Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

c h a p t e r

36
Role of the Adipocyte in
Metabolism and Endocrine
Function
Eric Ravussin and Steven R. Smith

CHAPTER OUTLINE
OBESITY, INSULIN RESISTANCE, INFLAMMATION, Integrative Biology of the Adipose Tissue, 639
ER STRESS, AND TYPE 2 DIABETES MELLITUS, 628 Endocrine Signals, 639
Link Between Obesity and Insulin Resistance, 628 Neural Signals to the Adipose Tissue, 640
Links Between Obesity, Inflammation, and ER Regulation of Lipolysis, 640
Stress, 631 Adipocyte as an Endocrine Organ, 641
Link Between Too Little Fat and Insulin The Adipocyte as a Target for the Treatment of
Resistance, 632 Obesity and Type 2 Diabetes, 646
Brown Adipose Tissue in Humans, 637

KEY POINTS
• A dipose tissue development and dysfunction plays a central role in several diseases
including type 2 diabetes, cardiovascular disease, and metabolic health.
• Once considered to be simply a passive fat storage organ, adipose tissue is a complex
endocrine organ and plays a central role in energy homeostasis and metabolic health.
• When the energy input exceeds the capacity for storage, adipose tissue becomes
dysfunctional and a liability for metabolic health.
• Adipose tissue dysfunction includes changes in hormone secretion, inflammation, and
alterations in lipid and glucose metabolism.
  

In an evolutionary sense, the adipose tissue represents the even in lean individuals, fat stores in the adipose tissue
most efficient way to store energy in periods of feast to alone can amount to approximately 10 kg, which is suf-
allow survival during periods of famine. There are two ficient to maintain bodily functions for weeks of survival
main reasons for this efficiency. First, on a weight basis, during total food deprivation. In obesity, fat stores can
triacylglycerol, commonly called triglycerides, yields be multiplied by as much as 10 to 20 times and provide
more than two times the amount of energy as glycogen energy for months of starvation. The energy content of
or proteins. Second, triglycerides are stored without asso- and the deficit required to lose 1 pound of body weight
ciated water whereas glycogen, which is hydrophilic, was originally determined to be 3500 kcal; however, more
binds as much as twice its weight to water. Similarly, recent calculations show that this is generally true but
proteins, the building blocks of cells, are associated with varies depending on the actual initial body weight, sex,
large amounts of water. As a result, the energy that can and body composition.1 The energy from triglycerides
be recovered from triglyceride stores by unit of weight is is stored in some 25 to 50 billion fat cells representing
more than four times larger than that from glycogen and/ the adipose tissue, mostly beneath the skin (subcutane-
or protein stores. An average 75-kg man can store only ous adipose tissue) and in the abdomen. Until only 20
approximately 500 g of carbohydrate in liver and muscle, years ago, the adipose tissue was primarily considered as
representing less than 1 day of energy stores. By contrast, an energy storage compartment providing energy fuel to
627
628 PART 5 DIABETES MELLITUS

the entire body in-between meals and during periods of tissue. We will also provide new information regarding
energy deficit. the potential importance of brown adipose tissue (BAT)
With the discovery of leptin in 19942 and the many in humans, a concept that was discarded over the past 2
other secreted proteins discovered since,3 adipose tissue decades because of the belief that such tissue was lack-
is now not only considered as an energy reservoir but ing in adult humans. In the next section, we will discuss
has reached the status of a true endocrine organ. Over the lipostatic theory, which led to the discovery of leptin
the past decade, effort has been concentrated on a better and many other hormones involved in health and disease.
understanding of the regulation of adipose tissue devel- Beside leptin, we will provide additional details on adipo-
opment and apoptosis/turnover throughout the life span nectin, resistin, TNF-α, apelin, and adipose. We conclude
and its consequence on health and diseases. In this chap- by proposing the adipocyte as a potential target for the
ter, we will review the role of the adipose as an energy treatment of obesity, dyslipidemia, and type 2 diabetes,
storage compartment, but more importantly we will sum- with emphasis on inducing BAT.
marize the current knowledge of what is now considered
as a finely tuned endocrine organ, which can influence
many facets of the conditions often referred as metabolic OBESITY, INSULIN RESISTANCE, INFLAMMATION, ER
syndrome. We will first describe the links between too STRESS, AND TYPE 2 DIABETES MELLITUS
much fat (obesity), inflammation, endoplasmic reticu-
lum (ER) stress, and insulin resistance and, alternatively, Link Between Obesity and Insulin Resistance
provide arguments why too little fat is equally and para- Numerous cross-sectional studies have shown an associa-
doxically associated with insulin resistance. Next, we tion between obesity and type 2 diabetes (Figs. 36-1 and
will review the pioneering work that led to the concept 36-2). Data from the third National Health Examination
of hypertrophic and hyperplastic obesity and the criti- Survey (NHANES III) provides unequivocal evidence that
cal periods during which the adipose tissue is thought to the prevalence of diabetes is almost three times higher
develop. The current understanding of the regulation of in overweight individuals than in nonoverweight indi-
adipogenesis is then reviewed, followed by a description viduals.4 Many prospective studies have confirmed this
of the afferent endocrine and neural signals to the adipose association. As an example, the likelihood of developing

CHRONIC ENERGY EXCESS

Obesity

Failure of Fetal Failure of


vascularization programming differentiation

↓ Adiponectin Adipocyte ER stress Inflammatory


↑ Leptin Hypertrophy cytokines

Decreased capillary
density and/or Chemokine
vasoconstriction secretion

Disordered lipid Macrophage


Hypoxia recruitment
metabolism Mitochondrial Chemokine
and activation
dysfunction secretion

Apoptosis

Figure 36-1 Obesity and adipocyte hypertrophy. Adipocyte hypertrophy is a key feature of the insulin-resistant state. The flux of free fatty acids
out of adipose tissue is disordered in obesity, especially when adipocyte hypertrophy is present. In addition, hypoxia, inflammation, and endoplasmic
reticulum (ER) stress have been implicated as important mechanistic links between obesity and insulin resistance. This figure attempts to reconcile the
available data on how obesity might lead to insulin resistance. In many instances, the directionality of the connections between mechanistic factors is
unclear. For example, it is clear that ER stress can lead to the secretion of inflammatory cytokines, but it is not entirely clear if inflammation and mac-
rophages might lead to ER stress. Similarly, reduced capillary density (rarefaction) leads to hypoxia, and hypoxia leads to the secretion of chemokines
and inflammatory cytokines and probably disordered adipokine secretion. Hypertrophic adipocytes secrete the inflammatory peptide SAA, which
may serve to amplify inflammation and chemotaxis of macrophages. At this time, it is difficult to disentangle the web of deleterious connections. It
appears that once the cascade is initiated, there are multiple interlocking pathways that may sustain the dysfunctional adipose tissue. (From Yang RZ,
Lee MJ, Hu H, et al. Acute-phase serum amyloid A: an inflammatory adipokine and potential link between obesity and its metabolic complications.
PLoS Med 3[6]:e287, 2006; and Jernas M, Palming J, Sjoholm K, et al. Separation of human adipocytes by size: hypertrophic fat cells display distinct
gene expression. FASEB J 20[9]:1540-1542, 2006.)
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 629

A B

C D

n
F
E
Figure 36-2 Angiogenesis and innervations in adipose tissue. Adipose tissue has the capacity to grow new blood vessels. A, Adipose tissue fragments
show outgrowth of new capillaries after 10 days of culture ex-vivo. B, In human adipose tissue, capillaries form a rich meshwork around adipocytes.
Adipocytes are stained green (BODIPY), and capillaries are stained red (Lectin UEA). In obesity, capillary density is decreased in subcutaneous adi-
pose tissue. Obese adipose tissue (C) and lean adipose tissue (D) have different capillary density. Adipocyte plasmalemma is stained green with Lectin-
GS, and capillaries are stained red with Lectin UEA. Human adipose tissue is innervated with sympathetic nervous system (SNS) nerve terminals that
are important for the activation of lipolysis. E, Fluorescent immunohistochemistry of nerve fibers (neural-specific antibody) in white adipose tissue.
F, SNS-specific staining with tyrosine hydroxylase. (A, Photomicrograph courtesy of Dr. Frank Greenway, Pennington Biomedical Research Center;
methods as in Greenway FL, Liu Z, Yu Y, et al. An assay to measure angiogenesis in human fat tissue. Obes Surg 17[4]:510-515, 2007. B, Photomi-
crograph courtesy of Drs. Magdalena Pasarica, David Burk, and Steven Smith, Pennington Biomedical Research Center. C, Methods are presented
in Trayhurn P, Wang B, Wood IS. Hypoxia and the endocrine and signalling role of white adipose tissue. Arch Physiol Biochem 114[4]:267-276,
2008. E and F, Reprinted with permission from Giordano A, Song CK, Bowers RR, et al. White adipose tissue lacks significant vagal innervation and
immunohistochemical evidence of parasympathetic innervation. Am J Physiol Regul Integr Comp Physiol 291[5]:R1243-R1255, 2006.)
630 PART 5 DIABETES MELLITUS

diabetes increases steeply with increasing body weight fasting reliance of skeletal muscle on fatty acids, there-
and fatness in Pima Indians.5 The reason for the asso- fore causing an accumulation of lipids into the muscle
ciation between obesity and diabetes is attributed mostly tissue. This “reversed Randle cycle” theory highlights the
to the increase in insulin resistance so common in obese primary role of impaired lipid oxidation in skeletal mus-
people.6 Insulin resistance is a clear predisposing factor cle rather than excessive lipolysis in the adipose tissue.
for the development of type 2 diabetes in individuals at Growing evidence suggests that the primary cause of the
risk for the disease.7-9 “metabolic inflexibility” in subjects susceptible to insulin
Obesity is usually characterized by increased circulat- resistance is impaired fat oxidation in the fasting state
ing plasma free-fatty acid (FFA) concentration. Studies in rather than a lack of increased carbohydrate oxidation
animals and humans support a causal effect of elevated in response to feeding.21 In support of this concept, there
FFA (produced by lipolysis of triglycerides from adipose is now evidence pointing toward decreased mitochon-
tissue) on impaired insulin-mediated glucose metabo- drial oxidative capacity in insulin-resistant subjects with
lism.10-13 Such studies reinforce the original hypothesis diabetes22 or family history of diabetes23 and in insulin-
proposed by Randle and colleagues in 196314,15 and later resistant older adults.24
by McGarry16 that altered fatty acid metabolism was the To prove or disprove the “Randle cycle” hypothesis,
key contributing factor to insulin resistance in obese and Shulman and co-workers went one step further. If the
diabetic patients. Randle and colleagues demonstrated Randle hypothesis was true, one would predict an accu-
that FFAs compete with glucose for substrate oxidation mulation of glucose-6-phosphate in the skeletal muscle
in isolated preparations of heart and diaphragm muscle of healthy subjects during glucose and insulin infusions
from rats. More specifically, they proposed that increased in the presence of high plasma FFA concentration.25
fatty acid availability in obesity causes an increase in the They directly tested this hypothesis using magnetic reso-
intramitochondrial acetyl-CoA/CoA and NADH/NAD+ nance spectroscopy. As expected, high concentrations of
ratios, leading to inactivation of the enzyme pyruvate circulating FFA caused a reduction in insulin-mediated
dehydrogenase. This, in turn, causes an intracellular glucose uptake with an approximately 50% decrease
increase in citrate concentration, leading to inhibition of in glucose storage and 50% decrease in glucose oxida-
phosphofructokinase, the rate-limiting enzyme for glycol- tion.12 However, in contrast to what would be predicted
ysis. As a consequence, glucose-6-phosphate accumulates by the Randle’s hypothesis, there was no accumulation of
in the cell and inhibits hexokinase II activity, leading to glucose-6-phosphate. Therefore, reduced glucose uptake
an increase in intracellular glucose and a decrease in glu- when FFAs are high is due to impaired glucose transport
cose uptake, oxidation, and storage. or impaired intracellular signaling. This series of stud-
Randle’s glucose–fatty acid cycle has been challenged ies shows that intramuscular fatty acids or fatty acid
by data from sophisticated clinical studies using stable metabolites seem to interfere with the transport of glu-
isotope turnover in conjunction with substrate leg bal- cose into the skeletal muscle cells. Studies have provided
ance methods or using 13C and 31P magnetic resonance some potential mechanisms of the effect of FFA-induced
spectroscopy. These studies provide evidence that insulin insulin resistance via an impact on insulin signaling at
resistance in obese individuals may be related to a pri- the level of protein kinase C θ.26-29 More recently how-
mary defect of fatty acid oxidation in skeletal muscle ever, Koves and colleagues, using targeted metabolomics,
rather than a competition between fat and carbohydrate found that obesity-related insulin resistance in skeletal
metabolism. muscle is characterized by excessive beta-oxidation,
First, Wolfe and associates identified some of the flaws impaired switching to carbohydrate during the fasted-
in the notion that fatty acid availability controls substrate to-fed transition, and simultaneous depletion of organic
oxidation in the fasting state.17 He provided evidence intermediates of the tricarboxylic acid cycle.30 There-
that fatty acid oxidation is largely controlled at the site of fore, an excessive rather than reduced beta-oxidation
oxidation, which is in turn determined by the availability may underlie the development of muscle insulin resis-
of glucose. In this model, the primary physiologic role tance by accumulation of acyl-CoAs and their respective
of increased adipose lipolysis in fasting conditions is to acyl-carnitines.30
provide the necessary glycerol as a gluconeogenic precur- While many studies have provided evidence for an
sor,18 and the rate of fatty acid oxidation is then regu- association between insulin sensitivity and visceral fat
lated by the rate of intracellular metabolism of glucose. mass (reviewed in reference 31), other studies provide evi-
Second, it is now well accepted that skeletal muscle pre- dence just as good for associations between the amount
dominantly relies on lipid oxidation during fasting and can of subcutaneous fat on the trunk and insulin resistance in
easily switch from lipid to increased glucose uptake and obese nondiabetic men32,33 and in men with type 2 dia-
oxidation in response to feeding and hyperinsulinemia.19 betes.20,34,35 Similarly, insulin resistance in obese women
This switch from fat to carbohydrate oxidation in skeletal is best related to overall elevated fat mass than just vis-
muscle has been called metabolic flexibility. Importantly, ceral fat mass.36,37 Thus, subcutaneous fat, which does
Kelley and Mandarino provided convincing evidence that not drain into the portal vein, causes insulin resistance
glucose oxidation after an overnight fast is increased in by a nonportal mechanism. The growing experimen-
the leg of subjects with type 2 diabetes, thereby decreasing tal evidence, which does not support the Randle/portal
its reliance on fat oxidation.20 In opposition to Randle’s hypothesis, therefore calls for a change in the scientific
glucose-fatty acid cycle, their series of studies suggest that paradigm to explain the insulin resistance so common in
hyperglycemia itself causes an impairment of the normal obesity. The bulk of the literature now provides evidence
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 631

that excessive total fat mass (rather than just visceral fat Two of the major unsolved questions regarding this
mass) and impaired muscle fat oxidation are associated observation are: Why does inflammation occur in parallel
with insulin resistance and increased risk for the develop- with obesity, and what is the consequence of this inflam-
ment of type 2 diabetes. mation? One of the growing hypotheses first proposed
In addition to the key role of total fat mass to the by Trayhurn’s group59 and more recently reviewed60,61 is
metabolic disturbances described earlier, fat distribution that inflammation is likely the result of reduced oxygen-
may play an important role in several aspects of meta- ation (hypoxia; see Figs. 36-1, 36-2, and 36-3) in adipose
bolic function. For example, some of our own studies tissue, which may provide cellular mechanisms for mac-
have clearly shown that the gluteal adipose tissue may be rophage infiltration, reduction in adiponectin secretion,
protective against insulin resistance rather than precipi- increased leptin secretion, adipocyte death, endoplasmic
tating it.38 In contrast, visceral omental adipose tissue reticulum (ER) stress, fibrosis, and mitochondrial dys-
exhibits more inflammation than subcutaneous adipose function62 in white adipose tissue (WAT) in obesity.63-65
tissue and intramuscular adipose tissue39-42 (the “mar- Inhibition of adipogenesis and triglyceride synthesis by
bled” adipose tissue in between the muscles) and both hypoxia may be a mechanism to elevate plasma FFA
are associated with a greater cardiovascular and diabe- and reduce adiponectin concentrations, both leading to
tes risks. The mechanisms underlying these regional dif- insulin resistance in obesity.63,64 The biological basis for
ferences in adipose tissue function and disease risk are adipose tissue hypoxia could be related to a reduction in
only now being elucidated and will be discussed later in arterial blood flow as a result of adipocytes outgrowing
the chapter. their blood supply (Fig. 36-3). A failure to maintain blood
flow or reduced capillary density (a lack of compensa-
Links Between Obesity, Inflammation, and ER Stress tory angiogenesis or vasoconstriction) may be the basis of
Studies in mice and humans suggested that adipose tis- the observed reduction in blood flow in human adipose
sue contains not only adipocytes and supporting cells, tissue.60 The hypothesis that increased adipocyte mass
but also macrophages (see Figs. 36-1 and 36-2).43,44 The
macrophage is responsible for consuming extracellular
bacteria, cellular debris, and lipids, whereas the adi-
pocyte is responsible for internalizing and sequestering Fat
excess lipids. The macrophages appear to be the major
site of TNF-α secretion and may also secrete other cyto-
kines such as IL-8,45,46 an atherogenic cytokine pro-
duced in adipose tissue. These studies also suggested Gain of fat mass
that the bone marrow was the major site of origin for (proliferation, differentiation, lipid storage)
the adipose tissue macrophages, suggesting that obesity
and diabetes might “recruit” these cells through the pro-
duction of one or more chemokines.47 The significance
of the inflammatory cells is that they are likely to acti- Decreased Adipose tissue
vate the NF-κb signaling cascade, like TNF-α. Iκκ-β, the capillary density vasoconstriction
upstream activator of NF-κb signaling, plays a key role
in insulin signaling and is necessary for the full expres-
sion of the insulin-resistant phenotype in the obese ob/
ob mouse.48 Upstream αα activators of this pathway Decreased adipose Insulin
tissue blood flow resistance
include not only TNF-α, but also fatty acids 49 and
bacterial lipopolysaccharide. Fatty acids activate this
pathway via the Toll-receptor 4, which also responds
Adipose tissue hypoxia Inflammation
to lipopolysaccharide.49 In vivo studies in mice48 and
humans50-52 demonstrate that salicylates, inhibitors of
the Iκκ-β pathway,53 play an important therapeutic role
Angiogenesis and/or
in insulin resistance and diabetes,54 in part through adi- vasodilation
pocyte-mediated pathways.55,56 The antidiabetic peroxi-
some proliferator-activated receptor γ (PPAR-γ) ligands
(e.g., TZDs) also decrease the gene transcriptional Adipose tissue remodeling
effects of the Iκκ-β pathway.57
Many more inflammatory cytokines such as monocyte Figure 36-3 Hypoxia, inflammation, and insulin resistance. Rap-
id growth of adipose tissue leads to an expansion that may not be
chemoattractant protein-1 (MCP-1) and plasminogen ­accompanied by a simultaneous maintenance of capillary density by in-
activator protein (PAI) have been linked to adiposity. The creased angiogenesis or vasodilation and therefore may cause a decrease
increase in inflammatory cytokines found in obesity ulti- in blood flow to the tissue. Decreased blood flow per unit of adipose tis-
mately characterizes excess adipose tissue as a state of sue leads to tissue hypoxia, which, in turn, induces inflammation. When
low-grade systemic inflammation, which may link obesity inflammation becomes out of control with increased systemic and local
cytokine concentrations, insulin resistance occurs. Adipose hypoxia is
to its comorbidities.58 Obesity therefore occurs in asso- therefore a signal for the remodeling of the adipose tissue. (Adapted
ciation with an increase in inflammatory cytokines and from Ye J. Emerging role of adipose tissue hypoxia in obesity and insu-
an infiltration of macrophages within the adipose tissue. lin resistance. Int J Obes [Lond] 33[1]:54-66, 2008.)
632 PART 5 DIABETES MELLITUS

