Download Orphan nuclear receptors: therapeutic opportunities in skeletal muscle

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Fatty acid synthesis wikipedia , lookup

Pharmacometabolomics wikipedia , lookup

Metabolic network modelling wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Ketosis wikipedia , lookup

Biochemical cascade wikipedia , lookup

Lipid signaling wikipedia , lookup

Paracrine signalling wikipedia , lookup

Fatty acid metabolism wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Basal metabolic rate wikipedia , lookup

Metabolism wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Signal transduction wikipedia , lookup

Glyceroneogenesis wikipedia , lookup

Myokine wikipedia , lookup

Transcript
Am J Physiol Cell Physiol 291: C203–C217, 2006;
doi:10.1152/ajpcell.00476.2005.
Invited Review
Orphan nuclear receptors: therapeutic opportunities in skeletal muscle
Aaron G. Smith and George E. O. Muscat
Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
metabolism; cardiovascular disease
CARDIOVASCULAR DISEASE (CVD) represents one of the most
critical global health threats, and accounts for ⬎16.7 million
deaths every year. A cluster of pathologies that constitute CVC
include heart disease, vascular disease, atherosclerosis, myocardial infarction, stroke, and hypertension. Furthermore, the
links between CVD and the ever-increasing obesity epidemic
suggests a grave prognosis for human health in the early
decades of the 21st century. Such grim predictions are supported by the realization that CVD contributed to one-third of
global morbidity in 2001 (3, 4, 140).
Dyslipidemia associated with anomalous levels of the lipid
triad [i.e., elevated levels of triglycerides (TG) and low-density
lipoprotein (LDL) cholesterol, low levels of high-density lipoprotein (HDL) cholesterol], hypertension, obesity, sedentary
lifestyle, diabetes, chronic inflammation, and smoking all constitute important independent risk factors for cardiovascular
disease. Importantly, diet, metabolism (genetic predisposition),
and physical activity directly influence these risk factors (3, 4,
140). Research indicates that ⬃62% of strokes and 49% of
heart attacks are caused by hypertension. High blood cholesterol (⬎5 mmol/l) accounts for ⬃18% of strokes and 50% of
heart disease. Aside from dietary and genetic factors, the
prevalence of cardiovascular disease is exacerbated by modern
Address for reprint requests and other correspondence: A. G. Smith, Institute
for Molecular Bioscience, Univ. of Queensland, St. Lucia 4072, Queensland,
Australia (e-mail: [email protected]).
http://www.ajpcell.org
sedentary lifestyles; current data suggest that ⬃60% of the
world’s population do not meet current physical activity guidelines (3, 4, 140).
Approximately 20% of the global population (up to 1 billion
adults) are deemed overweight and at least 300 million are
clinically obese. Obesity rates have tripled in North America,
Eastern Europe, the Middle East, Australia, and China. About
21% of coronary heart disease globally is attributable to an
elevated body mass index (⬎25) (4, 185). Obesity is the trigger
for the development of hypertension, dyslipidemia, and
diabetes, which ultimately leads to cardiovascular disease.
Furthermore, the increasing prevalence of diabetes in adults
(estimated at ⬃4.5% globally) leads to a 3- and 8-fold
higher coronary heart disease risk in men and women
respectively (185). Currently, 180 million people worldwide
suffer from diabetes, and at least 1 in 10 deaths among
adults between 35 and 64 years old is attributable to diabetes, primarily through the increased risk of cardiovascular
disease (122, 140, 185).
METABOLIC SYNDROME
Metabolic syndrome, or Syndrome X, represents a cluster of
metabolic abnormalities that all constitute known risk factors
for cardiovascular disease. The National Cholesterol Education
Program: Adult Treatment Panel III guidelines define metabolic syndrome as the presence of any three features listed in
Table 1 (1). The increased prevalence of metabolic syndrome
0363-6143/06 $8.00 Copyright © 2006 the American Physiological Society
C203
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
Smith, Aaron G., and George E. O. Muscat. Orphan nuclear receptors:
therapeutic opportunities in skeletal muscle. Am J Physiol Cell Physiol 291:
C203–C217, 2006; doi:10.1152/ajpcell.00476.2005.—Nuclear hormone receptors
(NRs) are ligand-dependent transcription factors that bind DNA and translate
physiological signals into gene regulation. The therapeutic utility of NRs is
underscored by the diversity of drugs created to manage dysfunctional hormone
signaling in the context of reproductive biology, inflammation, dermatology,
cancer, and metabolic disease. For example, drugs that target nuclear receptors
generate over $10 billion in annual sales. Almost two decades ago, gene products
were identified that belonged to the NR superfamily on the basis of DNA and
protein sequence identity. However, the endogenous and synthetic small molecules
that modulate their action were not known, and they were denoted orphan NRs.
Many of the remaining orphan NRs are highly enriched in energy-demanding major
mass tissues, including skeletal muscle, brown and white adipose, brain, liver, and
kidney. This review focuses on recently adopted and orphan NR function in skeletal
muscle, a tissue that accounts for ⬃35% of the total body mass and energy
expenditure, and is a major site of fatty acid and glucose utilization. Moreover, this
lean tissue is involved in cholesterol efflux and secretes that control energy
expenditure and adiposity. Consequently, muscle has a significant role in insulin
sensitivity, the blood lipid profile, and energy balance. Accordingly, skeletal
muscle plays a considerable role in the progression of dyslipidemia, diabetes, and
obesity. These are risk factors for cardiovascular disease, which is the the foremost
cause of global mortality (⬎16.7 million deaths in 2003). Therefore, it is not
surprising that orphan NRs and skeletal muscle are emerging as therapeutic
candidates in the battle against dyslipidemia, diabetes, obesity, and cardiovascular
disease.
Invited Review
C204
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
Table 1. Definitions of metabolic sndrome as outlined
by National Cholesterol Education Program:
Adult Treatment Panel III
Central obesity: waist circumference ⬎102 cm (men) or ⬎88 cm (women)
Impaired fasting glucose (⬎6.1 mmol/l)
Low HDL cholesterol (⬍1.0 mmol/l in men; ⬍1.3 mmol/l in women)
Hypertriglyceridemia (⬎1.7 mmol/l)
Hypertension: blood pressure ⬎135/85 mmHg (or medicated)
HDL, high-density lipoprotein.
SKELETAL MUSCLE CARBOHYDRATE AND
LIPID METABOLISM
Skeletal Muscle Metabolism
The three types of muscle tissues are skeletal, cardiac, and
smooth. Skeletal muscle is involved in movement, posture,
stability, metabolism, heat production, and cold tolerance.
Skeletal muscle fibers are classified by two major functional
characteristics: speed of contraction and the aerobic (oxidative)/anaerobic (glycolytic) production of ATP. The speed of
the fiber reflects how fast the fiber metabolizes ATP. Fast fibers
(also known as type IIb or white fibers) utilize anaerobic
glycolysis, are low in mitochondria and myoglobin, rich in
AJP-Cell Physiol • VOL
Skeletal Muscle Dysfunction in Metabolic Disease
Skeletal muscle insulin resistance comprises perturbations in
both glucose and fatty acid metabolism. It is well established in
the literature that pathophysiological conditions that are associated with impaired insulin resistance, such as type 2 diabetes
and obesity, correlate with elevations in circulating FFAs (15,
147). Considerable evidence obtained in both animal and
human studies have linked accumulation of intra-myocellular
lipids with insulin resistance in obesity and type 2 diabetes (49,
75, 109, 133). Investigations aimed at identifying the mechanisms underlying this association have implied an imbalance
between fatty acid uptake and the rate of fatty acid oxidation.
The direct link between intra-myocellular lipid accumulation
and insulin resistance is supported by the observation that
insulin sensitivity can be restored by treatments that promote
depletion of accumulated triglycerides, such as low-fat diets,
fasting, exercise, and leptin administration (125, 163). The
significance of this is exemplified by NMR spectroscopy studies (86, 157) that measured intramyocellular lipids and found
that levels correlated more closely with insulin resistance that
other commonly used indexes, such as body mass index and
waist-to-hip ratios.
Impaired mitochondrial function that directs fatty acids toward storage, as opposed to oxidation, and reduced oxidative
enzyme activities may in large part contribute to muscle lipid
accumulation in obesity and type 2 diabetes mellitus (76, 77,
165, 166). Moreover, the activity of carnitine palmitoyl transferase 1 (CPT1), a key step in the regulation of muscle fatty
acid oxidation, has been shown to be reduced in insulin
resistant muscle from obese subjects (165). In addition to this,
shunting of fatty acids toward storage in skeletal muscle as
opposed to oxidation triglyceride accumulation in skeletal
muscle of obese and type 2 diabetic subjects was shown to be
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
has arisen primarily as a result of the global obesity epidemic
with peripheral insulin resistance providing the most widely
accepted and unifying feature of the syndrome (40, 162). While
great inroads have been made in the identification of the relationship between the various clinical features of metabolic syndrome,
the underlying pathological mechanisms are not well understood.
Excessive circulating levels of saturated fatty acids is known
to be a major contributing factor to the development of insulin
resistance (40, 162). The development of insulin resistance in
response to increased dietary fatty acid intake is likely to be
exacerbated further by the ensuing inhibition of the antilipolytic effect of insulin action resulting in an increased
release of stored triglycerides into the circulation (84, 157).
Increased circulating fatty acids promotes insulin resistance by
inhibiting various components of insulin signaling pathways
such as protein kinase C in skeletal muscle (84) and insulin
receptor substrate phosphorylation in the liver (155). Increased
fatty acid delivery to the liver increases hepatic triglyceride
synthesis and promotes the production and secretion of ApoB
containing very low-density lipoprotein (VLDL) into the circulation contributing to the dyslipidemic features of Metabolic
Syndrome (97, 98). Defective glucose metabolism arises primarily from reduced ability of insulin to suppress hepatic
glucose production and promote glucose uptake by adipose and
muscle tissue. Subsequent compensatory mechanisms to maintain euglycemia result in the characteristic hyperinsulinemia
associated with metabolic syndrome (40, 157, 162). While
hyperinsulinemia will ultimately have feedback effects on
insulin secretion, prolonged exposure to circulating free fatty
acids (FFAs) are also implicated in perturbations in glucose
stimulated insulin secretion by pancreatic ␤-cells (92). Several
mechanisms have been proposed to underlie pancreatic lipotoxicity in metabolic syndrome (17, 69, 188). More recent
studies (93, 186) have suggested a relationship between elevated proinflammatory cytokines, such as TNF-␣ and the
pathogenesis of metabolic syndrome.
glycogen, and are suited to short-term intense activity. Slow
fibers (also known as type I or red fibers) utilize aerobic
metabolism, are rich in mitochondria and myoglobin, and are
suited to endurance activity. There is an intermediate fast fiber
type IIa, which combines fast twitch capacity with aerobic
fatigue-resistant metabolism and intermediate glycogen levels
(107).
Skeletal muscle is a “metabolically flexible” tissue and has
a paramount role in energy balance; for instance, it is the
primary tissue of insulin-stimulated glucose uptake (disposal),
and storage (4-fold the glycogen content of liver), regulates
ApoA1-dependent cholesterol efflux and strongly influences
metabolism via modulation of circulating and stored lipid flux
(57, 74, 78, 136). In fact, lipid catabolism supplies up to 70%
of the energy requirements in a fasted individual. During the
initial phase of aerobic exercise, skeletal muscle utilizes stored
glycogen; however, as exercise continues, glucose and stored
muscle triglycerides become important energy substrates (57,
148, 149). Prolonged exercise increasingly depends on fatty
acids and lipid mobilization from other tissues. Given the fact
that skeletal muscle can utilize both carbohydrate and lipid
energy substrates after appropriate mechanical and hormonal
cues and considering the relative mass of the tissue it is hardly
surprising that normal (and abnormal) skeletal muscle metabolism has a significant impact on insulin sensitivity, the blood
lipid profile and the pathogenesis of metabolic disease.
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
SKELETAL MUSCLE AS AN ENDOCRINE ORGAN: MYOKINES
Obesity-linked insulin resistance is known to be associated
with a chronic systemic inflammatory response (61, 183). In
recent years, it has become apparent that adipose tissue functions as more than just a reservoir for energy storage in the
form of triglycerides but rather functions as a vital endocrine
organ regulating systemic metabolic homeostasis via controlled storage/release of fatty acids and the secretion of biologically active “adipokines,” such as adiponectin (70), resistin
(169), leptin (106), adipsin (184), and TNF-␣ (62). Similarly,
the potential endocrine function of skeletal muscle is being
recognized with the release of bioactive molecules now termed
“myokines” (42, 132). Identification of these factors offers
support for a skeletal muscle endocrine axis that is potentially
pivotal to metabolic homeostasis.
AJP-Cell Physiol • VOL
Muscle tissue expresses and releases several cytokines into
the circulation, for example, IL-6, -8 and -15. The autocrine
actions of these factors are well documented; however, there is
accumulating evidence that these cytokines exert their effect in
other parts of the body. IL-15 is abundantly expressed in
skeletal muscle, is induced by acute exercise, and has anabolic
effects on skeletal muscle protein dynamics (141). Interestingly, administration of IL-15 in rats demonstrated a significant
reduction in adipose deposition (23). Studies (142) in adipogenic 3T3 cells revealed that despite little or no expression of
IL-15 mRNA, administration of IL-15 inhibited lipogenesis
and stimulated the release of adiponectin. Because skeletal
muscle expresses and secretes high levels of IL-15, such an
observation provides strong support for the hypothesis that a
skeletal muscle-adipose endocrine axis exists that modulates
lean body/adipose composition and insulin sensitivity.
