You are on page 1of 8

Huiyun Liang and Walter F.

Ward
Advan Physiol Educ 30:145-151, 2006. doi:10.1152/advan.00052.2006 You might find this additional information useful... This article cites 73 articles, 39 of which you can access free at: http://ajpadvan.physiology.org/cgi/content/full/30/4/145#BIBL This article has been cited by 12 other HighWire hosted articles, the first 5 are: PPAR{delta} and PGC1{alpha} act cooperatively to induce haem oxygenase-1 and enhance vascular endothelial cell resistance to stress F. Ali, N. S. Ali, A. Bauer, J. J. Boyle, S. S. Hamdulay, D. O. Haskard, A. M. Randi and J. C. Mason Cardiovasc Res, December 3, 2009; 0 (2009): cvp365v2-cvp365. [Abstract] [Full Text] [PDF] Exercise, sex, menstrual cycle phase, and 17{beta}-estradiol influence metabolism-related genes in human skeletal muscle M.-h. H. Fu, A. C. Maher, M. J. Hamadeh, C. Ye and M. A. Tarnopolsky Physiol Genomics, December 1, 2009; 40 (1): 34-47. [Abstract] [Full Text] [PDF] Is there an interaction between PPARD T294C and PPARGC1A Gly482Ser polymorphisms and human endurance performance? N. Eynon, Y. Meckel, A. J. Alves, C. Yamin, M. Sagiv, E. Goldhammer and M. Sagiv Exp Physiol, November 1, 2009; 94 (11): 1147-1152. [Abstract] [Full Text] [PDF] Functional genomic analysis of frataxin deficiency reveals tissue-specific alterations and identifies the PPAR{gamma} pathway as a therapeutic target in Friedreich's ataxia G. Coppola, D. Marmolino, D. Lu, Q. Wang, M. Cnop, M. Rai, F. Acquaviva, S. Cocozza, M. Pandolfo and D. H. Geschwind Hum. Mol. Genet., July 1, 2009; 18 (13): 2452-2461. [Abstract] [Full Text] [PDF] Mutual Dependence of Foxo3a and PGC-1{alpha} in the Induction of Oxidative Stress Genes Y. Olmos, I. Valle, S. Borniquel, A. Tierrez, E. Soria, S. Lamas and M. Monsalve J. Biol. Chem., May 22, 2009; 284 (21): 14476-14484. [Abstract] [Full Text] [PDF] Medline items on this article's topics can be found at http://highwire.stanford.edu/lists/artbytopic.dtl on the following topics: Biochemistry .. Mitochondria Biogenesis Cell Biology .. Peroxisomes Physiology .. Energy Metabolism Medicine .. Genetics Medicine .. Diabetes Genetics Medicine .. Obesity Updated information and services including high-resolution figures, can be found at: http://ajpadvan.physiology.org/cgi/content/full/30/4/145 Additional material and information about Advances in Physiology Education can be found at: http://www.the-aps.org/publications/advan

Downloaded from ajpadvan.physiology.org on January 28, 2010

This information is current as of January 28, 2010 .

Advances in Physiology Education is dedicated to the improvement of teaching and learning physiology, both in specialized courses and in the broader context of general biology education. It is published four times a year in March, June, September and December by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright 2005 by the American Physiological Society. ISSN: 1043-4046, ESSN: 1522-1229. Visit our website at http://www.the-aps.org/.

Adv Physiol Educ 30: 145151, 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
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 nal form 27 July 2006
1

Liang, Huiyun, and Walter F. Ward. PGC-1 : a key regulator of energy metabolism. Adv Physiol Educ 30: 145151, 2006; doi: 10.1152/advan.00052.2006.Peroxisome proliferator-activated receptor- coactivator (PGC)-1 is a member of a family of transcription coactivators that plays a central role in the regulation of cellular energy metabolism. It is strongly induced by cold exposure, linking this environmental stimulus to adaptive thermogenesis. PGC-1 stimulates mitochondrial biogenesis and promotes the remodeling of muscle tissue to a ber-type composition that is metabolically more oxidative and less glycolytic in nature, and it participates in the regulation of both carbohydrate and lipid metabolism. It is highly likely that PGC-1 is intimately involved in disorders such as obesity, diabetes, and cardiomyopathy. In particular, its regulatory function in lipid metabolism makes it an inviting target for pharmacological intervention in the treatment of obesity and Type 2 diabetes. adaptive thermogenesis; transcription coactivator; proliferator peroxisome-activated receptor; glucose metabolism; diabetes