in obesity without an adequate support of vasculariza- Link Between Too Little Fat and Insulin Resistance
tion might lead to hypoxia, macrophage infiltration, and At the other end of the spectrum from obesity, it is now
inflammation was confirmed in human studies66 (nicely recognized that a lack of adipose tissue is also associ-
reviewed in reference 67). ated with insulin resistance and increased risk for devel-
Over the past few years, publications have provided opment of type 2 diabetes. Lipodystrophy in humans is
growing evidence suggesting that ER stress may also play an acquired or hereditary syndrome characterized by
a role in the pathogenesis of type 2 diabetes.68 The ER is decreased adipose tissue mass, insulin resistance, and
a highly dynamic organelle with a central role in lipid and often diabetes mellitus.80-83 In these patients, insufficient
protein biosynthesis. It produces the transmembrane pro- adipose tissue mass leads to excess energy storage as tri-
teins and lipids for most cell organelles and is responsible glycerides in liver and skeletal muscle and causes insu-
for the synthesis of almost all secreted proteins. After lin resistance in these tissues.84,85 Genetic manipulation
the translation of proteins is performed by ribosomes on causing ablation of adipose tissue in mice supports the
the cytosolic surface of the ER, the unfolded polypeptide link between adipose deficiency and insulin resistance.
chains are translocated into the ER lumen where numer- Transgenic animals without adipose tissue store lipid in
ous chaperone proteins are crucial for the proper folding skeletal muscle and liver and develop insulin resistance,
of proteins and protein complexes.69 glucose intolerance, and eventually diabetes.86-88 This is
However, the ER is exquisitively sensitive to alterations identical to the fatty liver and muscle seen in obesity and
in homeostasis, and proteins formed in the ER may fail to type 2 diabetes. Furthermore, transplantation of adipose
attain proper conformation due to: (1) lack of chaperones tissue back into lipoatrophic animals reverses the elevated
or energy to promote chaperone-protein interactions, (2) glucose levels.89 However, transplantation of adipose tis-
calcium depletion, (3) disruption of the redox state, (4) sue from leptin-deficient mice (ob/ob) did not improve
protein mutations that hamper adequate folding, and (5) the metabolic abnormalities, indicating that the seques-
reduction of disulfide bonds.68 ER stress is also called the tration of triglyceride into adipose tissue is not entirely
unfolded protein response (UPR). Lipotoxicity and glu- sufficient to restore insulin sensitivity.90 In humans, treat-
cotoxicity as seen in prediabetes and diabetes are triggers ment of lipodystrophic patients with leptin can dramati-
of ER stress. cally reverse the fatty liver and insulin resistance,91-93 and
Excess FFAs as seen in obesity activate the ER stress recombinant leptin is now FDA approved for lipodystro-
response in beta cells, eventually causing apoptosis.70 phy.94 On the other hand, surgical removal of adipose
Data suggests that hypoxia in adipose tissue of obese tissue causes metabolic syndrome.95 Together these stud-
mice and individuals may contribute to the induction of ies demonstrate that, like in obesity, inadequate adipose
ER stress and thereby affect adipokine production. For tissue mass leads to “ectopic” fat storage and metabolic
example, adiponectin expression was decreased in adi- disturbances. Too little fat is therefore as deleterious
pose tissue from high-fat–fed KKAy mice.71 Evidence for as too much fat and predisposes to the development of
the role of ER stress in human tissues such as pancreas, the metabolic syndrome with insulin resistance and ulti-
muscle, and adipose is not currently available. Similarly, mately type 2 diabetes. Three new paradigms may there-
increase in expression and phosphorylation of stress- fore explain insulin resistance as follows:
activated kinases such as p38 and JNK were detected   
in omental adipose tissue from obese women compared 1. Inadequate lipid storage leading to “ectopic fat
with lean controls.72 In the next few years, research in storage syndrome” in which excess fat is deposited
this area should lead to a better understanding of the in tissues other than adipose tissue with functional
interaction between obesity, inflammation, hypoxia, and disturbances in these tissues
ER stress. 2. “Endocrine adipocyte” that secretes increased
In addition to disordered FFA release, inflammation, amounts of hormones involved in insulin resistance
ER stress, and hypoxia, fibrosis may play a pathophysi- and cardiovascular disease or secretes decreased
ologic role in obesity.73 Initial studies in mice and later amounts of beneficial hormones such as adiponectin
in humans10,74-76 demonstrated upregulation of genes 3. Inflammatory, fibrotic adipose tissue with adipose
encoding several collagens including collagen VI. Col- tissue macrophage infiltration and macrophage acti-
lagen VI is particularly interesting, as the knockout vation, leading to dysregulation of adipocyte lipid
of COLVIα3 combined with the absence of the leptin metabolism and disordered adipokine secretion
gene ob led to massive obesity and remarkable adipo-   
cyte hypertrophy. In contrast to the degree of obesity
and hypertrophy, these animals were insulin sensitive Obesity is Another Ectopic Fat Storage Syndrome
and metabolically normal. The hypertrophy suggested Positive energy balance in our “obesigenic” environment
that COLVIα3 acts as a “brake” on adipocyte growth. produces a pattern similar to lipodystrophy in humans,
Further study of the COLVIα3 protein revealed a novel that is, excess lipid storage in liver96 and skeletal mus-
peptide fragment that they named endotrophin.77-78 cle25,97,98 followed by insulin resistance, glucose intoler-
Importantly, neutralizing antibodies directed to this ance, and diabetes. However, in contrast to lipodystrophic
small peptide endotrophin normalized insulin action in patients, adipose tissue stores are adequate or even large
obese mice.79 This suggests that endotrophin is a key in obese patients, suggesting that the size of adipose tis-
autocrine, paracrine, or endocrine regulator of insulin sue becomes inadequate to sequester dietary lipid away
resistance in obesity. from liver, skeletal muscle, and pancreas. The adipocyte
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 633

becomes hypertrophic and unable to recruit and/or differ-


entiate new adipocytes to store the excessive dietary fat.99
This hypothesis is supported by the fact that, indepen-
dent of total fat mass, individuals with larger abdominal
fat cells are at higher risk of developing type 2 diabetes
than individuals with smaller fat cells.100 Furthermore,
thiazolidinediones (TZDs) improve insulin resistance par-
tially by promoting differentiation of new fat cells in sub-
cutaneous adipose tissue through activation of PPAR-γ,
therefore providing extra storage capacity for dietary
fat.101,102 When driven by PPAR-γ agonists, adipogenesis
translates into a gain in subcutaneous adipose tissue103
and a decrease in lipid infiltration in skeletal muscle and
liver.104,105 Through the upregulation of genes in the lipid
storage and synthesis pathways in adipose tissue, TZDs
also decrease FFA, providing a second mechanism to pro-
tect liver, muscle, and the beta cell from fatty acids.106,108
As discussed later, drugs may therefore be designed to A
decrease ectopic fat storage by increasing adipogenesis
and/or increasing fat oxidation leading to improved insu-
lin action. Weight gain with PPAR-γ agonists in humans
is probably due to an increase in food intake consistent
with the action of the PPAR-γ agonists to increase food
intake.109-111
Adipose Tissue: Hypertrophy vs. Hyperplasia
Historically, adipose tissue was viewed as an inert tissue
with a singular function: lipid storage. The main areas of
research in the field of adipose tissue were related to adi-
pocyte size and number as well as lipid synthesis, adrener-
gic regulation of lipolysis, and insulin signaling in isolated
adipocytes. In adults, obesity is associated with an
increase in both the number and size of adipocytes112,113
(Fig. 36-4). The increase in fat cell size (hypertrophy) is
thought to reflect an imbalance between adipocyte lipid
uptake or synthesis and the release of lipid via lipolytic B
pathways. In addition to increased adipocyte size, obese
individuals have an increase in the absolute number of Figure 36-4 Hyperplastic versus hypertrophic obesity. Adipocytes are
derived from mesenchymal stem cell precursors (see Figure 36-7). Both
adipocytes (hyperplasia). Early studies demonstrated het- the mesenchymal precursors and the preadipocyte, whose differentia-
erogeneity in fat cell size; some obese patients have adipo- tion potential is limited to the adipocyte lineage, can undergo mitosis.
cytes as large as 1 μl, and others have very small fat cells. Hyperplastic obesity (A) is defined by an increase in the number of adi-
This heterogeneity led to the concept of hypertrophic or pocytes, while adipocyte size remains small. Hypertrophic obesity (B) is
characterized by larger adipocytes. The reason for the increase in adi-
hyperplastic obesity based on the average size of fat cells pocyte size is not clear. Current hypotheses suggest that a failure of the
(see Fig. 36-4). In contrast to this dichotomous viewpoint, large adipocyte to recruit preadipocytes to differentiate may play a role
the reality is that obese individuals cannot be grouped into in the development of insulin resistance, a precursor to overt beta cell
such simple categories. There is a continuous distribution failure as manifested by diabetes. (Photomicrographs courtesy of Prof.
Saverio Cinti, MD, Institute of Anatomy, Faculty of Medicine, Univer-
of fat cell size, and most obese patients have both hyper- sity of Ancona, Italy.)
trophy and hyperplasia. Increased fat cell size is positively
correlated with fasting insulin and negatively with insulin
sensitivity.114 In Pima Indians, increased abdominal adi- mass is the result of increased lipid storage in adipocytes.
pocyte size is associated with insulin resistance,100,115 a A second view is that fat cells can continually be recruited
potential inherited trait116 that predicts the onset of type to differentiate into mature lipid-storing fat cells, and
2 diabetes.100 Together, these data suggest that increased large fat cells are an indication of a failure of this process.
fat cell size is important to whole-body metabolism and Several investigators have proposed that once adipocytes
insulin action. are filled to a certain degree, new fat cells are “recruited,”
There are several ways of thinking about why fat cells and lipid is then stored in these new insulin-sensitive and
might be large in obese individuals. First, adult adipose tis- lipid-hungry adipocytes.117 The cross-sectional data used
sue has been viewed as a non-mitotic tissue, and increases to support this model is presented in Figure 36-5. Aver-
in adipocyte size might simply reflect an imbalance age fat cell size increases as body fatness increases up to
between storage and lipolysis. If the number of adipo- a certain point, after which increased adiposity does not
cytes is considered “fixed,” any increase in adipose tissue result in an increase in fat cell size. Even if individuals
634 PART 5 DIABETES MELLITUS

with hyperplastic or hypertrophic obesity lose weight longitudinal data nor sufficiently precise measures of fat
equally, hyperplastic obese patients regain the weight cell number to confirm this model in humans. This is in
much more quickly than hypertrophic subjects, lending part due to our current inability to accurately quantify the
support for the concept of small lipid-hungry fat cells118 number of stem cells and preadipocytes in adipose tissue
(Figure 36-6). During weight loss, fat cell size decreases in vitro or in vivo, as well as the difficulties in quantify-
without a change in fat cell number but with a decrease ing the very smallest fat cells in adipose tissue.121 Studies
in fasting insulin.114 This has been interpreted as evidence using nuclear bomb–released 14C suggest that approxi-
that once a fat cell is formed, it is permanent. However, mately 10% of fat cells are renewed annually.122
studies have demonstrated a relatively high rate of adi- In contrast, the cross-sectional data illustrated in Fig-
pocyte turnover119 and evidence for regulation of apop- ure 36-6 could also be interpreted as evidence for the
tosis.120 It should be noted that there is neither sufficient recruitment of new adipocytes. If fat cell hypertrophy

GROWTH AND DEVELOPMENT OF ADIPOSE TISSUE IN CHILDHOOD


Fat cell number

Fat cell size

1m 6m 12m 24m 48m 8y adult 1m 6m 12m 24m 48m 8y adult


A Age B Age

Figure 36-5 Schematic relationship between age, body fat mass, and fat cell size/number in childhood. A, Whole-body fat cell number remains
constant during the first year of life and then increases over time. There is a wide range of fat cell sizes and numbers across individuals as indicated
by the dashed lines. B, The increase in body fat that is seen during the first year of life occurs primarily as a result of increased lipid storage and
hypertrophy of existing adipocytes rather than through the recruitment of preadipocytes. (Data from Hager A, Sjostrom L, Arvidsson B, et al. Body
fat and adipose tissue cellularity in infants: a longitudinal study. Metabolism 26[6]:607-614, 1977; and Soriguer Escofet FJ, Esteva de Antonio I,
Tinahones FJ, Pareja A. Adipose tissue fatty acids and size and number of fat cells from birth to 9 years of age—a cross-sectional study in 96 boys.
Metabolism 45[11]:1395-1401, 1996.)

ADIPOSE TISSUE CELLULARITY AND CELL SIZE IN ADULTHOOD


Fat cell number

Fat cell size

A BMI or fat mass B BMI or fat mass


Figure 36-6 Schematic relationship between body fat mass and fat cell size/number in adulthood. A, In adults, whole-body fat cell number increases
with increasing body mass (hyperplasia). B, Again in adults, cross-sectional data shows a positive correlation between body fat mass and fat cell size
at lower body fat mass (solid line) until fat cell size reaches a plateau at higher levels of fat mass. At this point, fat cell size cannot increase any further
due to (a) limitations on lipid storage for unknown reasons, and/or (b) recruitment of existing preadipocytes to differentiate and store lipid, and/or
c) proliferation and differentiation of mesenchymal precursor cells into mature lipid-storing adipocytes. If fat cell proliferation, differentiation, and/
or recruitment did not occur, fat cell size would continue to increase as fat mass increased. There is a wide range of fat cell sizes and numbers across
individuals as indicated by the dashed lines. Fat cell number does not appear to change with weight gain or loss; however, precise tools are not avail-
able to quantify small changes in adipocyte number in vivo in humans. (Adapted from Hirsch J, Batchelor B. Adipose tissue cellularity in human
obesity. Clin Endocrinol Metab 5[2]:299-311, 1976.)
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 635

was the only way to gain fat, then fat cell size would 4 billion observable fat cells, and this increases to approx-
increase linearly with fat mass. This is not the case. Based imately 10 to 40 billion in lean individuals and up to 50
on cross-sectional data, the point at which hypertrophy to 100 billion in obese patients,134 supporting the concept
recruits new fat cells probably occurs when average cell of ongoing adipocyte proliferation and/or recruitment
volume reaches 0.8 to 1.0 μl.123-125 Two additional pieces throughout life. In contrast to rodents, humans have a
of evidence support the view that recruitment of new long, slow growth and development period (neotony)
fat cells occurs in vivo in humans. First, when adipose and are likely to have several critical periods of adipocyte
tissue is separated into fat cells and the remaining cell development.135
populations (stromal-vascular fraction), adipocytic pre- Body fat mass increases during the first year of life
cursor cells from the stromal-vascular fraction are able mostly through fat cell hypertrophy. After the first year,
to differentiate in vitro into mature lipid-storing adipo- the number of adipocytes increases, the fat cell size
cytes throughout life and into old age. Obesity and age remains relatively constant, and whole-body adiposity
are determinants of the capacity to differentiate adipo- (% fat) decreases. At about 6 years of age, % body fat
cytes in vitro.126,127 Studies of in vivo DNA synthesis in begins to increase again. This has been termed the adi-
humans suggest that adipocyte turnover is high in adult posity rebound. Longitudinal body weight data in chil-
humans, with a t½ ranging from 240 to 425 days.119,128 dren demonstrates that an earlier adiposity rebound is
Other work suggests that the large fat cells secrete a factor associated with obesity in adulthood.136-138 Although no
or factor(s) that promote adipocyte proliferation and dif- detailed information exists on the relative role of hyper-
ferentiation, a finding that is consistent with the recruit- trophy versus hyperplasia for this age range, the adiposity
ment of “new” adipocytes by hypertrophic adipocytes, as rebound is considered a critical period for adiposity later
discussed earlier.129 in life.139
Studies in rodents by Hirsch and colleagues demon-
strated that animals that were calorically deprived before Regulation of Adipogenesis
weaning had a reduced total number of fat cells when Adipose tissue, unlike other tissues such as the brain, kid-
compared to animals that were suckled in smaller lit- ney, or liver, retains the ability to increase in size in adult-
ters with higher caloric intake. Similarly, about half of hood. Several processes control the mass of adipose tissue
the obesity in Zucker fatty rats can be prevented by the in the body as follows:
early restriction of energy.130 This gave rise to the concept   
that early overfeeding during adipose tissue development • A dipocyte precursor proliferation
might increase the population of adipocytes and their • Differentiation of these precursors into mature,
precursors that produce obesity over time. This model, insulin-sensitive, lipid-storing adipocytes
known as the adipose-cell or critical period hypothesis, • The balance of lipid storage, utilization, and release
predicts that a large number of adipocyte precursors early within each mature adipocyte
in life could lead to the development of obesity by provid- • Apoptosis of mature adipocytes
ing a “sink” destined to be filled with lipid. A corollary to   
this concept is that individuals with a reduction in adipo- Adipocytes can be classified based on anatomic loca-
cyte precursors (e.g., those with a failure of adipocyte dif- tion as subcutaneous, visceral (intraperitoneal), bone
ferentiation, described earlier) would be predisposed to marrow, and structural (periorbital, e.g., palms of hands
the development of diabetes when food intake is increased and soles of feet). The hereditary and acquired lipodys-
as their storage capacity for excess fat is diminished. trophies teach us that each of these “depots” of adipose
Although the concept of an early-life “critical period” tissue are developed or regulated differently, as each form
for adipocyte precursor development has been much dis- of lipodystrophy results in the loss or failure to differenti-
cussed, by comparison the actual data supporting this ate in specific depots. For example, in congenital gener-
concept is sparse. Consistent with this concept, obese alized lipodystrophy, “mechanical” adipose tissue of the
subjects with an early childhood onset of obesity tend to palms and soles is spared.140
have smaller fat cells and are less hyperplastic when com- Adipose tissue precursors are primarily mesenchymal in
pared to those with a later adult onset of obesity.131,132 origin (Figure 36-7). These precursor cells, also known as
Similarly, children of mothers who were energy-deprived preadipocyte or stromal cells, have the capacity to differ-
during the Dutch famine of 1945 had a lower incidence entiate into a limited number of cell types including adipo-
of obesity in adulthood.133 Although no prospective long- cytes, osteoblasts, and chondrocytes.141 There are at least
term data exists to support the adipose-cell hypothesis, two distinct subtypes of preadipocytes [reviewed in refer-
the cross-sectional data supports the concept that in many ence 142]. These two subtypes, probably distinct from the
cases of early-onset obesity, adipose tissue size tends to be WAT/BAT “switch” described later, differ in their capac-
hyperplastic rather than hypertrophic; the latter is more ity for replication, differentiation, and susceptibility for
often seen with late-onset obesity. TNFα-induced apoptosis. Subcutaneous adipose tissue
The original data and hypothesis presented by Hirsch preadipocyte precursors replicate faster and differentiate
suggested a single early “critical period”; later discussions better than omental precursors. There is some evidence
offered the concept that additional critical periods of adi- that omental and mesenteric adipose tissue preadipocytes
pocyte precursor proliferation might also exist113 with differ as well.143-144 These differences in preadipocyte
recruitment from the precursor pools into mature adipo- characteristics are postulated to influence the propen-
cytes throughout life. At birth, a typical infant has about sity of an individual to store fat in the visceral versus
636 PART 5 DIABETES MELLITUS

TABLE 36-1 Extracellular Factors That Regulate


Adipogenesis

Activating
• GH/IGF-1/insulin
• Cortisol (GR ligand)
Pleuripotent Pre-adipocyte Mature insulin • Thyroid hormone (TR ligand)
precursor sensitive, lipid storing • Retinoic acids (RXR ligands)
(MSC) adipocyte • “Endogenous” PPAR-γ ligands
i. PGJ2
ii. HODEs/HETE
Inhibiting
• TNF-α
Protein/activity

C/EBP δ
Phospho-STAT5 • IFN-γ
C/EBP β • Pref-1
PPAR γ • Resistin
C/EBP α • TGF-β

Days
For example, IGF-1, a growth factor under the control of
insulin and growth hormone (GH) in adipose tissue, pro-
motes the proliferation and differentiation of preadipo-
PPAR γ Differentiation cytes, while TGF inhibits proliferation.156 More is known
C/EBP β
Adipocyte gene expression about the processes whereby adipocyte precursors, par-
C/EBP δ
C/EBP α Insulin sensitivity ticularly 3T3-L1 preadipocytes, proceed along the path-
Figure 36-7 Adipocyte differentiation cascade. Adipocytes are derived
way from precursor to mature adipocyte. We now know
from mesenchymal stem cell precursors (MSC). Both the mesenchymal that a series of transcription factors coordinately regulate
precursors and the preadipocyte, whose differentiation potential is lim- multiple genes in a tightly regulated temporal fashion. As
ited to the adipocyte lineage, can undergo mitosis. The middle panel shown in Figure 36-7, each of these transcription factors
depicts the coordinate, sequential activation of the major nuclear tran- forms a nonredundant network that, once initiated, leads
scription factors thought to be involved in the adipogenic differentiation
process. PPAR-γ is considered an obligatory “master” regulator of adi- to the emergence of the adipocyte phenotype. Some of
pogenesis. The PPAR-γ system turns on genes involved in lipid synthesis the known “key” transcription factors include PPAR-γ;
and insulin action. STAT5; C/EBP/α, β, and δ; SREBP1c/ADD; CREB; and
Wnt/frizzled. In addition, a new family of proteins—the
subcutaneous depots. Transcriptome analysis of subcuta- Kruppel-like factors—has been identified as regulators of
neous and visceral adipose tissue revealed a distinct pat- adipocyte differentiation.157
tern of expression of many genes, not only between depots Multiple hormones, cytokines, growth factors, cell
but also across BMI.145-146 In addition, there were major cycle regulators, and adhesion molecules control this dif-
differences in the expression of developmental genes in ferentiation “cascade.” Classic studies by Green and col-
subcutaneous versus visceral adipose tissue. Similarly, leagues showed that when clonal cell lines such as 3T3-L1
gluteal-femoral subcutaneous adipocytes display a differ- and F442A are confluent, they differentiate into adipo-
ent pattern of HOX genes, microRNAs, and long non- cytes if exposed to a cocktail of insulin, dexamethasone,
coding RNAs,147,148 suggesting epigenetic control of the and isobutylmethylxanthine (IBMX).158-160 Emphasis has
known differences in the cell and tissue biology of upper also been placed on the role of cell cycle and the necessity
and lower body adipocytes.149-152 Taken together with the for proliferation prior to differentiation of precursors.
cellular phenotypes identified by Kirkland’s laboratory,153 However, more recent data suggest that this has more to
this suggests that the different cell types contribute to dif- do with the E2F transcription factors rather than the pro-
ferences in the function of each adipose tissue region and cess of mitosis per se.161-162
that these differences can be explained by developmental IBMX and other agents that increase cAMP act
programming. In addition to these developments in the through the transcription factor CREB.163 Several tran-
different types of preadipocytes, the origins of the pre- scription factors are critical for the conversion of cells
adipocyte are becoming clearer. Work in multiple tissues from a fibroblastic phenotype to an adipocytic pheno-
including adipose tissue suggests that pericytes, cells that type. PPAR-γ has received the most attention, and this is
lie just outside the capillary, are a major source of mesen- warranted since overexpression of PPAR-γ into fibroblas-
chymal stem cells and preadipocytes.154-155 tic cell types is sufficient to confer the adipocytic pheno-
We know very little about the systems that control type.164 There are several putative “endogenous” ligands
proliferation of adipose tissue precursors. Most of what for PPAR-γ including the prostaglandin PGJ2,165,166
we know is derived from the study of the cellularity of long-chain fatty acids, and 13-HODE and 15-HETE,
rodent adipose tissue or the behavior of stromal-vascu- which can be generated from linoleic and arachidonic
lar cultures in vitro. Some of the known activators and acids, respectively, by a 12/15-lipoxygenase.167 All of
inhibitors of adipogenesis are presented in Table 36-1. these compounds can activate the PPAR-γ transcription
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 637