Another cytokine released by skeletal muscle is IL-6 and has
been referred to as the “exercise factor” because its expression
and release was found to increase up to 100-fold during muscle
contraction and is postulated to underlie many of the metabolically beneficial effects of exercise (43). Circulating IL-6
stimulates AMPK activity, a fuel-sensing enzyme that is activated by changes in the energy state of the cell and in response
to adipokines, in adipose and lean tissue (80). IL-6 transcriptional regulation in skeletal muscle is influenced by glycogen
content via the p38 MAPK pathway that results in upregulated
expression in response to exercise-induced glycogen depletion
(26, 27). During exercise, IL-6 appears to play a glucoregulatory role via augmented hepatic glucose production and muscle
glucose uptake (43). IL-6 has been shown to be a potent
modulator of fatty acid metabolism via increased rates of fatty
acid oxidation and reesterification and attenuation of insulinmediated lipogenesis in both human subjects and isolated
rodent muscle samples (19, 173). More recently, IL-6-driven
increases in lipolysis were demonstrated in type 2 diabetic
patients without the induction of adverse side effects, such
as hypertriacylglyceridemia, suggesting a potential therapeutic utility of this cytokine in the treatment of metabolic
syndrome (135). Paradoxically, increased circulating IL-6
has been linked to type 2 diabetes, leading to speculation
that IL-6 may contribute to the induction or pathogenesis of
type 2 diabetes (137, 139, 152). However, firm evidence of
a causative link remains to be demonstrated. Furthermore,
IL-6-deficient transgenic mice are glucose intolerant, have
reduced endurance and energy expenditure, and develop
late-onset obesity (180).
Finally, myostatin, a member of the TGF-␤ superfamily, is
specifically expressed in vertebrate skeletal muscle. Mice lacking myostatin have a dramatic increase in skeletal muscle
growth and significantly reduced fat deposition with age (110,
111). Several mechanisms may account for the effect of myostatin on lipid metabolism, including a direct effect on adipocytes (81) or alternatively, anabolic effects in skeletal muscle
may produce a metabolic shift in energy consumption resulting
in a net reduction in adipose lipid storage. The latter hypothesis
is plausible, given the accumulating evidence that muscle fiber
composition has a favorable effect on the LDL/HDL cholesterol ratio (55) and the accepted tenet that muscle tissue has a
significantly higher metabolic rate than other major tissues.
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
linked to elevated rates of fatty acid transport via increased
sarcolemma fat/cd36 (16). Another hypothesis contends that
hyperinsulinemia may increase skeletal muscle and liver lipid
content due to increased lipogenesis via elevated expression of
SREBP-1C (164). This hypothesis is further supported by the
recent observation that SCD-1 expression and activity is elevated in the skeletal muscle of obese humans contributing to
the abnormal skeletal muscle lipid partitioning seen in these
individuals (63).
Impaired glucose utilization by skeletal muscle in metabolic
disease is also linked to elevated circulating FFAs. As early as
the 1960s, it was noted that FFA oxidation disrupts glucose
catabolism via the inhibition of phosphofructokinase and pyruvate dehydrogenase. In this context, the Randle hypothesis
proposed that FFAs compete with glucose for mitochondrial
oxidation (144). Similarly, studies support the concept that
FFAs inhibit glucose transport and key phosphorylation events
(38, 147). Alternatively, mounting evidence points to direct
effects of increase circulating and intracellular lipid content to
perturbations in insulin signaling pathways (38, 147, 175).
Excess triglyceride in insulin resistant muscle has been shown
to alter insulin signaling via consequent increases in lipid
intermediates, such as fatty-acyl CoA, diacylglycerol, and
ceramides. Activation of protein kinase C by diacylglycerol
inhibits the tyrosine kinase activity of the insulin receptor
leading to reduced insulin action (159). Ceramides activate a
protein phosphatase that dephosphorylates AKT/protein kinase
B, subsequently inhibiting glut4 translocation and glycogen
synthesis (28, 100). Lipid accumulation in skeletal muscle has
also been shown to inhibit phosphorylation of Irs-1, thus
inhibiting the binding and activation of phosphatidylinositol
3-kinase, a pivotal step in the normal insulin signaling cascade
(38, 190).
The metabolic flexibility of skeletal muscle allows it to
utilize both carbohydrate and lipid energy sources given the
appropriate hormonal and mechanical signals (74, 76, 77).
While the precise mechanistic basis underlying abnormal skeletal muscle function in obesity and diabetes remains unclear, it
is obvious that biochemical perturbations in this tissue have a
profound effect on systemic metabolic homeostasis. Therapeutic targeting of the metabolic derangements in skeletal muscle
will undoubtedly promote improvement in systemic metabolic
homeostasis and ultimately attenuate the pathogenesis associated with the metabolic syndrome.
C205
Invited Review
C206
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
NUCLEAR RECEPTOR REGULATION OF CARBOHYDRATE
AND LIPID METABOLISM
NUCLEAR RECEPTOR FUNCTION: GENERAL CONCEPTS
A decade ago, gene products were identified that belonged to
the NR superfamily on the basis of nucleic acid sequence
identity. However, the endogenous molecules which regulate
their activity were unknown and thus they were called orphan
NRs. On the basis of members of the NR superfamily that have
been characterized, the orphans forecast an enormous, yet
unexploited opportunity for the discovery of novel signaling
pathways and therapeutic agents. The potential impact of such
a discovery cannot be overstated, because every known NR has
been implicated in disease (14, 51, 172). All members of the
NR superfamily display a highly conserved structural organization with an amino terminal region AB (that encodes activation function 1; AF-1), followed by the COOH region, which
encodes the DNA binding domain; a linker region D and the
COOH-terminal E region. The DE region encodes the ligand
binding domain (LBD), and a transcriptional domain, denoted
as AF-2 (Fig. 1) (4, 105). Nuclear receptors trans-activate the
expression of target genes via the binding of DNA consensus
elements within regulatory regions as either monomers, homodimers, or heterodimers with the retinoid X receptors. DNA
binding is mediated by a highly conserved zinc finger DNA
binding domain and in the case of heterodimerization consensus elements have been found as direct repeats, inverted repeats,
or palindromic repeats (4, 105). A subgroup of the NR superfamily controls metabolism in an organ-specific manner (45).
The transcriptional activity of nuclear receptors is regulated
by coordinated interaction with nuclear cofactors. As a general
rule, the interaction of receptors with ligands promotes a
Fig. 1. A: schematic representation of nuclear receptor
functional domains. The variable NH2-terminal domain
possesses AF-1 ligand independent activity (A/B), the
conserved DNA binding domain (C), variable linker
region (D), ligand binding and dimerization domain (E)
and the AF-2 ligand-dependent transactivation domain
(F). B: co-regulator binding of nuclear receptors. Many
nuclear receptors are associated with co-repressor proteins in the absence of ligand. Upon ligand binding,
co-repressors are displaced in favor of co-activator
proteins. CoR, coenzyme receptor; CoA, coenzyme A.
AJP-Cell Physiol • VOL
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
The links between diets rich in free fatty acids and sugars
and the increasing prevalence of dyslipidemia, type II diabetes,
obesity, and cardiovascular disease are well established. Furthermore, it is becoming increasingly clear that the metabolic
regualtion of both lipid and carbohydrate homeostasis are
intrinsically connected. A pertinent example of this metabolic
interconnection is the regulation of triglyceride catabolism by
insulin, a signaling molecule known primarily as an essential
regulator of glucose disposal. Insulin regulates triglyceride
catabolism via inhibition of hormone sensitive lipase. The
significance of this regulatory mechanism is highlighted by the
demonstration that lipid accumulation and/or elevated lipoprotein lipase expression in nonadipose tissue (e.g., liver and
muscle) results in insulin resistance (82, 86). Perhaps paradoxically it has become evident that diets rich in unsaturated fatty
acids protect against metabolic disease prompting speculation
that dietary lipids function as signaling molecules that play
vital roles in controlling lipid homeostasis. The search for the
mechanisms by which various lipid molecules regulate lipid
homeostasis and the subsequent targets of such signaling have
become an intense area of investigation in recent years in an
effort to understand and control the increasing incidence of
metabolic disease in modern society.
Insights into the mechanisms regulating such processes have
significantly matured since the discovery that metabolism is, in
part, regulated by the nuclear hormone receptors (NRs) which
function as ligand (hormone)-regulated transcription factors
that bind DNA and control gene expression (14, 45). A ligand
or hormone can be considered any small molecule that can bind
to the receptor and affect its function. For example, steroids
(estrogen, testosterone, etc.), thyroid hormones, retinoids, dietary lipids, oxysterols, fatty and bile acids are signaling
molecules that bind to nuclear receptors and regulate gene
expression thereby controlling endocrine pathways directly
relevant to disease (14, 45). Essentially, NRs function as the
conduit between diet, metabolism (and environmental stimuli)
and gene expression mediating a unique physiological response
in a nutritionally dependent manner.
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
ORPHAN NRs AS THERAPEUTIC TARGETS
The fundamental importance of NRs in human health is
underscored by the gamut of drugs created to treat disorders
associated with dysfunctional hormone signaling ranging from
cancer and diabetes to reproductive, cardiovascular and inflammatory diseases (28a, 51, 52, 172).
The plethora of orphan NRs has been the catalyst for the
development of a new paradigm, called “reverse endocrinology” mediated by functional genomics, high throughput
screening and combinatorial chemistry. Reverse endocrinology
is the process whereby the orphan NR is used to search for a
previously unknown ligand/hormone and signaling pathway
(85). In particular, recent studies on previously denoted orphans [e.g., PPAR-␣/␥/␦, LXR, FXR, CAR, SXR] have uncovered a myriad of new signaling pathways that regulate
metabolism, and drug tolerance, and provided new insights/
treatments into many human diseases (i.e., diabetes, dyslipidemia, obesity, and metabolic syndrome) (45, 50, 90). The
identification of low-affinity endogenous and/or high affinity
synthetic ligands for orphan receptors has created an orphan
receptor subclass often referred to as “adopted orphan” receptors (28a). Low-affinity natural agonists for this class of receptor are commonly dietary lipids and their derivatives.
Drugs targeting nuclear receptors, including the estrogen,
thyroid, and glucocorticoid receptors, and PPAR-␣ and
PPAR-␥ are among the most commonly prescribed drugs on
the market and generate annual revenues in excess of $10
billion (131). Despite the proven therapeutic effectiveness of
these pharmaceuticals many are accompanied by undesirable
side effects prompting intense efforts to identify and develop
new agents that retain the positive attributes of current drugs
while avoiding unwanted side effects. Development of tissue
and gene specific nuclear receptor ligands (selective modulators) is a key focus of such efforts with this concept best
exemplified by the selective estrogen receptor modulators,
such as tamoxifen and raloxifene (50, 68). The identification of
AJP-Cell Physiol • VOL
endogenous and synthetic ligands to orphan NRs has generated
considerable excitement in recent years and highlights the
potential therapeutic opportunities given the wide range of
biological functions of these proteins. A pertinent example of
such promise was realized with the identification that the
anti-diabetic/insulin sensitizing thiazolidinediones (TZDs)
function largely as high-affinity ligands for PPAR-␥, whereas
the lipid lowering fibrate class of drugs signal via PPAR-␣ (91,
94). To date, several synthetic ligands have been identified for
other orphan nuclear receptors and are being intensively studied in laboratories, and in many cases, animal and clinical trials
are underway. While crystal structure analysis of orphan nuclear receptors suggest many will function as true orphan
receptors unable to accommodate molecules within their
LBDs, the identification of small molecules that bind to other
regions of the receptor and modulate coregulator interactions
and receptor function remain attractive avenues to explore (50,
114, 131, 160). In summary, the orphan receptors are key
functional regulators of metabolic homeostasis and provide a
scaffold with enormous scope and opportunity for the discovery of important new therapeutic agents.
REGULATION OF SKELETAL MUSCLE METABOLISM BY
ADOPTED AND ORPHAN NUCLEAR RECEPTORS
As discussed, skeletal muscle is a major mass peripheral
tissue accounts that for ⬎35% of the total body mass and
energy expenditure, and is the major site of fatty acid and
glucose oxidation. Moreover, this lean tissue mediates physical
activity and cholesterol efflux, and secretes myokines (myostatin, IL-6, -8, and -15) that control inflammation, energy
expenditure, and adiposity. Consequently, muscle has a significant role in insulin sensitivity, the blood lipid profile, inflammation, and energy balance, conferring a significant role in the
progression of dyslipidemia, diabetes, and obesity. These are
risk factors for cardiovascular disease, the foremost cause of
global mortality. These risk factors are recognized by the
World Health Organization as being in the top 10 global health
problems and are directly influenced by diet, metabolism, and
lifestyle.
Many of the recently adopted orphan NRs and skeletal
muscle are rapidly emerging as critical therapeutic targets in
the battle against dyslipidemia, diabetes, obesity, and cardiovascular disease. Along these lines it has been demonstrated
that “ligand-dependent” NRs in skeletal muscle enhance insulin-stimulated glucose disposal, lipid catabolism, energy expenditure, cholesterol efflux, and decrease triglycerides (see
below).
Surprisingly, the function of the remaining orphan NRs in
skeletal muscle metabolism has not been examined. Nevertheless, given the data on the orphan NR subgroup, abundant
expression in skeletal muscle, and their potential utility as
therapeutic targets for the treatment of disease, the contribution
of this tissue to orphan NR action requires further investigation. Along these lines, emerging stories from cell culture and
animal models illustrate that the orphan NRs, including estrogen-related receptor (ERR)-␣, retinoic acid-related orphan receptor (ROR)-␣, Rev-erb (␣ and ␤), and Nur77 regulate lipid
absorption, lipolysis, inflammation (I␬B␣), and myokine expression (71, 88, 102, 104, 108).
