Liang Ward 2006 Pgc 1α a Key Regulator of Energy Metabolism

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Adv Physiol Educ 30: 145–151, 2006;

doi:10.1152/advan.00052.2006. Staying Current

PGC-1␣: a key regulator of energy metabolism

Huiyun Liang1,2 and Walter F. Ward2,3


1
Department of Cellular and Structural Biology, 2Barshop Institute for Longevity and
Aging Studies, and 3Department of Physiology, Audie Murphy Veterans Administration
Medical Center and University of Texas Health Science Center, San Antonio, Texas
Submitted 22 June 2006; accepted in final form 27 July 2006

Liang, Huiyun, and Walter F. Ward. PGC-1␣: a key regulator of very low in white adipose tissue (WAT) (49). It is noteworthy
energy metabolism. Adv Physiol Educ 30: 145–151, 2006; doi: that PGC-1␣ does not exhibit histone acetyltransferase (HAT)
10.1152/advan.00052.2006.—Peroxisome proliferator-activated re- activity itself, but the conformational change it induces after
ceptor-␥ coactivator (PGC)-1␣ is a member of a family of transcrip- docking on specific transcription factors increases the affinity
tion coactivators that plays a central role in the regulation of cellular
energy metabolism. It is strongly induced by cold exposure, linking
of the transcription complex to additional coactivators possess-
this environmental stimulus to adaptive thermogenesis. PGC-1␣ stim- ing HAT activity, such as steroid receptor coactivator-1 and
ulates mitochondrial biogenesis and promotes the remodeling of cAMP response-binding element (CREB)-binding protein and
muscle tissue to a fiber-type composition that is metabolically more p300. The end result is acetylation of histone proteins, which
oxidative and less glycolytic in nature, and it participates in the produces conformation alterations that increase the accessibil-
regulation of both carbohydrate and lipid metabolism. It is highly ity of DNA to the transcription complex (46). Although there
likely that PGC-1␣ is intimately involved in disorders such as obesity, are three members of the PGC-1 family [PGC-1␣, PGC-1␤,
diabetes, and cardiomyopathy. In particular, its regulatory function in and PGC-1-related coactivator (48)], we will focus on PGC-1␣
lipid metabolism makes it an inviting target for pharmacological in this review. There is currently a great interest in PGC-1␣
intervention in the treatment of obesity and Type 2 diabetes. because rapidly growing evidence strongly suggests that it is a
adaptive thermogenesis; transcription coactivator; proliferator perox- powerful regulator of energy metabolism under conditions of
isome-activated receptor; glucose metabolism; diabetes both health and disease (12).

PGC-1␣ and Adaptive Thermogenesis


PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR (PPAR)-␥ coac-
tivator (PGC)-1␣ is a transcription coactivator that interacts PGC-1␣ was originally discovered as a cold-inducible tran-
with a broad range of transcription factors that are involved in scription coactivator of adaptive thermogenesis, a physiologi-
a wide variety of biological responses including adaptive cal process through which energy is dissipated as heat in
thermogenesis, mitochondrial biogenesis, glucose/fatty acid response to environmental conditions such as cold stress and
metabolism, fiber type switching in skeletal muscle, and heart overfeeding (8, 51). BAT and skeletal muscle are the two
development (Table 1). A transcription coactivator is defined major organs involved in this process. While rats and mice
as a protein or protein complex that increases the probability of have prominent brown fat depots, this isn’t the case in larger
a gene being transcribed by interacting with transcription mammals, including humans, although there may be brown fat
factors but does not itself bind to DNA in a sequence-specific cells dispersed among the adipocytes of WAT (49). Thus, in
manner (48). The PPARs (PPAR-␣, PPAR-␦, and PPAR-␥) are larger mammals, skeletal muscle is generally thought to be the
members of a relatively large family of nuclear receptors, all of major thermogenic tissue. The primary physiological function
which are subject to transcriptional coactivation by PGC-1␣. of WAT is energy storage, whereas the primary function of
PPAR-␥ is essential for adipogenesis and differentiation (31), BAT is energy dissipation, largely in the form of heat. In fact,
whereas PPAR-␣ and PPAR-␦ play important roles in the BAT produces ⬃60% of the heat generated by nonshivering,
control of fatty acid oxidation (61). In addition, PPAR-␥ is the adaptive thermogenesis. Spurred on in part by the prevalence
target of thiazolidinediones (TZDs), which are drugs that are of human obesity, interest in adaptive thermogenesis has in-
widely utilized for increasing insulin sensitivity in diabetic creased because of the possibility that it may provide a phys-
patients (50, 59). iological defense against obesity (68). This has also led to
The PGC-1␣ gene is located on chromosome 5 in mice increased attention being directed toward the regulation of
(chromosome 4 in humans) and encodes a protein containing adipose tissue differentiation. It was known that the transcrip-
797 (mouse) or 798 amino acids (human). PGC-1␣ has two tion factor PPAR-␥ is an important regulator of adipocyte
putative nuclear localization signals and is located in the cell differentiation, but, surprisingly, it is unable, by itself, to
nucleus (49). It is expressed at high levels in tissues where induce brown fat differentiation (45). It was during the search
mitochondria are abundant and oxidative metabolism is active, for the solution to this puzzle that PGC-1␣ was discovered
such as brown adipose tissue (BAT), the heart, and skeletal (49). PGC-1␣ binds to PPAR-␥ and coactivates PPAR-␥ to
muscle. PGC-1␣ expression is also high in the brain and stimulate the transcription of genes involved in the brown
kidney, whereas the expression level is low in the liver and adipocyte differentiation process (49).
The adaptive thermogenic program in both brown fat and
Address for reprint requests and other correspondence: W. F. Ward, Dept. of skeletal muscle involves the stimulation of mitochondria bio-
Physiology, Univ. of Texas Health Science Center, 7703 Floyd Curl Dr., San genesis, increased fatty acid oxidation, and the uncoupling of
Antonio, TX 78229-3900 (e-mail: wardw@uthscsa.edu). oxidative phosphorylation. In response to cold exposure,
145
Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
146 PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM

Table 1. Selected list of transcription factors for which endurance training in rodent models and human subjects (5, 15,
PGC-1␣ functions as a coactivator 41, 52). Our understanding of the biological role of PGC-1␣ in
skeletal muscle structure and function has been greatly im-
Transcription Factor Function Reference proved through the use of gain of function and loss of function
NRF1 Mitochondria biogenesis 68 mouse models. In a gain of function transgenic model, PGC-1␣
NRF2 Mitochondria biogenesis 36 is overexpressed in a skeletal muscle-specific manner under the
ERR-␣/␤/␥ Mitochondria biogenesis 35 control of the muscle creatine kinase (MCK) promoter (28).
PPAR-␣ Fatty acid oxidation 62 PGC-1␣ overexpression results in the conversion of fast-twitch
PPAR-␦ Fatty acid oxidation 64
TR-␤ CPT-I induction 67, 72 type IIb muscle fibers to type IIa and slow-twitch type I fibers
FXR Triglyceride metabolism 71 by 20% and 10%, respectively, in plantaris muscle. In addition,
LXR-␣/␤ Lipoprotein secretion 30 there is an activation of genes involved in mitochondrial
GR Gluconeogenesis 23 oxidative metabolism. The conversion to slow-twitch fibers is
HNF-4␣ Gluconeogenesis 39
FOXO1 Gluconeogenesis 47
also evidenced by the redder muscle color and the expression
PPAR-␥ Brown adipocyte differentiation; 16, 20 of contractile proteins characteristic of slow-twitch fibers such
UCP1 induction as slow troponin I and myoglobin. As would be predicted,
based on these alterations, muscles isolated from the MCK-
PGC-1␣, proliferator peroxisome-activated receptor (PPAR)-␥ coactivator-
1␣; NRF, nuclear respiratory factor; ERR, estrogen-related receptor; TR, PGC-1␣ transgenic mouse show increased resistance to elec-
thyroid receptor; CPT-I, carnitine palmitoyltransferase-I; FXR, farnesoid X trically stimulated fatigue (28). Consistent with this, Morten-
receptor; LXR, liver X receptor; GR, glucocorticoid receptor; HNF, hepatic son et al. (39) recently reported that overexpression of PGC-1␣
nuclear factor; FOXO1, forkhead box O1; UCP1, uncoupling protein-1. [Mod- in primary rat skeletal muscle cells leads to enhanced levels of
ified from Ref. 23.]
mRNA for the slow oxidative-associated myosin heavy chain
(MHC) isoform (MHCIb) and decreased mRNA levels for
PGC-1␣ expression is increased, with the induction being the fast glycolytic-associated MHC isoforms (MHCIIX and
mediated through the sympathetic nervous system’s ␤3-adren- MHCIIB) (39). In contrast, PGC-1␣-deficient mice exhibit
ergic receptor/cAMP pathway (49). Increased PGC-1␣ induces decreased mitochondrial number and decreased respiratory
the transcription of nuclear respiratory factor (NRF)1 and capacity in slow-twitch muscle as well as reduced exercise
NRF2, leading to the increased expression of mitochondrial capacity and a reduced fatigue resistance index (26).
transcription factor A (mtTFA) (68) as well as other nuclear- The upstream signaling involved in the activation of
encoded mitochondria subunits of the electron transport chain PGC-1␣ are unclear at present, but several pathways have been
complex such as ␤-ATP synthase, cytochrome c, and cyto- implicated, such as the calcineurin A and CaMK, p38 MAPK,
chrome c oxidase IV (56, 57). mtTFA translocates to the and AMP-activated protein kinase pathways (1, 67, 73). Al-
mitochondrion, where it stimulates mitochondrial biogenesis as though the relative importance of these kinases in controlling
manifested by stimulation of mitochondrial DNA replication PGC-1␣ is yet to be determined, there appears to be a consid-
and mitochondria gene expression (14, 24). PGC-1␣ also erable amount of evidence suggesting that the calcineurin and
interacts with other nuclear hormone receptors such as CaMK pathways play important roles in the regulation of
PPAR-␣, retinoic acid receptor, and thyroid receptor in BAT to PGC-1␣ expression. During exercise, the combination of in-
enhance the expression of brown fat-specific uncoupling pro- creased neuromuscular input and increased contractile activity
tein 1 (UCP1) (6, 9, 58). UCP1 action leads to dissipation of induces the expression of several transcription factors, e.g.,
the proton gradient and the uncoupling of oxidative phosphor- myocyte-specific enhancer factor (MEF)2 and CREB (10, 17).
ylation, thereby increasing heat production. Under these con- These induction processes appear to be mediated by cal-
ditions, there is a marked increase in the rate of energy cineurin and CaMK (67). Increased expression of MEF2 leads
metabolism. The essential role of PGC-1␣ in adaptive thermo- to increased MEF2 binding to the promoter region of the
genesis is convincingly demonstrated by the observation that PGC-1␣ gene, increasing PGC-1␣ expression. PGC-1␣ can
the PGC-1␣-deficient mice are unable to withstand a cold then bind directly to MEF2 to coactivate the transcription of
stress (4°C) for longer than 6 h due to a continuous decrease of those genes involved in the determination of slow-twitch fiber
core body temperature. Wild-type control mice, on the other types and mitochondrial oxidative metabolism (10, 27). Fi-
hand, were able to tolerate the cold stress by keeping their core nally, it should be pointed out that recent work from Holloszy’s
body temperature at ⬃36.5°C, after an initial drop of ⬃1.5°C laboratory (13) suggests that calcineurin may not be required
(26, 29). for exercise-induced increases in PGC-1␣ and a range of
mitochondrial proteins. Obviously, further work is needed to
PGC-1␣ and Skeletal Muscle Fiber Conversion
elucidate the roles of calcineurin and calcium signaling path-
Skeletal muscle fibers are classified into three types: type I, ways in the regulation of PGC-1␣.
type IIa, and type IIb. Slow-twitch type I and fast-twitch type
PGC-1␣ and Heart Development
IIa fibers contain more mitochondria and exhibit relatively
higher rates of oxidative metabolism. In contrast, type IIb The heart, an organ with a very high and dynamic demand
fibers have fewer mitochondria and are metabolically glyco- for ATP, derives most of the required ATP from fatty acid
lytic. It is now well established that PGC-1␣ induces a remod- oxidation. In the developing mouse heart, as the energy source
eling of skeletal muscle fiber composition. In general, the ratio turns toward mitochondria fatty acid oxidation after birth, there
of glycolytic type IIb fibers to the more oxidative type I and is a large burst of mitochondria biogenesis and oxidative
type IIa fibers decreases. The expression of PGC-1␣ in skeletal metabolism that is preceded by a dramatically increased ex-
muscle is readily inducible by both short-term exercise and pression of PGC-1␣ (25). Short-term fasting, another condition