factor that heterodimerizes with the RXR transcription Given their central role in lipid storage and synthesis,
factor to turn on genes in the glucose uptake,168,169 lipid the PAT proteins are being actively explored as a means
uptake170 and lipid synthesis pathways.171,172 The true to better understand the regulation of these critical cel-
endogenous ligand(s) are unknown, but their synthesis/ lular processes.
activity appears to be downstream of the C/EBP-β tran-
scription factor.173 C/EBP-α is expressed contempora- Brown Adipose Tissue in Humans
neously with PPAR-γ and facilitates the full adipocytic BAT is an exquisitely designed tissue/organ system
phenotype. Immediately upstream of PPAR-γ lies the C/ evolved for the maintenance of body temperature. It is
EBP transcription factors C/EBPβ and C/EBPδ, which characterized by smaller cells with large amounts of mito-
upregulate PPAR-γ. Other transcriptional promoters of chondria and small lipid droplets providing a potential for
adipogenesis include STAT 5,174 the glucocorticoid recep- high cellular metabolism. At the metabolic, protein, and
tor, and ADD/SREBP-1c.175 Transcriptional inhibitors transcription levels, the BAT is upregulated principally
include GATA 3,176 TCF/LEF, and the Wnt pathway.177 by the sympathetic nervous system when production of
Combined with the transcriptional activators, they coop- heat is needed to maintain body temperature. Years ago,
erate in an orchestrated cascade of transcriptional events Nicholls and Ricquier described a mechanism for heat
leading to a mature adipocyte. production based upon a specific highly abundant protein
Lastly, PPAR-γ cofactors may regulate the ultimate (uncoupling protein 1; Ucp1) in the inner mitochondria
transcriptional program in adipocytes. For example, adi- of the brown adipocytes that uncouples the production of
pocytes can be converted from energy storage to energy chemical energy as ATP from oxidative phosphorylation
consumers by the PPAR-γ cofactor PGC-1a.178 Similarly, and instead produces heat.183,184
the PPAR-γ cofactors SRC-1 and TIF2 may determine Until quite recently, BAT has been thought to be
the responses of adipose tissue to high-fat diets, SRC-1– mostly important in small mammals and infants to main-
activating fatty acid oxidation, and TIF2-promoting lipid tain body temperature, but a function in the physiology
storage.179 These two examples highlight a growing under- of adult humans was dismissed because of low numbers
standing that not only the ligand but also the transcrip- of brown adipocytes.185,186 However, unrelated pursuits
tion factors and cofactors are important in whole-body in nuclear medicine using PET/CT scanning techniques
metabolism. The intracellular transcriptional control sys- have revealed the presence of BAT in adult humans espe-
tem is regulated by extracellular signals from cytokines, cially after cold exposure.187-188 The question is now how
hormones, neural inputs, and the autocrine/paracrine pro- can we induce this amazing organ not only to generate
duction of ligands for these transcription factors. heat but also to enhance fat oxidation, take up glucose,
Adipocytes and indeed virtually all other cells store and therefore reduce obesity.
neutral lipids, such as triglycerides, in droplets of varying Although the normal function of brown fat thermo-
sizes. These droplets are formed as triglyceride is synthe- genesis may be specific for the regulation of body temper-
sized in the ER or taken into the cell from the plasma ature, many genetic and pharmacologic studies in rodents
membrane. Lipid droplets are coated by a layer of phos- have shown that constitutive overexpression of Ucp1 in
pholipids and proteins that serve to sequester the neutral white fat and skeletal muscle can drastically reduce both
lipids from the cytosol and to regulate the access of lipases genetic and diet-induced obesity, offering therefore a new
to the surface of the lipid droplet. Substantial progress has safe molecular target for the treatment of obesity.189-191
been made in the identification of these lipid coat proteins This potential for brown fat adaptive thermogenesis as
since the initial discovery of perilipin and other members a drug target for obesity has not been ignored by the
of the perilipin family (perilipin, adipophilin, S3-12, and pharmaceutical industry. Unfortunately, many candidate
TIP-47).180 Consistent with their functional role, the agonists of the beta-3 adrenergic receptor have failed in
structure of these proteins is highly conserved with both human clinical trials, even though these drugs have been
hydrophilic and hydrophobic domains. In adipose tis- efficacious in rodent models of obesity. What is different
sue, each PAT protein plays a distinct role contributing in the human and mouse?
to lipid synthesis or lipolysis. For example, perilipin-A is Most of the effects of genetic, pharmaceutical or cold-
phosphorylated by PKA and PKG, which allows docking induced upregulation of Ucp1 in the mouse models result
of lipases and the initiation of lipolysis.181 On the synthe- in the emergence of new brown adipocytes in white fat
sis side, the repertoire of lipid droplet proteins changes as depots with levels of Ucp1 upregulated up to several
the lipid droplet is formed and matures, demonstrating hundred fold (Figure 36-8). Unfortunately, human WAT
the on-off exchange of these proteins and the dynamic does not appear to be able to mount such transient induc-
nature of the process for formation and movement of tion of brown adipocytes, at least not in subcutaneous
lipid droplets. Once again, the biology shows that the adi- adipose tissue. The failure of brown fat thermogenesis
pocyte is not a static, inert tissue but rather participates as in humans appears to be based upon the lack of funda-
an active organ in the regulation of metabolism via lipid mental information on the mechanisms controlling the
droplets, which are dynamic organelles in their own right. developmental origins of brown adipocytes in the discrete
Lipid droplet proteins may also play an important role in brown fat depots (e.g., interscapular brown fat) and in
lipid oxidation independent of their role in lipolysis. A the small number of diffusely localized brown adipocytes
novel PAT protein LDRP5 (also called OXPAT or Mldp) in various white fat depots. However, the transcription
appears to be important to activate lipid oxidation in oxi- factor PRDM16, whose presence can promote the dif-
dative tissues such as the cardiac myocytes.182 ferentiation of preadipocytes and myoblasts into brown
638 PART 5 DIABETES MELLITUS

? BMP7
Mesenchymal stem cells

Preadipocyte Progenitor cell


In utero
? +PRDM16 PRDM16

Discrete BAT Skeletal


muscle
WAT
Post-natal
Adrenergic
signaling
Neck
Supraclavicular
Diffuse BAT
Mediastinum
(para-aortic)
Paravertebral

?
Suprarenal

Figure 36-8 Brown adipose tissue in humans. Mesenchymal stem cells can have two major pathways, one into preadipocytes and another into
progenitor cells of brown adipocytes or skeletal muscle cells. Bone morphorgenetic protein 7 (BMP7) activates a full program of brown adipogenesis
including induction of early regulators of brown fat PRDM16 and PGC-1α. In absence of PRDM16, these progenitor cells are transformed into
skeletal muscle cells. Preadipocytes are differentiated into white adipocytes in subcutaneous and visceral adipose tissue. By stimulation of adrenergic
signaling, some of these white adipocytes can be transformed into brown adipocytes defused in the white adipose tissue (WAT). On the other hand,
discrete brown adipoctyes are concentrated in brown adipose tissue (BAT) depots such as in the neck, supraclavicular, paravertebral, and s­ uprarenal.
The BAT regulator PRDM16 is responsible, at least in rodents, only for the generation of discrete BAT. (From Seale P, Bjork B, Yang W, et al.
PRDM16 controls a brown fat/skeletal muscle switch. Nature 454[7207]:961-967, 2008; and Tseng YH, Kokkotou E, Schulz TJ, et al. New role of
bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454[7207]:1000-1004, 2008.)

adipocytes and whose absence promotes the myogenic white fat depots since at least in rodents. Diffuse BAT
differentiation program192 (see Figure 36-8), plays a key adipogenesis is more closely related to increased ther-
role in the development of BAT. Importantly, the abil- mogenesis and with reduced obesity.146 Enthusiasm
ity of PRDM16 to induce the brown adipocyte lineage is for the promise of PRDM16 as a drug target needs to
restricted to the discrete brown fat depots, such as that be tempered by the caveat that mice with an inactivated
found in the interscapular and supraclavicular region, PRDM16 gene die at birth, suggesting that PRDM16 is
but it does not participate in the induction of the dif- a transcription factor with additional unknown functions
fuse brown adipocytes located in the white fat depots. in mammalian development. It has long been known
The data support the concept that interscapular BAT and that chronic increases in circulating catecholamines in
brown adipocytes in white fat have separate independent patients with pheochromocytoma lead to large brown
developmental origins.193 PRDM16 is clearly an impor- fat depots.194 This historical data together with the more
tant player in brown adipogenesis but may not be suf- recent findings of discrete brown fat depots uncovered by
ficient, since PRDM16 KO mice have significant levels PET technologies187 should stimulate a renewed effort to
of interscapular fat with Ucp1 expression.192 However, find strategies to induce more brown adipocytes and to
one does not know whether upregulation of PRDM16 in ask why the many previous studies with beta-3 agonists
humans can induce increased discrete BAT and/or diffuse failed to significantly stimulate thermogenesis in humans.
brown adipocytes. Maybe the lack of beta-3 adrenergic receptors in human
While the regulator PRDM16 has provided important white adipocytes is something that needs to be overcome
insights into the developmental origins of discrete brown to facilitate conversion of white to BAT and to stimulate
fat depots, the next important step will be to determine thermogenesis. It is important also to evaluate the effects
the origin(s) of diffusely localized brown adipocytes in of bone morphogenetic protein 7 (BMP7) on stimulating
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 639

the enhanced expression of brown adipocytes as shown leads to chemotaxis of macrophages and inflammation.
by Tseng and colleagues in 2008.195 Similarly, the natri- These findings were recapitulated in human adipose tis-
uretic peptides also stimulate the expansion and activa- sue.66 Under these circumstances, hypoxia should turn
tion of BAT.196 Natriuretic peptides are released during on the transcription factor HIF-1 leading to angiogenesis
exercise, activate adipocyte lipolysis197 and therefore fat and a reversal of the hypoxia. This does not appear to be
oxidation,198 and when infused into humans increase the case in mature adipose tissue,213,214 where VEGF, a
thermogenesis.199 Combined with the newly discovered primary downstream transcriptional target of the HIF-1
capacity for BAT activation and interaction with the pO2 sensing system, is not activated. Anatomically, there
beta-3 adrenoreceptor, natriuretic peptides therefore pro- is good evidence that the new blood vessels sprout from
vide an attractive target for reversing metabolic distur- existing endothelial cells,215 and this has been co-opted in
bances of obesity. Lastly, the neuropeptide orexin-A has the development of ex vivo assays to clearly demonstrate
been shown in mice to expand and active BAT200 and is that human adipose tissue is capable of sprouting/angio-
necessary for BAT development. Irisin,201-203 metorin,204 genesis.216 Why the angiogenic signals are not increased
and FGF-21205,206 are other key peptides in the regulation when pO2 is decreased is a paradox that needs to be
of BAT mass and activity, the latter in a paracrine and investigated.
endocrine system involving the sympathetic nervous sys- In murine adipose tissue development, angiogenesis,
tem. Together, these results raise the intriguing possibility adipogenesis and the stromal cells interact in a way that is
that BAT might be activated pharmacologically through coordinated217,218 and relies on VEGF.219 This is different
non-adrenergic peptides such as orexin-A, irisin, FGF-21, from the situation in mature “hypoxic” adipose tissue,
and the natriuretic peptides. highlighting the differences in mechanisms and the rela-
Since the reemergence of BAT as a potential player in tive importance of growth factors in growth and devel-
the regulation of energy homeostasis, there has been a opment as opposed to the events occurring in mature
lingering question regarding the absolute effects in terms adipose tissue. Adiponectin, secreted by small but not
of calories that can be burned by the small amount of hypertrophic inflammatory adipocytes, stimulates angio-
BAT in humans. Recent studies using chronic cold to acti- genesis, highlighting an interplay of adipokines, which
vate BAT, analogous to using physical exercise to train we typically consider a “metabolic” hormone working in
muscle, showed that BAT can be expanded and activated concert with the growth factors classically implicated in
in vivo in humans.207,208 The overall thermogenic capac- angiogenesis.220
ity appears to be between 100 and 300 kcal/day,209-211 Endocrine Signals
although much work remains to be done to confirm
this.212 Glucocorticoids
The discovery of previously uncovered BAT/cells in Glucocorticoid treatment of laboratory animals results
adult humans and its potential physiologic significance in in the development of obesity. Animal models of obesity
cold- and dietary-induced thermogenesis should revamp invariably have increased levels of corticosterone. Adre-
our effort to target the molecular development of brown nalectomy results in the reversal or prevention of obesity.
adipogenesis in the treatment of obesity. Activation of the glucocorticoid receptor results in dif-
ferentiation of preadipocyte precursors158-160 and lipid
Integrative Biology of the Adipose Tissue storage in adipocytes. In humans, overproduction of
To maintain energy homeostasis, the brain has two ave- cortisol (Cushing’s syndrome) results in a phenotype of
nues of communication with the periphery, hormones, central (abdominal) obesity, hypertension, and diabetes.
and neurons. The adipose tissue is no exception to this Of the many investigations into the role of adrenal gluco-
rule, since it is well established that both hormones and corticoids in human obesity, most show normal urinary
neurons control its life cycle and metabolism. Under- free cortisol, normal circadian variation in cortisol val-
standing how adipose tissue operates and is regulated ues, and normal plasma cortisol values, although meta-
in vivo is a prime example of integrative biology. Imaging bolic clearance rate and production are increased.221 The
studies of the different adipose depots as well as studies enzyme 11βHSD-1 is present in human adipose tissue and
of adipose tissue metabolism and secretary function using converts inactive cortisone into active cortisol.222 Impor-
arterial-venous differences and microdialysis in conjunc- tantly, there is a strong positive correlation between
tion with measures of adipose tissue blood flow have all adipocyte size and the activity of 11βHSD-1 to convert
improved our integrative view of the metabolism and cortisone into cortisol. This fits with the idea that adipo-
function of the adipose tissue. cyte hypertrophy generates signals, like cortisol, to recruit
There is increasing appreciation for the importance of new adipocytes.
the vasculature to the proper functioning of the adipose The most compelling data for an association between
tissue. Nutrients such as glucose, fatty acids, and proteins human obesity and cortisol comes from studies that clas-
are supplied from the vasculature and are critical to the sify obese women into central and peripheral types of
growth and maintenance of adipose tissue. Recent studies obesity. By stratifying volunteers on this basis, Marin and
have focused on the supply of oxygen for oxidative metab- coworkers demonstrated an increase in urinary and serum
olism in adipocytes. Current data suggest that the delivery cortisol as the waist-to-hip ratio increased.223 Serum cor-
of oxygen may be limiting in adipose tissue of obese mice. tisol responses to stress were greater in women with high
The consequences of the hypoxia are unclear, but in vitro waist-to-hip ratio, suggesting a role of response to envi-
data59 support the in vivo data,63 suggesting that hypoxia ronmental stressors as a potential factor in abdominal
640 PART 5 DIABETES MELLITUS

obesity.223,224 Other evidence in humans suggests that In human adipose tissue, ERβ was higher in abdominal
cortisol values within normal concentrations are some- compared to gluteal femoral adipose tissue, and regional
times related to fat patterning, possibly via increased sen- differences in adipose tissue expression of ERα and ERβ
sitivity to exogenous stressors. Genetic factors may also were described by Pedersen and colleagues.246 But not all
determine the susceptibility of the adipose tissue to these of the effects of estrogen are in the adipose tissue. Admin-
exogenous stressors.225 istering estrogen directly into the brain is able to reverse
the gain in visceral fat seen with ovariectomy, suggesting
Growth Hormone/IGF-1 that much of the fat patterning attributed to peripheral
GH is a potent lipolytic hormone.226 GH receptors acti- effects might actually be mediated through hypothalamic
vate classic cAMP lipolytic systems in adipose tissue. In signaling.247
addition to stimulating lipolysis, GH increases IGF-1 In summary, estradiol is an important sex steroid, both
production in adipose tissue.227 IGF-1 potently activates for the proliferation of adipocyte precursors and for the
preadipocyte proliferation and differentiation of precur- regulation of lipid storage in a regional-specific fashion.
sors into mature lipid-storing adipocytes.156 Deficiency of
GH is associated with central obesity, and replacement of Neural Signals to the Adipose Tissue
GH reduces visceral adiposity.228 Despite early reports of As discussed earlier, the human adipose tissue can be
therapeutic efficacy of GH in men with central obesity,229 divided into two major compartments, subcutaneous and
other studies do not show this effect and in fact show visceral (approximately 80% and 10%, respectively),
an increase in body fat after GH withdrawal compat- whereas other depots such as intramuscular adipose tis-
ible with the effects of IGF-1 to promote adipoctye dif- sue and retroperitoneal, perirenal, and orbital fat account
ferentiation.213 GH treatment in the absence of clear-cut for the remainder.248 The two major compartments
GH deficiency cannot be recommended as the side-effect have clearly different rates of lipid synthesis and lipoly-
profile includes edema, carpal tunnel syndrome, glucose sis, probably due to differences in hormonal exposure
intolerance, and many others. GH-like peptides that and innervation. The brain needs to transmit messages
increase lipolysis without upregulation of IGF-1 synthesis to different parts of the body in a selective manner. For
have been discovered230 and may be beneficial without that reason, the sympathetic nervous system innervates
the adverse effects of GH. different adipose tissues in different ways, influencing
not only regional blood flow but also functions such as
Estrogen in Adipose Tissue lipolysis and lipid synthesis. By viral injection in fat pads
Men and women have a different distribution of body of Siberian hamsters, Youngstrom and Bartness showed
fat: a gluteal-femoral pattern in women and an abdomi- the presence of sympathetic projections from central sym-
nal pattern in men. This sexual dimorphism is thought pathetic ganglia, which were confirmed by injection of
to be due to differences in the sex steroids estrogen and fluorescent anterograde tract tracers into the sympathetic
testosterone. Lipoprotein lipase (LPL) activity, indicative chain ganglia249 and viral tracing studies.250-252 In addi-
of lipid storage, is increased in the gluteal-femoral region tion, denervated fat depots weigh 10% more than the
of women as compared to men. After menopause, LPL intact contralateral depot, implying impaired lipid mobi-
activity is equivalent across all adipose tissue depots, sug- lization in fat pads deprived of their innervation.253 From
gesting that estrogen upregulates LPL in a depot-specific such studies, it was hypothesized that catecholamines
fashion.231 In support of this concept, treatment of post- not only increase lipolysis but also inhibit adipose tissue
menopausal women with estradiol increased LPL activity hyperplasia from preadipocytes, which is supported by
in the gluteal-femoral region,232 which was reversed by in vitro data.250,254,255
the addition of a progestin.233 In vitro in human abdomi-
nal subcutaneous adipocytes, low-dose estradiol increased Regulation of Lipolysis
LPL protein, and higher-dose estradiol decreased LPL.234 Adipose tissue lipolysis, that is, the catabolic process
These dose-dependent effects of estradiol to decrease LPL leading to the breakdown of triglycerides into fatty acids
were also observed in a cross-sectional study235 and after and glycerol, is often considered as a simple and well-
local transdermal application of estradiol.236 In addi- understood metabolic pathway (Figure 36-9). However,
tion to systemic estradiol, the stromal-vascular fraction we continue to discover new layers of complexity in the
of adipose tissue is able to convert estrogenic precursors system. Hormone-sensitive lipase is a major determinant
to estrogen vis-à-vis the enzyme aromatase. In men, tes- of fatty acid mobilization in adipose tissue. Transloca-
tosterone, but not the non-aromatizable steroid dihy- tion of hormone-sensitive lipase to the lipid droplet seems
drotestosterone, increases adipose tissue lipid turnover, to be an important step during lipolytic activation. Reor-
suggesting that testosterone acts in adipose tissue via ganization of the lipid droplet coating by perilipin may
local conversion of testosterone into estrogen by aroma- also facilitate the access of the enzyme. In humans, altera-
tase.237,238 In vitro, estradiol increases the proliferation tions of hormone-sensitive lipase expression are associ-
of stromal-vascular cell cultures of both human 239 and ated with changes in lipolysis in various physiologic
rodent preadipocytes.240 and pathologic states. The major hormones controlling
Several investigators241-244 have demonstrated estradiol the lipolytic process are catecholamines (stimulation of
binding and ERα mRNA in adipose tissue extracts. After lipolysis) and insulin (inhibition of lipolysis). It is well
the cloning of the ERβ gene, both mRNA and protein for accepted that the adrenergic system is the major regulator
ERβ were subsequently described in adipose tissue.243-245 of lipolysis via a cAMP pathway. In turn, cAMP increases
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 641