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
dissociation of co-repressor proteins and a recruitment of
coactivator proteins and associated complexes, due primarily
to structural changes in the LBD, eliciting a transcriptional
response and target gene expression (Fig. 1B). In addition to
ligand-induced activation, the function many nuclear receptors
is modulated by signaling cascades that phosphorylate the
receptor, further controlling activity in a ligand-independent or
ligand-dependent manner (34, 72, 161). In the case of true
orphan receptors, this level of regulation may prove to be
pivotal to the design of small molecules that target the function
of these receptors.
Genetic and pharmacological studies have demonstrated that
the liver X receptors (LXR)-␣ and -␤, farnesoid X receptor,
peroxisome proliferator-activated receptors (PPAR)-␣, -␤/␦,
and -␥, liver receptor homolog-1 and the small heterodimeric
partner regulate lipid, glucose, and energy homeostasis (45).
Concordantly, dysfunctional NR signaling results in dyslipidemia, diabetes, obesity, and inflammation. Recently, additional
orphan NRs have been implicated in the regulation of lipid and
carbohydrate metabolism, for example, Nur77, Rev-erb-␣ and
-␤, that control lipolysis, and lipid absorption. Moreover,
evidence for cross-talk between the ␤-adrenergic and the
Nur77 signaling pathways has appeared in the literature.
C207
Invited Review
C208
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
Clearly, improvements in skeletal muscle lipid and carbohydrate metabolism mediated by nuclear receptor function and
pharmacological activation will yield concomitant improvements in systemic metabolic homeostasis. numerous metabolic
genes regulated by nuclear receptors have been identified
ranging from transport molecules, enzymes involves in lipid
and carbohydrate anabolism/catabolism, lipid and carbohydrate storage, thermogenesis, and signaling pathways. For
brevity, the key genes presented and discussed in this review
are outlined in Table 2 with a brief explanation of their
respective functional roles. This review will focus on the
orphan and adopted nuclear receptors in the regulation of
skeletal muscle lipid and carbohydrate metabolism and the
implications for the treatment of metabolic disease (Fig. 2).
PPAR functions in skeletal muscle are emerging as critical
targets in the battle against obesity (39), metabolic syndrome X
(171), type II diabetes (58), and dyslipidemia (39, 54, 58). For
example PPAR-␣ (116), PPAR-␦ (171) (112, 134, 181), and
PPAR-␥ (58), have been shown to be involved in enhancing
insulin-stimulated glucose disposal rate, decreasing triglycerides, and increasing lipid catabolism, energy expenditure, cholesterol efflux and plasma HDL-C levels. Recently, two studies
that focused on PPAR-␦ action in skeletal muscle cells (39) and
tissue (171) demonstrated that this NR induces genes involved
in cholesterol efflux, lipid catabolism, and energy expenditure.
Moreover, PPAR␦ activation leads to a predominant type I/slow/
oxidative fiber phenotype, and dramatically increased endurance,
increased insulin sensitivity, and resistance to obesity (103, 181).
Table 2. Key metabolic genes discussed in this review as targets of nuclear hormone receptor signaling in skeletal muscle
ABCA1
ACS4
Adiponectin receptors
AMPK
ApoA1
ApoE
CD36
CEBP␣
CPT-1
⌬-6-Desaturase
FAS
FABP3
GLUT-4 and -5
Glycogenin
IRS-1
LPL
MCAD
PDK-2 and -4
PGC1
SCD-1 and -2
SREBP-1c
UCP1, -2 and -3
ATP binding cassette: transporters that transfer cholesterol to the HDL acceptors, i.e., reverse cholesterol efflux.
Acyl-CoA synthetase-4. Enhances uptake of fatty acids by catalyzing their activation to acyl-CoA esters for subsequent use in
catabolic fatty acid oxidation pathways.
Adiponectin receptors 1 and 2. Cell surface membrane receptors for adiponectin that regulate glucose uptake and fatty acid
oxidation
5⬘AMP-activated protein kinase. A fuel-sensing enzyme that responds to cellular stress by regulating carbohydrate and fat
metabolism
The major apolipoprotein of HDL.
Apolipoprotein E: facilitates cholesterol and lipid efflux.
Scavenger receptor involved in the uptake of oxidized LDL.
CCAAT/enhancer-binding protein transcription factor known to be important for adipogenesis.
Carnitine palmitoyl transferase-1. Transfers the long-chain fatty acyl group from CoA to carnitine, the initial reaction of
mitochondrial import of LCFAs and their subsequent oxidation.
Involved in desaturation of fatty acids via introduction of double bonds between defined carbons of the fatty acyl chain.
Fatty acid synthase. Involved in de novo fatty acid production.
Fatty acid translocase, and fatty acid binding protein. Facilitate uptake of LCFAs and LDLs.
Glucose transporters. GLUT4 facilitates glucose uptake in response to insulin stimulation. GLUT5 catalyzes uptake of fructose.
Initiates the synthesis of glycogen, the principal storage form of glucose in skeletal muscle.
Docking protein involved in binding and activating other signal transduction molecules after being phosphorylated by the insulin
receptor kinase.
Lipoprotein lipase. Hydrolysis of lipoprotein triglycerides into free fatty acids and responsible for the uptake of free fatty acids.
Medium chain acyl CoA dehydrogenase, a key enzyme involved in fatty acid oxidation.
Pyruvate dehydrogenase kinases: inhibiting the pyruvate dehydrogenase complex, thereby controlling glucose oxidation and
maintaining pyruvate for gluconeogenesis.
Co-activator of a number of nuclear receptors involved in adaptive thermogenesis.
Stearoyl CoA desaturase-1 and -2. Enzymes associated with adiposity, i.e., storage and esterification of cholesterol, and
responsible for cis saturation of stearoyl and palmitoyl-CoA, converting them to oleate and palmitoleate (monounsaturated fatty
acids of triglycerides).
Sterol regulatory element binding protein-1c, the hierarchical transcriptional activator of lipogenesis.
Uncoupling proteins. Mitochondrial proteins that uncouple metabolic fuel oxidation from ATP synthesis, regulating energy
expenditure.
LDL, low-density lipoprotein; CoA, coenzyme A; LCFA, long-chain fatty acid.
AJP-Cell Physiol • VOL
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
PPARs
PPARs primarily function as systemic lipid sensors that
regulate a variety of homeostatic functions, including metabolism, inflammation and development. Three mammalian
PPAR subtypes, designated PPAR-␣ (NR1C1), PPAR-␥
(NR1C3), and PPAR-␦ (NR1C2) have been identified (12, 41,
90). PPAR activity is regulated by an array of dietary lipids
including saturated and unsaturated fatty acids and their respective metabolites derived through the lipoxygenase and
cyclooxygenase pathways. Furthermore, PPAR activity is targeted by selective synthetic compounds including hypolipidemic fibrates (PPAR-␣), the insulin-sensitizing TZDs
(PPAR-␥) (12, 41, 90), and the antisyndrome X phenoxy-acetic
acid compound GW-501516 currently in clinical trials (PPAR␦). Cell culture and animal studies over the last decade have
revealed discrete functions for the various PPAR subtypes in
the regulation of genes that control lipid and carbohydrate
metabolism in a range of tissues. PPAR-␣ and PPAR-␥ are
predominantly, though not exclusively, expressed in liver and
adipose tissue, respectively, whereas PPAR-␦ expression is
relatively ubiquitous, although it is abundantly expressed in
brain, intestine, skeletal muscle, spleen, macrophages, lung,
fat, and adrenals (12, 41, 90). Until recently, relatively little
was known about the specific function of PPAR-␦. Prostanoids,
produced by the conversion of polyunsaturated fatty acids,
have been proposed as potential endogenous ligands for
PPAR-␦. The development and analysis of a potent and selective PPAR-␦ agonist (GW-501516), a phenoxyacetic acid derivative, has generated intense interest by provided evidenced
for a role of this PPAR subtype in the coordination of glucose
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
C209
tolerance, fatty acid oxidation, and energy expenditure in
adipose and skeletal muscle (128).
PPAR-␣
PPAR-␣ is known to regulate fatty acid oxidation gene
expression in the liver, a function which correlates with the
lipid lowering effect of the fibrate class of drugs (12, 41, 90).
PPAR-␣-specific agonists stimulate mitochondrial ␤-oxidation
in vivo in both liver and muscle via the upregulation of genes
such as mCPT1, medium chain acyl CoA dehydrogenase, and
MTE1 (113, 116, 168). Furthermore, PPAR-␣ activation promotes thermogenesis in skeletal muscle via the induction of
uncoupling proteins-1, -2, and -3 (20, 79). PPAR-␣-null mice
exhibit low rates of ␤-oxidation and lipid accumulation in
hepatic and cardiac tissues (36, 96). However, these animals
exhibited minimal alteration in the expression of PPAR-␣
target genes UCP-3, CPT-1, and PDK4 after a period of
starvation or exertion primarily due to a functional redundancy
with PPAR-␦ (116). Furthermore, while metabolism was significantly impaired in liver and cardiac muscle, in contrast,
skeletal muscle appeared refractory to the deleterious effects of
PPAR-␣ ablation. Myocardial lipid accumulation in PPAR-␣
null mice highlights the importance of PPAR-␣ in mitochondrial fatty acid uptake and oxidation in this tissue (36, 176).
PPAR-␣ plays a critical role in normal cardiac lipid homeostasis and its activity is downregulated during cardiac hypertrophy, resulting in diminished fatty acid oxidation and a reversion to glucose and lactate as primary energy sources (32, 153).
Ultimately, such this metabolic shift results in lipid accumulation and associated lipotoxicity of the myocardium (11).
PPAR-␥
PPAR-␥ is most abundantly expressed in adipose tissue,
intestine and macrophages and studies using rodent cell lines
demonstrate its essential role in normal fat cell development
(150, 151, 154). Mouse PPAR-␥ nulls die in utero due to
AJP-Cell Physiol • VOL
cardiac and placental defects; however, embryonic transfer of
mutant embryos onto wild-type placentas allowed the generation of a single live null animal, which exhibited no detectable
adipose tissue (9). Human patients with loss of function mutations in the PPAR-␥ gene present with a novel syndrome,
including lipodystrophy, insulin resistance, type 2 diabetes,
and hypertension (2, 13, 52, 156, 158). PPAR-␥ has generated
intense interest with the identification that the insulin sensitizing TZDs are high-affinity PPAR-␥ ligands (94). Subsequently
studies aimed at elucidating the functional mechanisms underlying the PPAR-␥ function have demonstrated a pivotal role for
PPAR-␥ in the regulation of systemic lipid and glucose (52,
145, 150, 151). While many studies have focused on the role of
PPAR-␥ in adipogenesis and the regulation of adipocyte metabolic function accumulating evidence suggests PPAR-␥ plays
important roles in the regulation of systemic homeostasis via
activities in other key tissues such as liver and skeletal muscle
(187). While low levels of detectable PPAR-␥ expression in
skeletal muscle had led to the long-held assumption that the
augmentation of insulin sensitivity by TZDs was facilitated
primarily via PPAR-␥ activity in adipose tissue, TZDs have
been shown to exert their insulin-sensitizing effects in skeletal
muscle independently of their activity in adipose tissue (21).
While the activity of PPAR-␥ in adipose tissue undoubtedly
contributes to systemic, and specifically skeletal muscle, metabolic homeostasis via storage of circulating fatty acids and the
release of adipokines (many of which act directly on muscle
cells), numerous studies offer support for a direct role for
PPAR-␥ in muscle cells. Studies to date support the contention
that augmentation of insulin sensitivity in skeletal muscle by
PPAR-␥ arises primarily via activation of FFA oxidation (25,
30). Furthermore, PPAR-␥ can directly activate components of
the insulin signaling pathway in skeletal muscle (3, 73, 83,
174). PPAR-␥ signaling activates expression of a number of
metabolic genes in isolated muscle tissue and muscle cell lines,
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
Fig. 2. Schematic representation of nuclear receptors
involved in skeletal muscle lipid and carbohydrate homeostasis. Studies investigating normal and abnormal
function of several orphan nuclear receptors illustrate
the importance of these receptors in metabolic homeostasis. Specifically, regulation of metabolic homeostasis in skeletal muscle by these receptors may
have profound effects on systemic metabolic balance,
adiposity and the development of metabolic and cardiovascular disease. In this context, these receptors can be
considered a physiological conduit between diet, metabolism, activity, and gene expression. PPAR, peroxisome proliferator activator receptor; LXR, liver X receptor; ERR, estrogen-related receptor, ROR, retinoic
acid-related orphan receptor.
Invited Review
C210
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
including UCP-2, UCP-3, IRS-1, ⌬-6-desaturase, and C/EBP␣
(53, 101, 167, 179) and promotes GLUT4 translocation (189).
More recently, muscle-specific disruption of PPAR-␥ expression in mice resulted in severe insulin resistance with
skeletal muscle remaining refractory to the insulin-sensitizing
effects of TZDs, despite normal TZD responses in liver and
adipose tissue and decreased plasma FFA levels (58). Paradoxically another muscle specific mouse knockout model was
similarly shown to be insulin resistant, despite apparently
normal skeletal muscle insulin sensitivity. In this study, it was
proposed that insulin resistance and increased adiposity arose
from a failure of insulin to suppress hepatic glucose production
(124).