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM 147

that produces an increased reliance on mitochondrial fatty acid During fasting, the expression of PGC-1␣ is activated by
oxidation, also activates myocardial PGC-1␣ (25). Under in glucagon and catecholamines via the stimulation of the cAMP
vitro conditions, ectopic expression of PGC-1␣ in cultured pathway and the CREB transcription factor. PGC-1␣ then
cardiac myocytes has been shown to drive mitochondrial bio- coactivates a variety of transcription factors such as hepatic
genesis, and oxidative phosphorylation in these mitochondria nuclear factor-4␣, glucocorticoid receptor, and forkhead box
is tightly coupled with ATP production (25). In contrast, in O1 (FOXO1). These transcription factors bind to the promoter
adipocyte and C2C12 mouse myoblast cell lines, PGC-1␣ regions of those genes encoding key gluconeogenic enzymes
expression leads to the uncoupling of oxidative phosphoryla- such as phosphoenolpyruvate carboxykinase (PEPCK) and
tion, resulting in decreased ATP formation and increased heat glucose-6-phosphatase (G-6-Pase).
production (68). When the heart undergoes hypertrophy in Tissue culture studies have revealed that forced overexpres-
response to pressure overload or cyclin T1-induced activation sion of PGC-1␣ in primary hepatocytes is sufficient to drive the
of Cdk9, PGC-1␣ expression is markedly decreased, and this expression of key gluconeogenic genes (69). In contrast,
reduction of PGC-1␣ levels is associated with a conversion knockdown of PGC-1␣ expression by short interfering RNA in
from fatty acid oxidation to glycolytic metabolism (54, 55, 55). the mouse liver dramatically reduced the expression of PEPCK
To further investigate the role of PGC-1␣ in the heart, two and G-6-Pase (22). The essential role of PGC-1␣ in the control
independent lines of PGC-1␣-deficient mouse models have of hepatic gluconeogenesis has been further reinforced by
been established (26, 29). Although myocardial mitochondrial studies in the PGC-1␣-deficient mouse model (26, 29). One
volume density didn’t change significantly in either PGC-1␣ strain of PGC-1␣ knockout mice exhibited fasting hypoglyce-
knockout model, the expression of genes of oxidative phos- mia in response to impaired gluconeogenic gene expression
phorylation was markedly blunted in the PGC-1␣-deficient and hepatic glucose production (26, 29). Although the expres-
heart. This was accompanied with reduced mitochondrial en- sion of PGC-1␣ target genes involved in gluconeogenesis was
zymatic activities and decreased levels of ATP and phospho- not altered in another strain of PGC-1␣-deficient mice, the
creatine (2). The PGC-1␣-deficient heart also exhibits a dimin- hepatic glucose production was reduced secondary to dimin-
ished response to exercise and ␤-adrenergic stimulation in vivo ished fatty acid ␤-oxidation and tricarboxylic acid cycle flux
(26). Consistent with this, it has been reported that there is a (7). Interestingly, after short-term starvation, one line of PGC-
decreased ability to increase both the heart rate and rate of 1␣-deficient mice developed hepatic steatosis due to a combi-
change of pressure in response to chemical or electrical stim- nation of reduced mitochondrial fatty acid oxidation capacity
ulation in a isolated perfused heart preparation of PGC-␣- and an increased expression of lipogenic genes (26). Fasting
deficient mice (2). In addition, starting from 7 to 8 mo of age, also induces PPAR-␣ expression, which, when coactivated by
PGC-1␣-deficient mice exhibit significant cardiac dysfunction PGC-1␣, results in the expression of genes involved in hepatic
(2). In this context, it has been recently reported that PGC-1␣- fatty acid oxidation. The gene expression changes that have
deficient mice underwent accelerated heart failure when chal- been described, under conditions of nutrient deprivation, cause
lenged by transverse aortic constriction (3). These data indicate a shift in fuel usage from glucose utilization to fatty acid
that PGC-1␣ is essential for the heart to be able to meet the oxidation, which contributes to the conservation of glucose for
increased demands for both ATP and work output in response use by the central nervous system (19, 69).
to physiological stimuli. Surprisingly, the in vivo gain of Under fed conditions, skeletal muscle is a major site for
function studies, carried out using two independent transgenic glucose disposal. Skeletal muscle takes up glucose using glu-
mouse models that overexpress PGC-1␣ specifically in the cose transporters (GLUTs) located in the cell membrane. There
heart (MHC-PGC-1␣ mice and tet-on PGC-1␣ mice), revealed are two types of skeletal muscle GLUTs: constitutive, insulin-
modest to dramatic mitochondria biogenesis associated with insensitive GLUT1 and GLUT3 and insulin-sensitive GLUT4.
cardiomyopathy (25, 53). The development of this cardiomy- PGC-1␣ has been shown to increase the expression of GLUT
opathy is thought to be associated with mitochondrial ultra- 4 in cultured muscle cells and skeletal muscle (5, 33, 33).
structural abnormalities and the disruption of sarcomere struc- Whereas MEF2C has been shown to mediate PGC-1␣-induced
ture by overwhelmingly increased mitochondria biogenesis GLUT4 expression in cultured muscle cells (33), the MEF2A
(25, 53). isoform appears to be responsible for GLUT4 upregulation
under other conditions (4, 37). Surprisingly, PGC-1␣ has not
PGC-1␣ and Glucose Metabolism
been shown to increase glucose uptake in skeletal muscle in
Maintenance of plasma glucose homeostasis is vital for the vivo, although it induced an increased glucose uptake into
survival of mammalian organisms, and, as a result, glucose skeletal muscle cells (33). In addition, it has been reported that
levels are tightly regulated in response to nutrient conditions GLUT4 expression is downregulated in a muscle-selective
and hormonal signals. Insulin suppresses plasma glucose level PGC-1␣ transgenic mouse model (34). Resolution of the role
by the inhibition of glucose production in the liver and by of PGC-1␣ in the regulation of skeletal muscle glucose uptake
promotion of glucose disposal into skeletal muscle and white thus requires further investigation.
fat tissue. Conversely, glucagon and glucocorticoids increase It is now well established that insulin also stimulates the
glucose levels through the activation of gluconeogenesis, in recruitment of GLUT4 to the cell membrane of insulin-sensi-
addition to the activation of glycogenolysis, in the liver and tive tissues. A very important area of future research is the
reduction of glucose utilization in skeletal muscle. Under elucidation of the interaction between insulin and PGC-1␣ in
normal, fed conditions, PGC-1␣ is expressed at very low levels the regulation of glucose metabolism, an interaction about
in the liver (49). However, fasting produces a robust increase which there is very little known at the present time. There is
of PGC-1␣ expression, which, in turn, stimulates hepatic some evidence suggesting that insulin has a suppressive effect
gluconeogenesis and fatty acid oxidative metabolism (19, 69). on the expression of PGC-1␣, i.e., PGC-1␣ promotor activity