α2-agonists peptide activation of the cGMP system plays an important


Adenosine role in exercise-activated lipolysis.197
NPY/PYY Insulin ANP
Norepinephrine Prostaglandins BNP It was once thought that hormone-sensitive lipase was
Rs Ri the first step in the lipolytic cascade. The recent discovery
Plasma membrane of patatin-like phospholipase domain containing 2, also
Gs Gi known as Adipose TriGlyceride Lipase (ATGL), changed
PDE3 GMP
ATP
AC
cAMP 5'AMP PDE5 GC GTP that view, and we now know that ATGL is a key molecule
for the first step in triglyceride hydrolysis, TAG hydrolase
cGMP
activity.263 ATGL and its co-activator protein CGi-58/
P-perilipin ABHD5 are required for the full activation of the lipolytic
Perilipin cAMP cGMP Perilipin
PrK PrK
cascade (see Figure 36-9), with HSL acting to hydrolyze
Cgi-58 HSL P-HSL HSL
(PKA) (cGKI) DAGs into MAGs and monoglyceride lipase finishing the
cascade. New data show that this sequential model may
ATG
be too simplified. When perilipin-A is phosphorylated
Cgi-58
by PKA, CGi-58 is released and “assists” in the recruit-
LIPOLYSIS
ment of ATGL to the lipid droplet.264 Taken together,
Figure 36-9 Regulators of human adipocyte lipolysis. Hormone-sen- the discovery of ATGL, along with the discovery of the
sitive lipase and perilipin are rate-limiting steps in the regulation of adi- natriuretic peptide–driven cGMP lipolytic pathway,262
pocyte lipolysis. Both enzymes need to be phosphorylated to be active
and allow the breakdown of triglycerides into glycerol and free fatty has dramatically changed our view of the regulation of
acid. Part of this process involves the release of Cgi-58 when perilipin lipolysis. We now know that translocation of HSL to the
is phosphorylated; it can then bind and activate ATGL, the specific tri- lipid droplet is not the only regulatory step in the activa-
glyceride hydrolase. The adrenergic systems (β-receptors and α2 recep- tion of lipolysis; there is a network of controlled signal-
tors) are major regulators of lipolysis via cyclic AMP (cAMP) pathways.
β-receptors stimulate lipolysis, whereas α-2 receptors inhibit lipolysis.
ing that includes interactions between the lipases and the
There are other novel Gi-coupled G protein–coupled receptors; see text PAT proteins. This is a hot area of research, and a better
for details. cAMP increases the activity of protein kinase A, which in understanding of the interactions between the PAT pro-
turn phosphorylates the hormone-sensitive lipase and perilipin. Insulin teins, lipases, and their signaling systems will hopefully
inhibits lipolysis via inhibition of phosphodiesterase (PDE), therefore lead to new therapies to prevent the dysregulated lipolysis
decreasing cAMP and lipolysis. A novel lipolytic system using natriuret-
ic peptides stimulates lipolysis through a cGMP-dependent pathway, observed in hypertrophic adipocytes.
which is not influenced or suppressed by insulin action. In adipose tis-
sue, cGMP is degraded by PDE5 (385). AC, Adenlyate cyclase; AMP, Adipocyte as an Endocrine Organ
adenosine monophosphate; ANP, atrial natriuretic protein; ATGL, The study of the biology of adipose tissue including the
adipose triglyceride lipase; BNP, brain natriuretic protein; cAMP, cyclic
AMP; cGMP, cyclic GMP; GC, guanylate cyclase; Gi, inhibitory G-protein;
mechanisms of adipogenesis has enjoyed an explosive
GMP, guanine monophosphate; Gs, stimulatory G-protein; NPY/PYY, growth over the past 15 years. Unarguably, the trigger
neuropeptide Y, peptide YY; PDE, phosphodiesterase; Ri, receptor, for this renewed interest came from the cloning of the ob
inhibitory; Rs, receptor, stimulating. (From Moro C, Klimcakova E, (obese) gene and the discovery of leptin in 1994.2 This
Lafontan M, et al. Phosphodiesterase-5A and neutral endopeptidase seminal discovery initiated a period of intense research
activities in human adipocytes do not control atrial natriuretic peptide-
mediated lipolysis. Br J Pharmacol 152[7]:1102-1110, 2007.) for uncovering the endocrine and paracrine roles of the
adipose tissue and its role in the regulation of energy bal-
ance and the development of obesity and its related dis-
eases. The steps that led to the discovery of leptin were
the activity of protein kinase-A, which phosphorylates summarized in the original description of the cloning of
both the hormone-sensitive lipase and perilipin. As a the leptin gene.2 In brief, the original notion of a homeo-
counteracting hormone, insulin binds to its receptor and static regulation of energy balance (and therefore adipose
activates the various elements of the insulin signaling mass) dates back to Lavoisier and Laplace.265-267 The key
cascade by stimulation of Type 3 cGMP-inhibited phos- role of the brain in this regulation came later from clini-
phodiesterase (PDE3B), therefore decreasing cAMP and cal observations and was confirmed by stereotaxic lesions
suppressing lipolysis.256 The antilipolytic effect of insu- of different regions of the brain.268 It was therefore pos-
lin is reduced in the insulin-resistant state.257 Progress on tulated that energy balance was regulated by a feedback
the hormonal regulation and molecular mechanisms of loop in which the body energy stores were sensed by the
beta-lipolytic and alpha2-antilipolytic adrenergic control hypothalamus, which, in turn, sent signals to control
of lipolysis has improved our understanding of the rela- both food intake and energy expenditure. The nature
tive contribution of the two types of receptors.258 Genetic of the signal inputs to the hypothalamus was, however,
studies show that polymorphisms in genes coding for dif- not clear. Jean Mayer proposed a “glucostatic theory” in
ferent beta-adrenoceptor subtypes and hormone-sensitive which blood glucose was the sensed signal.269 Kennedy
lipase may participate in the polygenic background of postulated the presence of a “fat metabolism factor”
obesity.259 and proposed what is now accepted as the lipostatic the-
More recently, a novel lipolytic system has been char- ory.270 In this model, a signal coming from the fat stores
acterized in human fat cells. Natriuretic peptides stimulate in the adipose tissue is read by the central nervous system
lipolysis through a cGMP-dependent pathway, which is to regulate feeding and energy homeostasis. Subsequent
not influenced or suppressed by insulin action.260-262 Along parabiosis studies performed by Hervey confirmed that
with catecholamine stimulation of cAMP, natriuretic blood-borne signals were coming from the adipose tissue
642 PART 5 DIABETES MELLITUS

and regulated food intake and body weight.271 Not too TNF-α
long after, Coleman performed the seminal parabiosis Interleukins
TGF β
studies using single-gene models of obesity and diabetes IGF-1 FGF
(ob/ob and db/db mice) and concluded that the product Bone morphogenic protein IGFBP EGF
MCP-1
of the ob gene was secreted by the adipose tissue, trans- Fatty acids
ported by the blood, and received in the hypothalamus by Visfatin Lysophospholipid
Lactate
the receptor encoded by the db gene.272 This interaction Resistin Adenosine
between a factor produced by the adipose tissue and a Adipose
Apelin Prostaglandins
receptor in the hypothalamus became the foundation for Glutamine
Adiponectin Adipose
Leibel and colleagues to undertake the positioning clon- UNKNOWN
tissue
ing effort of the ob and db genes, leading to the publica- Adipsin FACTORS
tion of the discovery of leptin in Nature in 19942 and of
Estrogen Agouti protein
its receptor in Cell 1 year later.273
Retinol
Since the discovery of leptin, the simple paradigm of Angiotensin-II
adipose tissue as a fat-storage tank has evolved into a Plasminogen activator
inhibitor-1
complex paradigm. First, the size of the adipose tissue Angiotensin Leptin
is not only controlled by the filling of pre-existing adi- Acylation-stimulating protein
pocytes but involves finely tuned mechanisms control-
ling differentiation and apoptosis of the tissue. Second, Figure 36-10 Proteins secreted by the adipose tissue. Adipose tissue
is an endocrine gland that secretes numerous factors, many of which
adipose tissue depots are multipotential secretory organs are implicated in affecting energy homeostasis, insulin sensitivity, and
with different secretory capacities for different depots. nutrient-sensing pathways.
These adipose tissues are composed mostly of adipocytes
but also of fibroblasts and immune cells such as macro-
phages and mast cells, which all use endocrine, paracrine, metabolism since they act on different tissues including the
and autocrine pathways to secrete multiple bioactive brain, the liver, the skeletal muscle, and the adipose tissue
proteins called adipokines or adipocytokines. The adipo- itself.
cytes respond to various stimuli such as circulating hor-
mones, circulating metabolites, neural input, and cellular Leptin
energy signals by releasing hormones and substrates as Leptin is a highly conserved 16-kD hormone secreted
shown in Figure 36-10.274-275 The molecular revolution principally but not exclusively by adipocytes and which
brought to light many adipocyte-secreted factors, some of acts both centrally and peripherally. Plasma leptin con-
which are secreted into the bloodstream such as IL-6 and centrations are positively correlated with body fat
leptin, whereas others, such as TNF-α, exert their effects mass.277 Leptin crosses the blood-brain barrier by a satu-
in an autocrine/paracrine fashion.276 Although adipose rable active transport system and serves as a signal to the
tissue has a similar histological appearance throughout central nervous system originating in the adipose tissue.
the body, it is now obvious that there are fundamental Even though it was originally described as the hormone
regional differences in the quality and the amount of regulating energy balance, available data now suggest
secreted adipokines by these different depots. that a relative lack of leptin or resistance to its action are
A major emphasis in adipose tissue biology research probably not causal of most cases of human obesity. The
is the understanding of the molecular mechanisms con- main biological function of leptin seems to be the mainte-
trolling the secretion of adipokines by different depots nance of a minimum level of energy stores during periods
and their implication in a variety of chronic diseases. of caloric restriction.278-279 Low leptin concentration can
These secreted proteins have been grouped in molecules therefore be seen as a “starvation signal” when energy
regulating physiologic and pathophysiologic functions
­ stores become insufficient, commanding the body to seek
such as:275 food and become thrifty. As part of such a protective
   mechanism, leptin plays a role in reproduction, angiogen-
1. Energy homeostasis (leptin, adiponectin, resistin, esis, bone architecture, and immune function, and it may
omentin, apelin) also influence processes such as beta-cell insulin secre-
2. Innate immune system (TNF-α, IL-6, IL-8) tion, carbohydrate transport, and platelet aggregation.280
3. Vasculature (VEGF, monobutyrin, ESM-1) Low levels of circulating leptin trigger strong biological
4. Acute phase reactant response (alpha1 acid glyco- responses to protect the organism against the deleterious
protein, SAA3, PTX-3) effect of starvation, whereas high levels of leptin (as seen
5. Molecules involved in lipoprotein metabolism/insu- in obesity) engender rather weak biological responses.278
lin action such as LPL or components of extracel- This asymmetric biological effect of leptin is illustrated in
lular matrix (type VI collagen) the upper panel of Figure 36-12.
6. Recruitment of immune cells (MCP-1, MIP-1) Studies of caloric restriction in animals and humans
   provide information regarding the importance of leptin
In this chapter, we have chosen to present the cur- as a mediator of neuroendocrine responses. Shimokawa
rent knowledge on only five of these adipokines: leptin, reviewed the endocrine changes associated with short-
adiponectin, resistin, TNF-α, and apelin. As shown in term caloric deprivation in rodent models.281 Many of
­Figure 36-11, these adipokines are involved in whole-body these alterations have been described in humans as well
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 643

↓ Food intake
↑ Energy expenditure ↑ Triglyceride content
↑ Glucose output

↑ Insulin resistance
↑ Lipolysis
TNFα

Leptin

Free-fatty acids

Adiponectin ↓

Adipose tissue
Resistin

↑ Triglyceride content
↓ Glucose uptake

Figure 36-11 Central role of adipose tissue in insulin-resistant syndrome. The production by adipocytes of substrates such as free fatty acids; hor-
mones such as leptin, adiponectin, and resistin; and cytokines such as TNF-α positions the adipocyte as a central mediator of insulin-resistant syn-
drome in obese individuals. In response to weight gain, free-fatty acids, leptin, resistin, and TNF-α are all increased, whereas adiponectin concentra-
tion is decreased. These changes impact the insulin sensitivity of skeletal muscle and liver and the central nervous system control of energy expenditure
and food intake. Positive feedbacks are shown in solid lines, whereas negative feedbacks are shown in dotted lines. (Adapted from Farmer SR. In:
Alberti KGM, DeFronzo RA, Keen H, Zimmet P, eds. International textbook of diabetes mellitus. 3rd ed. Hoboken, NJ: John Wiley & Sons, 2004.)

and include a decrease in T3,282 an increase in cortisol leptin seems to exert its peripheral metabolic effects is
secretion,283 and a decrease in gonadal function.279 It has by activating 5′-AMP-activated protein kinase (AMPK)
long been hypothesized that the neuroendocrine system in muscle and liver.290-291 As a consequence of AMPK
coordinates and integrates some of the antiaging actions activation, ATP-consuming anabolic pathways are inhib-
of calorie restriction.284-287 In a 48-hour prolonged star- ited, whereas ATP-producing catabolic pathways are
vation study in mice, Ahima and associates provided evi- activated. The activated mechanisms include glucose
dence that the reduction in leptin with starvation caused transport, beta-oxidation, glycolysis, and mitochondrial
a decrease in the activity of the gonadal and thyroid axes biogenesis. The relevance of leptin in normal human
and an increase in the activity of the adrenal axis.279 The metabolic function is provided by leptin replacement in
changes in activity of these axes during fasting were pre- individuals with genetic leptin deficiency,292 deficiency
vented by leptin administration, suggesting that leptin is a due to weight loss,293 or lipodystrophy.294 The effects
master regulator of the neuroendocrine system and possi- of recombinant leptin therapy in children with congeni-
bly the endocrine candidate of the “disposable soma the- tal leptin deficiency was investigated and clearly showed
ory” of aging stating that longevity requires investment in a spectacular effect on reducing food intake and body
somatic maintenance by reducing the resources available weight but almost no effect on energy expenditure and
for reproduction281-288 (see Fig. 36-12, bottom panel). fat oxidation.292 In adult patients with a similar congeni-
In the obese state in which the circulating leptin con- tal deficiency, leptin replacement not only impacted food
centration is already high (see Fig. 36-12), the hormone intake but also prevented the drop in energy expenditure
is a rather weak signal to prevent overconsumption of usually observed with weight loss and increased 24-hour
food and does not appear to be a viable treatment for fat oxidation by more than three times.295 Interestingly,
obesity.289 However, if provided in sufficient amounts weight loss seems to be amplified in a synergetic manner
in obese individuals or to organisms deficient in circu- when leptin is administered in conjunction with pramlint-
lating leptin, injection of the hormone can reduce body ide, an analogue of amylin.296
weight and fat mass by decreasing food intake and HIV and HIV therapy are also associated with altera-
increasing energy expenditure. The mechanism by which tions in body composition, including lipoatrophy, lipid
644 PART 5 DIABETES MELLITUS

5 to 30 nM.307 Adiponectin, a 30-kD protein consisting


Biological effect of leptin
of an N-terminal collagenous domain and a C-terminal
globular domain, was discovered almost simultaneously
by four separate groups using different methods.306,308-310
Adiponectin exists in the blood in monomer, trimer, hex-
Asymmetrical amer, and very high molecular weight forms.311 This
biological response protein is closely related to complement factor Cq1,308
but the folded crystal structure and gene organization
show close similarity to TNF-α.312,313 Arguably, the
most interesting observation is that unlike other adipo-
cytokines whose expression increases with increasing fat
mass, adiponectin is inversely related to fat mass.314-316
A Plasma leptin concentration or body fat (%) How the increased mass of the tissue from which the gene
is expressed reduces expression and/or secretion of the
protein is still an unanswered question. Consistent with
Energy restriction the observation that lipodystrophic patients have very
low concentrations of adiponectin317 and increased ecto-
↓Fat mass
pic fat, a reduction in circulating adiponectin (as well as
leptin) may facilitate the ectopic storage of fat.
Adiponectin is clearly an insulin-sensitizing hormone,
↓Leptin and administration of recombinant adiponectin in rodents
increases glucose uptake and fat oxidation in muscle,
reduces fatty acid uptake and hepatic glucose production
↑Energy available for: ↓Energy available for: in liver, and improves whole-body insulin resistance. Two
• Maintenance of soma • Reproduction receptors for adiponectin have been discovered, with the
• Adrenalcortical axis • Growth first isoform (AdipoR1) mostly expressed in skeletal mus-
B • Energy metabolism
cle and the second (AdipoR2) mostly in liver.318 Unlike in
Figure 36-12 Leptin as the master neuroendocrine signal. As shown in mice, gene expression profiling in humans indicates that
panel A, the biological responses to changes in body energy stores and both isoforms are highly expressed in skeletal muscle.319
circulating leptin are asymmetric and are much more robust when plas- Interestingly, in individuals with normal glucose toler-
ma leptin concentration decreases with caloric restriction and significant
weight loss rather than when it increases with obesity (278). Leptin’s
ance, muscle expression levels of AdipoR1 and AdipoR2
main function may be to provide the appropriate metabolic responses were lower in subjects with a family history of type 2
to decreased energy stores (by acting to increase feeding and by sparing diabetes when compared to those without a family his-
energy/fat expenditure) rather than protecting the body against excess tory, and the expression level of both receptors correlated
energy stores (i.e., by increasing energy/fat expenditure and decreasing positively with insulin sensitivity.319 These data indicate
food intake). Panel B describes the role of leptin as a master regula-
tor of neuroendocrine pathways involved in response to the effects of that both isoforms of the receptor may play a role in the
caloric restriction. It is probably a major signal for the antiaging effects insulin-sensitizing effect of adiponectin probably via a
of dietary restriction (281). (From Leibel RL. The role of leptin in the stimulation of mitochondrial biogenesis, mitochondrial
control of body weight. Nutr Rev [10 Pt 260]:S15-S19, 2002; discussion function, and fat oxidation.320
S68-S84, 85-17; and Shimokawa I, Higami Y. Leptin and anti-aging
action of caloric restriction. J Nutr Health Aging 5[1]:43-48, 2001.)
Even if the signaling cascade for adiponectin is unclear,
there is growing evidence that adiponectin may activate
AMPK, the putative master metabolic regulator described
storage as abdominal adipose tissue, or buffalo hump.297 earlier. Thus, excitement surrounds the potential for
The constellation of metabolic findings in these patients is adiponectin (or mimetics of adiponectin) to represent
consistent with the ectopic fat storage hypothesis as pre- pharmacologic agents for patients suffering from insulin
sented earlier. Several studies suggest that activation and resistance and type 2 diabetes. As for leptin, many func-
rebound of the immune system during antiretroviral ther- tions have already been attributed to adiponectin; it has
apy are associated with lipodystrophy.298,299 This is anal- been linked to cardiovascular disease and endothelial
ogous to defects seen in congenital partial lipodystrophy, and immune dysfunctions.321 However, we will focus
which are due to mutations in the lamin A/C gene.300-302 mostly on the role of adiponectin as an insulin-sensitizing
There is also evidence of increased apoptosis in adipose hormone.
tissue from HIV lipodystrophy patients.303 Interestingly, Adiponectin acts peripherally to improve insulin sen-
patients with congenital lypodystrophy or patients with sitivity in rodents,322-325 although the proposed mecha-
AIDS and lipoatrophy induced by antiretroviral therapy nisms differ. Combs and colleagues found that increasing
have shown dramatic improvement of their ectopic fat circulating adiponectin concentrations in mice during a
storage syndrome with leptin replacement therapy.304,305 euglycemic clamp increased the rate of glucose infusion by
73%.326 The rates of glucose uptake, glycolysis, and gly-
Adiponectin cogen synthesis were unchanged, but the rate of glucose
Adiponectin (also known as AdipoQ, Acrp30, APM1, and production was suppressed by 65%. Chronic infusion
GBP28) is exclusively expressed in adipose tissue306 and of a proteolytic product of adiponectin prevents weight
circulates in human serum at very high concentrations of gain in mice fed a high-fat diet, whereas mice infused with
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 645

full-length adiponectin or saline gained weight.322 The normalized glucose and insulin concentrations and dra-
prevention of weight gain was associated with increased matically improved glucose tolerance as well as positively
fat oxidation. The same investigators went on to show affected serum triglyceride levels despite morbid obesity.
that adiponectin induces fatty acid oxidation in muscle The authors propose that adiponectin acts as a peripheral
in vitro and reduces FFA flux following a high-fat meal “starvation” signal promoting the storage of triglycerides
or intralipid infusion in vivo.322 Similarly, Yamauchi and preferentially in adipose tissue, therefore protecting the
co-workers observed that adiponectin treatment dur- animals from ectopic fat depots.332
ing high-fat feeding prevents adipose tissue deposition
in wild-type mice by increasing energy expenditure and Resistin
decreasing ectopic fat deposition.323 To determine the Resistin is a putative adipocyte-derived “insulin resis-
cellular mechanisms underlying this observation, they tance” hormone that was identified during an in vitro
measured the expression of genes involved in fatty acid screen for genes upregulated during adipocyte differentia-
transport, oxidation, and energy dissipation in both mus- tion and downregulated by PPAR-γ agonists.333 In mice,
cle and liver. Adiponectin treatment increased the mRNA serum resistin and resistin mRNA expression in adipose
of genes involved in fatty acid uptake and β-oxidation tissue are increased by high-fat diet, and decreased after
(specifically, CD36, acyl-CoA oxidase, and uncoupling rosiglitazone treatment.333 Importantly, blocking resistin
protein 2) in muscle and decreased the expression of (by antibodies) increases glucose uptake in fat cells and
genes involved in fatty acid transport in the liver, result- increases insulin sensitivity. In vitro, resistin decreases
ing in decreased storage of triglycerides in non-adipose glucose uptake in skeletal muscle cells but does not affect
tissues and indirectly improving insulin sensitivity. Taken the “classic” insulin-signaling pathways.334 As expected
together, the results support the theory that adiponectin from these results, intraperitoneal administration of resis-
is a regulator of insulin sensitivity through the reduction tin increased blood glucose following a glucose tolerance
of ectopic fat deposition. test in mice.333 Taken together, these results led to the
Like leptin, adiponectin was shown to directly activate hypothesis that resistin promotes insulin resistance. At
AMPK in muscle, thereby increasing the phosphorylation about the same time, two other groups independently
of acetyl coenzyme A carboxylase.325 In turn, malonyl identified resistin.335,336 Kim and colleagues335 observed
CoA content is reduced, increasing carnitine palmitoyl- that resistin inhibited adipocyte differentiation in 3T3-
transferase 1 activity and stimulating fat oxidation. In the L1 cells, suggesting that resistin may promote insulin
liver, AMPK stimulates fatty acid oxidation and ketogen- resistance by increasing storage of triglycerides in muscle
esis and inhibits cholesterol synthesis, lipogenesis, and tri- and liver instead of adipose tissue. Resistin-deficient mice
glyceride synthesis, whereas it modulates insulin secretion have low blood glucose after a fast, decreased hepatic
in pancreatic beta cells (reviewed in reference 327). glucose production, and less hyperglycemia when obese,
Human studies in Pima Indians and Japanese indi- suggesting a key role in the regulation of hepatic glucose
viduals demonstrated an association between low plasma production.337 Ahima and Lazar reviewed the role of adi-
adiponectin concentrations and obesity or type 2 dia- pokines, including resistin, on the control of energy and
betes.314,316 Furthermore, positive correlations between carbohydrate metabolism.338
plasma adiponectin concentrations and insulin sensitiv- In humans, the role of resistin in regulating insulin
ity have been reported in several studies.315,316,328 More sensitivity is unclear. In one study, serum resistin was
importantly, low adiponectin concentrations are pre- related to fat mass in young, healthy subjects and was
dictive of type 2 diabetes incidence rates over a 5-year significantly higher in women than in men,339 but it was
follow-up in Pima Indians.329 Other indicators that low not related to BMI, percent body fat, or insulin sensitiv-
levels of adiponectin may be involved in the development ity in other studies.340,341 Resistin mRNA expression was
of insulin resistance are derived from intervention studies shown to be higher in morbidly obese subjects as com-
showing that adiponectin is decreased by behaviors lead- pared with lean control subjects 342 and higher in individu-
ing to obesity and diabetes323 and increased in situations als with a promoter mutation and high levels of oxidative
of reduced body fat or increased insulin sensitivity.330,331 stress.343 However, in that same study, serum resistin was
At present, adiponectin remains a validated target for not different between non-obese, obese, or obese diabetic
the potential treatment of insulin resistance and type 2 groups. Data in humans indicates that resistin is derived
diabetes. However, many questions regarding adiponec- mainly from macrophages.344 Given the emerging interre-
tin remained to be resolved before contemplating the use lationship between inflammation and metabolic disease,
of adiponectin (or mimetics) as therapeutic agents. These hyperresistinemia may be a biomarker and/or a mediator
include: (1) which circulating form of adiponectin is bio- of metabolic and inflammatory disease in humans.
logically active; (2) what are the post-translational mech-
anisms that regulate adiponectin concentration/secretion; TNF-α
(3) what are the exact sites of action of adiponectin in TNF-α is a cytokine that is produced by macrophages,
central and peripheral tissues; and (4) what is the signal- monocytes, endothelial cells, neutrophils, smooth muscle
ing cascade of adiponectin after binding to its receptor? cells, activated lymphocytes, astrocytes, and adipocytes.345
Kim and associates332 in Scherer’s laboratory made a big TNF-α has a variety of functions, such as mediating expres-
step toward a better understanding of the function of sion of genes for growth factors, cytokines, transcription
adiponectin by creating mice lacking leptin while over- factors, and receptors. TNF-α is synthesized as a 26-kD
expressing adiponectin. Surprisingly, these mice have transmembrane protein found on the surface or processed
646 PART 5 DIABETES MELLITUS