very low in white adipose tissue (WAT) (49). It is noteworthy that PGC-1 does not exhibit histone acetyltransferase (HAT) activity itself, but the conformational change it induces after docking on specic transcription factors increases the afnity of the transcription complex to additional coactivators possessing HAT activity, such as steroid receptor coactivator-1 and cAMP response-binding element (CREB)-binding protein and p300. The end result is acetylation of histone proteins, which produces conformation alterations that increase the accessibility of DNA to the transcription complex (46). Although there are three members of the PGC-1 family [PGC-1 , PGC-1 , and PGC-1-related coactivator (48)], we will focus on PGC-1 in this review. There is currently a great interest in PGC-1 because rapidly growing evidence strongly suggests that it is a powerful regulator of energy metabolism under conditions of both health and disease (12). PGC-1 and Adaptive Thermogenesis PGC-1 was originally discovered as a cold-inducible transcription coactivator of adaptive thermogenesis, a physiological process through which energy is dissipated as heat in response to environmental conditions such as cold stress and overfeeding (8, 51). BAT and skeletal muscle are the two major organs involved in this process. While rats and mice have prominent brown fat depots, this isnt the case in larger mammals, including humans, although there may be brown fat cells dispersed among the adipocytes of WAT (49). Thus, in larger mammals, skeletal muscle is generally thought to be the major thermogenic tissue. The primary physiological function of WAT is energy storage, whereas the primary function of BAT is energy dissipation, largely in the form of heat. In fact, BAT produces 60% of the heat generated by nonshivering, adaptive thermogenesis. Spurred on in part by the prevalence of human obesity, interest in adaptive thermogenesis has increased because of the possibility that it may provide a physiological defense against obesity (68). This has also led to increased attention being directed toward the regulation of adipose tissue differentiation. It was known that the transcription factor PPAR- is an important regulator of adipocyte differentiation, but, surprisingly, it is unable, by itself, to induce brown fat differentiation (45). It was during the search for the solution to this puzzle that PGC-1 was discovered (49). PGC-1 binds to PPAR- and coactivates PPAR- to stimulate the transcription of genes involved in the brown adipocyte differentiation process (49). The adaptive thermogenic program in both brown fat and skeletal muscle involves the stimulation of mitochondria biogenesis, increased fatty acid oxidation, and the uncoupling of oxidative phosphorylation. In response to cold exposure,
145

Downloaded from ajpadvan.physiology.org on January 28, 2010

(PPAR)- coactivator (PGC)-1 is a transcription coactivator that interacts with a broad range of transcription factors that are involved in a wide variety of biological responses including adaptive thermogenesis, mitochondrial biogenesis, glucose/fatty acid metabolism, ber type switching in skeletal muscle, and heart development (Table 1). A transcription coactivator is dened as a protein or protein complex that increases the probability of a gene being transcribed by interacting with transcription factors but does not itself bind to DNA in a sequence-specic manner (48). The PPARs (PPAR- , PPAR- , and PPAR- ) are members of a relatively large family of nuclear receptors, all of which are subject to transcriptional coactivation by PGC-1 . PPAR- is essential for adipogenesis and differentiation (31), whereas PPAR- and PPAR- play important roles in the control of fatty acid oxidation (61). In addition, PPAR- is the target of thiazolidinediones (TZDs), which are drugs that are widely utilized for increasing insulin sensitivity in diabetic patients (50, 59). The PGC-1 gene is located on chromosome 5 in mice (chromosome 4 in humans) and encodes a protein containing 797 (mouse) or 798 amino acids (human). PGC-1 has two putative nuclear localization signals and is located in the cell nucleus (49). It is expressed at high levels in tissues where mitochondria are abundant and oxidative metabolism is active, such as brown adipose tissue (BAT), the heart, and skeletal muscle. PGC-1 expression is also high in the brain and kidney, whereas the expression level is low in the liver and
Address for reprint requests and other correspondence: W. F. Ward, Dept. of Physiology, Univ. of Texas Health Science Center, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900 (e-mail: wardw@uthscsa.edu).

PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR

Staying Current
146 PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM

Table 1. Selected list of transcription factors for which PGC-1 functions as a coactivator
Transcription Factor Function Reference

NRF1 NRF2 ERR- / / PPARPPARTRFXR LXR- / GR HNF-4 FOXO1 PPAR-

Mitochondria biogenesis Mitochondria biogenesis Mitochondria biogenesis Fatty acid oxidation Fatty acid oxidation CPT-I induction Triglyceride metabolism Lipoprotein secretion Gluconeogenesis Gluconeogenesis Gluconeogenesis Brown adipocyte differentiation; UCP1 induction

68 36 35 62 64 67, 72 71 30 23 39 47 16, 20

PGC-1 , proliferator peroxisome-activated receptor (PPAR)- coactivator1 ; NRF, nuclear respiratory factor; ERR, estrogen-related receptor; TR, thyroid receptor; CPT-I, carnitine palmitoyltransferase-I; FXR, farnesoid X receptor; LXR, liver X receptor; GR, glucocorticoid receptor; HNF, hepatic nuclear factor; FOXO1, forkhead box O1; UCP1, uncoupling protein-1. [Modied from Ref. 23.]

PGC-1 expression is increased, with the induction being mediated through the sympathetic nervous systems 3-adrenergic receptor/cAMP pathway (49). Increased PGC-1 induces the transcription of nuclear respiratory factor (NRF)1 and NRF2, leading to the increased expression of mitochondrial transcription factor A (mtTFA) (68) as well as other nuclearencoded mitochondria subunits of the electron transport chain complex such as -ATP synthase, cytochrome c, and cytochrome c oxidase IV (56, 57). mtTFA translocates to the mitochondrion, where it stimulates mitochondrial biogenesis as manifested by stimulation of mitochondrial DNA replication and mitochondria gene expression (14, 24). PGC-1 also interacts with other nuclear hormone receptors such as PPAR- , retinoic acid receptor, and thyroid receptor in BAT to enhance the expression of brown fat-specic uncoupling protein 1 (UCP1) (6, 9, 58). UCP1 action leads to dissipation of the proton gradient and the uncoupling of oxidative phosphorylation, thereby increasing heat production. Under these conditions, there is a marked increase in the rate of energy metabolism. The essential role of PGC-1 in adaptive thermogenesis is convincingly demonstrated by the observation that the PGC-1 -decient mice are unable to withstand a cold stress (4C) for longer than 6 h due to a continuous decrease of core body temperature. Wild-type control mice, on the other hand, were able to tolerate the cold stress by keeping their core body temperature at 36.5C, after an initial drop of 1.5C (26, 29). PGC-1 and Skeletal Muscle Fiber Conversion Skeletal muscle bers are classied into three types: type I, type IIa, and type IIb. Slow-twitch type I and fast-twitch type IIa bers contain more mitochondria and exhibit relatively higher rates of oxidative metabolism. In contrast, type IIb bers have fewer mitochondria and are metabolically glycolytic. It is now well established that PGC-1 induces a remodeling of skeletal muscle ber composition. In general, the ratio of glycolytic type IIb bers to the more oxidative type I and type IIa bers decreases. The expression of PGC-1 in skeletal muscle is readily inducible by both short-term exercise and