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
148 PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM

is inhibited by insulin (19). Consistent with this, hepatic PGC-1␣ in mitochondria biogenesis and glucose/fatty acid
PGC-1␣ expression levels have been reported to be increased metabolism. Evidence supporting this hypothesis has been
in animal models exhibiting insulin deficiencies (69). On the found in a number of studies. For example, it has been
other hand, there is evidence showing a suppressive effect of observed that expression of PGC-1␣ is downregulated in mus-
insulin downstream of PGC-1␣. The mechanism for this effect cles of Type 2 diabetic subjects (36, 42, 44). In addition, a
may involve the transcription factor FOXO1. Activation of the common polymorphism of the PGC-1␣ gene (Gly482Ser),
insulin signaling pathway leads to phosphorylation of FOXO1, expressing reduced PGC-1␣ activity, has been linked to an
inhibiting its translocation to the nucleus and increasing its increased risk of Type 2 diabetes (18, 40). These observations
susceptibility to degradation. Because PGC-1␣ requires would suggest that either reduced levels or compromised
FOXO1 to bind to the promotor regions of gluconeogenic activity of PGC-1␣ can be associated with the development of
genes, this action of insulin could have a suppressive effect on insulin resistance and Type 2 diabetes (Fig. 1). In this context,
PGC-1␣ action (47). it is interesting to note that TZDs, an important class of
antidiabetic drugs, also act to increase insulin sensitivity coin-
PGC-1␣ and Type 2 Diabetes
cident with the activation of PGC-1␣ (66). The effects of TZDs
Type 2 diabetes is the most common metabolic disease in the are likely mediated through the ability of PGC-1␣ to activate
world, reaching epidemic proportions. Although there are mul- mitochondria biogenesis and increase mitochondrial function.
tiple complicated genetic elements involved in the pathogene- As described above, PGC-1␣ stimulates the conversion of
sis of Type 2 diabetes, lifestyle and age seem to be the two muscle fiber type toward more oxidative type I and IIa fibers,
major risk factors triggering the disease. A sedentary lifestyle favoring fatty acid oxidative metabolism (28). The promotion
and overeating can contribute to excessive weight gain and of fatty acid oxidative metabolism would be expected to lead to
obesity, with the latter being closely associated with insulin a reduction of fat accumulation in muscle, which, in turn, could
resistance. Insulin resistance is considered to be present when increase insulin sensitivity because there is a close relationship
the biological effects of insulin are less than expected for both between triglyceride accumulation and insulin resistance (43).
glucose disposal in skeletal muscle and suppression of glucose One manifestation of increased insulin sensitivity would be
production by the liver (11). When the insulin level is not increased glucose uptake by insulin-sensitive tissues. PGC-1␣
sufficient to overcome this insulin-resistant state, Type 2 dia- has, as previously noted, been reported to activate the expres-
betes ensues. In this regard, there is there is increasing evi- sion of insulin-sensitive GLUT4 in skeletal muscle (5, 33).
dence suggesting that mitochondrial dysfunction plays a role in Taken together, the evidence presented supports a role for
the pathogenesis of insulin resistance and Type 2 diabetes. For PGC-1␣ in preventing insulin resistance and Type 2 diabetes
example, it has been reported that the activities of both mito- mellitus.
chondrial oxidative enzymes and mitochondria complex I are However, it should be pointed out that PGC-1␣ expression
decreased in Type 2 diabetic patients (21, 63). In keeping with has been reported to be increased in the liver of both Type 1
the observations that both obesity and mitochondrial dysfunc- and Type 2 diabetic mouse models, in contrast to the reported
tion are risk factors for the development of insulin resistance, decrease in PGC-1␣ expression observed in the muscle of
obese individuals have been reported to have smaller mito- human Type 2 diabetic subjects described above (48). Further-
chondria that exhibit a compromised bioenergetic capacity more, PGC-1␣ has been shown to inhibit the insulin signaling
(21). Insulin resistance also develops with age, and, in this pathway in the liver (22), inhibit glucose utilization in cultured
case, a potentially important defect has been found in the myotubes (65), and suppress ␤-cell energy metabolism and
mitochondrial fatty acid oxidation pathway. There appears to insulin release in mice (70). Increased hepatic PGC-1␣ expres-
be a reduction in the rate of fatty acid oxidation, leading to the sion could be expected to stimulate hepatic glucose output,
accumulation of intracellular triglycerides in elderly patients
compared with matched young controls (43). Furthermore, the
accumulation of triglycerides has been directly correlated with
the development of insulin resistance in skeletal muscle and the
liver. Thus, a specific type of mitochondrial dysfunction, i.e.,
decreased fatty acid oxidation, may play a significant role in
the development of insulin resistance in both aging and obesity
(43). It is interesting to note that triglycerides can also accu-
mulate in pancreatic ␤-cells, resulting in decreased rates of
insulin secretion, which would further exacerbate the problem
of glucose disposal (32, 60). As noted above, the accumulation
of intracellular triglycerides could be the result of a deficiency
in fatty acid oxidation related to mitochondrial dysfunction. As
a result, the cause of insulin resistance may well be due to
mitochondrial dysfunction rather than the accumulation of
triglycerides, a mechanistic concept that will be important to
establish in gaining an understanding of the etiology of Type 2
diabetes.
Fig. 1. Potential role of decreased peroxisome proliferator-activated recep-
It has been hypothesized that a close relationship exists tor-␥ coactivator (PGC)-1␣ expression in skeletal muscle as it relates to the
among PGC-1␣ function, insulin sensitivity, and Type 2 dia- development of the metabolic phenotype of insulin resistance and Type 2
betes, which is most likely related to the essential roles of diabetes mellitus. [Revised from Ref. 34.]