to release the 17-kD soluble form.346 Some adipocytokines confirming that the apelin/APJ signaling pathway plays
are secreted and transported into the blood (i.e., leptin, a critical role in the development of the functional vascu-
PAI-1, and IL-6), whereas TNF-α is secreted and probably lar network in adipose tissue.363 The adipokine apelin is
acts locally in an autocrine-paracrine fashion.276 not only upregulated by hypoxia and insulin but also by
Initial reports implicated TNF-α as an adipocyte- the transcriptional co-activator PGC-1α in human white
derived cytokine that was able to block adipocyte differ- adipocytes.364 Finally and importantly, the same labora-
entiation347,348 and was upregulated in human obesity/ tory presented a convincing argument that acute intrave-
insulin resistance.349,350 As a pluripotent cytokine, the nous injection of apelin had a powerful effect on lowering
mechanisms by which TNF-α might decrease insulin plasma glucose by enhancing glucose utilization in skel-
action and affect adipocyte functioning are numerous. etal muscle and adipose tissue.365 Therefore, apelin will
As one example, TNF-α suppresses adipocyte-specific probably soon be considered as an important target for
genes with NF-κB being an obligatory signaling inter- the treatment of obesity-related insulin resistance.
mediate55 and decreases the expression of transcription
factors necessary for adipocyte differentiation.347 In an The Adipocyte as a Target for the Treatment of Obesity
autocrine-paracrine fashion, TNF-α blocks further energy and Type 2 Diabetes
accumulation in adipocytes through “deactivation” of the The brain currently serves as the main therapeutic target
insulin-signaling pathway (i.e., insulin resistance),55,351 for the treatment of obesity. Given the central role of the
increased lipolysis, and decreased lipid uptake.352 TNF-α adipocyte in the regulation of body weight and energy
may be a homeostatic mechanism that may prevent fur- metabolism, the adipocyte should not be discarded as
ther fat deposition by regulating LPL activity and leptin a target. Classic adipocyte biology, emphasizing the
production.353 adrenergic signaling systems, provided the rationale for
TNF-α has been termed an adipostat because its adi- the development of beta-3 adrenoreceptor agonists as a
pose tissue expression is, like leptin, more or less propor- means to increase energy expenditure in muscle and pro-
tional to the degree of adiposity. TNF-α has also been mote lipolysis in adipose tissue. These efforts have been
proposed to link obesity with insulin resistance, with ser- hampered by the failure of the drug discovery systems to
ine phosphorylation of the insulin receptor substrate-1 identify “clean” beta-3–selective agonists.366 Alternate
being a prominent mechanism for TNF-α–induced insu- lipolytic systems such as the activation of the growth
lin resistance.354 TNF-α increases 11β HSD1 mRNA and hormone receptor, blockade of the antilipolytic alpha2
enzyme activity and therefore local cortisol production in adrenergic receptor, or activation of the recently discov-
human adipocytes,355 thus potentially linking TNF-α to ered natriuretic peptide signaling pathway may provide
visceral adiposity. The mechanism by which transcription alternate strategies for increasing adipose tissue lipolysis.
is upregulated during energy excess is not entirely clear. Increasing lipolysis and lipid delivery to peripheral tissue
Insulin upregulates TNF-α mRNA, and TZDs appear to will produce weight loss only in the presence of increased
downregulate TNF-α,356 whereas environmental toxins energy expenditure or fat oxidation in liver and skeletal
(such as TCCD) upregulate TNF-α.357 muscle or a reduction in energy intake. The concern with
Clinically, higher plasma levels of TNF-α are also asso- the lipolytic approach is whether the increased lipid sup-
ciated with insulin resistance, higher BMI, higher fasting ply to liver and skeletal muscle will produce or exacerbate
glucose levels, and higher LDL-cholesterol levels.358 Using ectopic fat and insulin resistance.
confirmatory factor analysis and structural equation mod- Another avenue to impact insulin action might be
eling, it was shown that obesity, dyslipidemia, and TNF-α to increase lipid storage in adipose tissue by recruit-
were the principal explanatory variables for the various ing “new” adipocytes from preadipocytes. At first, this
components of the metabolic syndrome.359 TNF-α has approach might be counterintuitive since body weight
also been implicated in HIV-associated lipodystrophy.360 is likely to increase. The effectiveness of the antidiabetic
TZDs in animals and humans provide a strong rationale
Apelin for this approach. In humans and animals, TZDs increase
Apelin is a novel bioactive peptide first identified in 1998 lipid storage in adipose tissue, decrease FFA prior to an
as the endogenous ligand of the orphan G protein–cou- improvement in insulin action, and reduce hepatic and
pled receptor (GPR) APJ.361 A 77–amino acid precursor skeletal muscle fat. Such data suggest that sequestration
can be cleaved into a 55–amino acid fragment and then of lipid in newly recruited or existing adipocytes away
into shorter, less-characterized forms. The physiologically from liver and muscle might be an effective therapeu-
active form of apelin is thought to be apelin-36, although tic strategy. Unfortunately, the acceptability by patients
shorter C-terminal sequences also elicit biological activ- of the weight gain that occurs in this setting makes this
ity.361 It was only 10 years later that a putative role for approach less attractive.
apelin in the etiology of obesity was described. Rayalam Studies suggest that angiogenesis might precede and
and co-workers first described that apelin’s major role was drive adipogenesis.367 This is logical that adipose tissue
to promote angiogenesis in adipose tissue.362 At the same might need nutrients and oxygen to develop properly.
time, Kunduzova and associates proposed that the angio- However, the opposite concept, namely that anti-angio-
genic response to apelin in adipose tissue—­endothelial genic agents might prevent weight gain or result in weight
cells migration, proliferation, and capillary f­ormation— loss, has been demonstrated in animals367 and, along with
was dose-dependent.363 Furthermore, hypoxia upreg- targeting adipocytes with a lytic peptide,368 holds promise
ulates the expression of apelin in adipocytes, thus as a way to modulate adipose tissue mass and function.369
36 ROLE OF THE ADIPOCYTE IN METABOLISM AND ENDOCRINE FUNCTION 647

The secretion of potent endocrine hormones from conceptually permanent. Studies in vivo in humans suggest
adipose tissue is another approach to treat insulin resis- that adipocytes are constantly being formed and undergo
tance or obesity. As an example, antidiabetic TZDs apoptosis.119,128 These high turnover rates for adipose
upregulate the expression of the insulin-sensitizing hor- tissue suggest that a reduction in adipocyte recruitment
mone adiponectin and increase blood concentrations up and/or an increase in adipocyte apoptosis might lead to
to threefold.370 Given that body weight is regulated by a reduction in adipocyte mass. As noted for the lipolytic
leptin-dependent and leptin-independent signals, and adi- pathways, if the excess energy is not completely oxidized,
pose tissue communicates with the brain to regulate food then a possible outcome might be accumulation of lipid in
intake, additional therapeutic targets and therapeutic skeletal muscle and liver as occurs in lipodystrophy syn-
opportunities are likely. dromes. This approach would make sense in the setting
Exogenous cortisol administration or overproduction of weight loss achieved by other means as a way to reduce
of cortisol in conditions such as Cushing’s disease increase the number of lipid-storing small adipocytes.
the accumulation of lipid in visceral depots.371,372 The Another approach to reverse excess lipid supply to
local production of cortisol within adipose tissue by con- the liver and skeletal muscle is to modulate lipolysis.
version of cortisone to bioactive cortisol by the enzyme Lipolysis is disordered in obesity and type 2 diabetes
11 beta HSD-1 also increases the accumulation of lipid where insulin fails to fully suppress lipolysis. Decreased
in visceral depots.373-375 Acting through both adipogenic lipolysis associated with the decrease in FFA that occurs
and lipogenic pathways, blockade of the local production with daytime meals might improve insulin action.
of cortisol is likely to reduce visceral adipose tissue mass, Indeed, reducing lipolysis for only 7 days improved
and therapeutic agents are currently in clinical studies. insulin action in vivo.104,180 Preserving the increase in
Beta-3 receptor agonists not only increase energy lipolysis during sleep and exercise might be necessary
expenditure in rodents but also increase the number of to normalize the meal-related and circadian pattern of
brown adipocytes.376-377 It has been suggested that this lipolysis.
occurs as a result of trans-differentiation of lipid-storing In addition to exercise, which improves blood flow and
white adipocytes into energy-consuming brown adipo- insulin sensitivity in adipose tissue, it might be possible
cytes. The hallmark of brown adipocytes is the expres- to pharmacologically manipulate lipolysis. Nicotinic acid
sion of the thermogenic uncoupling protein UCP-1. In therapy is a well-established treatment for hypertriglyc-
rodents, the conversion of WAT into BAT is under genetic eridemia and low HDL cholesterol, and the discovery of
control, and the resulting weight loss improves the fea- a nicotinic acid receptor (HM74) opens the door for the
tures of metabolic syndrome. Overexpression of the development of small-molecule inhibitors of lipolysis.380
transcriptional factor enhancer PGC-1 in human adipo- Other previously “orphan” GPRs such as the antilipolytic
cytes in vitro also increases UCP-1 mRNA and protein GPR43 are ripe as targets to improve insulin sensitivity
and serves as an example of how this might also occur via suppression of lipolysis.381 GPR43 belongs to a sub-
in humans in vivo.178 Increased energy expenditure and family of GPRs that include GPR40 and GPR41 and are
fat oxidation would result in a decrease in body weight, all fatty acid receptors. Given the modulation of lipolysis
although the potential mechanisms remain elusive.378,379 across the day, the rational design of these drugs should
The findings of discrete brown fat depots uncovered by consider dosing and PK in an attempt to mimic the nor-
PET technologies187 should stimulate a renewed effort to mal variation in FFA.
find strategies to induce more brown adipocytes. As noted   
earlier, peptide hormones such as natriuretic peptides, • F
 or your free Expert Consult eBook with biblio-
orexin, meteorin, irisin, and FGF-21 are attractive targets. graphic citations as well as the ability to take notes,
Lastly, as discussed earlier, once precursor cells are highlight important content, search the full text, and
recruited to differentiate into white adipocytes, they are more, visit http://www.ExpertConsult.Inkling.com.  
REFERENCES 25. Shulman GI. Cellular mechanisms of insulin resistance. J Clin In-
1. Hall KD. What is the required energy deficit per unit weight loss? vest. 2000;(2106):171–176.
Int J Obes (Lond). 2008;32(3):573–576. 26. Cortright RN, Azevedo Jr JL, Zhou Q, et al. Protein kinase C
2. Zhang Y, Proenca R, Maffei M, et al. Positional cloning of the modulates insulin action in human skeletal muscle. Am J Physiol
mouse obese gene and its human homologue. [Published erratum Endocrinol Metab. 2000;(3278):E553–E562.
appears in Nature 1995;374(6521):479] [see comments] Nature. 27. Itani SI, Zhou Q, Pories WJ, et al. Involvement of protein kinase C
1994;372(6505):425–432. in human skeletal muscle insulin resistance and obesity. Diabetes.
3. Deng Y, Scherer PE. Adipokines as novel biomarkers and regulators 2000;(849):1353–1358.
of the metabolic syndrome. Ann N Y Acad Sci. 2010;1212:E1–E19. 28. Schmitz-Peiffer C, Browne CL, Oakes ND, et al. Alterations in the
4. Harris MI, Flegal KM, Cowie CC, et al. Prevalence of diabe- expression and cellular localization of protein kinase C isozymes
tes, impaired fasting glucose, and impaired glucose tolerance epsilon and theta are associated with insulin resistance in skeletal
in U.S. adults. The Third National Health and Nutrition Ex- muscle of the high-fat-fed rat. Diabetes. 1997;(246):169–178.
amination Survey, 1988-1994 [see comments]. Diabetes Care. 29. Schmitz-Peiffer C, Oakes ND, Browne CL, et al. Reversal of
1998;(421):518–524. chronic alterations of skeletal muscle protein kinase C from fat-fed
5. Knowler WC, Pettitt DJ, Savage PJ, Bennett PH. Diabetes inci- rats by BRL-49653. Am J Physiol. 1997;(5 Pt 1273):E915–E921.
dence in Pima Indians: contributions of obesity and parental dia- 30. Koves TR, Ussher JR, Noland RC, et al. Mitochondrial overload
betes. Am J Epidemiol. 1981;(2113):144–156. and incomplete fatty acid oxidation contribute to skeletal muscle
6. Olefsky JM. Lilly lecture 1980. Insulin resistance and in- insulin resistance. Cell Metab. 2008;7(1):45–56.
sulin action. An in vitro and in vivo perspective. Diabetes. 31. Kissebah AH, Krakower GR. Regional adiposity and morbidity.
1981;(230):148–162. Physiol Rev. 1994;(474):761–811.
7. Reaven GM. Banting lecture 1988. Role of insulin resistance in 32. Abate N, Garg A, Peshock RM, et al. Relationships of generalized
human disease. Diabetes. 1988;(1237):1595–1607. and regional adiposity to insulin sensitivity in men. J Clin Invest.
8. Martin BC, Warram JH, Krolewski AS, et al. Role of glucose 1995;(196):88–98.
and insulin resistance in development of type 2 diabetes melli- 33. Goodpaster BH, Thaete FL, Simoneau JA, Kelley DE. Sub-
tus: results of a 25-year follow-up study [see comments]. Lancet. cutaneous abdominal fat and thigh muscle composition pre-
1992;(8825340):925–929. dict insulin sensitivity independently of visceral fat. Diabetes.
9. Lillioja S, Mott DM, Spraul M, et al. Insulin resistance and insulin 1997;(1046):1579–1585.
secretory dysfunction as precursors of non-insulin-dependent dia- 34. Smith SR, Lovejoy JC, Greenway F, et al. Contributions of total
betes mellitus. Prospective studies of Pima Indians. N Engl J Med. body fat, abdominal subcutaneous adipose tissue compartments,
1993;27329:1988-1992. and visceral adipose tissue to the metabolic complications of obe-
10. Boden G, Chen X, Ruiz J, White JV, Rossetti L. Mechanisms of sity. Metabolism. 2001;(450):425–435.
fatty acid-induced inhibition of glucose uptake. J Clin Invest. 35. Abate N, Garg A, Peshock RM, et al. Relationship of generalized
1994;(693):2438–2446. and regional adiposity to insulin sensitivity in men with NIDDM.
11. Kelley DE, Mokan M, Simoneau JA, Mandarino LJ. Interaction Diabetes. 1996;(1245):1684–1693.
between glucose and free fatty acid metabolism in human skeletal 36. Marcus MA, Murphy L, Pi-Sunyer FX, Albu JB. Insulin sensitiv-
muscle. J Clin Invest. 1993;(192):91–98. ity and serum triglyceride level in obese white and black women:
12. Roden M, Price TB, Perseghin G, et al. Mechanism of free fat- relationship to visceral and truncal subcutaneous fat. Metabolism.
ty acid-induced insulin resistance in humans. J Clin Invest. 1999;(248):194–199.
1996;(1297):2859–2865. 37. Albu JB, Kovera AJ, Johnson JA. Fat distribution and health in
13. Boden G, Chen X. Effects of fat on glucose uptake and utiliza- obesity. Ann N Y Acad Sci. 2000;(904):491–501.
tion in patients with non-insulin-dependent diabetes. J Clin Invest. 38. Azuma K, Heilbronn LK, Albu JB, et al. Adipose tissue distribu-
1995;(396):1261–1268. tion in relation to insulin resistance in type 2 diabetes mellitus. Am
14. Randle PJ, Garland PB, Hales CN, et al. The glucose fatty acid J Physiol Endocrinol Metab. 2007;293(1):E435–E442.
cycle: its role in insulin sensitivity and metabolic disturbances of 39. Goodpaster BH, Kelley DE, Thaete FL, et al. Skeletal muscle at-
diabetes mellitus. Lancet. 1963;(i):7285–7289. tenuation determined by computed tomography is associated
15. Randle PJ, Garland PB, Newsholme EA, Hales CN. The glucose with skeletal muscle lipid content. J Appl Physiol. 2000;89(1):
fatty acid cycle in obesity and maturity onset diabetes mellitus. 104–110.
Ann N Y Acad Sci. 1965;(1131):324–333. 40. Goodpaster BH, Thaete FL, Kelley DE. Thigh adipose tissue dis-
16. McGarry JD. What if Minkowski had been ageusic? An alterna- tribution is associated with insulin resistance in obesity and in
tive angle on diabetes. Science. 1992;258(5083):766–770. type 2 diabetes mellitus. Am J Clin Nutr. 2000;71(4):885–892.
17. Wolfe RR. Metabolic interactions between glucose and fatty acids 41. Gallagher D, Kuznia P, Heshka S, Albu J, et al. Adipose tissue in
in humans. Am J Clin Nutr. 1998;(3 Suppl 67):519S–526S. muscle: a novel depot similar in size to visceral adipose tissue. Am
18. Baba H, Zhang XJ, Wolfe RR. Glycerol gluconeogenesis in fasting J Clin Nutr. 2005;81(4):903–910.
humans. Nutrition. 1995;11(2):149–153. 42. Yim JE, Heshka S, Albu JB, et al. Femoral-gluteal subcutaneous
19. Kelley DE, Goodpaster B, Wing RR, Simoneau JA. Skeletal muscle and intermuscular adipose tissues have independent and oppos-
fatty acid metabolism in association with insulin resistance, obesity, ing relationships with CVD risk. J Appl Physiol. 2008;104(3):
and weight loss. Am J Physiol. 1999;(6 Pt 1 277):E1130–E1141. 700–707.
20. Kelley DE, Mandarino LJ. Fuel selection in human skeletal mus- 43. Weisberg SP, McCann D, Desai M, et al. Obesity is associated
cle in insulin resistance: a reexamination. Diabetes. 2000;(549): with macrophage accumulation in adipose tissue. J Clin Invest.
677–683. 2003;112(12):1796–1808.
21. Galgani JE, Heilbronn LK, Azuma K, et al. Metabolic flexibil- 44. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat
ity in response to glucose is not impaired in people with type plays a crucial role in the development of obesity-related insulin
2 diabetes after controlling for glucose disposal rate. Diabetes. resistance. J Clin Invest. 2003;112(12):1821–1830.
2008;57(4):841–845. 45. Bruun JM, Pedersen SB, Richelsen B. Regulation of Interleukin 8
22. Petersen KF, Dufour S, Befroy D, et al. Impaired mitochondrial production and gene expression in human adipose tissue in vitro.
activity in the insulin-resistant offspring of patients with type 2 J Clin Endocrinol Metab. 2001;86(3):1267–1273.
diabetes. N Engl J Med. 2004;350(7):664–671. 46. Bruun JM, Pedersen SB, Richelsen B. Interleukin-8 production
23. Ukropcova B, Sereda O, de Jonge L, et al. Family history of diabe- in human adipose tissue. Inhibitory effects of anti-diabetic com-
tes links impaired substrate switching and reduced mitochondrial pounds, the thiazolidinedione ciglitazone and the biguanide met-
content in skeletal muscle. Diabetes. 2007;56(3):720–727. formin. Horm Metab Res. 2000;32(11-12):537–541.
24. Petersen KF, Befroy D, Dufour S, et al. Mitochondrial dysfunc- 47. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein
tion in the elderly: possible role in insulin resistance. Science. 1 in obesity and insulin resistance. Proc Natl Acad Sci U S A.
2003;(5622300):1140–1142. 2003;100(12):7265–7270.