endurance training in rodent models and human subjects (5, 15, 41, 52). Our understanding of the biological role of PGC-1 in skeletal muscle structure and function has been greatly improved through the use of gain of function and loss of function mouse models. In a gain of function transgenic model, PGC-1 is overexpressed in a skeletal muscle-specic manner under the control of the muscle creatine kinase (MCK) promoter (28). PGC-1 overexpression results in the conversion of fast-twitch type IIb muscle bers to type IIa and slow-twitch type I bers by 20% and 10%, respectively, in plantaris muscle. In addition, there is an activation of genes involved in mitochondrial oxidative metabolism. The conversion to slow-twitch bers is also evidenced by the redder muscle color and the expression of contractile proteins characteristic of slow-twitch bers such as slow troponin I and myoglobin. As would be predicted, based on these alterations, muscles isolated from the MCKPGC-1 transgenic mouse show increased resistance to electrically stimulated fatigue (28). Consistent with this, Mortenson et al. (39) recently reported that overexpression of PGC-1 in primary rat skeletal muscle cells leads to enhanced levels of mRNA for the slow oxidative-associated myosin heavy chain (MHC) isoform (MHCIb) and decreased mRNA levels for the fast glycolytic-associated MHC isoforms (MHCIIX and MHCIIB) (39). In contrast, PGC-1 -decient mice exhibit decreased mitochondrial number and decreased respiratory capacity in slow-twitch muscle as well as reduced exercise capacity and a reduced fatigue resistance index (26). The upstream signaling involved in the activation of PGC-1 are unclear at present, but several pathways have been implicated, such as the calcineurin A and CaMK, p38 MAPK, and AMP-activated protein kinase pathways (1, 67, 73). Although the relative importance of these kinases in controlling PGC-1 is yet to be determined, there appears to be a considerable amount of evidence suggesting that the calcineurin and CaMK pathways play important roles in the regulation of PGC-1 expression. During exercise, the combination of increased neuromuscular input and increased contractile activity induces the expression of several transcription factors, e.g., myocyte-specic enhancer factor (MEF)2 and CREB (10, 17). These induction processes appear to be mediated by calcineurin and CaMK (67). Increased expression of MEF2 leads to increased MEF2 binding to the promoter region of the PGC-1 gene, increasing PGC-1 expression. PGC-1 can then bind directly to MEF2 to coactivate the transcription of those genes involved in the determination of slow-twitch ber types and mitochondrial oxidative metabolism (10, 27). Finally, it should be pointed out that recent work from Holloszys laboratory (13) suggests that calcineurin may not be required for exercise-induced increases in PGC-1 and a range of mitochondrial proteins. Obviously, further work is needed to elucidate the roles of calcineurin and calcium signaling pathways in the regulation of PGC-1 . PGC-1 and Heart Development The heart, an organ with a very high and dynamic demand for ATP, derives most of the required ATP from fatty acid oxidation. In the developing mouse heart, as the energy source turns toward mitochondria fatty acid oxidation after birth, there is a large burst of mitochondria biogenesis and oxidative metabolism that is preceded by a dramatically increased expression of PGC-1 (25). Short-term fasting, another condition
30 DECEMBER 2006