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM 149

and, when coupled with the reported inhibitory effect of proteins and uncoupling protein gene promoter reveal several putative
PGC-1␣ on insulin signaling and secretion, the combined transactivating factors including Ets1, retinoid X receptor, thyroid hor-
mone receptor, and a CACCC box-binding protein. J Biol Chem 269:
effects would be to contribute to the hyperglycemic state, a 24335–24342, 1994.
prodiabetes effect. This prodiabetic effect of PGC-1␣ has been 10. Czubryt MP, McAnally J, Fishman GI, and Olson EN. Regulation of
further supported by the manifestation of improved insulin peroxisome proliferator-activated receptor gamma coactivator 1 alpha
sensitivity in PGC-1␣-deficient mouse models (22, 26, 29). (PGC-1 alpha) and mitochondrial function by MEF2 and HDAC5. Proc
Moreover, another recent report (38) has indicated that Natl Acad Sci USA 100: 1711–1716, 2003.
11. Dinneen S, Gerich J, and Rizza R. Carbohydrate metabolism in non-
PGC-1␣ expression is normal in insulin-resistant subjects de- insulin-dependent diabetes mellitus. N Engl J Med 327: 707–713, 1992.
spite severe impairments in mitochondrial function. These 12. Finck BN and Kelly DP. PGC-1 coactivators: inducible regulators of
apparently paradoxical actions of PGC-1␣ in different tissues energy metabolism in health and disease. J Clin Invest 116: 615– 622,
are intriguing. The underlining mechanisms for the interactions 2006.
between these tissues are currently unknown and deserve 13. Garcia-Roves PM, Huss J, and Holloszy JO. Role of calcineurin in
exercise-induced mitochondrial biogenesis. Am J Physiol Endocrinol
further intensive investigations. Metab 290: E1172–E1179, 2006.
14. Garesse R and Vallejo CG. Animal mitochondrial biogenesis and func-
Summary tion: a regulatory cross-talk between two genomes. Gene 263: 1–16, 2001.
As an inducible transcription coactivator, PGC-1␣ is en- 15. Goto M, Terada S, Kato M, Katoh M, Yokozeki T, Tabata I, and
Shimokawa T. cDNA Cloning and mRNA analysis of PGC-1 in epitroch-
riched in metabolically active tissues. It is intimately involved learis muscle in swimming-exercised rats. Biochem Biophys Res Commun
in adaptive thermogenesis, skeletal muscle fiber type switch- 274: 350 –354, 2000.
ing, glucose/fatty acid metabolism, and heart development. 16. Guan HP, Ishizuka T, Chui PC, Lehrke M, and Lazar MA. Corepres-
Among these varied biological responses, a common mecha- sors selectively control the transcriptional activity of PPARgamma in
nism of action appears to be the promotion of oxidative adipocytes. Genes Dev 19: 453– 461, 2005.
17. Handschin C, Rhee J, Lin J, Tarr PT, and Spiegelman BM. An
metabolism accompanying the stimulation of mitochondria autoregulatory loop controls peroxisome proliferator-activated receptor
biogenesis. Furthermore, there is growing evidence that gamma coactivator 1alpha expression in muscle. Proc Natl Acad Sci USA
PGC-1␣ plays a role in disorders such as obesity, diabetes, and 100: 7111–7116, 2003.
cardiomyopathy. Because of the involvement of PGC-1␣ in so 18. Hara K, Tobe K, Okada T, Kadowaki H, Akanuma Y, Ito C, Kimura
many important biological processes, in conjunction with the S, and Kadowaki T. A genetic variation in the PGC-1 gene could confer
insulin resistance and susceptibility to Type II diabetes. Diabetologia 45:
rapid improvement in our understanding of its mechanisms of 740 –743, 2002.
action, it has become an inviting target for the design of 19. Herzig S, Long F, Jhala US, Hedrick S, Quinn R, Bauer A, Rudolph
pharmacological interventions for treatment of these disorders. D, Schutz G, Yoon C, Puigserver P, Spiegelman B, and Montminy M.
CREB regulates hepatic gluconeogenesis through the coactivator PGC-1.
REFERENCES Nature 413: 179 –183, 2001.
20. Huss JM, Kopp RP, and Kelly DP. Peroxisome proliferator-activated
1. Akimoto T, Pohnert SC, Li P, Zhang M, Gumbs C, Rosenberg PB, receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched
Williams RS, and Yan Z. Exercise stimulates Pgc-1alpha transcription in nuclear receptors estrogen-related receptor-alpha and -gamma. Identifica-
skeletal muscle through activation of the p38 MAPK pathway. J Biol tion of novel leucine-rich interaction motif within PGC-1alpha. J Biol
Chem 280: 19587–19593, 2005. Chem 277: 40265– 40274, 2002.
2. Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, Ahmad F, 21. Kelley DE, He J, Menshikova EV, and Ritov VB. Dysfunction of
Matsui T, Chin S, Wu PH, Rybkin II, Shelton JM, Manieri M, Cinti mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51:
S, Schoen FJ, Bassel-Duby R, Rosenzweig A, Ingwall JS, and 2944 –2950, 2002.
Spiegelman BM. Transcriptional coactivator PGC-1 alpha controls the
22. Koo SH, Satoh H, Herzig S, Lee CH, Hedrick S, Kulkarni R, Evans
energy state and contractile function of cardiac muscle. Cell Metab 1:
RM, Olefsky J, and Montminy M. PGC-1 promotes insulin resistance in
259 –271, 2005.
liver through PPAR-alpha-dependent induction of TRB-3. Nat Med 10:
3. Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, and Spiegelman
530 –534, 2004.
BM. Transverse aortic constriction leads to accelerated heart failure in
23. Kressler D, Schreiber SN, Knutti D, and Kralli A. The PGC-1-related
mice lacking PPAR-gamma coactivator 1alpha. Proc Natl Acad Sci USA
protein PERC is a selective coactivator of estrogen receptor alpha. J Biol
103: 10086 –10091, 2006.
4. Baar K, Song Z, Semenkovich CF, Jones TE, Han DH, Nolte LA, Chem 277: 13918 –13925, 2002.
Ojuka EO, Chen M, and Holloszy JO. Skeletal muscle overexpression 24. Larsson NG, Wang J, Wilhelmsson H, Oldfors A, Rustin P, Lewan-
of nuclear respiratory factor 1 increases glucose transport capacity. FASEB doski M, Barsh GS, and Clayton DA. Mitochondrial transcription factor
J 17: 1666 –1673, 2003. A is necessary for mtDNA maintenance and embryogenesis in mice. Nat
5. Baar K, Wende AR, Jones TE, Marison M, Nolte LA, Chen M, Kelly Genet 18: 231–236, 1998.
DP, and Holloszy JO. Adaptations of skeletal muscle to exercise: rapid 25. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, and
increase in the transcriptional coactivator PGC-1. FASEB J 16: 1879 – Kelly DP. Peroxisome proliferator-activated receptor gamma coactiva-
1886, 2002. tor-1 promotes cardiac mitochondrial biogenesis. J Clin Invest 106:
6. Barbera MJ, Schluter A, Pedraza N, Iglesias R, Villarroya F, and 847– 856, 2000.
Giralt M. Peroxisome proliferator-activated receptor alpha activates tran- 26. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina
scription of the brown fat uncoupling protein-1 gene. A link between S, Courtois M, Wozniak DF, Sambandam N, Bernal-Mizrachi C,
regulation of the thermogenic and lipid oxidation pathways in the brown Chen Z, Holloszy JO, Medeiros DM, Schmidt RE, Saffitz JE, Abel ED,
fat cell. J Biol Chem 276: 1486 –1493, 2001. Semenkovich CF, and Kelly DP. PGC-1alpha deficiency causes multi-
7. Burgess SC, Leone TC, Wende AR, Croce MA, Chen Z, Sherry AD, system energy metabolic derangements: muscle dysfunction, abnormal
Malloy CR, and Finck BN. Diminished hepatic gluconeogenesis via weight control and hepatic steatosis. PLoS Biol 3: e101, 2005.
defects in tricarboxylic acid cycle flux in peroxisome proliferator-activated 27. Lin J, Handschin C, and Spiegelman BM. Metabolic control through the
receptor␥ coactivator-1␣ (PGC-1␣)-deficient mice. J Biol Chem 281: PGC-1 family of transcription coactivators. Cell Metab 1: 361–370, 2005.
19000 –19008, 2006. 28. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael
8. Cannon B, Houstek J, and Nedergaard J. Brown adipose tissue. More LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel-Duby R,
than an effector of thermogenesis? Ann NY Acad Sci 856: 171–187, 1998. and Spiegelman BM. Transcriptional co-activator PGC-1 alpha drives
9. Cassard-Doulcier AM, Larose M, Matamala JC, Champigny O, the formation of slow-twitch muscle fibres. Nature 418: 797– 801,
Bouillaud F, and Ricquier D. In vitro interactions between nuclear 2002.