Loss-of-function studies in the mouse have revealed a multitude of biological processes in which PPAR-␦ is active during
development. PPAR-␦-null embryos die at an early stage due
to a placental defect, with the few that survive exhibiting a
lower body weight than wild-type littermates, reduced body fat
mass, skin defects, and abnormal myelination (112, 134).
Paradoxically, this phenotype was absent in an adipocyte
specific PPAR-␦ null model, suggesting a complex regulation
of systemic lipid metabolism, as opposed to specific adipocyte
functions (8).
PPAR-␦ has generated a great deal of interest as a potential
therapeutic target in obesity after the identification of potent
synthetic phenoxy-acetic acid ligands. Treatment of insulinresistant and obese rhesus monkeys and Db/Db mice with
PPAR-␦ agonists lowered fasting insulin levels, raised serum
HDL cholesterol, and lowered fasting triglycerides. Although it
was unclear which tissue was the major target for this activity,
the classification of PPAR-␦ as sensor of dietary triglyceride in
native VLDLs released by lipoprotein lipase activity suggested
skeletal muscle was a potential target tissue (29, 95, 128).
Elevation of HDL cholesterol involved increases in the expression of ABCA1 in macrophages, and skeletal muscle and was
accompanied by an induction of ApoA1-specific cholesterol
efflux (to an even greater extent than agonists for PPAR-␣ or
PPAR-␥). Given the relative mass of skeletal muscle this
observation is entirely consistent with the profound effects of
this drug on systemic HDL-cholesterol (39, 177). Furthermore,
the PPAR-␦ agonist induced the expression of acyl-CoA synthetase (ACS), CPT1, UCP2, and UCP3 in rodent neonatal
cardiomyocytes, which correlated with an increase in fatty acid
oxidation (48). PPAR-␦ has been further implicated in the
regulation of skeletal muscle metabolism with reported modulation of expression of a range of genes involved in lipid
absorption [LPL, fatty acid binding protein 3 (FABP3), ACS4],
lipid catabolism (CPT1, PDK4), and energy expenditure
(UCP2 and -3) in cultured C2C12 muscle cells treated with
PPAR-␦ agonists (39). This study also demonstrated that
PPAR␦ agonists (but not PPAR-␣) primarily regulate the
CPT-1 promoter in skeletal muscle cells. Similarly, microarray
expression profiling in GW-501516-treated rat skeletal muscle
cells (L6 myotubes) and skeletal muscle of treated mice (171)
identified a large panel of genes involved lipid homeostasis
including CPT-1. The authors went on to demonstrate that
GW-501516 treatment of mice stimulates fatty acid oxidation
in skeletal muscle; in contrast, ␤-oxidation was not induced in
AJP-Cell Physiol • VOL
LXRs
Recent evidence suggests the Liver X Receptors (LXR␣ and
LXR␤) regulate lipid metabolism in skeletal muscle. Analysis
of gene expression changes in both rodent quadricep muscle
and cultured skeletal muscle myotubes treated with the synthetic LXR agonist T0901317 revealed an upregulation of key
genes involved in lipid and cholesterol efflux and lipogenesis
such as ABCA1, ApoE and SREBP-1c (119). While LXR
agonists have proven utility in the treatment of hypercholesterolemia their development into an efficacious treatment for
this condition has been impaired by the deleterious side effects
consequential to the activation of lipogenesis. Significantly, the
induction of the SREBP-1c target genes FAS, and SCD-1
involved in lipogenesis was observed in livers but not skeletal
muscle of rodents treated with LXR agonists. This observation
is believed to stem from the fact the LXR␤ is the predominant
isoform expressed in skeletal muscle as opposed to LXR␣ in
liver and prompted speculation that LXR␤ specific agonists
may retain the desired cholesterol lowering activity while
avoiding the undesirable hypertriglyceridaemia. The observation that synthetic LXR agonists increase ABCA1 mediated
cholesterol efflux in skeletal muscle cells underscores the
potential of this tissue as a target of reverse cholesterol transport and HDL cholesterol regulation (119).
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
PPAR-␦
the liver. Starvation induces PPAR␦ mRNA expression in
murine gastrocnemius muscle with concomitant activation of
fatty acid translocase/CD36 (FAT/CD36), muscle carnitine
palmitoyl transferase (M-CPT1 or CPT-1␤) and heart FABP
(59).
PPAR-␦ is predominantly expressed in mitochondrial rich
type I (oxidative, slow twitch) relative to type II (glycolytic,
fast twitch) skeletal muscle. Endurance training promotes conversion into type I muscle, accompanied by an increased
expression of PPAR-␦ (103). Muscle-specific expression of
activated PPAR-␦ in mice leads to an increase in type I muscle
fibers. Concordantly, increased activity of enzymes involved in
oxidative (not glycolytic) metabolism has been reported. Expression analysis also revealed an induction of the genes
involved in increased oxidative metabolism, glucose tolerance,
preferential lipid utilization, energy expenditure (i.e., troponin-I slow, cytochrome c, UCP2, UCP3, and CPT1) (103), and
increased endurance (181).
Numerous studies demonstrated that GW-501516 treatment
and skeletal muscle-specific PPAR-␦ overexpression in mice
ameliorated diet induced obesity, enhanced metabolic rate,
lipid oxidation, reduced intramuscular triglycerides, and increased mitochondria in skeletal muscle. Moreover, anatomical
analysis revealed the resistance to increased body weight was
largely due to reduced mass of visceral and epidermal fat
depots. The drug treatment ameliorated the diet induced 1)
hypertrophy in epidermal white adipose, and brown fat, 2)
hepatic steatosis, and 3) accumulation of intramuscular lipid
droplets (103, 171, 181). Interestingly, these mice have profoundly increased endurance capabilities, and resistance to
fatigue relative to their wild-type littermates. Selective
PPAR-␦ agonists (i.e., GW-501516) are currently in clinical
trials for the treatment of dyslipidemia, insulin resistance, and
obesity.
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
ESTROGEN-RELATED RECEPTOR
RORS AND REV-ERBS
RORs and Rev-erbs belong to the NR1F, and NR1D subgroups subgroups of nuclear hormone receptors, respectively.
These are closely related families of orphan nuclear receptors
that operate in opposing manners. Three ROR genes have been
identified; ROR␣ encodes four alternatively spliced isoforms
(ROR␣1, ROR␣2, ROR␣3, and ROR␣␣), which are predominantly expressed in blood, brain, skeletal muscle, and fat cells.
ROR␤ is expressed specifically in the brain and ROR␥ is found
at high levels in skeletal muscle and thymocytes (67). Two
Rev-erb genes have been identified: Rev-erb␣ and Rev-erb␤.
The Rev-erb␤ gene encodes two alternatively spliced isoforms,
Rev-erb␤1 and Rev-erb␤2. The mRNAs are expressed in most
tissues, although very abundant expression is seen in energydemanding tissues, including skeletal and cardiac muscle,
liver, kidney, and brown fat.
Analysis of the spontaneously generated “staggerer” (sg/sg)
mutant mouse, which carries a loss of function mutation in the
ROR-␣ gene, has been extremely informative on the role of
ROR-␣ in lipid homeostasis and skeletal muscle function. The
most obvious staggerer phenotype is ataxia associated with a
cerebellar atrophy, aberrant vascular physiology, hypersensitive inflammatory response, dyslipidemia, and enhanced susceptibility to atherosclerosis (104). In the context of lipid
homeostasis, the sg/sg mice exhibit a dyslipidemia, characterized by lower circulating plasma levels of HDL-C, ApoA1,
AII, and CIII and plasma triglycerides. Notably, ApoAI is a
AJP-Cell Physiol • VOL
major constituent of HDL, whereas ApoCIII is a component of
HDL and VLDL that plays a role in regulation of triglyceride
levels and lipoprotein lipase activity (146, 178). This observation is supported by cell culture studies demonstrating that
ROR-␣, and Rev-erb-␣ activate and repress the expression of
ApoAI and CIII in cell culture, respectively (18, 66).
Futhermore, sg/sg mice have been shown to have a reduced
muscular strength, a finding supported by earlier work conducted in our laboratory, demonstrating that ROR-␣ potentiates skeletal muscle myogenesis, whereas the Rev-erbs antagonize muscle cell differentiation in culture (22, 37, 87). Ectopic
expression of a dominant negative form of ROR-␣ in skeletal
muscle cells attenuated ROR-dependent gene expression, repressed the endogenous levels of ROR-␣ and ROR-␥ mRNA,
and also resulted in attenuated expression of many genes
involved in lipogenesis and energy homeostasis, including
SREBP-1c, SCD1/2 and FAS, a finding consistent with the
lowered TG levels observed in sg/sg mice (88). Furthermore,
previous studies have demonstrated that MCPT-1 an established target for PPAR-␣ in cardiac muscle and PPAR-␦ in
skeletal muscle cells, is also directly regulated by ROR␣1
raising the suggesting a potential tissue-specific cross-talk
between PPAR-␣ and ROR-␣. Cell culture studies and analysis
of the sg/sg mouse highlights the importance of ROR-␣ in the
regulation of lipid homeostasis, and we hypothesize that
ROR-␣1 in skeletal muscle programs a cascade of gene expression designed to regulate lipid and energy homeostasis in this
tissue (88).
Similarly, mice homozygous (⫺/⫺) for the Rev-erb-␣ null
allele have a dyslipidemic phenotype and a 20% lower body
weight. Male Rev-erb-␣-deficient mice show elevated serum
VLDL triglyceride levels and increased serum and liver expression of apoCIII mRNA (146). Elevated ApoCIII and
VLDL triglycerides in Rev-erb-␣⫺/⫺ is in contrast to reduced
ApoCIII and triglycerides observed in ROR-␣ mutant sg/sg
mice and concurs with the negative regulation of ApoCIII by
Rev-erb-␣. Interestingly, homozygous Rev-erb-␣-null mice
display a significant shift from fast (type IIA) to slow (type I)
myosin heavy chain expression in the slow twitch, mitochondrial rich oxidative fibres which encodes the thick filament of
the sarcomere. This is in concordance with the selective expression of Rev-erb-␣ in type IIB glycolytic and intermediate
type IIA oxidative fibers (138). This suggests that this orphan
NR regulates muscle fiber type, which will have concomitant
effects on insulin sensitivity and aerobic/anaerobic metabolism.
Ectopic expression of a dominant negative mutant Rev-erb␤⌬E (lacking the LBD) in skeletal muscle cells attenuated a
range of genes involved in fatty acid/lipid absorption including
CD36, FABP3, and FABP4 (143). Increased expression of
SREBP-1c and SCD1 was also observed and is consistent with
elevated TG levels in Rev-erb-␣⫺/⫺ mice. Moreover, a dramatic decrease in the myostatin expression and elevated IL-6
expression further highlights a potential role of this orphan
receptor in the regulation of muscle mass, adiposity, and
inflammation (143). Modulation of these myokines coupled
with observed changes in genes controlling muscle lipid absorption and metabolism suggest that Rev-erb-␤ activity plays
and important role in muscle growth and lipid homeostasis
(143).
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
Estrogen-related receptors (␣, ␤, and ␥) are orphan receptors
with close homology to the estrogen receptor that have implicated roles in the regulation of cellular energy metabolism.
Accordingly ERRs are abundantly expressed in tissues with a
high capacity for fatty acid ␤-oxidation (47, 65). Homozygous
knock-out ERR␣⫺/⫺ mice were found to have a significant
reduction in fat mass and a commensurate resistance to high fat
induced obesity (102). This phenotype was attributable to the
altered expression of genes involved in lipid metabolism and
adipocyte development. Somewhat paradoxically, a synthetic
ERR␣ agonist was found to increase energy expenditure and
thus antagonize the development of obesity (71).
ERR regulation of cellular metabolism appears to involve
interactions with the PGC1 co-regulator. PGC1 has been
shown to be a potent co-activator of ERR␣ with a synthetic
inhibitor of ERR␣ significantly attenuating PGC1␣ mediated
regulation of oxidative phosphorylation and mitochondrial respiration in skeletal and cardiac muscle cells (115). ERR␣ has
also been found to activate PPAR␣ in skeletal muscle cells
subsequently regulating an array of genes involved in fatty acid
uptake, fatty acid oxidation, and oxidative phosphorylation
(64). Recent evidence that an ERR␣-dependent PGC1␣ program mediates the induction of mitofusin-2 helps explain the
link between exercise and PGC1␣/ERR␣ mediated modulation
of mitochondrial function (24). Mitofusin-2 is a key regulator
of mitochondrial biogenesis and is pivotal to the increase in
mitochondrial size, content and oxidative capacity in response
to exericse. In light of established links between insulin resistance and impaired mitochondrial function synthetic modulation of ERR function may prove to be an effective therapeutic
target in the treatment of metabolic disease.
C211
Invited Review
C212
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
These studies underscore the potential therapeutic utility of
ROR-␣1, and Rev-erb agonists, that is further highlighted by
genetic and biochemical studies demonstrating the anti-atherogenic and anti-inflammatory properties of ROR-␣, and proinflammatory properties of Rev-erb␣. The mechanism involves
the modulation of the NF-␬B signaling pathway and regulation
of p65 translocation, and of I␬B␣ activity. Moreover, we
propose that the atherogenic and dyslipidemic phenotype of the
sg/sg mouse is related to aberrant ROR␣ function in skeletal
muscle and surmise that ROR␣1 agonists may have therapeutic
utility in the treatment of hypercholesterolemia and atherosclerosis.