647.e1
647.e2 REFERENCES

48. Yuan M, Konstantopoulos N, Lee J, et al. Reversal of obesity- and 73. Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose
diet-induced insulin resistance with salicylates or targeted disrup- tissue dysfunction. Cell Metab. 2013;18(4):470–477.
tion of Ikkbeta. Science. 2001;293(5535):1673–1677. 74. Pasarica M, Sereda OR, Redman LM, et al. Reduced adipose tis-
49. Lee JY, Plakidas A, Lee WH, et al. Differential modulation of sue oxygenation in human obesity: evidence for rarefaction, mac-
Toll-like receptors by fatty acids: preferential inhibition by n-3 rophage chemotaxis, and inflammation without an angiogenic
polyunsaturated fatty acids. J Lipid Res. 2003;44(3):479–486. response. Diabetes. 2009;58(3):718–725.
50. Hundal RS, Petersen KF, Mayerson AB, et al. Mechanism by 75. Divoux A, Tordjman J, Lacasa D, et al. Fibrosis in human adipose
which high-dose aspirin improves glucose metabolism in type 2 tissue: composition, distribution, and link with lipid metabolism
diabetes. J Clin Invest. 2002;109(10):1321–1326. and fat mass loss. Diabetes. 2010;59(11):2817–2825.
51. Goldfine AB, Fonseca V, Jablonski KA, et al. The effects of sal- 76. Tam CS, Tordjman J, Divoux A, et al. Adipose tissue remodel-
salate on glycemic control in patients with type 2 diabetes: a ran- ing in children: the link between collagen deposition and age-
domized trial. Ann Intern Med. 2010;152(6):346–357. related adipocyte growth. J Clin Endocrinol Metab. 2012;97(4):
52. Goldfine AB, Buck JS, Desouza C, et al. Targeting inflamma- 1320–1327.
tion using salsalate in patients with type 2 diabetes: effects 77. Park J, Scherer PE. Endotrophin—a novel factor linking obesity
on flow-mediated dilation (TINSAL-FMD). Diabetes Care. with aggressive tumor growth. Oncotarget. 2012;3(12):1487–1488.
2013;36(12):4132–4139. 78. Park J, Scherer PE. Adipocyte-derived endotrophin promotes
53. Kopp E, Ghosh S. Inhibition of NF-kappa B by sodium salicylate malignant tumor progression. J Clin Invest. 2012;122(11):
and aspirin. Science. 1994;265(5174):956–959. 4243–4256.
54. Shoelson SE, Lee J, Yuan M. Inflammation and the IKK beta/I kap- 79. Sun K, Park J, Gupta OT, et al. Endotrophin triggers adipose tissue
pa B/NF-kappa B axis in obesity- and diet-induced insulin resis- fibrosis and metabolic dysfunction. Nat Commun. 2014;5:3485.
tance. Int J Obes Relat Metab Disord. 2003;27(Suppl 3):S49–S52. 80. Garg A. Lipodystrophies. Am J Med. 2000;108(2):143–152.
55. Ruan H, Miles PD, Ladd CM, et al. Profiling gene transcription 81. Agarwal AK, Garg A. Genetic disorders of adipose tissue devel-
in vivo reveals adipose tissue as an immediate target of tumor ne- opment, differentiation, and death. Annu Rev Genomics Hum
crosis factor-alpha: implications for insulin resistance. Diabetes. Genet. 2006;7:175–199.
2002;51(11):3176–3188. 82. Agarwal AK, Garg A. Genetic basis of lipodystrophies and
56. Wellen KE, Hotamisligil GS. Obesity-induced inflammatory management of metabolic complications. Annu Rev Med.
changes in adipose tissue. J Clin Invest. 2003;112(12):1785–1788. 2006;57:297–311.
57. Ruan H, Pownall HJ, Lodish HF. Troglitazone antagonizes tumor 83. Simha V, Garg A. Lipodystrophy: lessons in lipid and energy me-
necrosis factor-alpha-induced reprogramming of adipocyte gene tabolism. Curr Opin Lipidol. 2006;17(2):162–169.
expression by inhibiting the transcriptional regulatory functions 84. Robbins DC, Danforth Jr E, Horton ES, et al. The effect of
of NF-kappaB. J Biol Chem. 2003;278(30):28181–28192. diet on thermogenesis in acquired lipodystrophy. Metabolism.
58. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 1979;(928):908–916.
2006;444(7121):860–867. 85. Robbins DC, Horton ES, Tulp O, Sims EA. Familial partial lipo-
59. Trayhurn P, Wang B, Wood IS. Hypoxia and the endocrine and dystrophy: complications of obesity in the non-obese? Metabo-
signalling role of white adipose tissue. Arch Physiol Biochem. lism. 1982;(531):445–452.
2008;114(4):267–276. 86. Reitman ML, Mason MM, Moitra J, et al. Transgenic mice lack-
60. Ye J. Emerging role of adipose tissue hypoxia in obesity and insu- ing white fat: models for understanding human lipoatrophic dia-
lin resistance. Int J Obes (Lond). 2008;33(1):54–66. betes. Ann N Y Acad Sci. 1999;(892):289–296.
61. Wang B, Wood IS, Trayhurn P. Dysregulation of the expression 87. Shimomura I, Hammer RE, Ikemoto S, et al. Leptin reverses insu-
and secretion of inflammation-related adipokines by hypoxia in lin resistance and diabetes mellitus in mice with congenital lipo-
human adipocytes. Pflugers Arch. 2007;455(3):479–492. dystrophy. Nature. 1999;(6748401):73–76.
62. Bogacka I, Xie H, Bray GA, Smith SR. Pioglitazone induces mi- 88. Kim JK, Gavrilova O, Chen Y, et al. Mechanism of insulin resistance
tochondrial biogenesis in human subcutaneous adipose tissue in A-ZIP/F-1 fatless mice. J Biol Chem. 2000;(12275):8456–8460.
in vivo. Diabetes. 2005;54(5):1392–1399. 89. Gavrilova O, Marcus-Samuels B, Graham D, et al. Surgical im-
63. Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for plantation of adipose tissue reverses diabetes in lipoatrophic mice.
chronic inflammation and adiponectin reduction in adipose tissue J Clin Invest. 2000;(3105):271–278.
of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab. 90. Colombo C, Cutson JJ, Yamauchi T, et al. Transplantation of
2007;293(4):E1118–E1128. adipose tissue lacking leptin is unable to reverse the metabolic
64. Yin J, Gao Z, He Q, et al. Role of hypoxia in obesity-induced abnormalities associated with lipoatrophy. Diabetes. 2002;51(9):
disorders of glucose and lipid metabolism in adipose tissue. 2727–2733.
Am J Physiol Endocrinol Metab. 2009;296(2):E333–E342. 91. Ebihara K, Kusakabe T, Hirata M, et al. Efficacy and safety of
65. Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines leptin-replacement therapy and possible mechanisms of leptin ac-
macrophage localization and function in adipose tissue of obese tions in patients with generalized lipodystrophy. J Clin Endocrinol
mice and humans. J Lipid Res. 2005;46(11):2347–2355. Metab. 2007;92(2):532–541.
66. Pasarica M, Sereda OR, Redman LM, et al. Reduced Adipose 92. Javor ED, Ghany MG, Cochran EK, et al. Leptin reverses non-
tissue oxygenation in human obesity—evidence for rarefaction, alcoholic steatohepatitis in patients with severe lipodystrophy.
macrophage chemotaxis and inflammation without an angiogenic Hepatology. 2005;41(4):753–760.
response. Diabetes. 2009;58(3):718–725. 93. Park JY, Javor ED, Cochran EK, et al. Long-term efficacy of leptin
67. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and replacement in patients with Dunnigan-type familial partial lipo-
obesity. J Clin Invest. 2011;121(6):2094–2101. dystrophy. Metabolism. 2007;56(4):508–516.
68. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic retic- 94. Prescribing information for recombinant leptin in the treatment
ulum stress in diabetes mellitus. Endocr Rev. 2008;29(1):42–61. of lipodystrophy. 2014. Available at http://www1.astrazeneca-
69. Gething MJ, Sambrook J. Protein folding in the cell. Nature. us.com/pi/pi_myalept.pdf.
1992;355(6355):33–45. 95. Weber RV, Buckley MC, Fried SK, Kral JG. Subcutaneous lipecto-
70. Laybutt DR, Preston AM, Akerfeldt MC, et al. Endoplasmic re- my causes a metabolic syndrome in hamsters. Am J Physiol Regul
ticulum stress contributes to beta cell apoptosis in type 2 diabetes. Integr Comp Physiol. 2000;279(3):R936–R943.
Diabetologia. 2007;50(4):752–763. 96. Ryysy L, Hakkinen AM, Goto T, et al. Hepatic fat content and
71. Hosogai N, Fukuhara A, Oshima K, et al. Adipose tissue hypoxia insulin action on free fatty acids and glucose metabolism rather
in obesity and its impact on adipocytokine dysregulation. Diabe- than insulin absorption are associated with insulin requirements
tes. 2007;56(4):901–911. during insulin therapy in type 2 diabetic patients. Diabetes.
72. Bashan N, Dorfman K, Tarnovscki T, et al. Mitogen-activated 2000;(549):749–758.
protein kinases, inhibitory-kappaB kinase, and insulin signaling 97. Goodpaster BH, Thaete FL, Kelley DE. Thigh adipose tissue dis-
in human omental versus subcutaneous adipose tissue in obesity. tribution is associated with insulin resistance in obesity and in
Endocrinology. 2007;148(6):2955–2962. type 2 diabetes mellitus. Am J Clin Nutr. 2000;(471):885–892.
REFERENCES 647.e3

98. McGarry JD. Banting Lecture 2001: Dysregulation of fatty 122. Spalding KL, Arner E, Westermark PO, et al. Dynamics of fat cell
acid metabolism in the etiology of type 2 diabetes. Diabetes. turnover in humans. Nature. 2008;453(7196):783–787.
2002;(151):7–18. 123. Hager A. Adipose cell size and number in relation to obesity. Post-
99. Danforth Jr E. Failure of adipocyte differentiation causes type II grad Med J. 1977;53(Suppl 2):101–110.
diabetes mellitus? Nat Genet. 2000;24(4):377–380. 124. Hager A, Sjostrom L, Arvidsson B, et al. Body fat and adipose
100. Weyer C, Foley JE, Bogardus C, et al. Enlarged subcutane- tissue cellularity in infants: a longitudinal study. Metabolism.
ous abdominal adipocyte size, but not obesity itself, predicts 1977;26(6):607–614.
type II diabetes independent of insulin resistance. Diabetologia. 125. Soriguer Escofet FJ, Esteva de Antonio I, Tinahones FJ, Pareja
2000;43(12):1498–1506. A. Adipose tissue fatty acids and size and number of fat cells
101. Adams M, Montague CT, Prins JB, et al. Activators of peroxi- from birth to 9 years of age—a cross-sectional study in 96 boys.
some proliferator-activated receptor gamma have depot-specific ­Metabolism. 1996;45(11):1395–1401.
effects on human preadipocyte differentiation. J Clin Invest. 126. Hauner H, Entenmann G, Wabitsch M, et al. Promoting effect of
1997;(12100):3149–3153. glucocorticoids on the differentiation of human adipocyte precur-
102. Smith SR, Xie H, Baghian S, et al. Pioglitazone changes the sor cells cultured in a chemically defined medium. J Clin Invest.
distribution of adipocyte size in Type 2 diabetics. Adipocytes. 1989;84(5):1663–1670.
2006;2(1):11–22. 127. van Harmelen V, Skurk T, Rohrig K, et al. Effect of BMI and
103. Akazawa S, Sun F, Ito M, et al. Efficacy of troglitazone on body age on adipose tissue cellularity and differentiation capac-
fat distribution in type 2 diabetes. Diabetes Care. 2000;(823): ity in women. Int J Obes Relat Metab Disord. 2003;27(8):
1067–1071. 889–895.
104. Bajaj M, Suraamornkul S, Kashyap S, et al. Sustained reduction 128. Pilyugin SS, Ganusov VV, Murali-Krishna K, et al. The rescaling
in plasma free fatty acid concentration improves insulin action method for quantifying the turnover of cell populations. J Theor
without altering plasma adipocytokine levels in subjects with Biol. 2003;225(2):275–283.
strong family history of type 2 diabetes. J Clin Endocrinol Metab. 129. Marques BG, Hausman DB, Martin RJ. Association of fat cell
2004;89(9):4649–4655. size and paracrine growth factors in development of hyperplastic
105. Bajaj M, Suraamornkul S, Pratipanawatr T, et al. Pioglitazone re- obesity. Am J Physiol. 1998;275(6 Pt 2):R1898–R1908.
duces hepatic fat content and augments splanchnic glucose uptake 130. Johnson PR, Stern JS, Greenwood MR, et al. Effect of early nutri-
in patients with type 2 diabetes. Diabetes. 2003;52(6):1364–1370. tion on adipose cellularity and pancreatic insulin release in the
106. Qi N, Kazdova L, Zidek V, et al. Pharmacogenetic evidence that Zucker rat. J Nutr. 1973;103(5):738–743.
cd36 is a key determinant of the metabolic effects of pioglitazone. 131. Salans LB, Cushman SW, Weismann RE. Studies of human adi-
J Biol Chem. 2002;277(50):48501–48507. pose tissue. Adipose cell size and number in nonobese and obese
107. Deleted in page proofs. patients. J Clin Invest. 1973;52(4):929–941.
108. Boden G, Cheung P, Mozzoli M, Fried SK. Effect of thiazolidin- 132. Sjostrom L, Bjorntorp P. Body composition and adipose cellular-
ediones on glucose and fatty acid metabolism in patients with type ity in human obesity. Acta Med Scand. 1974;195(3):201–211.
2 diabetes. Metabolism. 2003;52(6):753–759. 133. Ravelli GP, Stein ZA, Susser MW. Obesity in young men af-
109. Diano S, Liu ZW, Jeong JK, et al. Peroxisome proliferation-associ- ter famine exposure in utero and early infancy. N Engl J Med.
ated control of reactive oxygen species sets melanocortin tone and 1976;295(7):349–353.
feeding in diet-induced obesity. Nat Med. 2011;17(9):1121–1127. 134. Kirtland J, Gurr MI. Adipose tissue cellularity: a review. 2.
110. Lu M, Sarruf DA, Talukdar S, et al. Brain PPAR-gamma promotes The relationship between cellularity and obesity. Int J Obes.
obesity and is required for the insulin-sensitizing effect of thiazoli- 1979;3(1):15–55.
dinediones. Nat Med. 2011;17(5):618–622. 135. Hirsch J. Obesity: matter over mind? In: Nevins JR, ed. Cerebrum:
111. Sarruf DA, Yu F, Nguyen HT, et al. Expression of peroxisome the Dana Forum on brain science. Vol 5. New York: Dana Press;
proliferator-activated receptor-gamma in key neuronal subsets 2003:7–18.
regulating glucose metabolism and energy homeostasis. Endocri- 136. Rolland-Cachera MF, Deheeger M, Bellisle F, et al. Adiposity
nology. 2009;150(2):707–712. ­rebound in children: a simple indicator for predicting obesity. Am
112. Noppa H, Bengtsson C, Isaksson B, Smith U. Adipose tissue cel- J Clin Nutr. 1984;39(1):129–135.
lularity in adulthood and its relation to childhood obesity. Int J 137. Prokopec M, Bellisle F. Adiposity in Czech children followed from
Obes. 1980;4(3):253–263. 1 month of age to adulthood: analysis of individual BMI patterns.
113. Hirsch J, Batchelor B. Adipose tissue cellularity in human obesity. Ann Hum Biol. 1993;20(6):517–525.
Clin Endocrinol Metab. 1976;5(2):299–311. 138. Siervogel RM, Roche AF, Guo SM, Mukherjee D, Chumlea WC.
114. Stern JS, Batchelor BR, Hollander N, et al. Adipose-cell size and Patterns of change in weight/stature2 from 2 to 18 years: findings
immunoreactive insulin levels in obese and normal-weight adults. from long-term serial data for children in the Fels longitudinal
Lancet. 1972;2(7784):948–951. growth study. Int J Obes. 1991;15(7):479–485.
115. Paolisso G, Tataranni PA, Foley JE, et al. A high concentration 139. Dietz WH. Periods of risk in childhood for the development of adult
of fasting plasma non-esterified fatty acids is a risk factor for the obesity—what do we need to learn? J Nutr. 1997;127(9):1884S–
development of NIDDM. Diabetologia. 1995;(1038):1213–1217. 1886S.
116. Weyer C, Wolford JK, Hanson RL, et al. Subcutaneous abdominal 140. Premkumar A, Chow C, Bhandarkar P, et al. Lipoatrophic-lipo-
adipocyte size, a predictor of type 2 diabetes, is linked to chromo- dystrophic syndromes: the spectrum of findings on MR imaging.
some 1q21–q23 and is associated with a common polymorphism in Am J Roentgenol. 2002;178(2):311–318.
LMNA in Pima Indians. Mol Genet Metab. 2001;72(3):231–238. 141. Gimble J, Guilak F. Adipose-derived adult stem cells: isolation,
117. Hirsch J. Editorial: The adipose-cell hypothesis. N Engl J Med. characterization, and differentiation potential. Cytotherapy.
1976;295(7):389–390. 2003;5(5):362–369.
118. Krotkiewski M, Sjostrom L, Bjorntorp P, et al. Adipose tissue cel- 142. Cartwright MJ, Tchkonia T, Kirkland JL. Aging in adipocytes:
lularity in relation to prognosis for weight reduction. Int J Obes. potential impact of inherent, depot-specific mechanisms. Exp
1977;1(4):395–416. Gerontol. 2007;42(6):463–471.
119. Strawford A, Antelo F, Christiansen M, Hellerstein MK. Adipose 143. Tchkonia T, Giorgadze N, Pirtskhalava T, et al. Fat depot-specific
tissue triglyceride turnover, de novo lipogenesis and cell prolifera- characteristics are retained in strains derived from single human
tion in humans measured with 2H2O. Am J Physiol Endocrinol preadipocytes. Diabetes. 2006;55(9):2571–2578.
Metab. 2004;286(4):E577–E588. 144. Tchkonia T, Lenburg M, Thomou T, et al. Identification of depot-
120. Domingo P, Matias-Guiu X, Pujol RM, et al. Subcutaneous adipo- specific human fat cell progenitors through distinct expression
cyte apoptosis in HIV-1 protease inhibitor-associated lipodystro- profiles and developmental gene patterns. Am J Physiol Endocri-
phy. Aids. 1999;13(16):2261–2267. nol Metab. 2007;292(1):E298–E307.
121. Julien P, Despres JP, Angel A. Scanning electron microscopy of 145. Gesta S, Bluher M, Yamamoto Y, et al. Evidence for a role of
very small fat cells and mature fat cells in human obesity. J Lipid developmental genes in the origin of obesity and body fat distribu-
Res. 1989;30(2):293–299. tion. Proc Natl Acad Sci U S A. 2006;103(17):6676–6681.
647.e4 REFERENCES

146. Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: track- 170. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, et al. PPARal-
ing obesity to its source. Cell. 2007;131(2):242–256. pha and PPARgamma activators direct a distinct tissue-specific
147. Karastergiou K, Fried SK, Xie H, et al. Distinct developmental transcriptional response via a PPRE in the lipoprotein lipase gene.
signatures of human abdominal and gluteal subcutaneous adipose Embo J. 1996;15(19):5336–5348.
tissue depots. J Clin Endocrinol Metab. 2013;98(1):362–371. 171. Picard F, Auwerx J. PPAR(gamma) and glucose homeostasis.
148. Divoux A, Karastergiou K, Xie H, et al. Identification of a novel Annu Rev Nutr. 2002;22:167–197.
lncRNA in gluteal adipose tissue and evidence for its positive 172. Glorian M, Duplus E, Beale EG, et al. A single element in the phos-
effect on preadipocyte differentiation. Obesity (Silver Spring). phoenolpyruvate carboxykinase gene mediates thiazolidinedione
2014;22(8):1781–1785. action specifically in adipocytes. Biochimie. 2001;83(10):933–943.
149. Votruba SB, Jensen MD. Insulin sensitivity and regional fat gain in re- 173. Hamm JK, Park BH, Farmer SR. A role for C/EBPbeta in regu-
sponse to overfeeding. Obesity (Silver Spring). 2011;19(2):269–275. lating peroxisome proliferator-activated receptor gamma activ-
150. Tchoukalova YD, Votruba SB, Tchkonia T, et al. Regional differ- ity during adipogenesis in 3T3-L1 preadipocytes. J Biol Chem.
ences in cellular mechanisms of adipose tissue gain with overfeed- 2001;276(21):18464–18471.
ing. Proc Natl Acad Sci U S A. 2010;107(42):18226–18231. 174. Floyd ZE, Stephens JM. STAT5A promotes adipogenesis in non-
151. Tchoukalova YD, Koutsari C, Votruba SB, et al. Sex- and depot- precursor cells and associates with the glucocorticoid receptor
dependent differences in adipogenesis in normal-weight humans. during adipocyte differentiation. Diabetes. 2003;52(2):308–314.
Obesity (Silver Spring). 2010;18(10):1875–1880. 175. Tontonoz P, Kim JB, Graves RA, Spiegelman BM. ADD1: a novel
152. Mundi MS, Karpyak MV, Koutsari C, et al. Body fat distribu- helix-loop-helix transcription factor associated with adipocyte deter-
tion, adipocyte size, and metabolic characteristics of nondiabetic mination and differentiation. Mol Cell Biol. 1993;13(8):4753–4759.
adults. J Clin Endocrinol Metab. 2010;95(1):67–73. 176. Tong Q, Dalgin G, Xu H, et al. Function of GATA transcrip-
153. Tchkonia T, Tchoukalova YD, Giorgadze N, et al. Abundance tion factors in preadipocyte-adipocyte transition. Science.
of two human preadipocyte subtypes with distinct capacities for 2000;290(5489):134–138.
replication, adipogenesis, and apoptosis varies among fat depots. 177. Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC, Erickson
Am J Physiol Endocrinol Metab. 2005;288(1):E267–E277. RL, MacDougald OA. Inhibition of adipogenesis by Wnt signal-
154. Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mes- ing. Science. 2000;289(5481):950–953.
enchymal stem cells in multiple human organs. Cell Stem Cell. 178. Tiraby C, Tavernier G, Lefort C, et al. Acquirement of brown
2008;3(3):301–313. fat cell features by human white adipocytes. J Biol Chem.
155. Traktuev DO, Merfeld-Clauss S, Li J, et al. A population of mul- 2003;278(35):33370–33376.
tipotent CD34-positive adipose stromal cells share pericyte and 179. Picard F, Gehin M, Annicotte J, et al. SRC-1 and TIF2 control
mesenchymal surface markers, reside in a periendothelial location, energy balance between white and brown adipose tissues. Cell.
and stabilize endothelial networks. Circ Res. 2008;102(1):77–85. 2002;111(7):931–941.
156. Richardson RL, Hausman GJ, Gaskins HR. Effect of transform- 180. Ducharme NA, Bickel PE. Lipid droplets in lipogenesis and lipoly-
ing growth factor-beta on insulin-like growth factor 1- and dexa- sis. Endocrinology. 2008;149(3):942–949.
methasone-induced proliferation and differentiation in primary 181. Londos C, Sztalryd C, Tansey JT, Kimmel AR. Role of PAT pro-
cultures of pig preadipocytes. Acta Anat. 1992;145(4):321–326. teins in lipid metabolism. Biochimie. 2005;87(1):45–49.
157. Banerjee SS, Feinberg MW, Watanabe M, et al. The Kruppel- 182. Wolins NE, Quaynor BK, Skinner JR, et al. OXPAT/PAT-1 is a
like factor KLF2 inhibits peroxisome proliferator-activated PPAR-induced lipid droplet protein that promotes fatty acid utili-
receptor-gamma expression and adipogenesis. J Biol Chem. zation. Diabetes. 2006;55(12):3418–3428.
2003;278(4):2581–2584. 183. Heaton GM, Wagenvoord RJ, Kemp Jr A, Nicholls DG. Brown-
158. Green H, Kehinde O. An established preadipose cell line and its adipose-tissue mitochondria: photoaffinity labelling of the regu-
differentiation in culture. II. Factors affecting the adipose conver- latory site of energy dissipation. Eur J Biochem. 1978;82(2):
sion. Cell. 1975;5(1):19–27. 515–521.
159. Green H, Kehinde O. Spontaneous heritable changes leading to in- 184. Ricquier D, Kader JC. Mitochondrial protein alteration in active
creased adipose conversion in 3T3 cells. Cell. 1976;7(1):105–113. brown fat: a sodium dodecyl sulfate-polyacrylamide gel electropho-
160. Russell TR, Ho R. Conversion of 3T3 fibroblasts into adipose retic study. Biochem Biophys Res Commun. 1976;73(3):577–583.
cells: triggering of differentiation by prostaglandin F2alpha 185. Astrup A, Bulow J, Christensen NJ, Madsen J. Ephedrine-induced
and 1-methyl-3-isobutyl xanthine. Proc Natl Acad Sci U S A. thermogenesis in man: no role for interscapular brown adipose
1976;73(12):4516–4520. tissue. Clin Sci (Lond). 1984;66(2):179–186.
161. Fajas L, Landsberg RL, Huss-Garcia Y, et al. E2Fs regulate adipo- 186. Astrup A, Bulow J, Madsen J, Christensen NJ. Contribution of
cyte differentiation. Dev Cell. 2002;3(1):39–49. BAT and skeletal muscle to thermogenesis induced by ephedrine
162. Janderova L, McNeil M, Murrell AN, et al. Human mesenchymal in man. Am J Physiol. 1985;248(5 Pt 1):E507–E515.
stem cells as an in vitro model for human adipogenesis. Obes Res. 187. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for
2003;11(1):65–74. active brown adipose tissue in adult humans. Am J Physiol Endo-
163. Reusch JE, Colton LA, Klemm DJ. CREB activation induces adi- crinol Metab. 2007;293(2):E444–E452.
pogenesis in 3T3-L1 cells. Mol Cell Biol. 2000;20(3):1008–1020. 188. Tatsumi M, Engles JM, Ishimori T, et al. Intense (18)F-FDG up-
164. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis take in brown fat can be reduced pharmacologically. J Nucl Med.
in fibroblasts by PPAR gamma 2, a lipid-activated transcription 2004;45(7):1189–1193.
factor. Cell. 1994;79(7):1147–1156. 189. Cummings DE, Brandon EP, Planas JV, et al. Genetically lean
165. Yu K, Bayona W, Kallen CB, et al. Differential activation of per- mice result from targeted disruption of the RII beta subunit of
oxisome proliferator-activated receptors by eicosanoids. J Biol protein kinase A. Nature. 1996;382(6592):622–626.
Chem. 1995;270(41):23975–23983. 190. Ghorbani M, Himms-Hagen J. Appearance of brown adipocytes
166. Kliewer SA, Lenhard JM, Willson TM, et al. A prostaglandin J2 in white adipose tissue during CL 316,243-induced reversal of
metabolite binds peroxisome proliferator-activated receptor gamma obesity and diabetes in Zucker fa/fa rats. Int J Obes Relat Metab
and promotes adipocyte differentiation. Cell. 1995;83(5):813–819. Disord. 1997;21(6):465–475.
167. Huang JT, Welch JS, Ricote M, et al. Interleukin-4-dependent 191. Kopecky J, Clarke G, Enerback S, et al. Expression of the mito-
production of PPAR-gamma ligands in macrophages by 12/15-li- chondrial uncoupling protein gene from the aP2 gene promoter
poxygenase. Nature. 1999;400(6742):378–382. prevents genetic obesity. J Clin Invest. 1995;96(6):2914–2923.
168. Wu Z, Xie Y, Morrison RF, et al. PPARgamma induces the insu- 192. Seale P, Bjork B, Yang W, et al. PRDM16 controls a brown fat/
lin-dependent glucose transporter GLUT4 in the absence of C/EB- skeletal muscle switch. Nature. 2008;454(7207):961–967.
Palpha during the conversion of 3T3 fibroblasts into adipocytes. 193. Xue B, Rim JS, Hogan JC, et al. Genetic variability affects the
J Clin Invest. 1998;101(1):22–32. development of brown adipocytes in white fat but not in inter-
169. Baumann CA, Chokshi N, Saltiel AR, Ribon V. Cloning and scapular brown fat. J Lipid Res. 2007;48(1):41–51.
characterization of a functional peroxisome proliferator activator 194. Ricquier D, Nechad M, Mory G. Ultrastructural and biochemical
receptor-gamma-responsive element in the promoter of the CAP characterization of human brown adipose tissue in pheochromo-
gene. J Biol Chem. 2000;275(13):9131–9135. cytoma. J Clin Endocrinol Metab. 1982;54(4):803–807.
REFERENCES 647.e5

195. Tseng YH, Kokkotou E, Schulz TJ, et al. New role of bone mor- 220. Shibata R, Ouchi N, Kihara S, et al. Adiponectin stimulates
phogenetic protein 7 in brown adipogenesis and energy expendi- angiogenesis in response to tissue ischemia through stimula-
ture. Nature. 2008;454(7207):1000–1004. tion of amp-activated protein kinase signaling. J Biol Chem.
196. Bordicchia M, Liu D, Amri EZ, et al. Cardiac natriuretic peptides 2004;279(27):28670–28674.
act via p38 MAPK to induce the brown fat thermogenic program 221. Smith SR. The endocrinology of obesity. In: Bray G, ed. Endocri-
in mouse and human adipocytes. J Clin Invest. 2012;122(3): nology and metabolism clinics of north america. Vol 25. Saun-
1022–1036. ders; 1996:921–942.
197. Moro C, Pillard F, de Glisezinski I, et al. Exercise-induced lipid 222. Lee MJ, Fried SK, Mundt SS, et al. Depot-specific regulation of
mobilization in subcutaneous adipose tissue is mainly related to the conversion of cortisone to cortisol in human adipose tissue.
natriuretic peptides in overweight men. Am J Physiol Endocrinol Obesity (Silver Spring). 2008;16(6):1178–1185.
Metab. 2008;295(2):E505–E513. 223. Marin P, Darin N, Amemiya T, et al. Cortisol secretion in relation
198. Moro C, Polak J, Hejnova J, et al. Atrial natriuretic peptide stimu- to body fat distribution in obese premenopausal women. Metabo-
lates lipid mobilization during repeated bouts of endurance exer- lism. 1992;41(8):882–886.
cise. Am J Physiol Endocrinol Metab. 2006;290(5):E864–E869. 224. Pasquali R, Cantobelli S, Casimirri F, et al. The hypothalamic-pitu-
199. Moro C, Smith SR. Natriuretic peptides: new players in energy itary-adrenal axis in obese women with different patterns of body
homeostasis. Diabetes. 2009;58(12):2726–2728. fat distribution. J Clin Endocrinol Metab. 1993;77(2):341–346.
200. Sellayah D, Bharaj P, Sikder D. Orexin is required for brown 225. Rosmond R, Chagnon YC, Holm G, et al. A glucocorticoid recep-
adipose tissue development, differentiation, and function. Cell tor gene marker is associated with abdominal obesity, leptin, and
Metab. 2011;14(4):478–490. dysregulation of the hypothalamic-pituitary-adrenal axis. Obes
201. Wu J, Spiegelman BM. Irisin ERKs the fat. Diabetes. Res. 2000;8(3):211–218.
2014;63(2):381–383. 226. Galton DJ, Bray GA. Studies on lipolysis in human adipose cells.
202. Spiegelman BM. Banting Lecture 2012: Regulation of adipogen- J Clin Invest. 1967;46(4):621–629.
esis: toward new therapeutics for metabolic disease. Diabetes. 227. Ramsay TG, Chung IB, Czerwinski SM, et al. Tissue IGF-I protein
2013;62(6):1774–1782. and mRNA responses to a single injection of somatotropin. Am J
203. Bostrom P, Wu J, Jedrychowski MP, et al. A PGC1-alpha-depen- Physiol. 1995;269(4 Pt 1):E627–E635.
dent myokine that drives brown-fat-like development of white fat 228. Snel YE, Brummer RJ, Doerga ME, et al. Adipose tissue assessed by
and thermogenesis. Nature. 2012;481(7382):463–468. magnetic resonance imaging in growth hormone-deficient adults:
204. Rao RR, Long JZ, White JP, et al. Meteorin-like is a hormone the effect of growth hormone replacement and a comparison with
that regulates immune-adipose interactions to increase beige fat control subjects. Am J Clin Nutr. 1995;61(6):1290–1294.
thermogenesis. Cell. 2014;157(6):1279–1291. 229. Johannsson G, Marin P, Lonn L, et al. Growth hormone treat-
205. Owen BM, Ding X, Morgan DA, et al. FGF21 acts centrally to ment of abdominally obese men reduces abdominal fat mass,
induce sympathetic nerve activity, energy expenditure, and weight improves glucose and lipoprotein metabolism, and reduces dia-
loss. Cell Metab. 2014. Accessed at http://dx.doi.org/10.1016/j.c stolic blood pressure [see comments]. J Clin Endocrinol Metab.
met.2014.07.012. 1997;82(3):727–734.
206. Lee P, Linderman JD, Smith S, et al. Irisin and FGF21 are cold- 230. Heffernan MA, Jiang WJ, Thorburn AW, Ng FM. Effects of
induced endocrine activators of brown fat function in humans. oral administration of a synthetic fragment of human growth
Cell Metab. 2014;19(2):302–309. hormone on lipid metabolism. Am J Physiol Endocrinol Metab.
207. Lee P, Smith S, Linderman J, et al. Temperature-acclimated brown 2000;279(3):E501–E507.
adipose tissue modulates insulin sensitivity in humans. Diabetes. 231. Rebuffe-Scrive M, Eldh J, Hafstrom LO, Bjorntorp P. Metabolism
2014. Jun 22. pii: DB_140513. of mammary, abdominal, and femoral adipocytes in women be-
208. Chen KY, Brychta RJ, Linderman JD, et al. Brown fat activation fore and after menopause. Metabolism. 1986;35(9):792–797.
mediates cold-induced thermogenesis in adult humans in response 232. Rebuffe-Scrive M, Lonnroth P, Marin P, et al. Regional adipose
to a mild decrease in ambient temperature. J Clin Endocrinol tissue metabolism in men and postmenopausal women. Int J
Metab. 2013;98(7):E1218–E1223. Obes. 1987;11(4):347–355.
209. Ravussin E. The presence and role of brown fat in adult humans. 233. Lindberg UB, Crona N, Silfverstolpe G, et al. Regional adipose
Curr Diab Rep. 2010;10(2):90–92. tissue metabolism in postmenopausal women after treatment with
210. Ravussin E, Kozak LP. Have we entered the brown adipose tissue exogenous sex steroids. Horm Metab Res. 1990;22(6):345–351.
renaissance? Obes Rev. 2009;10(3):265–268. 234. Palin SL, McTernan PG, Anderson LA, et al. 17beta-estradiol and
211. Chen YC, Cypess AM, Chen YC, et al. Measurement of human anti-estrogen ICI: Compound 182,780 regulate expression of lipo-
brown adipose tissue volume and activity using anatomic MR protein lipase and hormone-sensitive lipase in isolated subcutane-
imaging and functional MR imaging. J Nucl Med. 2013;54(9): ous abdominal adipocytes. Metabolism. 2003;52(4):383–388.
1584–1587. 235. Iverius PH, Brunzell JD. Relationship between lipoprotein li-
212. Cypess AM, Haft CR, Laughlin MR, Hu HH. Brown fat in pase activity and plasma sex steroid level in obese women. J Clin
humans: consensus points and experimental guidelines. Cell
­ ­Invest. 1988;82(3):1106–1112.
Metab. 2014;20(3):408–415. 236. Price TM, O’Brien SN, Welter BH, et al. Estrogen regulation of
213. Pasarica M, Zachwieja JJ, Dejonge L, et al. Effect of growth adipose tissue lipoprotein lipase—possible mechanism of body fat
hormone on body composition and visceral adiposity in mid- distribution. Am J Obstet Gynecol. 1998;178(1 Pt 1):101–107.
dle-aged men with visceral obesity. J Clin Endocrinol Metab. 237. Marin P, Krotkiewski M, Bjorntorp P. Androgen treatment of
2007;92(11):4265–4270. middle-aged, obese men: effects on metabolism, muscle and adi-
214. Voros G, Maquoi E, Demeulemeester D, et al. Modulation of pose tissues. Eur J Med. 1992;1(6):329–336.
­angiogenesis during adipose tissue development in murine models 238. Marin P, Oden B, Bjorntorp P. Assimilation and mobilization of
of obesity. Endocrinology. 2005;146(10):4545–4554. triglycerides in subcutaneous abdominal and femoral adipose tis-
215. Neels JG, Thinnes T, Loskutoff DJ. Angiogenesis in an in vivo sue in vivo in men: effects of androgens. J Clin Endocrinol Metab.
model of adipose tissue development. FASEB J. 2004;18(9): 1995;80(1):239–243.
983–985. 239. Anderson LA, McTernan PG, Barnett AH, Kumar S. The effects
216. Greenway FL, Liu Z, Yu Y, et al. An assay to measure angiogen- of androgens and estrogens on preadipocyte proliferation in hu-
esis in human fat tissue. Obes Surg. 2007;17(4):510–515. man adipose tissue: influence of gender and site. J Clin Endocrinol
217. Cao Y. Angiogenesis modulates adipogenesis and obesity. J Clin Metab. 2001;86(10):5045–5051.
Invest. 2007;117(9):2362–2368. 240. Dieudonne MN, Pecquery R, Leneveu MC, Giudicelli Y. Opposite
218. Fukumura D, Ushiyama A, Duda DG, et al. Paracrine regulation effects of androgens and estrogens on adipogenesis in rat preadi-
of angiogenesis and adipocyte differentiation during in vivo adi- pocytes: evidence for sex and site-related specificities and possible
pogenesis. Circ Res. 2003;93(9):e88–e97. involvement of insulin-like growth factor 1 receptor and peroxi-
219. Nishimura S, Manabe I, Nagasaki M, et al. Adipogenesis in obe- some proliferator-activated receptor gamma2. Endocrinology.
sity requires close interplay between differentiating adipocytes, 2000;141(2):649–656.
stromal cells, and blood vessels. Diabetes. 2007;56(6):1517–1526.
647.e6 REFERENCES

241. Price TM, O’Brien SN. Determination of estrogen receptor mes- 264. Granneman JG, Moore HP. Location, location: protein traf-
senger ribonucleic acid (mRNA) and cytochrome P450 aromatase ficking and lipolysis in adipocytes. Trends Endocrinol Metab.
mRNA levels in adipocytes and adipose stromal cells by competi- 2008;19(1):3–9.
tive polymerase chain reaction amplification. J Clin Endocrinol 265. Rubner M. Die Quelle der thierischen Warme. Ztschr Biol. 1894;
Metab. 1993;77(4):1041–1045. (30):73–142.
242. Mizutani T, Nishikawa Y, Adachi H, et al. Identification of es- 266. Adolph E. Urges to eat and drink in rats. Am J Physiol. 1947;
trogen receptor in human adipose tissue and adipocytes. J Clin (151):110–125.
Endocrinol Metab. 1994;78(4):950–954. 267. Hervey GR. Regulation of energy balance. Nature. 1969;
243. Pedersen SB, Fuglsig S, Sjogren P, Richelsen B. Identification (222):629–631.
of steroid receptors in human adipose tissue. Eur J Clin Invest. 268. Brobeck J. Mechanism of the development of obesity in animals
1996;26(12):1051–1056. with hypothalamic lesions. Physiol Rev. 1946;(25):541–559.
244. Pedersen SB, Hansen PS, Lund S, et al. Identification of oestrogen 269. Mayer J. Regulation of energy intake and the body weight: the
receptors and oestrogen receptor mRNA in human adipose tissue. glucostatic theory and the lipostatic hypothesis. Ann N Y Acad
Eur J Clin Invest. 1996;26(4):262–269. Sci. 1955;(163):15–43.
245. Crandall DL, Busler DE, Novak TJ, et al. Identification of es- 270. Kennedy GC. The role of depot fat in the hypothalamic con-
trogen receptor beta RNA in human breast and abdominal trol of food intake in the rat. Proc R Soc Lond B Biol Sci.
subcutaneous adipose tissue. Biochem Biophys Res Commun. 1953;(901140):578–596.
1998;248(3):523–526. 271. Hervey GR. The effects of lesions in the hypothalamus in parabi-
246. Pedersen SB, Bruun JM, Hube F, et al. Demonstration of estrogen otic rats. J Physiol. 1959;(2145):336–352.
receptor subtypes alpha and beta in human adipose tissue: influ- 272. Coleman DL. Effects of parabiosis of obese with diabetes and nor-
ences of adipose cell differentiation and fat depot localization. mal mice. Diabetologia. 1973;9(4):294–298.
Mol Cell Endocrinol. 2001;182(1):27–37. 273. Tartaglia LA, Dembski M, Weng X, et al. Identification and ­expression
247. Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones cloning of a leptin receptor. OB-R. Cell. 1995;(783):1263–1271.
determine sensitivity to central leptin and insulin. Diabetes. 274. Heilbronn LK, Smith SR, Ravussin E. The insulin-sensitizing
2006;55(4):978–987. role of the fat derived hormone adiponectin. Curr Pharm Des.
248. Arner P. Free fatty acids—do they play a central role in type 2 2003;9(17):1411–1418.
diabetes? Diabetes Obes Metab. 2001;(3 Suppl 1):11–19. 275. Rajala MW, Scherer PE. Minireview: The adipocyte—at the
249. Youngstrom TG, Bartness TJ. Catecholaminergic innerva- crossroads of energy homeostasis, inflammation. atherosclerosis.
tion of white adipose tissue in Siberian hamsters. Am J Physiol. ­Endocrinology. 2003;(9144):3765–3773.
1995;268(3 Pt 2):R744–R751. 276. Mohamed-Ali V, Goodrick S, Rawesh A, et al. Subcutaneous adi-
250. Bowers RR, Festuccia WT, Song CK, et al. Sympathetic innervation pose tissue releases interleukin-6, but not tumor necrosis factor-
of white adipose tissue and its regulation of fat cell number. Am J alpha, in vivo. J Clin Endocrinol Metab. 1997;(1282):4196–4200.
Physiol Regul Integr Comp Physiol. 2004;286(6):R1167–R1175. 277. Maffei M, Halaas J, Ravussin E, et al. Leptin levels in human and
251. Giordano A, Song CK, Bowers RR, et al. White adipose tissue rodent: measurement of plasma leptin and ob RNA in obese and
lacks significant vagal innervation and immunohistochemical evi- weight-reduced subjects. Nat Med. 1995;(111):1155–1161.
dence of parasympathetic innervation. Am J Physiol Regul Integr 278. Leibel RL. The role of leptin in the control of body weight. Nutr
Comp Physiol. 2006;291(5):R1243–R1255. Rev. 2002;(10 Pt 260):S15–S19. discussion S68-S84, 85-17.
252. Song CK, Schwartz GJ, Bartness TJ. Anterograde transneu- 279. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in
ronal viral tract tracing reveals central sensory circuits from the neuroendocrine response to fasting. Nature. 1996;(6588382):
white adipose tissue. Am J Physiol Regul Integr Comp Physiol. 250–252.
2009;296(3):R501–R511. 280. Huang L, Li C. Leptin: a multifunctional hormone. Cell Res.
253. Cousin B, Casteilla L, Lafontan M, et al. Local sympathetic de- 2000;(210):81–92.
nervation of white adipose tissue in rats induces preadipocyte pro- 281. Shimokawa I, Higami Y. Leptin and anti-aging action of caloric
liferation without noticeable changes in metabolism. Endocrinol- restriction. J Nutr Health Aging. 2001;5(1):43–48.
ogy. 1993;133(5):2255–2262. 282. Roti E, Minelli R, Salvi M. Thyroid hormone metabolism in obesi-
254. Jones DD, Ramsay TG, Hausman GJ, Martin RJ. Norepinephrine ty. Int J Obes Relat Metab Disord. 2000;(24 Suppl 2):S113–S115.
inhibits rat pre-adipocyte proliferation. Int J Obes Relat Metab 283. Fichter MM, Pirke KM, Holsboer F. Weight loss causes neuroen-
Disord. 1992;16(5):349–354. docrine disturbances: experimental study in healthy starving sub-
255. Hodgson AJ, Abolhasan P, Kubbinga A, Llewellyn-Smith IJ. jects. Psychiatry Res. 1986;(117):61–72.
Sympathetic control of pacemaker adipocytes. Int J Obes. 284. Everitt AV, Seedsman NJ, Jones F. The effects of hypophysectomy
2002;26(Suppl 1). A659. and continuous food restriction, begun at ages 70 and 400 days,
256. Summers SA, Whiteman EL, Birnbaum MJ. Insulin signaling in on collagen aging, proteinuria, incidence of pathology and longev-
the adipocyte. Int J Obes Relat Metab Disord. 2000;24(Suppl ity in the male rat. Mech Ageing Dev. 1980;(212):161–172.
4):S67–S70. 285. Meites J. Evidence that underfeeding acts via the neuroendo-
257. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. crine system to influence aging processes. Prog Clin Biol Res.
2000;106(4):473–481. 1989;(287):169–180.
258. Langin D, Lucas S, Lafontan M. Millennium fat-cell lipoly- 286. Masoro EJ. Food restriction in rodents: an evaluation of its role in
sis ­reveals unsuspected novel tracks. Horm Metab Res. 2000; the study of aging. J Gerontol. 1988;(343):B59–B64.
32(11-12):443–452. 287. Nelson JF. Neuroendocrine involvement in the retardation of ag-
259. Arner P, Hoffstedt J. Adrenoceptor genes in human obesity. ing by food restriction: A hypothesis. In: Yu BP, ed. Modulation of
J Intern Med. 1999;245(6):667–672. aging processes by dietary restriction. Boca Raton, FL: CRC Press;
260. Sengenes C, Bouloumie A, Hauner H, et al. Involvement of a 1994:37–55.
cGMP-dependent pathway in the natriuretic peptide-mediated 288. Kirkwood TB. Evolution of ageing. Nature. 1977;(5635270):
hormone-sensitive lipase phosphorylation in human adipocytes. J 301–304.
Biol Chem. 2003;278(49):48617–48626. 289. Heymsfield SB, Greenberg AS, Fujioka K, et al. Recombinant
261. Moro C, Crampes F, Sengenes C, et al. Atrial natriuretic peptide leptin for weight loss in obese and lean adults: a randomized, con-
contributes to the physiological control of lipid mobilization in trolled, dose-escalation trial. JAMA. 1999;(16282):1568–1575.
humans. FASEB J. 2004;18(7):908–910. 290. Minokoshi Y, Kim Y, Peroni O, et al. Leptin stimulates fatty-acid
262. Lafontan M, Moro C, Berlan M, et al. Control of lipolysis by oxidation by activating AMP-activated protein kinase. Nature.
natriuretic peptides and cyclic GMP. Trends Endocrinol Metab. 2002;(415):339–343.
2008;19(4):130–137. 291. Minokoshi Y, Kahn BB. Role of AMP-activated protein kinase in
263. Zechner R, Kienesberger PC, Haemmerle G, et al. Adipose tri- leptin-induced fatty acid oxidation in muscle. Biochem Soc Trans.
glyceride lipase and the lipolytic catabolism of cellular fat stores. 2003;(Pt 131):196–201.
J Lipid Res. 2009;50(1):3–21.
REFERENCES 647.e7