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
150 PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM

29. Lin J, Wu PH, Tarr PT, Lindenberg KS, St Pierre J, Zhang CY, 46. Puigserver P, Adelmant G, Wu Z, Fan M, Xu J, O’Malley B, and
Mootha VK, Jager S, Vianna CR, Reznick RM, Cui L, Manieri M, Spiegelman BM. Activation of PPARgamma coactivator-1 through tran-
Donovan MX, Wu Z, Cooper MP, Fan MC, Rohas LM, Zavacki AM, scription factor docking. Science 286: 1368 –1371, 1999.
Cinti S, Shulman GI, Lowell BB, Krainc D, and Spiegelman BM. 47. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente F,
Defects in adaptive energy metabolism with CNS-linked hyperactivity in Kitamura Y, Altomonte J, Dong H, Accili D, and Spiegelman BM.
PGC-1alpha null mice. Cell 119: 121–135, 2004. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha
30. Lin J, Yang R, Tarr PT, Wu PH, Handschin C, Li S, Yang W, Pei L, interaction. Nature 423: 550 –555, 2003.
Uldry M, Tontonoz P, Newgard CB, and Spiegelman BM. Hyperlip- 48. Puigserver P and Spiegelman BM. Peroxisome proliferator-activated
idemic effects of dietary saturated fats mediated through PGC-1beta receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coac-
coactivation of SREBP. Cell 120: 261–273, 2005. tivator and metabolic regulator. Endocr Rev 24: 78 –90, 2003.
31. Lowell BB. PPARgamma: an essential regulator of adipogenesis and 49. Puigserver P, Wu Z, Park CW, Graves R, Wright M, and Spiegelman
modulator of fat cell function. Cell 99: 239 –242, 1999. BM. A cold-inducible coactivator of nuclear receptors linked to adaptive
32. Lowell BB and Shulman GI. Mitochondrial dysfunction and type 2 thermogenesis. Cell 92: 829 – 839, 1998.
diabetes. Science 307: 384 –387, 2005. 50. Rangwala SM and Lazar MA. Peroxisome proliferator-activated recep-
33. Michael LF, Wu Z, Cheatham RB, Puigserver P, Adelmant G, Leh- tor gamma in diabetes and metabolism. Trends Pharmacol Sci 25: 331–
man JJ, Kelly DP, and Spiegelman BM. Restoration of insulin-sensitive 336, 2004.
glucose transporter (GLUT4) gene expression in muscle cells by the 51. Rothwell NJ and Stock MJ. A role for brown adipose tissue in diet-
transcriptional coactivator PGC-1. Proc Natl Acad Sci USA 98: 3820 – induced thermogenesis. Nature 281: 31–35, 1979.
3825, 2001. 52. Russell AP, Feilchenfeldt J, Schreiber S, Praz M, Crettenand A,
34. Miura S, Kai Y, Ono M, and Ezaki O. Overexpression of peroxisome Gobelet C, Meier CA, Bell DR, Kralli A, Giacobino JP, and Deriaz O.
proliferator-activated receptor gamma coactivator-1alpha down-regulates Endurance training in humans leads to fiber type-specific increases in
GLUT4 mRNA in skeletal muscles. J Biol Chem 278: 31385–31390, levels of peroxisome proliferator-activated receptor-gamma coactivator-1
2003. and peroxisome proliferator-activated receptor-alpha in skeletal muscle.
35. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S, Yang Diabetes 52: 2874 –2881, 2003.
W, Altshuler D, Puigserver P, Patterson N, Willy PJ, Schulman IG, 53. Russell LK, Mansfield CM, Lehman JJ, Kovacs A, Courtois M, Saffitz
Heyman RA, Lander ES, and Spiegelman BM. Err␣ and Gabpa/b JE, Medeiros DM, Valencik ML, McDonald JA, and Kelly DP.
specify PGC-1␣ -dependent oxidative phosphorylation gene expression Cardiac-specific induction of the transcriptional coactivator peroxisome
that is altered in diabetic muscle. Proc Natl Acad Sci USA 101: 6570 – proliferator-activated receptor gamma coactivator-1alpha promotes mito-
6575, 2004. chondrial biogenesis and reversible cardiomyopathy in a developmental
36. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, stage-dependent manner. Circ Res 94: 525–533, 2004.
Lehar J, Puigserver P, Carlsson E, Ridderstrale M, Laurila E, Houstis 54. Sack MN, Disch DL, Rockman HA, and Kelly DP. A role for Sp and
N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, nuclear receptor transcription factors in a cardiac hypertrophic growth
Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, and Groop program. Proc Natl Acad Sci USA 94: 6438 – 6443, 1997.
LC. PGC-1alpha-responsive genes involved in oxidative phosphorylation 55. Sano M, Wang SC, Shirai M, Scaglia F, Xie M, Sakai S, Tanaka T,
are coordinately downregulated in human diabetes. Nat Genet 34: 267– Kulkarni PA, Barger PM, Youker KA, Taffet GE, Hamamori Y,
273, 2003. Michael LH, Craigen WJ, and Schneider MD. Activation of cardiac