NR4A FAMILY
AJP-Cell Physiol • VOL
CHICKEN OVALBUMIN UPSTREAM PROMOTERTRANSCRIPTION FACTOR
The chicken ovalbumin upstream promotor (COUP)-transcription factors (TFs) are most related to the retinoid X
receptor/retinoid acid receptor, and are generally considered to
be repressors of other nuclear receptors, such as PPARs,
retinoic acid, thyroid hormone, and vitamin D receptors (130).
COUP-TFs are involved in the regulation of several fundamental biological processes, such as neurogenesis, organogenesis,
and metabolic homeostasis. COUP-TFs can modulate the activity of genes that are fundamental in the orchestration of
glucose and lipid metabolism, such as bile acid synthesis
(CYP7A-cholesterol 7a-hydroxylase) (31), ketogenesis (mitochondrial HMG-CoA synthase) (56), cholesterol transport
(cholesterol ester transfer gene and apolipoprotein AI) (46,
121), fatty-acid ␤-oxidation (medium-chain acyl CoA dehydrogenase) (35), and glucose homeostasis and insulin sensitivity (10). Moreover, COUP-TF can inhibit preadipocyte differentiation, and its ortholog in Drosophila (svp) is required for
fat cell differentiation (60). Previous studies (7) in muscle cell
cultures have demonstrated a role for COUP-TFs in muscle
development showing COUP-TFII inhibits the expression and
function of MyoD. Despite COUP-TFII involvement in muscle
differentiation and its implication in the regulation of carbohydrate and lipid metabolism in other tissues the specific
contribution of COUP-TFs in skeletal muscle metabolic homeostasis remains unexplored.
In conclusion, it is increasingly obvious that orphan and
adopted nuclear receptors are key regulators of lipid and
carbohydrate homeostasis in skeletal muscle. The efficacy of a
number of pharmacological agents that target a number of
these adopted orphan receptors such as PPAR-␥ (TZD) and
PPAR-␣ (fibrates) underscores the immense potential of orphan nuclear receptors as therapeutic targets in the treatment of
metabolic and cardiovascular disease. The relative mass and
metabolic flexibility of skeletal muscle, and the observed
metabolic and biochemical perturbations in diabetic and obese
individuals, attests to the importance of this tissue in systemic
metabolic homeostasis. For this reason, NRs and skeletal
muscle are valid targets in the battle to ameliorate metabolic
perturbations associated with metabolic syndrome, obesity, and
cardiovascular disease. Despite recent advances in our understanding of NR function in metabolic homeostasis, considerable future effort is required to realise the potential of NR
agonists in the battle against cardiovascular disease.
Dyslipidemia, insulin resistance, blood glucose levels (and
glucose tolerance), diabetes, inflammation, and obesity are all
serious risk factors for cardiovascular and metabolic disease.
NRs and skeletal muscle are validated targets in the battle to
ameliorate metabolic perturbations; however, the real promise
and potential of NR agonists in the battle against cardiovascular disease has not yet been realized.
REFERENCES
1. Adult Treatment Panel III. Executive Summary of The Third Report of
The National Cholesterol Education Program (NCEP) Expert Panel on
Detection, Evaluation, and Treatment of High Blood Cholesterol In
Adults. JAMA 285: 2486 –2497, 2001.
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
The NR4A subgroup of orphan nuclear receptors comprises
three members Nurr1, Nur77, and NOR-1, which largely function as immediate-early genes, whose expression and activation is regulated in a cell-type specific manner in response to a
range of signals, such as mitogenic and apoptotic stimuli.
Nur77 is expressed in a range of tissues, including the thymus,
muscle, lung, liver, testis, ovary, and prostate, and adrenal,
thyroid and pituitary glands (33, 120, 126). NOR-1 is expressed in the pituitary and adrenal glands, cardiac and skeletal
muscle, thymus, and kidney (44, 127, 191), whereas Nurr1 is
expressed in the central nervous system, thymus, liver, and
pituitary gland (89, 117, 118). Crystalographic studies suggest
the NR4A subgroup are perhaps true orphan receptors that
have little space available for ligand interaction within their
respective LBDs, but when expressed, are constitutively active
and highly responsive to various signaling pathways. Despite
the fact that these genes may not afford agonist (or antagonist)
modulation of activity in the classic sense, recent evidence has
demonstrated the potential for the identification of small molecule regulators of these orphan nuclear receptors. The finding
that the purine anti-metabolite 6-mercaptopurine modulated the
activity of NR4A family members in an AF1 dependent manner suggests such molecules may provide a platform for the
pharmacological and ultimately therapeutic exploitation of
these genes (129, 182).
The Nur77 orphan nuclear receptor has recently been linked
to adrenergic control of energy balance/homeostasis. In cultured muscle cells ␤-adrenergic receptor activation by isoprenaline elicited a rapid, striking, and transient upregulation of
Nur77 expression (99, 108). Concordantly, it was found that
small interfering RNA-mediated knockdown of Nur77 expression in this cell model resulted in attenuation of gene expression pathways associated with preferential lipid utilization and
energy balance, such as UCP-3 and AMPK␥3 (108). Similarly,
UCP-3 was found to be downregulated in mouse tibialis
muscle after injection and electro-transfer of siNur77 in concordance with decreased expression of UCP-3 in the C2C12
culture model (108). In addition, attenuation of Nur77 expression resulted in decreased expression of genes involved in
carbohydrate and lipid homeostasis, such as adiponectin receptor 2, CD36 and GLUT4, which correlated with decreased
lypolysis in these cells (108). These data are concordant with
the known effects of adrenergic agonists on glucose uptake
(123) and the links between UCP3 and AMPK in skeletal
muscle cells (170). Increased lipolysis and energy expenditure
upon Nur77 activation makes Nur77 a highly attractive potential therapeutic target in the treatment of obesity.
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
AJP-Cell Physiol • VOL
22. Burke L, Downes M, Carozzi A, Giguere V, and Muscat GE.
Transcriptional repression by the orphan steroid receptor RVR/Rev-erb ␤
is dependent on the signature motif and helix 5 in the E region: functional
evidence for a biological role of RVR in myogenesis. Nucleic Acids Res
24: 3481–3489, 1996.
23. Carbo N, Lopez-Soriano J, Costelli P, Busquets S, Alvarez B, Baccino FM, Quinn LS, Lopez-Soriano FJ, and Argiles JM. Interleukin-15 antagonizes muscle protein waste in tumour-bearing rats. Br J
Cancer 83: 526 –531, 2000.
24. Cartoni R, Leger B, Hock MB, Praz M, Crettenand A, Pich S,
Ziltener JL, Luthi F, Deriaz O, Zorzano A, Gobelet C, Kralli A, and
Russell AP. Mitofusins 1/2 and ERR␣ expression are increased in human
skeletal muscle after physical exercise. J Physiol 567: 349 –358, 2005.
25. Cha BS, Ciaraldi TP, Carter L, Nikoulina SE, Mudaliar S, Mukherjee R, Paterniti JR Jr, and Henry RR. Peroxisome proliferatoractivated receptor (PPAR) ␥ and retinoid X receptor (RXR) agonists have
complementary effects on glucose and lipid metabolism in human skeletal muscle. Diabetologia 44: 444 – 452, 2001.
26. Chan MH, Carey AL, Watt MJ, and Febbraio MA. Cytokine gene
expression in human skeletal muscle during concentric contraction:
evidence that IL-8, like IL-6, is influenced by glycogen availability. Am J
Physiol Regul Integr Comp Physiol 287: R322–R327, 2004.
27. Chan MH, McGee SL, Watt MJ, Hargreaves M, and Febbraio MA.
Altering dietary nutrient intake that reduces glycogen content leads to
phosphorylation of nuclear p38 MAP kinase in human skeletal muscle:
association with IL-6 gene transcription during contraction. FASEB J 18:
1785–1787, 2004.
28. Chavez JA, Knotts TA, Wang LP, Li G, Dobrowsky RT, Florant GL,
and Summers SA. A role for ceramide, but not diacylglycerol, in the
antagonism of insulin signal transduction by saturated fatty acids. J Biol
Chem 278: 10297–10303, 2003.
28a.Chawla A, Repa JJ, Evans RM, and Mangelsdorf DJ. Nuclear
receptors and lipid physiology: opening the X-files. Science 294: 1866 –
1870, 2001.
29. Chawla A, Lee CH, Barak Y, He W, Rosenfeld J, Liao D, Han J,
Kang H, and Evans RM. PPAR␦ is a very low-density lipoprotein
sensor in macrophages. Proc Natl Acad Sci USA 100: 1268 –1273, 2003.
30. Ciaraldi TP, Cha BS, Park KS, Carter L, Mudaliar SR, and Henry
RR. Free fatty acid metabolism in human skeletal muscle is regulated by
PPAR␥ and RXR agonists. Ann NY Acad Sci 967: 66 –70, 2002.
31. Crestani M, Sadeghpour A, Stroup D, Galli G, and Chiang JY.
Transcriptional activation of the cholesterol 7␣-hydroxylase gene
(CYP7A) by nuclear hormone receptors. J Lipid Res 39: 2192–2200,
1998.
32. Davila-Roman VG, Vedala G, Herrero P, de las Fuentes L, Rogers
JG, Kelly DP, and Gropler RJ. Altered myocardial fatty acid and
glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll
Cardiol 40: 271–277, 2002.
33. Davis IJ and Lau LF. Endocrine and neurogenic regulation of the
orphan nuclear receptors Nur77 and Nurr-1 in the adrenal glands. Mol
Cell Biol 14: 3469 –3483, 1994.
34. Diradourian C, Girard J, and Pegorier JP. Phosphorylation of PPARs:
from molecular characterization to physiological relevance. Biochimie
87: 33–38, 2005.
35. Disch DL, Rader TA, Cresci S, Leone TC, Barger PM, Vega R, Wood
PA, and Kelly DP. Transcriptional control of a nuclear gene encoding a
mitochondrial fatty acid oxidation enzyme in transgenic mice: role for
nuclear receptors in cardiac and brown adipose expression. Mol Cell Biol
16: 4043– 4051, 1996.
36. Djouadi F, Weinheimer CJ, Saffitz JE, Pitchford C, Bastin J, Gonzalez FJ, and Kelly DP. A gender-related defect in lipid metabolism and
glucose homeostasis in peroxisome proliferator-activated receptor ␣-deficient mice. J Clin Invest 102: 1083–1091, 1998.
37. Downes M, Carozzi AJ, and Muscat GE. Constitutive expression of the
orphan receptor, Rev-erbA ␣, inhibits muscle differentiation and abrogates the expression of the myoD gene family. Mol Endocrinol 9:
1666 –1678, 1995.
38. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline
GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, Petersen
KF, and Shulman GI. Effects of free fatty acids on glucose transport
and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest
103: 253–259, 1999.
39. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, and Muscat GE.
The peroxisome proliferator-activated receptor ␤/␦ agonist, GW501516,
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
2. Agostini M, Gurnell M, Savage DB, Wood EM, Smith AG, Rajanayagam O, Garnes KT, Levinson SH, Xu HE, Schwabe JW, Willson
TM, O’Rahilly S, and Chatterjee VK. Tyrosine agonists reverse the
molecular defects associated with dominant-negative mutations in human
peroxisome proliferator-activated receptor ␥. Endocrinology 145: 1527–
1538, 2004.
3. American Heart Association. Heart Disease and Stroke Statistics–2004
Update, 2004.
4. American Heart Association. International Cardiovascular Disease
Statistics, 2005.
5. Arakawa K, Ishihara T, Aoto M, Inamasu M, Saito A, and Ikezawa
K. Actions of novel antidiabetic thiazolidinedione, T-174, in animal
models of non-insulin-dependent diabetes mellitus (NIDDM) and in
cultured muscle cells. Br J Pharmacol 125: 429 – 436, 1998.
6. Aranda A and Pascual A. Nuclear hormone receptors and gene expression. Physiol Rev 81: 1269 –1304, 2001.
7. Bailey P, Sartorelli V, Hamamori Y, and Muscat GE. The orphan
nuclear receptor, COUP-TF II, inhibits myogenesis by post-transcriptional regulation of MyoD function: COUP-TF II directly interacts with
p300 and myoD. Nucleic Acids Res 26: 5501–5510, 1998.
8. Barak Y, Liao D, He W, Ong ES, Nelson MC, Olefsky JM, Boland R,
and Evans RM. Effects of peroxisome proliferator-activated receptor
delta on placentation, adiposity, and colorectal cancer. Proc Natl Acad
Sci USA 99: 303–308, 2002.
9. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR,
Koder A, and Evans RM. PPAR gamma is required for placental,
cardiac, and adipose tissue development. Mol Cell 4: 585–595, 1999.
10. Bardoux P, Zhang P, Flamez D, Perilhou A, Lavin TA, Tanti JF,
Hellemans K, Gomas E, Godard C, Andreelli F, Buccheri MA, Kahn
A, Le Marchand-Brustel Y, Burcelin R, Schuit F, and VasseurCognet M. Essential role of chicken ovalbumin upstream promotertranscription factor II in insulin secretion and insulin sensitivity revealed
by conditional gene knockout. Diabetes 54: 1357–1363, 2005.
11. Barger PM and Kelly DP. PPAR signaling in the control of cardiac
energy metabolism. Trends Cardiovasc Med 10: 238 –245, 2000.
12. Barish GD and Evans RM. PPARs and LXRs: atherosclerosis goes
nuclear. Trends Endocrinol Metab 15: 158 –165, 2004.
13. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos
MA, Maslen GL, Williams TD, Lewis H, Schafer AJ, Chatterjee VK,
and O’Rahilly S. Dominant negative mutations in human PPAR␥
associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 402: 880 – 883, 1999.