Downloaded from ajpadvan.physiology.org on January 28, 2010

Advances in Physiology Education VOL

Staying Current
PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM 147

that produces an increased reliance on mitochondrial fatty acid oxidation, also activates myocardial PGC-1 (25). Under in vitro conditions, ectopic expression of PGC-1 in cultured cardiac myocytes has been shown to drive mitochondrial biogenesis, and oxidative phosphorylation in these mitochondria is tightly coupled with ATP production (25). In contrast, in adipocyte and C2C12 mouse myoblast cell lines, PGC-1 expression leads to the uncoupling of oxidative phosphorylation, resulting in decreased ATP formation and increased heat production (68). When the heart undergoes hypertrophy in response to pressure overload or cyclin T1-induced activation of Cdk9, PGC-1 expression is markedly decreased, and this reduction of PGC-1 levels is associated with a conversion from fatty acid oxidation to glycolytic metabolism (54, 55, 55). To further investigate the role of PGC-1 in the heart, two independent lines of PGC-1 -decient mouse models have been established (26, 29). Although myocardial mitochondrial volume density didnt change signicantly in either PGC-1 knockout model, the expression of genes of oxidative phosphorylation was markedly blunted in the PGC-1 -decient heart. This was accompanied with reduced mitochondrial enzymatic activities and decreased levels of ATP and phosphocreatine (2). The PGC-1 -decient heart also exhibits a diminished response to exercise and -adrenergic stimulation in vivo (26). Consistent with this, it has been reported that there is a decreased ability to increase both the heart rate and rate of change of pressure in response to chemical or electrical stimulation in a isolated perfused heart preparation of PGC- decient mice (2). In addition, starting from 7 to 8 mo of age, PGC-1 -decient mice exhibit signicant cardiac dysfunction (2). In this context, it has been recently reported that PGC-1 decient mice underwent accelerated heart failure when challenged by transverse aortic constriction (3). These data indicate that PGC-1 is essential for the heart to be able to meet the increased demands for both ATP and work output in response to physiological stimuli. Surprisingly, the in vivo gain of function studies, carried out using two independent transgenic mouse models that overexpress PGC-1 specically in the heart (MHC-PGC-1 mice and tet-on PGC-1 mice), revealed modest to dramatic mitochondria biogenesis associated with cardiomyopathy (25, 53). The development of this cardiomyopathy is thought to be associated with mitochondrial ultrastructural abnormalities and the disruption of sarcomere structure by overwhelmingly increased mitochondria biogenesis (25, 53). PGC-1 and Glucose Metabolism Maintenance of plasma glucose homeostasis is vital for the survival of mammalian organisms, and, as a result, glucose levels are tightly regulated in response to nutrient conditions and hormonal signals. Insulin suppresses plasma glucose level by the inhibition of glucose production in the liver and by promotion of glucose disposal into skeletal muscle and white fat tissue. Conversely, glucagon and glucocorticoids increase glucose levels through the activation of gluconeogenesis, in addition to the activation of glycogenolysis, in the liver and reduction of glucose utilization in skeletal muscle. Under normal, fed conditions, PGC-1 is expressed at very low levels in the liver (49). However, fasting produces a robust increase of PGC-1 expression, which, in turn, stimulates hepatic gluconeogenesis and fatty acid oxidative metabolism (19, 69).

During fasting, the expression of PGC-1 is activated by glucagon and catecholamines via the stimulation of the cAMP pathway and the CREB transcription factor. PGC-1 then coactivates a variety of transcription factors such as hepatic nuclear factor-4 , glucocorticoid receptor, and forkhead box O1 (FOXO1). These transcription factors bind to the promoter regions of those genes encoding key gluconeogenic enzymes such as phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G-6-Pase). Tissue culture studies have revealed that forced overexpression of PGC-1 in primary hepatocytes is sufcient to drive the expression of key gluconeogenic genes (69). In contrast, knockdown of PGC-1 expression by short interfering RNA in the mouse liver dramatically reduced the expression of PEPCK and G-6-Pase (22). The essential role of PGC-1 in the control of hepatic gluconeogenesis has been further reinforced by studies in the PGC-1 -decient mouse model (26, 29). One strain of PGC-1 knockout mice exhibited fasting hypoglycemia in response to impaired gluconeogenic gene expression and hepatic glucose production (26, 29). Although the expression of PGC-1 target genes involved in gluconeogenesis was not altered in another strain of PGC-1 -decient mice, the hepatic glucose production was reduced secondary to diminished fatty acid -oxidation and tricarboxylic acid cycle ux (7). Interestingly, after short-term starvation, one line of PGC1 -decient mice developed hepatic steatosis due to a combination of reduced mitochondrial fatty acid oxidation capacity and an increased expression of lipogenic genes (26). Fasting also induces PPAR- expression, which, when coactivated by PGC-1 , results in the expression of genes involved in hepatic fatty acid oxidation. The gene expression changes that have been described, under conditions of nutrient deprivation, cause a shift in fuel usage from glucose utilization to fatty acid oxidation, which contributes to the conservation of glucose for use by the central nervous system (19, 69). Under fed conditions, skeletal muscle is a major site for glucose disposal. Skeletal muscle takes up glucose using glucose transporters (GLUTs) located in the cell membrane. There are two types of skeletal muscle GLUTs: constitutive, insulininsensitive GLUT1 and GLUT3 and insulin-sensitive GLUT4. PGC-1 has been shown to increase the expression of GLUT 4 in cultured muscle cells and skeletal muscle (5, 33, 33). Whereas MEF2C has been shown to mediate PGC-1 -induced GLUT4 expression in cultured muscle cells (33), the MEF2A isoform appears to be responsible for GLUT4 upregulation under other conditions (4, 37). Surprisingly, PGC-1 has not been shown to increase glucose uptake in skeletal muscle in vivo, although it induced an increased glucose uptake into skeletal muscle cells (33). In addition, it has been reported that GLUT4 expression is downregulated in a muscle-selective PGC-1 transgenic mouse model (34). Resolution of the role of PGC-1 in the regulation of skeletal muscle glucose uptake thus requires further investigation. It is now well established that insulin also stimulates the recruitment of GLUT4 to the cell membrane of insulin-sensitive tissues. A very important area of future research is the elucidation of the interaction between insulin and PGC-1 in the regulation of glucose metabolism, an interaction about which there is very little known at the present time. There is some evidence suggesting that insulin has a suppressive effect on the expression of PGC-1 , i.e., PGC-1 promotor activity
30 DECEMBER 2006