37. Mora S and Pessin JE. The MEF2A isoform is required for striated Cdk9 represses PGC-1 and confers a predisposition to heart failure. EMBO
muscle-specific expression of the insulin-responsive GLUT4 glucose J 23: 3559 –3569, 2004.
transporter. J Biol Chem 275: 16323–16328, 2000. 56. Scarpulla RC. Nuclear activators and coactivators in mammalian mito-
38. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes N, chondrial biogenesis. Biochim Biophys Acta 1576: 1–14, 2002.
Neschen S, White MF, Bilz S, Sono S, Pypaert M, and Shulman GI. 57. Scarpulla RC. Transcriptional activators and coactivators in the nuclear
Reduced mitochondrial density and increased IRS-1 serine phosphoryla- control of mitochondrial function in mammalian cells. Gene 286: 81– 89,
tion in muscle of insulin-resistant offspring of type 2 diabetic parents. 2002.
J Clin Invest 115: 3587–3593, 2005. 58. Sears IB, MacGinnitie MA, Kovacs LG, and Graves RA. Differentia-
39. Mortensen OH, Frandsen L, Schjerling P, Nishimura E, and Grunnet tion-dependent expression of the brown adipocyte uncoupling protein
N. PGC-1␣ and PGC-1␤ have both similar and distinct effects upon gene: regulation by peroxisome proliferator-activated receptor gamma.
myofiber switching towards an oxidative phenotype. Am J Physiol Endo- Mol Cell Biol 16: 3410 –3419, 1996.
crinol Metab 291: E807–E816, 2006. 59. Spiegelman BM. PPAR-gamma: adipogenic regulator and thiazolidinedi-
40. Muller YL, Bogardus C, Pedersen O, and Baier L. A Gly482Ser one receptor. Diabetes 47: 507–514, 1998.
missense mutation in the peroxisome proliferator-activated receptor 60. Unger RH. Lipotoxicity in the pathogenesis of obesity-dependent
gamma coactivator-1 is associated with altered lipid oxidation and early NIDDM. Genetic and clinical implications. Diabetes 44: 863– 870, 1995.
insulin secretion in Pima Indians. Diabetes 52: 895– 898, 2003. 61. van Raalte DH, Li M, Pritchard PH, and Wasan KM. Peroxisome
41. Norrbom J, Sundberg CJ, Ameln H, Kraus WE, Jansson E, and proliferator-activated receptor (PPAR)-alpha: a pharmacological target
Gustafsson T. PGC-1␣ mRNA expression is influenced by metabolic with a promising future. Pharm Res 21: 1531–1538, 2004.
perturbation in exercising human skeletal muscle. J Appl Physiol 96: 62. Vega RB, Huss JM, and Kelly DP. The coactivator PGC-1 cooperates
189 –194, 2004. with peroxisome proliferator-activated receptor alpha in transcriptional
42. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, control of nuclear genes encoding mitochondrial fatty acid oxidation
Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldfine enzymes. Mol Cell Biol 20: 1868 –1876, 2000.
AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, and Mandarino LJ. 63. Vondra K, Rath R, Bass A, Slabochova Z, Teisinger J, and Vitek V.
Coordinated reduction of genes of oxidative metabolism in humans with Enzyme activities in quadriceps femoris muscle of obese diabetic male
insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc patients. Diabetologia 13: 527–529, 1977.
Natl Acad Sci USA 100: 8466 – 8471, 2003. 64. Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, and Evans RM.
43. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, Peroxisome-proliferator-activated receptor delta activates fat metabolism
DiPietro L, Cline GW, and Shulman GI. Mitochondrial dysfunction in to prevent obesity. Cell 113: 159 –170, 2003.
the elderly: possible role in insulin resistance. Science 300: 1140 –1142, 65. Wende AR, Huss JM, Schaeffer PJ, Giguere V, and Kelly DP.
2003. PGC-1alpha coactivates PDK4 gene expression via the orphan nuclear
44. Petersen KF, Dufour S, Befroy D, Garcia R, and Shulman GI. receptor ERRalpha: a mechanism for transcriptional control of muscle
Impaired mitochondrial activity in the insulin-resistant offspring of pa- glucose metabolism. Mol Cell Biol 25: 10684 –10694, 2005.
tients with type 2 diabetes. N Engl J Med 350: 664 – 671, 2004. 66. Wilson-Fritch L, Nicoloro S, Chouinard M, Lazar MA, Chui PC,
45. Puigserver P. Tissue-specific regulation of metabolic pathways through Leszyk J, Straubhaar J, Czech MP, and Corvera S. Mitochondrial
the transcriptional coactivator PGC1-alpha. Int J Obes (Lond) 29, Suppl 1: remodeling in adipose tissue associated with obesity and treatment with
S5–S9, 2005. rosiglitazone. J Clin Invest 114: 1281–1289, 2004.