14. Berkenstam A and Gustafsson JA. Nuclear receptors and their relevance to diseases related to lipid metabolism. Curr Opin Pharmacol 5:
171–176, 2005.
15. Bierman EL, Dole VP, and Roberts TN. An abnormality of nonesterified fatty acid metabolism in diabetes mellitus. Diabetes 6: 475– 479,
1957.
16. Bonen A, Parolin ML, Steinberg GR, Calles-Escandon J, Tandon
NN, Glatz JF, Luiken JJ, Heigenhauser GJ, and Dyck DJ. Triacylglycerol accumulation in human obesity and type 2 diabetes is associated
with increased rates of skeletal muscle fatty acid transport and increased
sarcolemmal FAT/CD36. FASEB J 18: 1144 –1146, 2004.
17. Boucher A, Lu D, Burgess SC, Telemaque-Potts S, Jensen MV,
Mulder H, Wang MY, Unger RH, Sherry AD, and Newgard CB.
Biochemical mechanism of lipid-induced impairment of glucose-stimulated insulin secretion and reversal with a malate analogue. J Biol Chem
279: 27263–27271, 2004.
18. Boukhtouche F, Mariani J, and Tedgui A. The “CholesteROR” protective pathway in the vascular system. Arterioscler Thromb Vasc Biol
24: 637– 643, 2004.
19. Bruce CR and Dyck DJ. Cytokine regulation of skeletal muscle fatty
acid metabolism: effect of interleukin-6 and tumor necrosis factor-␣.
Am J Physiol Endocrinol Metab 287: E616 –E621, 2004.
20. Brun S, Carmona MC, Mampel T, Vinas O, Giralt M, Iglesias R, and
Villarroya F. Activators of peroxisome proliferator-activated receptor-␣
induce the expression of the uncoupling protein-3 gene in skeletal
muscle: a potential mechanism for the lipid intake-dependent activation
of uncoupling protein-3 gene expression at birth. Diabetes 48: 1217–
1222, 1999.
21. Burant CF, Sreenan S, Hirano K, Tai TA, Lohmiller J, Lukens J,
Davidson NO, Ross S, and Graves RA. Troglitazone action is independent of adipose tissue. J Clin Invest 100: 2900 –2908, 1997.
C213
Invited Review
C214
40.
41.
42.
43.
44.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
regulates the expression of genes involved in lipid catabolism and energy
uncoupling in skeletal muscle cells. Mol Endocrinol 17: 2477–2493,
2003.
Eckel RH, Grundy SM, and Zimmet PZ. The metabolic syndrome.
Lancet 365: 1415–1428, 2005.
Evans RM, Barish GD, and Wang YX. PPARs and the complex
journey to obesity. Nat Med 10: 355–361, 2004.
Febbraio MA and Pedersen BK. Contraction-induced myokine production and release: is skeletal muscle an endocrine organ? Exerc Sport Sci
Rev 33: 114 –119, 2005.
Febbraio MA and Pedersen BK. Muscle-derived interleukin-6: mechanisms for activation and possible biological roles. FASEB J 16: 1335–
1347, 2002.
Fernandez PM, Brunel F, Jimenez MA, Saez JM, Cereghini S, and
Zakin MM. Nuclear receptors Nor1 and NGFI-B/Nur77 play similar,
albeit distinct, roles in the hypothalamo-pituitary-adrenal axis. Endocrinology 141: 2392–2400, 2000.
Francis GA, Fayard E, Picard F, and Auwerx J. Nuclear receptors and
the control of metabolism. Annu Rev Physiol 65: 261–311, 2003.
Gaudet F and Ginsburg GS. Transcriptional regulation of the cholesteryl ester transfer protein gene by the orphan nuclear hormone
receptor apolipoprotein AI regulatory protein-1. J Biol Chem 270:
29916 –29922, 1995.
Giguere V, Yang N, Segui P, and Evans RM. Identification of a new
class of steroid hormone receptors. Nature 331: 91–94, 1988.
Gilde AJ, van der Lee KA, Willemsen PH, Chinetti G, van der Leij
FR, van der Vusse GJ, Staels B, and van Bilsen M. Peroxisome
proliferator-activated receptor (PPAR)␣ and PPAR␤/␦, but not PPARgamma, modulate the expression of genes involved in cardiac lipid
metabolism. Circ Res 92: 518 –524, 2003.
Goodpaster BH and Wolf D. Skeletal muscle lipid accumulation in
obesity, insulin resistance, and type 2 diabetes. Pediatr Diabetes 5:
219 –226, 2004.
Gronemeyer H, Gustafsson JA, and Laudet V. Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov 3:
950 –964, 2004.
Gurnell M and Chatterjee VK. Nuclear receptors in disease: thyroid
receptor beta, peroxisome-proliferator-activated receptor gamma and
orphan receptors. Essays Biochem 40: 169 –189, 2004.
Gurnell M, Savage DB, Chatterjee VK, and O’Rahilly S. The metabolic syndrome: peroxisome proliferator-activated receptor ␥ and its
therapeutic modulation. J Clin Endocrinol Metab 88: 2412–2421, 2003.
Hammarstedt A and Smith U. Thiazolidinediones (PPAR␥ ligands)
increase IRS-1, UCP-2 and C/EBP␣ expression, but not transdifferentiation, in L6 muscle cells. Diabetologia 46: 48 –52, 2003.
He W, Barak Y, Hevener A, Olson P, Liao D, Le J, Nelson M, Ong
E, Olefsky JM, and Evans RM. Adipose-specific peroxisome proliferator-activated receptor ␥ knockout causes insulin resistance in fat and
liver but not in muscle. Proc Natl Acad Sci USA 100: 15712–15717,
2003.
Hedman A, Byberg L, Reneland R, and Lithell HO. Muscle morphology, self-reported physical activity and insulin resistance syndrome. Acta
Physiol Scand 175: 325–332, 2002.
Hegardt FG. Transcriptional regulation of mitochondrial HMG-CoA
synthase in the control of ketogenesis. Biochimie 80: 803– 806, 1998.
Henriksson J. Muscle fuel selection: effect of exercise and training.
Proc Nutr Soc 54: 125–138, 1995.
Hevener AL, He W, Barak Y, Le J, Bandyopadhyay G, Olson P,
Wilkes J, Evans RM, and Olefsky J. Muscle-specific Pparg deletion
causes insulin resistance. Nat Med 9: 1491–1497, 2003.
Holst D, Luquet S, Nogueira V, Kristiansen K, Leverve X, and
Grimaldi PA. Nutritional regulation and role of peroxisome proliferatoractivated receptor delta in fatty acid catabolism in skeletal muscle.
Biochim Biophys Acta 1633: 43–50, 2003.
Hoshizaki DK, Blackburn T, Price C, Ghosh M, Miles K, Ragucci M,
and Sweis R. Embryonic fat-cell lineage in Drosophila melanogaster.
Development 120: 2489 –2499, 1994.
Hotamisligil GS. Inflammatory pathways and insulin action. Int J Obes
Relat Metab Disord 27, Suppl 3: S53–55, 2003.
Hotamisligil GS. The role of TNF␣ and TNF receptors in obesity and
insulin resistance. J Intern Med 245: 621– 625, 1999.
Hulver MW, Berggren JR, Carper MJ, Miyazaki M, Ntambi JM,
Hoffman EP, Thyfault JP, Stevens R, Dohm GL, Houmard JA, and
Muoio DM. Elevated stearoyl-CoA desaturase-1 expression in skeletal
AJP-Cell Physiol • VOL
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
muscle contributes to abnormal fatty acid partitioning in obese humans.
Cell Metab 2: 251–261, 2005.
Huss JM, Torra IP, Staels B, Giguere V, and Kelly DP. Estrogenrelated receptor ␣ directs peroxisome proliferator-activated receptor ␣
signaling in the transcriptional control of energy metabolism in cardiac
and skeletal muscle. Mol Cell Biol 24: 9079 –9091, 2004.
Ichida M, Nemoto S, and Finkel T. Identification of a specific molecular repressor of the peroxisome proliferator-activated receptor gamma
coactivator-1␣ (PGC-1␣). J Biol Chem 277: 50991–50995, 2002.
Jarvis CI, Staels B, Brugg B, Lemaigre-Dubreuil Y, Tedgui A, and
Mariani J. Age-related phenotypes in the staggerer mouse expand the
ROR␣ nuclear receptor’s role beyond the cerebellum. Mol Cell Endocrinol 186: 1–5, 2002.
Jetten AM, Kurebayashi S, and Ueda E. The ROR nuclear orphan
receptor subfamily: critical regulators of multiple biological processes.
Prog Nucleic Acid Res Mol Biol 69: 205–247, 2001.
Jordan VC. Selective estrogen receptor modulation: concept and consequences in cancer. Cancer Cell 5: 207–213, 2004.
Joseph JW, Koshkin V, Saleh MC, Sivitz WI, Zhang CY, Lowell BB,
Chan CB, and Wheeler MB. Free fatty acid-induced ␤-cell defects are
dependent on uncoupling protein 2 expression. J Biol Chem 279: 51049 –
51056, 2004.
Kadowaki T and Yamauchi T. Adiponectin and adiponectin receptors.
Endocr Rev 26: 439 – 451, 2005.
Kamei Y, Ohizumi H, Fujitani Y, Nemoto T, Tanaka T, Takahashi
N, Kawada T, Miyoshi M, Ezaki O, and Kakizuka A. PPARgamma
coactivator 1␤/ERR ligand 1 is an ERR protein ligand, whose expression
induces a high-energy expenditure and antagonizes obesity. Proc Natl
Acad Sci USA 100: 12378 –12383, 2003.
Kato S. Estrogen receptor-mediated cross-talk with growth factor signaling pathways. Breast Cancer 8: 3–9, 2001.
Kausch C, Krutzfeldt J, Witke A, Rettig A, Bachmann O, Rett K,
Matthaei S, Machicao F, Haring HU, and Stumvoll M. Effects of
troglitazone on cellular differentiation, insulin signaling, and glucose
metabolism in cultured human skeletal muscle cells. Biochem Biophys
Res Commun 280: 664 – 674, 2001.
Kelley DE. Skeletal muscle fat oxidation: timing and flexibility are
everything. J Clin Invest 115: 1699 –1702, 2005.
Kelley DE. Skeletal muscle triglycerides: an aspect of regional adiposity
and insulin resistance. Ann NY Acad Sci 967: 135–145, 2002.
Kelley DE, Goodpaster B, Wing RR, and Simoneau JA. Skeletal
muscle fatty acid metabolism in association with insulin resistance,
obesity, and weight loss. Am J Physiol Endocrinol Metab 277: E1130 –
E1141, 1999.
Kelley DE, He J, Menshikova EV, and Ritov VB. Dysfunction of
mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51:
2944 –2950, 2002.
Kelley DE and Mandarino LJ. Fuel selection in human skeletal muscle
in insulin resistance: a reexamination. Diabetes 49: 677– 683, 2000.
Kelly LJ, Vicario PP, Thompson GM, Candelore MR, Doebber TW,
Ventre J, Wu MS, Meurer R, Forrest MJ, Conner MW, Cascieri
MA, and Moller DE. Peroxisome proliferator-activated receptors ␥ and
␣ mediate in vivo regulation of uncoupling protein (UCP-1, UCP-2,
UCP-3) gene expression. Endocrinology 139: 4920 – 4927, 1998.
Kelly M, Keller C, Avilucea PR, Keller P, Luo Z, Xiang X, Giralt M,
Hidalgo J, Saha AK, Pedersen BK, and Ruderman NB. AMPK
activity is diminished in tissues of IL-6 knockout mice: the effect of
exercise. Biochem Biophys Res Commun 320: 449 – 454, 2004.
Kim HS, Liang L, Dean RG, Hausman DB, Hartzell DL, and Baile
CA. Inhibition of preadipocyte differentiation by myostatin treatment in
3T3-L1 cultures. Biochem Biophys Res Commun 281: 902–906, 2001.
Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M, Lutz EP,
Kako Y, Velez-Carrasco W, Goldberg IJ, Breslow JL, and Shulman
GI. Tissue-specific overexpression of lipoprotein lipase causes tissuespecific insulin resistance. Proc Natl Acad Sci USA 98: 7522–7527, 2001.
Kim YB, Ciaraldi TP, Kong A, Kim D, Chu N, Mohideen P,
Mudaliar S, Henry RR, and Kahn BB. Troglitazone but not metformin
restores insulin-stimulated phosphoinositide 3-kinase activity and increases p110␤ protein levels in skeletal muscle of type 2 diabetic
subjects. Diabetes 51: 443– 448, 2002.
Kim YB, Shulman GI, and Kahn BB. Fatty acid infusion selectively
impairs insulin action on Akt1 and protein kinase C ␭/zeta but not on
glycogen synthase kinase-3. J Biol Chem 277: 32915–32922, 2002.
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
45.
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
AJP-Cell Physiol • VOL
105. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G,
Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, and
Evans RM. The nuclear receptor superfamily: the second decade. Cell
83: 835– 839, 1995.
106. Margetic S, Gazzola C, Pegg GG, and Hill RA. Leptin: a review of its
peripheral actions and interactions. Int J Obes Relat Metab Disord 26:
1407–1433, 2002.
107. Martini F. Fundamentals of Anatomy and Physiology. Englewood
Cliffs, NJ: Prentice-Hall, 1998.
108. Maxwell MA, Cleasby ME, Harding A, Stark A, Cooney GJ, and
Muscat GE. Nur77 regulates lipolysis in skeletal muscle cells. Evidence
for cross-talk between the ␤-adrenergic and an orphan nuclear hormone
receptor pathway. J Biol Chem 280: 12573–12584, 2005.
109. McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 51: 7–18, 2002.
110. McPherron AC and Lee SJ. Double muscling in cattle due to mutations
in the myostatin gene. Proc Natl Acad Sci USA 94: 12457–12461, 1997.
111. McPherron AC and Lee SJ. Suppression of body fat accumulation in
myostatin-deficient mice. J Clin Invest 109: 595– 601, 2002.
112. Michalik L, Desvergne B, Tan NS, Basu-Modak S, Escher P, Rieusset
J, Peters JM, Kaya G, Gonzalez FJ, Zakany J, Metzger D, Chambon
P, Duboule D, and Wahli W. Impaired skin wound healing in peroxisome proliferator-activated receptor (PPAR)␣ and PPAR␤ mutant mice.
J Cell Biol 154: 799 – 814, 2001.
113. Minnich A, Tian N, Byan L, and Bilder G. A potent PPAR␣ agonist
stimulates mitochondrial fatty acid ␤-oxidation in liver and skeletal
muscle. Am J Physiol Endocrinol Metab 280: E270 –E279, 2001.
114. Mohan R and Heyman RA. Orphan nuclear receptor modulators. Curr
Top Med Chem 3: 1637–1647, 2003.
115. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S,
Yang W, Altshuler D, Puigserver P, Patterson N, Willy PJ, Schulman
IG, Heyman RA, Lander ES, and Spiegelman BM. Erralpha and
Gabpa/b specify PGC-1␣-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc Natl Acad Sci USA 101:
6570 – 6575, 2004.
116. Muoio DM, MacLean PS, Lang DB, Li S, Houmard JA, Way JM,
Winegar DA, Corton JC, Dohm GL, and Kraus WE. Fatty acid
homeostasis and induction of lipid regulatory genes in skeletal muscles of
peroxisome proliferator-activated receptor (PPAR)␣ knock-out mice.
Evidence for compensatory regulation by PPAR␦. J Biol Chem 277:
26089 –26097, 2002.
117. Murphy EP and Conneely OM. Neuroendocrine regulation of the
hypothalamic pituitary adrenal axis by the nurr1/nur77 subfamily of
nuclear receptors. Mol Endocrinol 11: 39 – 47, 1997.
118. Murphy EP, Dobson AD, Keller C, and Conneely OM. Differential
regulation of transcription by the NURR1/NUR77 subfamily of nuclear
transcription factors. Gene Expr 5: 169 –179, 1996.
119. Muscat GE, Wagner BL, Hou J, Tangirala RK, Bischoff ED, Rohde
P, Petrowski M, Li J, Shao G, Macondray G, and Schulman IG.
Regulation of cholesterol homeostasis and lipid metabolism in skeletal
muscle by liver X receptors. J Biol Chem 277: 40722– 40728, 2002.
120. Nakai A, Kartha S, Sakurai A, Toback FG, and DeGroot LJ. A
human early response gene homologous to murine nur77 and rat
NGFI-B, and related to the nuclear receptor superfamily. Mol Endocrinol
4: 1438 –1443, 1990.
121. Nakamura T, Fox-Robichaud A, Kikkawa R, Kashiwagi A, Kojima
H, Fujimiya M, and Wong NC. Transcription factors and age-related
decline in apolipoprotein A-I expression. J Lipid Res 40: 1709 –1718,
1999.
122. Narayan KM, Boyle JP, Thompson TJ, Sorensen SW, and Williamson DF. Lifetime risk for diabetes mellitus in the United States. JAMA
290: 1884 –1890, 2003.
123. Nevzorova J, Bengtsson T, Evans BA, and Summers RJ. Characterization of the ␤-adrenoceptor subtype involved in mediation of glucose
transport in L6 cells. Br J Pharmacol 137: 9 –18, 2002.
124. Norris AW, Chen L, Fisher SJ, Szanto I, Ristow M, Jozsi AC,
Hirshman MF, Rosen ED, Goodyear LJ, Gonzalez FJ, Spiegelman
BM, and Kahn CR. Muscle-specific PPAR␥-deficient mice develop
increased adiposity and insulin resistance but respond to thiazolidinediones. J Clin Invest 112: 608 – 618, 2003.
125. Oakes ND, Bell KS, Furler SM, Camilleri S, Saha AK, Ruderman
NB, Chisholm DJ, and Kraegen EW. Diet-induced muscle insulin
resistance in rats is ameliorated by acute dietary lipid withdrawal or a
single bout of exercise: parallel relationship between insulin stimulation
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
85. Kliewer SA, Lehmann JM, and Willson TM. Orphan nuclear receptors: shifting endocrinology into reverse. Science 284: 757–760, 1999.
86. Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM,
Rothman DL, Roden M, and Shulman GI. Intramyocellular lipid
concentrations are correlated with insulin sensitivity in humans: a 1H
NMR spectroscopy study. Diabetologia 42: 113–116, 1999.
87. Lau P, Bailey P, Dowhan DH, and Muscat GE. Exogenous expression
of a dominant negative ROR␣1 vector in muscle cells impairs differentiation: ROR␣1 directly interacts with p300 and myoD. Nucleic Acids
Res 27: 411– 420, 1999.
88. Lau P, Nixon SJ, Parton RG, and Muscat GE. RORalpha regulates the
expression of genes involved in lipid homeostasis in skeletal muscle
cells: caveolin-3 and CPT-1 are direct targets of ROR. J Biol Chem 279:
36828 –36840, 2004.
89. Law SW, Conneely OM, DeMayo FJ, and O’Malley BW. Identification of a new brain-specific transcription factor, NURR1. Mol Endocrinol
6: 2129 –2135, 1992.
90. Lee CH, Olson P, and Evans RM. Minireview: lipid metabolism,
metabolic diseases, and peroxisome proliferator-activated receptors. Endocrinology 144: 2201–2207, 2003.
91. Lee SS, Pineau T, Drago J, Lee EJ, Owens JW, Kroetz DL, Fernandez-Salguero PM, Westphal H, and Gonzalez FJ. Targeted disruption
of the ␣ isoform of the peroxisome proliferator-activated receptor gene in
mice results in abolishment of the pleiotropic effects of peroxisome
proliferators. Mol Cell Biol 15: 3012–3022, 1995.
92. Lee Y, Hirose H, Ohneda M, Johnson JH, McGarry JD, and Unger
RH. Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent
diabetes mellitus of obese rats: impairment in adipocyte-␤-cell relationships. Proc Natl Acad Sci USA 91: 10878 –10882, 1994.
93. Lee YH and Pratley RE. The evolving role of inflammation in obesity
and the metabolic syndrome. Curr Diab Rep 5: 70 –75, 2005.
94. Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson
TM, and Kliewer SA. An antidiabetic thiazolidinedione is a high affinity
ligand for peroxisome proliferator-activated receptor ␥ (PPAR␥). J Biol
Chem 270: 12953–12956, 1995.
95. Leibowitz MD, Fievet C, Hennuyer N, Peinado-Onsurbe J, Duez H,
Bergera J, Cullinan CA, Sparrow CP, Baffic J, Berger GD, Santini
C, Marquis RW, Tolman RL, Smith RG, Moller DE, and Auwerx J.
Activation of PPAR␦ alters lipid metabolism in db/db mice. FEBS Lett
473: 333–336, 2000.
96. Leone TC, Weinheimer CJ, and Kelly DP. A critical role for the
peroxisome proliferator-activated receptor alpha (PPAR␣) in the cellular
fasting response: the PPAR␣-null mouse as a model of fatty acid
oxidation disorders. Proc Natl Acad Sci USA 96: 7473–7478, 1999.
97. Lewis GF and Steiner G. Acute effects of insulin in the control of
VLDL production in humans. Implications for the insulin-resistant state.
Diabetes Care 19: 390 –393, 1996.
98. Lewis GF, Uffelman KD, Szeto LW, Weller B, and Steiner G.
Interaction between free fatty acids and insulin in the acute control of
very low density lipoprotein production in humans. J Clin Invest 95:
158 –166, 1995.
99. Lim RW, Yang WL, and Yu H. Signal-transduction-pathway-specific
desensitization of expression of orphan nuclear receptor TIS1. Biochem
J 308: 785–789, 1995.
100. Long SD and Pekala PH. Lipid mediators of insulin resistance: ceramide signalling down-regulates GLUT4 gene transcription in 3T3–L1
adipocytes. Biochem J 319: 179 –184, 1996.
101. Lopez-Solache I, Marie V, Vignault E, Camirand A, and Silva JE.
Regulation of uncoupling protein-2 mRNA in L6 myotubules. I: thiazolidinediones stimulate uncoupling protein-2 gene expression by a mechanism requiring ongoing protein synthesis and an active mitogen-activated protein kinase. Endocrinology 19: 197–208, 2002.
102. Luo J, Sladek R, Carrier J, Bader JA, Richard D, and Giguere V.
Reduced fat mass in mice lacking orphan nuclear receptor estrogenrelated receptor alpha. Mol Cell Biol 23: 7947–7956, 2003.
103. Luquet S, Lopez-Soriano J, Holst D, Fredenrich A, Melki J, Rassoulzadegan M, and Grimaldi PA. Peroxisome proliferator-activated
receptor ␦ controls muscle development and oxidative capability. FASEB
J 17: 2299 –2301, 2003.
104. Mamontova A, Seguret-Mace S, Esposito B, Chaniale C, Bouly M,
Delhaye-Bouchaud N, Luc G, Staels B, Duverger N, Mariani J, and
Tedgui A. Severe atherosclerosis and hypoalphalipoproteinemia in the
staggerer mouse, a mutant of the nuclear receptor ROR␣. Circulation 98:
2738 –2743, 1998.
C215
Invited Review
C216
126.
127.
128.
129.
131.
132.
133.
134.
135.
136.
137.
138.
139.
140.
141.
142.
143.
144.
145.
146.
of glucose uptake and suppression of long-chain fatty acyl-CoA. Diabetes 46: 2022–2028, 1997.
Ohkubo T, Sugawara Y, Sasaki K, Maruyama K, Ohkura N, and
Makuuchi M. Early induction of nerve growth factor-induced genes
after liver resection-reperfusion injury. J Hepatol 36: 210 –217, 2002.
Ohkura N, Ito M, Tsukada T, Sasaki K, Yamaguchi K, and Miki K.
Structure, mapping and expression of a human NOR-1 gene, the third
member of the Nur77/NGFI-B family. Biochim Biophys Acta 1308:
205–214, 1996.
Oliver WR Jr, Shenk JL, Snaith MR, Russell CS, Plunket KD,
Bodkin NL, Lewis MC, Winegar DA, Sznaidman ML, Lambert MH,
Xu HE, Sternbach DD, Kliewer SA, Hansen BC, and Willson TM. A
selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc Natl Acad Sci USA 98: 5306 –
5311, 2001.
Ordentlich P, Yan Y, Zhou S, and Heyman RA. Identification of the
antineoplastic agent 6-mercaptopurine as an activator of the orphan
nuclear hormone receptor Nurr1. J Biol Chem 278: 24791–24799, 2003.
Park JI, Tsai SY, and Tsai MJ. Molecular mechanism of chicken
ovalbumin upstream promoter-transcription factor (COUP-TF) actions.
Keio J Med 52: 174 –181, 2003.
Pearce KH, Iannone MA, Simmons CA, and Gray JG. Discovery of
novel nuclear receptor modulating ligands: an integral role for peptide
interaction profiling. Drug Discov Today 9: 741–751, 2004.
Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard P, Wolsk-Petersen E, and Febbraio M. The metabolic role of
IL-6 produced during exercise: is IL-6 an exercise factor? Proc Nutr Soc
63: 263–267, 2004.
Perseghin G. Muscle lipid metabolism in the metabolic syndrome. Curr
Opin Lipidol 16: 416 – 420, 2005.
Peters JM, Lee SS, Li W, Ward JM, Gavrilova O, Everett C,
Reitman ML, Hudson LD, and Gonzalez FJ. Growth, adipose, brain,
and skin alterations resulting from targeted disruption of the mouse
peroxisome proliferator-activated receptor ␤(␦). Mol Cell Biol 20: 5119 –
5128, 2000.
Petersen EW, Carey AL, Sacchetti M, Steinberg GR, Macaulay SL,
Febbraio MA, and Pedersen BK. Acute IL-6 treatment increases fatty
acid turnover in elderly humans in vivo and in tissue culture in vitro.
Am J Physiol Endocrinol Metab 288: E155–E162, 2005.
Petersen KF and Shulman GI. Pathogenesis of skeletal muscle insulin
resistance in type 2 diabetes mellitus. Am J Cardiol 90: 11G-18G, 2002.
Pickup JC, Mattock MB, Chusney GD, and Burt D. NIDDM as a
disease of the innate immune system: association of acute-phase reactants
and interleukin-6 with metabolic syndrome X. Diabetologia 40: 1286 –
1292, 1997.
Pircher P, Chomez P, Yu F, Vennström B, and Larsson L. Aberrant
expression of myosin isoforms in skeletal muscles from mice lacking the
rev-erb␣ orphan receptor gene. Am J Physiol Regul Integr Comp Physiol
288: R482–R490, 2004.
Pradhan AD, Manson JE, Rifai N, Buring JE, and Ridker PM.
C-reactive protein, interleukin 6, and risk of developing type 2 diabetes
mellitus. JAMA 286: 327–334, 2001.
Prentice T. World Health Organization: World Health Report. Overview. 2002.
