang X, Heckmann BL, Lu X & Liu J 2011 Relative
contribution of adipose triglyceride lipase and hormone-sensitive
lipase to tumor necrosis factor-a (TNF-a)-induced lipolysis in
adipocytes. Journal of Biological Chemistry 286 4047740485.
(doi:10.1074/jbc.M111.257923)
Yang X, Heckmann BL, Zhang X, Smas CM & Liu J 2013 Distinct
mechanisms regulate ATGL-mediated adipocyte lipolysis by lipid
droplet coat proteins. Molecular Endocrinology 27 116126.
(doi:10.1210/me.2012-1178)
Yao-Borengasser A, Varma V, Coker RH, Ranganathan G, Phanavanh B,
Rasouli N & Kern PA 2011 Adipose triglyceride lipase expression in
human adipose tissue and muscle. Role in insulin resistance and
response to training and pioglitazone. Metabolism 60 10121020.
(doi:10.1016/j.metabol.2010.10.005)
Yonezawa T, Kurata R, Kimura M & Inoko H 2011 Which CIDE are you on?
Apoptosis and energy metabolism Molecular BioSystems 7 91100.
(doi:10.1039/c0mb00099j)
Zandbergen F, Mandard S, Escher P, Tan NS, Patsouris D, Jatkoe T,
Rojas-Caro S, Madore S, Wahli W, Tafuri S et al. 2005 The G0/G1
switch gene 2 is a novel PPAR target gene. Biochemical Journal 392
313324. (doi:10.1042/BJ20050636)
Zechner R, Zimmermann R, Eichmann TO, Kohlwein SD, Haemmerle G,
Lass A & Madeo F 2012 FAT SIGNALS lipases and lipolysis in lipid
metabolism and signaling. Cell Metabolism 15 279291. (doi:10.1016/
j.cmet.2011.12.018)
Zhang HH, Halbleib M, Ahmad F, Manganiello VC & Greenberg AS
2002 Tumor necrosis factor-a stimulates lipolysis in differentiated

human adipocytes through activation of extracellular signal-related


kinase and elevation of intracellular cAMP. Diabetes 51 29292935.
(doi:10.2337/ diabetes.51.10.2929)
Zhang X, Xie X, Heckmann BL, Saarinen AM, Czyzyk TA & Liu J 2013
Target disruption of G0/G1 switch gene 2 enhances adipose
lipolysis, alters hepatic energy balance, and alleviates high fat dietinduced liver steatosis. Diabetes 63 934946. (doi:10.2337/db131422)
Zhou Z, Yon TS, Chen Z, Guo K, Ng CP, Ponniah S, Lin SC, Hong W & Li
P 2003 Cidea-deficient mice have lean phenotype and are resistant to
obesity. Nature Genetics 35 4956. (doi:10.1038/ng1225)
Zhou L, Xu L, Ye J, Li D, Wang W, Li X, Wu L, Wang H, Guan F & Li P
2012 Cidea promotes hepatic steatosis by sensing dietary fatty acids.
Hepatology 56 95107. (doi:10.1002/hep.25611)
Zierler KA, Jaeger D, Pollak NM, Eder S, Rechberger GN, Radner FP,
Woelkart G, Kolb D, Schmidt A, Kumari M et al. 2013 Functional
cardiac lipolysis in mice critically depends on comparative gene
identification58. Journal of Biological Chemistry 288 98929904. (doi:10.1074/jbc.
M112.420620)
Zimmermann R, Haemmerle G, Wagner EM, Strauss JG, Kratky D &
Zechner R 2003 Decreased fatty acid esterification compensates for the
reduced lipolytic activity in hormone-sensitive lipase-deficient white
adipose tissue. Journal of Lipid Research 44 20892099. (doi:10.1194/jlr.
M300190-JLR200)
Zimmermann R, Strauss JG, Haemmerle G, Schoiswohl G, BirnerGruenberger R, Riederer M, Lass A, Neuberger G, Eisenhaber F,
Hermetter A et al. 2004 Fat mobilization in adipose tissue is promoted
by adipose triglyceride lipase. Science 306 13831386. (doi:10.1126/
science.1100747)

Received in final form 14 February 2014


Accepted 25 February 2014
Accepted Preprint published online 27 February 2014

Вам также может понравиться