Downloaded from ajpadvan.physiology.org on January 28, 2010

Advances in Physiology Education VOL

Staying Current
148 PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM

is inhibited by insulin (19). Consistent with this, hepatic PGC-1 expression levels have been reported to be increased in animal models exhibiting insulin deciencies (69). On the other hand, there is evidence showing a suppressive effect of insulin downstream of PGC-1 . The mechanism for this effect may involve the transcription factor FOXO1. Activation of the insulin signaling pathway leads to phosphorylation of FOXO1, inhibiting its translocation to the nucleus and increasing its susceptibility to degradation. Because PGC-1 requires FOXO1 to bind to the promotor regions of gluconeogenic genes, this action of insulin could have a suppressive effect on PGC-1 action (47). PGC-1 and Type 2 Diabetes Type 2 diabetes is the most common metabolic disease in the world, reaching epidemic proportions. Although there are multiple complicated genetic elements involved in the pathogenesis of Type 2 diabetes, lifestyle and age seem to be the two major risk factors triggering the disease. A sedentary lifestyle and overeating can contribute to excessive weight gain and obesity, with the latter being closely associated with insulin resistance. Insulin resistance is considered to be present when the biological effects of insulin are less than expected for both glucose disposal in skeletal muscle and suppression of glucose production by the liver (11). When the insulin level is not sufcient to overcome this insulin-resistant state, Type 2 diabetes ensues. In this regard, there is there is increasing evidence suggesting that mitochondrial dysfunction plays a role in the pathogenesis of insulin resistance and Type 2 diabetes. For example, it has been reported that the activities of both mitochondrial oxidative enzymes and mitochondria complex I are decreased in Type 2 diabetic patients (21, 63). In keeping with the observations that both obesity and mitochondrial dysfunction are risk factors for the development of insulin resistance, obese individuals have been reported to have smaller mitochondria that exhibit a compromised bioenergetic capacity (21). Insulin resistance also develops with age, and, in this case, a potentially important defect has been found in the mitochondrial fatty acid oxidation pathway. There appears to be a reduction in the rate of fatty acid oxidation, leading to the 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 specic type of mitochondrial dysfunction, i.e., decreased fatty acid oxidation, may play a signicant role in the development of insulin resistance in both aging and obesity (43). It is interesting to note that triglycerides can also accumulate 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 deciency 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. It has been hypothesized that a close relationship exists among PGC-1 function, insulin sensitivity, and Type 2 diabetes, which is most likely related to the essential roles of