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.
Staying Current
PGC-1␣: A KEY REGULATOR OF ENERGY METABOLISM 151

67. Wu H, Kanatous SB, Thurmond FA, Gallardo T, Isotani E, Bassel- energy metabolism and insulin release by PGC-1alpha. Dev Cell 5: 73– 83,
Duby R, and Williams RS. Regulation of mitochondrial biogenesis in 2003.
skeletal muscle by CaMK. Science 296: 349 –352, 2002. 71. Zhang Y, Castellani LW, Sinal CJ, Gonzalez FJ, and Edwards PA.
68. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Peroxisome proliferator-activated receptor-gamma coactivator 1alpha
Troy A, Cinti S, Lowell B, Scarpulla RC, and Spiegelman BM. (PGC-1alpha) regulates triglyceride metabolism by activation of the nu-
Mechanisms controlling mitochondrial biogenesis and respiration through clear receptor FXR. Genes Dev 18: 157–169, 2004.
the thermogenic coactivator PGC-1. Cell 98: 115–124, 1999. 72. Zhang Y, Ma K, Song S, Elam MB, Cook GA, and Park EA. Peroxisomal
69. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, Adelmant proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1 alpha) en-
G, Stafford J, Kahn CR, Granner DK, Newgard CB, and Spiegelman hances the thyroid hormone induction of carnitine palmitoyltransferase I
BM. Control of hepatic gluconeogenesis through the transcriptional coac- (CPT-I alpha). J Biol Chem 279: 53963–53971, 2004.
tivator PGC-1. Nature 413: 131–138, 2001. 73. Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ, and
70. Yoon JC, Xu G, Deeney JT, Yang SN, Rhee J, Puigserver P, Levens Shulman GI. AMP kinase is required for mitochondrial biogenesis in
AR, Yang R, Zhang CY, Lowell BB, Berggren PO, Newgard CB, skeletal muscle in response to chronic energy deprivation. Proc Natl Acad
Bonner-Weir S, Weir G, and Spiegelman BM. Suppression of beta cell Sci USA 99: 15983–15987, 2002.

Advances in Physiology Education • VOL 30 • DECEMBER 2006


Downloaded from journals.physiology.org/journal/advances (078.083.126.042) on February 20, 2024.

You might also like