292. Farooqi IS, Jebb SA, Langmack G, et al. Effects of recombinant 313. Schaffler A, Orso E, Palitzsch KD, et al. The human apM-1,
leptin therapy in a child with congenital leptin deficiency. N Engl an adipocyte-specific gene linked to the family of TNF’s and to
J Med. 1999;(12341):879–884. genes expressed in activated T cells, is mapped to chromosome
293. Rosenbaum M, Murphy EM, Heymsfield SB, et al. Low dose 1q21.3-q23, a susceptibility locus identified for familial com-
leptin administration reverses effects of sustained weight-re- bined hyperlipidaemia (FCH). Biochem Biophys Res Commun.
duction on energy expenditure and circulating concentrations 1999;(2260):416–425.
of thyroid hormones. J Clin Endocrinol Metab. 2002;(587): 314. Hotta K, Funahashi T, Arita Y, et al. Plasma concentrations of
2391–2394. a novel, adipose-specific protein, adiponectin, in type 2 diabetic
294. Petersen KF, Oral EA, Dufour S, et al. Leptin reverses insulin resis- patients. Arterioscler Thromb Vasc Biol. 2000;20(6):1595–1599.
tance and hepatic steatosis in patients with severe lipodystrophy. 315. Arita Y, Kihara S, Ouchi N, et al. Paradoxical decrease of an ad-
J Clin Invest. 2002;(10109):1345–1350. ipose-specific protein, adiponectin, in obesity. Biochem Biophys
295. Ravussin E, Caglayan S, Williamson DA, et al. Effects of human Res Commun. 1999;(1257):79–83.
leptin replacement of food intake and energy metabolism in 3 316. Weyer C, Funahashi T, Tanaka S, et al. Hypoadiponectinemia
leptin-deficient adults (Abstract). Int J Obes. 2002;(S126):S136. in obesity and type 2 diabetes: close association with insu-
296. Roth JD, Roland BL, Cole RL, et al. Leptin responsiveness re- lin resistance and hyperinsulinemia. J Clin Endocrinol Metab.
stored by amylin agonism in diet-induced obesity: evidence 2001;(586):1930–1935.
from nonclinical and clinical studies. Proc Natl Acad Sci U S A. 317. Haque WA, Shimomura I, Matsuzawa Y, Garg A. Serum adipo-
2008;105(20):7257–7262. nectin and leptin levels in patients with lipodystrophies. J Clin
297. Carr A, Samaras K, Thorisdottir A, et al. Diagnosis, prediction, Endocrinol Metab. 2002;87(5):2395.
and natural course of HIV-1 protease-inhibitor-associated lipo- 318. Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin
dystrophy, hyperlipidaemia, and diabetes mellitus: a cohort study receptors that mediate antidiabetic metabolic effects. Nature.
­
[see comments]. Lancet. 1999;353(9170):2093–2099. 2003;(6941423):762–769.
298. Ledru E, Christeff N, Patey O, et al. Alteration of tumor necro- 319. Civitarese AE, Jenkinson CP, Richardson D, et al. Adiponecting
sis factor-alpha T-cell homeostasis following potent antiretrovi- receptors gene expression and insulin sensitivity in non-diabetic
ral therapy: contribution to the development of human immu- Mexican Americans with or without family history of Type 2 dia-
nodeficiency virus-associated lipodystrophy syndrome. Blood. betes. Diabetologia. 2004;47(5):816–820.
2000;95(10):3191–3198. 320. Civitarese AE, Carling S, Heilbronn LK, et al. Calorie restriction
299. Mynarcik DC, McNurlan MA, Steigbigel RT, et al. Association increases muscle mitochondrial biogenesis in healthy humans.
of severe insulin resistance with both loss of limb fat and elevated PLoS Med. 2007;4(3):e76.
serum tumor necrosis factor receptor levels in HIV lipodystrophy. 321. Okamoto Y, Arita Y, Nishida M, et al. An adipocyte-derived plas-
J Acquir Immune Defic Syndr. 2000;25(4):312–321. ma protein, adiponectin, adheres to injured vascular walls. Horm
300. Cao H, Hegele RA. Nuclear lamin A/C R482Q mutation in Cana- Metab Res. 2000;(232):47–50.
dian kindreds with Dunnigan-type familial partial lipodystrophy. 322. Fruebis J, Tsao T-S, Javorschi S, et al. Proteolytic cleavage product
Hum Mol Genet. 2000;9(1):109–112. of 30-kDa adipocyte complement-related protein increases fatty
301. Genschel J, Schmidt HH. Mutations in the LMNA gene encoding acid oxidation in muscle and causes weight loss in mice. PNAS.
lamin A/C. Hum Mutat. 2000;16(6):451–459. 2001;(498):2005–2010.
302. Shackleton S, Lloyd DJ, Jackson SN, et al. LMNA, encod- 323. Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone
ing lamin A/C, is mutated in partial lipodystrophy. Nat Genet. adiponectin reverses insulin resistance associated with both lipoat-
2000;24(2):153–156. rophy and obesity. Nat Med. 2001;(87):941–946.
303. Lloreta J, Domingo P, Pujol RM, et al. Ultrastructural features of 324. Berg AH, Combs TP, Du X, et al. The adipocyte-secreted pro-
highly active antiretroviral therapy-associated partial lipodystro- tein Acrp30 enhances hepatic insulin action. Nat Med. 2001;(87):
phy. Virchows Arch. 2002;441(6):599–604. 947–953.
304. Lee JH, Chan JL, Sourlas E, et al. Recombinant methionyl human 325. Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stim-
leptin therapy in replacement doses improves insulin resistance ulates glucose utilization and fatty-acid oxidation by acti-
and metabolic profile in patients with lipoatrophy and metabolic vating AMP-activated protein kinase. Nat Med. 2002;(118):
syndrome induced by the highly active antiretroviral therapy. 1288–1295.
J Clin Endocrinol Metab. 2006;91(7):2605–2611. 326. Combs TP, Berg AH, Obici S, et al. Endogenous glucose produc-
305. Oral EA, Simha V, Ruiz E, et al. Leptin-replacement therapy for tion is inhibited by the adipose-derived protein Acrp30. J Clin
lipodystrophy. N Engl J Med. 2002;346(8):570–578. ­Invest. 2001;(12108):1875–1881.
306. Maeda K, Okubo K, Shimomura I, et al. cDNA cloning and ex- 327. Winder WW, Hardie DG. AMP-activated protein kinase, a meta-
pression of a novel adipose specific collagen-like factor, apM1 bolic master switch: possible roles in Type 2 diabetes. Am J Physi-
(AdiPose Most abundant Gene transcript 1). Biochem Biophys ol Endocrinol Metab. 1999;(1277):E1–E10.
Res Commun. 1996;(2221):286–289. 328. Yamamoto Y, Hirose H, Saito I, et al. Correlation of the adi-
307. Combs TP, Berg AH, Rajala MW, et al. Sexual differentiation, pocyte-derived protein adiponectin with insulin resistance in-
pregnancy, calorie restriction, and aging affect the adipocyte-spe- dex and serum high-density lipoprotein-cholesterol, indepen-
cific secretory protein adiponectin. Diabetes. 2003;(252):268–276. dent of body mass index, in the Japanese population. Clin Sci.
308. Scherer PE, Williams S, Fogliano M, et al. A novel serum protein 2002;(2103):137–142.
similar to C1q, produced exclusively in adipocytes. J Biol Chem. 329. Lindsay RS, Funahashi T, Hanson RL, et al. Adiponectin and de-
1995;(45270):26746–26749. velopment of type 2 diabetes in the Pima Indian population. Lan-
309. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-spe- cet. 2002;(9326360):57–58.
cific gene dysregulated in obesity. J Biol Chem. 1996;(18271): 330. Yang W-S, Lee W-J, Funahashi T, et al. Weight reduc-
10697–10703. tion increases plasma levels of an adipose-derived anti-in-
310. Nakano Y, Tobe T, ChoiMiura NH, et al. Isolation and charac- flammatory protein, adiponectin. J Clin Endocrinol Metab.
terization of GBP28, a novel gelatin-binding protein purified from 2001;(886):3815–3819.
human plasma. J Biochem (Tokyo). 1996;(4120):803–812. 331. Yu JG, Javorschi S, Hevener AL, et al. The effect of thiazolidin-
311. Pajvani UB, Du X, Combs TP, et al. Structure-function studies ediones on plasma adiponectin levels in normal, obese, and type 2
of the adipocyte-secreted hormone Acrp30/Adiponectin. Impli- diabetic subjects. Diabetes. 2002;(1051):2968–2974.
cations for metabolic regulation and bioactivity. J Biol Chem. 332. Kim JY, van de Wall E, Laplante M, et al. Obesity-associated im-
2003;278(11):9073–9085. provements in metabolic profile through expansion of adipose tis-
312. Shapiro L, Scherer PE. The crystal structure of a complement-1q sue. J Clin Invest. 2007;117(9):2621–2637.
family protein suggests an evolutionary link to tumor necrosis 333. Steppan CM, Bailey ST, Bhat S, et al. The hormone resistin links
­factor. Curr Biol. 1998;(68):335–338. obesity to diabetes. Nature. 2001;409(6818):307–312.
647.e8 REFERENCES

334. Moon B, Kwan JJ, Duddy N, et al. Resistin inhibits glucose 356. McTernan PG, Harte AL, Anderson LA, et al. Insulin and rosi-
uptake in L6 cells independently of changes in insulin signal- glitazone regulation of lipolysis and lipogenesis in human adipose
ing and GLUT4 translocation. Am J Physiol Endocrinol Metab. tissue in vitro. Diabetes. 2002;51(5):1493–1498.
2003;285(1):E106–E115. 357. Kern PA, Dicker-Brown A, Said ST, et al. The stimulation of
335. Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue- tumor necrosis factor and inhibition of glucose transport and li-
specific secretory factor inhibits adipocyte differentiation. J Biol poprotein lipase in adipose cells by 2,3,7,8-tetrachlorodibenzo-p-
Chem. 2001;276(14):11252–11256. dioxin. Metabolism. 2002;51(1):65–68.
336. Holcomb IN, Kabakoff RC, Chan B, et al. FIZZ1, a novel 358. Nilsson J, Jovinge S, Niemann A, et al. Relation between plas-
cysteine-rich secreted protein associated with pulmonary in- ma tumor necrosis factor-alpha and insulin sensitivity in elderly
flammation, defines a new gene family. Embo J. 2000;19(15): men with non-insulin-dependent diabetes mellitus. Arterioscler
4046–4055. Thromb Vasc Biol. 1998;18(8):1199–1202.
337. Banerjee RR, Rangwala SM, Shapiro JS, et al. Regulation of 359. Chan JC, Cheung JC, Stehouwer CD, et al. The central roles of
fasted blood glucose by resistin. Science. 2004;303(5661): obesity-associated dyslipidaemia, endothelial activation and cy-
1195–1198. tokines in the Metabolic Syndrome—an analysis by structural
338. Ahima RS, Lazar MA. Adipokines and the peripheral and neural equation modelling. Int J Obes Relat Metab Disord. 2002;26(7):
control of energy balance. Mol Endocrinol. 2008;22(5):1023–1031. 994–1008.
339. Yannakoulia M, Yiannakouris N, Bluher S, et al. Body fat mass 360. Mynarcik DC, McNurlan MA, Steigbigel RT, et al. Association
and macronutrient intake in relation to circulating soluble leptin of severe insulin resistance with both loss of limb fat and elevated
receptor, free leptin index, adiponectin, and resistin concentra- serum tumor necrosis factor receptor levels in HIV lipodystrophy.
tions in healthy humans. J Clin Endocrinol Metab. 2003;88(4): J Acquir Immune Defic Syndr. 2000;25(4):312–321.
1730–1736. 361. Tatemoto K, Hosoya M, Habata Y, et al. Isolation and charac-
340. Lee S, Batzoglou S. Application of independent component analy- terization of a novel endogenous peptide ligand for the human
sis to microarrays. Genome Biology. 2003;4. R76(11). APJ receptor. Biochem Biophys Res Commun. 1998;251(2):
341. Heilbronn LK, Janderova L, Rood J, et al. Relationship between 471–476.
serum resistin concentrations and insulin resistance in non-obese, 362. Rayalam S, Della-Fera MA, Krieg PA, et al. A putative role for
obese and obese-diabetic subjects. J Clin Endocrinol Metab. apelin in the etiology of obesity. Biochem Biophys Res Commun.
2004;89(4):1844–1848. 2008;368(3):815–819.
342. Savage DB, Sewter CP, Klenk ES, et al. Resistin/Fizz3 expres- 363. Kunduzova O, Alet N, Delesque-Touchard N, et al. Apelin/APJ
sion in relation to obesity and peroxisome proliferator-activat- signaling system: a potential link between adipose tissue and endo-
ed receptor-gamma action in humans. Diabetes. 2001;50(10): thelial angiogenic processes. FASEB J. 2008;22(12):4146–4153.
2199–2202. 364. Mazzucotelli A, Ribet C, Castan-Laurell I, et al. The transcrip-
343. Smith SR, Bai F, Charbonneau C, et al. A promoter genotype and tional co-activator PGC-1alpha up regulates apelin in human and
oxidative stress potentially link resistin to human insulin resis- mouse adipocytes. Regul Pept. 2008;150(1-3):33–37.
tance. Diabetes. 2003;52(7):1611–1618. 365. Dray C, Knauf C, Daviaud D, et al. Apelin stimulates glucose uti-
344. Lazar MA. Resistin- and obesity-associated metabolic diseases. lization in normal and obese insulin-resistant mice. Cell Metab.
Horm Metab Res. 2007;39(10):710–716. 2008;8(5):437–445.
345. Smith AD. Oxford dictionary of biochemistry and molecular biol- 366. Weyer C, Gautier JF, Danforth Jr E. Development of beta 3-ad-
ogy, revised ed. Oxford University Press; 2000. renoceptor agonists for the treatment of obesity and diabetes—an
346. Xu H, Hirosumi J, Uysal KT, et al. Exclusive action of trans- update. Diabetes Metab. 1999;25(1):11–21.
membrane TNF alpha in adipose tissue leads to reduced adipose 367. Rupnick MA, Panigrahy D, Zhang CY, et al. Adipose tissue mass
mass and local but not systemic insulin resistance. Endocrinology. can be regulated through the vasculature. Proc Natl Acad Sci
2002;143(4):1502–1511. U S A. 2002;99(16):10730–10735.
347. Stephens JM, Butts M, Stone R, et al. Regulation of transcrip- 368. Kolonin MG, Saha PK, Chan L, et al. Reversal of obesity by tar-
tion factor mRNA accumulation during 3T3-L1 preadipocyte geted ablation of adipose tissue. Nat Med. 2004;10(6):625–632.
differentiation by antagonists of adipogenesis. Mol Cell Biochem. 369. Kahn CR. Medicine. Can we nip obesity in its vascular bud? Sci-
1993;123(1-2):63–71. ence. 2008;322(5901):542–543.
348. Xing H, Northrop JP, Grove JR, et al. TNF alpha-mediated inhi- 370. Hirose H, Kawai T, Yamamoto Y, et al. Effects of pioglitazone on
bition and reversal of adipocyte differentiation is accompanied by metabolic parameters, body fat distribution, and serum adiponec-
suppressed expression of PPARgamma without effects on Pref-1 tin levels in Japanese male patients with type 2 diabetes. Metabo-
expression. Endocrinology. 1997;138(7):2776–2783. lism. 2002;51(3):314–317.
349. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression 371. Bjorntorp P, Rosmond R. Obesity and cortisol. Nutrition.
of tumor necrosis factor-alpha: direct role in obesity-linked insulin 2000;16(10):924–936.
resistance. Science. 1993;259(5091):87–91. 372. Bjorntorp P, Rosmond R. The metabolic syndrome—a neuroen-
350. Tsigos C, Kyrou I, Chala E, et al. Circulating tumor necrosis fac- docrine disorder? Br J Nutr. 2000;83(Suppl 1):S49–S57.
tor alpha concentrations are higher in abdominal versus periph- 373. Bujalska IJ, Kumar S, Stewart PM. Does central obesity re-
eral obesity. Metabolism. 1999;48(10):1332–1335. flect “Cushing’s disease of the omentum”? Lancet. 1997;349
351. Lofgren P, van Harmelen V, Reynisdottir S, et al. Secretion of tu- (9060):1210–1213.
mor necrosis factor-alpha shows a strong relationship to insulin- 374. Masuzaki H, Paterson J, Shinyama H, et al. A transgenic mod-
stimulated glucose transport in human adipose tissue. Diabetes. el of visceral obesity and the metabolic syndrome. Science.
2000;49(5):688–692. 2001;294(5549):2166–2170.
352. Fried SK, Zechner R. Cachectin/tumor necrosis factor decreases 375. Kotelevtsev Y, Holmes MC, Burchell A, et al. 11beta -Hydroxys-
human adipose tissue lipoprotein lipase mRNA levels, synthesis, teroid dehydrogenase type 1 knockout mice show attenuated
and activity. J Lipid Res. 1989;30(12):1917–1923. glucocorticoid-inducible responses and resist hyperglycemia on
353. Bullo M, Garcia-Lorda P, Peinado-Onsurbe J, et al. TNFalpha obesity or stress. PNAS. 1997;94(26):14924–14929.
expression of subcutaneous adipose tissue in obese and morbid 376. Kozak LP. Genetic studies of brown adipocyte induction. J Nutr.
obese females: relationship to adipocyte LPL activity and leptin 2000;130(12):3132S–3133S.
synthesis. Int J Obes Relat Metab Disord. 2002;26(5):652–658. 377. Guerra C, Koza RA, Yamashita H, et al. Emergence of brown
354. Sykiotis GP, Papavassiliou AG. Serine phosphorylation of insulin adipocytes in white fat in mice is under genetic control. Ef-
receptor substrate-1: a novel target for the reversal of insulin resis- fects on body weight and adiposity. J Clin Invest. 1998;102(2):
tance. Mol Endocrinol. 2001;15(11):1864–1869. 412–420.
355. Friedberg M, Zoumakis E, Hiroi N, et al. Modulation of 11 beta- 378. Tiraby C, Langin D. Conversion from white to brown adipocytes:
hydroxysteroid dehydrogenase type 1 in mature human subcuta- a strategy for the control of fat mass? Trends Endocrinol Metab.
neous adipocytes by hypothalamic messengers. J Clin Endocrinol 2003;14(10):439–441.
Metab. 2003;88(1):385–393.
REFERENCES 647.e9

379. Walczak R, Tontonoz P. Setting fat on fire. Nat Med. 2003;9(11): 381. Ge H, Li X, Weiszmann J, et al. Activation of G protein-cou-
1348–1349. pled receptor 43 in adipocytes leads to inhibition of lipoly-
380. Tunaru S, Kero J, Schaub A, et al. PUMA-G and HM74 are re- sis and suppression of plasma free fatty acids. Endocrinology.
ceptors for nicotinic acid and mediate its anti-lipolytic effect. Nat 2008;149(9):4519–4526.
Med. 2003;9(3):352–355.

You might also like