Quinn LS, Haugk KL, and Grabstein KH. Interleukin-15: a novel
anabolic cytokine for skeletal muscle. Endocrinology 136: 3669 –3672,
1995.
Quinn LS, Strait-Bodey L, Anderson BG, Argiles JM, and Havel PJ.
Interleukin-15 stimulates adiponectin secretion by 3T3–L1 adipocytes:
evidence for a skeletal muscle-to-fat signaling pathway. Cell Biol Int 29:
449 – 457, 2005.
Ramakrishnan SN, Lau P, Burke LJ, and Muscat GE. Rev-erb␤
regulates the expression of genes involved in lipid absorption in skeletal
muscle cells: evidence for cross-talk between orphan nuclear receptors
and myokines. J Biol Chem 280: 8651– 8659, 2005.
Randle PJ, Garland PB, Hales CN, and Newsholme EA. The glucose
fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1: 785–789, 1963.
Rangwala SM and Lazar MA. Peroxisome proliferator-activated receptor-␥ in diabetes and metabolism. Trends Pharmacol Sci 25: 331–336,
2004.
Raspe E, Duez H, Gervois P, Fievet C, Fruchart JC, Besnard S,
Mariani J, Tedgui A, and Staels B. Transcriptional regulation of
AJP-Cell Physiol • VOL
147.
148.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
159.
160.
161.
162.
163.
164.
165.
166.
167.
apolipoprotein C-III gene expression by the orphan nuclear receptor
ROR␣. J Biol Chem 276: 2865–2871, 2001.
Roden M, Krssak M, Stingl H, Gruber S, Hofer A, Furnsinn C,
Moser E, and Waldhausl W. Rapid impairment of skeletal muscle
glucose transport/phosphorylation by free fatty acids in humans. Diabetes 48: 358 –364, 1999.
Romijn JA, Coyle EF, Sidossis LS, Gastaldelli A, Horowitz JF,
Endert E, and Wolfe RR. Regulation of endogenous fat and carbohydrate metabolism in relation to exercise intensity and duration. Am J
Physiol Endocrinol Metab 265: E380 –E391, 1993.
Romijn JA, Klein S, Coyle EF, Sidossis LS, and Wolfe RR. Strenuous
endurance training increases lipolysis and triglyceride-fatty acid cycling
at rest. J Appl Physiol 75: 108 –113, 1993.
Rosen ED and Spiegelman BM. PPAR␥: a nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem 276: 37731–37734,
2001.
Rosen ED, Walkey CJ, Puigserver P, and Spiegelman BM. Transcriptional regulation of adipogenesis. Genes Dev 14: 1293–1307, 2000.
Rotter V, Nagaev I, and Smith U. Interleukin-6 (IL-6) induces insulin
resistance in 3T3–L1 adipocytes and is, like IL-8 and tumor necrosis
factor-␣, overexpressed in human fat cells from insulin-resistant subjects.
J Biol Chem 278: 45777– 45784, 2003.
Sack MN, Rader TA, Park S, Bastin J, McCune SA, and Kelly DP.
Fatty acid oxidation enzyme gene expression is downregulated in the
failing heart. Circulation 94: 2837–2842, 1996.
Saladin R, Fajas L, Dana S, Halvorsen YD, Auwerx J, and Briggs M.
Differential regulation of peroxisome proliferator activated receptor ␥1
(PPAR␥1) and PPAR␥2 messenger RNA expression in the early stages
of adipogenesis. Cell Growth Differ 10: 43– 48, 1999.
Samuel VT, Liu ZX, Qu X, Elder BD, Bilz S, Befroy D, Romanelli
AJ, and Shulman GI. Mechanism of hepatic insulin resistance in
non-alcoholic fatty liver disease. J Biol Chem 279: 32345–32353, 2004.
Savage DB, Agostini M, Barroso I, Gurnell M, Luan J, Meirhaeghe
A, Harding AH, Ihrke G, Rajanayagam O, Soos MA, George S,
Berger D, Thomas EL, Bell JD, Meeran K, Ross RJ, Vidal-Puig A,
Wareham NJ, O’Rahilly S, Chatterjee VK, and Schafer AJ. Digenic
inheritance of severe insulin resistance in a human pedigree. Nat Genet
31: 379 –384, 2002.
Savage DB, Petersen KF, and Shulman GI. Mechanisms of insulin
resistance in humans and possible links with inflammation. Hypertension
45: 828 – 833, 2005.
Savage DB, Tan GD, Acerini CL, Jebb SA, Agostini M, Gurnell M,
Williams RL, Umpleby AM, Thomas EL, Bell JD, Dixon AK, Dunne
F, Boiani R, Cinti S, Vidal-Puig A, Karpe F, Chatterjee VK, and
O’Rahilly S. Human metabolic syndrome resulting from dominantnegative mutations in the nuclear receptor peroxisome proliferatoractivated receptor-␥. Diabetes 52: 910 –917, 2003.
Schmitz-Peiffer C. Protein kinase C and lipid-induced insulin resistance
in skeletal muscle. Ann NY Acad Sci 967: 146 –157, 2002.
Schulman IG and Heyman RA. The flip side: identifying small molecule regulators of nuclear receptors. Chem Biol 11: 639 – 646, 2004.
Shao D and Lazar MA. Modulating nuclear receptor function: may the
phos be with you. J Clin Invest 103: 1617–1618, 1999.
Sharma AM and Chetty VT. Obesity, hypertension and insulin resistance. Acta Diabetol 42, Suppl 1: S3–S8, 2005.
Shimabukuro M, Koyama K, Chen G, Wang MY, Trieu F, Lee Y,
Newgard CB, and Unger RH. Direct antidiabetic effect of leptin
through triglyceride depletion of tissues. Proc Natl Acad Sci USA 94:
4637– 4641, 1997.
Shimomura I, Matsuda M, Hammer RE, Bashmakov Y, Brown MS,
and Goldstein JL. Decreased IRS-2 and increased SREBP-1c lead to
mixed insulin resistance and sensitivity in livers of lipodystrophic and
ob/ob mice. Mol Cell 6: 77– 86, 2000.
Simoneau JA, Colberg SR, Thaete FL, and Kelley DE. Skeletal
muscle glycolytic and oxidative enzyme capacities are determinants of
insulin sensitivity and muscle composition in obese women. FASEB J 9:
273–278, 1995.
Simoneau JA, Veerkamp JH, Turcotte LP, and Kelley DE. Markers
of capacity to utilize fatty acids in human skeletal muscle: relation to
insulin resistance and obesity and effects of weight loss. FASEB J 13:
2051–2060, 1999.
Son C, Hosoda K, Matsuda J, Fujikura J, Yonemitsu S, Iwakura H,
Masuzaki H, Ogawa Y, Hayashi T, Itoh H, Nishimura H, Inoue G,
Yoshimasa Y, Yamori Y, and Nakao K. Up-regulation of uncoupling
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
130.
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
Invited Review
METABOLIC DISEASE, RECEPTOR, AND MUSCULAR LINKS
168.
169.
170.
171.
173.
174.
175.
176.
177.
178.
AJP-Cell Physiol • VOL
179.
180.
181.
182.
183.
184.
185.
186.
187.
188.
189.
190.
191.
scriptional regulation of apolipoprotein A-I gene expression by the
nuclear receptor ROR␣. J Biol Chem 272: 22401–22404, 1997.
Wahl HG, Kausch C, Machicao F, Rett K, Stumvoll M, and Haring
HU. Troglitazone downregulates ␦-6 desaturase gene expression in
human skeletal muscle cell cultures. Diabetes 51: 1060 –1065, 2002.
Wallenius V, Wallenius K, Ahren B, Rudling M, Carlsten H, Dickson
SL, Ohlsson C, and Jansson JO. Interleukin-6-deficient mice develop
mature-onset obesity. Nat Med 8: 75–79, 2002.
Wang YX, Zhang CL, Yu RT, Cho HK, Nelson MC, BayugaOcampo CR, Ham J, Kang H, and Evans RM. Regulation of muscle
fiber type and running endurance by PPAR␦. PLoS Biol 2: E294, 2004.
Wansa KD, Harris JM, Yan G, Ordentlich P, and Muscat GE. The
AF-1 domain of the orphan nuclear receptor NOR-1 mediates transactivation, coactivator recruitment, and activation by the purine antimetabolite 6-mercaptopurine. J Biol Chem 278: 24776 –24790, 2003.
Wellen KE and Hotamisligil GS. Obesity-induced inflammatory
changes in adipose tissue. J Clin Invest 112: 1785–1788, 2003.
White RT, Damm D, Hancock N, Rosen BS, Lowell BB, Usher P,
Flier JS, and Spiegelman BM. Human adipsin is identical to complement factor D and is expressed at high levels in adipose tissue. J Biol
Chem 267: 9210 –9213, 1992.
WHOIDF. Diabetes Action Now. 2004.
Wisse BE. The inflammatory syndrome: the role of adipose tissue
cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol 15:
2792–2800, 2004.
Wolf G. Tissue-specific knockout defines peroxisome proliferator-activated receptor ␥ function in muscle and liver. Nutr Rev 62: 253–255,
2004.
Yaney GC and Corkey BE. Fatty acid metabolism and insulin secretion
in pancreatic beta cells. Diabetologia 46: 1297–1312, 2003.
Yonemitsu S, Nishimura H, Shintani M, Inoue R, Yamamoto Y,
Masuzaki H, Ogawa Y, Hosoda K, Inoue G, Hayashi T, and Nakao
K. Troglitazone induces GLUT4 translocation in L6 myotubes. Diabetes
50: 1093–1101, 2001.
Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Bergeron R,
Kim JK, Cushman SW, Cooney GJ, Atcheson B, White MF, Kraegen
EW, and Shulman GI. Mechanism by which fatty acids inhibit insulin
activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J Biol Chem 277: 50230 –50236,
2002.
Zetterstrom RH, Solomin L, Mitsiadis T, Olson L, and Perlmann T.
Retinoid X receptor heterodimerization and developmental expression
distinguish the orphan nuclear receptors NGFI-B, Nurr1, and Nor1. Mol
Endocrinol 10: 1656 –1666, 1996.
291 • AUGUST 2006 •
www.ajpcell.org
Downloaded from http://ajpcell.physiology.org/ by 10.220.32.247 on April 30, 2017
172.
protein 3 gene expression by fatty acids and agonists for PPARs in L6
myotubes. Endocrinology 142: 4189 – 4194, 2001.
Stavinoha MA, RaySpellicy JW, Essop MF, Graveleau C, Abel ED,
Hart-Sailors ML, Mersmann HJ, Bray MS, and Young ME. Evidence
for mitochondrial thioesterase 1 as a peroxisome proliferator-activated
receptor-␣-regulated gene in cardiac and skeletal muscle. Am J Physiol
Endocrinol Metab 287: E888 –E895, 2004.
Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright
CM, Patel HR, Ahima RS, and Lazar MA. The hormone resistin links
obesity to diabetes. Nature 409: 307–312, 2001.
Suwa M, Nakano H, and Kumagai S. Effects of chronic AICAR
treatment on fiber composition, enzyme activity, UCP3, and PGC-1 in rat
muscles. J Appl Physiol 95: 960 –968, 2003.
Tanaka T, Yamamoto J, Iwasaki S, Asaba H, Hamura H, Ikeda Y,
Watanabe M, Magoori K, Ioka RX, Tachibana K, Watanabe Y,
Uchiyama Y, Sumi K, Iguchi H, Ito S, Doi T, Hamakubo T, Naito M,
Auwerx J, Yanagisawa M, Kodama T, and Sakai J. Activation of
peroxisome proliferator-activated receptor delta induces fatty acid betaoxidation in skeletal muscle and attenuates metabolic syndrome. Proc
Natl Acad Sci USA 100: 15924 –15929, 2003.
Tenbaum S and Baniahmad A. Nuclear receptors: structure, function
and involvement in disease. Int J Biochem Cell Biol 29: 1325–1341,
1997.
Van Hall G, Steensberg A, Sacchetti M, Fischer C, Keller C, Schjerling P, Hiscock N, Moller K, Saltin B, Febbraio MA, and Pedersen
BK. Interleukin-6 stimulates lipolysis and fat oxidation in humans. J Clin
Endocrinol Metab 88: 3005–3010, 2003.
Verma NK, Singh J, and Dey CS. PPAR-␥ expression modulates
insulin sensitivity in C2C12 skeletal muscle cells. Br J Pharmacol 143:
1006 –1013, 2004.
Vettor R, Fabris R, Serra R, Lombardi AM, Tonello C, Granzotto
M, Marzolo MO, Carruba MO, Ricquier D, Federspil G, and Nisoli
E. Changes in FAT/CD36, UCP2, UCP3 and GLUT4 gene expression
during lipid infusion in rat skeletal and heart muscle. Int J Obes Relat
Metab Disord 26: 838 – 847, 2002.
Vosper H, Khoudoli GA, Graham TL, and Palmer CN. Peroxisome
proliferator-activated receptor agonists, hyperlipidaemia, and atherosclerosis. Pharmacol Ther 95: 47– 62, 2002.
Vosper H, Patel L, Graham TL, Khoudoli GA, Hill A, Macphee CH,
Pinto I, Smith SA, Suckling KE, Wolf CR, and Palmer CN. The
peroxisome proliferator-activated receptor ␦ promotes lipid accumulation
in human macrophages. J Biol Chem 276: 44258 – 44265, 2001.
Vu-Dac N, Gervois P, Grotzinger T, De Vos P, Schoonjans K,
Fruchart JC, Auwerx J, Mariani J, Tedgui A, and Staels B. Tran-
C217