PGC-1 in mitochondria biogenesis and glucose/fatty acid metabolism. Evidence supporting this hypothesis has been found in a number of studies. For example, it has been observed that expression of PGC-1 is downregulated in muscles of Type 2 diabetic subjects (36, 42, 44). In addition, a common polymorphism of the PGC-1 gene (Gly482Ser), expressing reduced PGC-1 activity, has been linked to an increased risk of Type 2 diabetes (18, 40). These observations would suggest that either reduced levels or compromised activity of PGC-1 can be associated with the development of insulin resistance and Type 2 diabetes (Fig. 1). In this context, it is interesting to note that TZDs, an important class of antidiabetic drugs, also act to increase insulin sensitivity coincident with the activation of PGC-1 (66). The effects of TZDs are likely mediated through the ability of PGC-1 to activate mitochondria biogenesis and increase mitochondrial function. As described above, PGC-1 stimulates the conversion of muscle ber type toward more oxidative type I and IIa bers, favoring fatty acid oxidative metabolism (28). The promotion of fatty acid oxidative metabolism would be expected to lead to a reduction of fat accumulation in muscle, which, in turn, could increase insulin sensitivity because there is a close relationship between triglyceride accumulation and insulin resistance (43). One manifestation of increased insulin sensitivity would be increased glucose uptake by insulin-sensitive tissues. PGC-1 has, as previously noted, been reported to activate the expression of insulin-sensitive GLUT4 in skeletal muscle (5, 33). Taken together, the evidence presented supports a role for PGC-1 in preventing insulin resistance and Type 2 diabetes mellitus. However, it should be pointed out that PGC-1 expression has been reported to be increased in the liver of both Type 1 and Type 2 diabetic mouse models, in contrast to the reported decrease in PGC-1 expression observed in the muscle of human Type 2 diabetic subjects described above (48). Furthermore, PGC-1 has been shown to inhibit the insulin signaling pathway in the liver (22), inhibit glucose utilization in cultured myotubes (65), and suppress -cell energy metabolism and insulin release in mice (70). Increased hepatic PGC-1 expression could be expected to stimulate hepatic glucose output,

Downloaded from ajpadvan.physiology.org on January 28, 2010

Fig. 1. Potential role of decreased peroxisome proliferator-activated receptor- coactivator (PGC)-1 expression in skeletal muscle as it relates to the development of the metabolic phenotype of insulin resistance and Type 2 diabetes mellitus. [Revised from Ref. 34.]
30 DECEMBER 2006

Advances in Physiology Education VOL

Staying Current
PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM 149

and, when coupled with the reported inhibitory effect of PGC-1 on insulin signaling and secretion, the combined effects would be to contribute to the hyperglycemic state, a prodiabetes effect. This prodiabetic effect of PGC-1 has been further supported by the manifestation of improved insulin sensitivity in PGC-1 -decient mouse models (22, 26, 29). Moreover, another recent report (38) has indicated that PGC-1 expression is normal in insulin-resistant subjects despite severe impairments in mitochondrial function. These apparently paradoxical actions of PGC-1 in different tissues are intriguing. The underlining mechanisms for the interactions between these tissues are currently unknown and deserve further intensive investigations. Summary As an inducible transcription coactivator, PGC-1 is enriched in metabolically active tissues. It is intimately involved in adaptive thermogenesis, skeletal muscle ber type switching, glucose/fatty acid metabolism, and heart development. Among these varied biological responses, a common mechanism of action appears to be the promotion of oxidative metabolism accompanying the stimulation of mitochondria biogenesis. Furthermore, there is growing evidence that PGC-1 plays a role in disorders such as obesity, diabetes, and cardiomyopathy. Because of the involvement of PGC-1 in so many important biological processes, in conjunction with the rapid improvement in our understanding of its mechanisms of action, it has become an inviting target for the design of pharmacological interventions for treatment of these disorders.
REFERENCES 1. Akimoto T, Pohnert SC, Li P, Zhang M, Gumbs C, Rosenberg PB, Williams RS, and Yan Z. Exercise stimulates Pgc-1alpha transcription in skeletal muscle through activation of the p38 MAPK pathway. J Biol Chem 280: 1958719593, 2005. 2. Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, Ahmad F, Matsui T, Chin S, Wu PH, Rybkin II, Shelton JM, Manieri M, Cinti S, Schoen FJ, Bassel-Duby R, Rosenzweig A, Ingwall JS, and Spiegelman BM. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab 1: 259 271, 2005. 3. Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, and Spiegelman BM. Transverse aortic constriction leads to accelerated heart failure in mice lacking PPAR-gamma coactivator 1alpha. Proc Natl Acad Sci USA 103: 10086 10091, 2006. 4. Baar K, Song Z, Semenkovich CF, Jones TE, Han DH, Nolte LA, Ojuka EO, Chen M, and Holloszy JO. Skeletal muscle overexpression of nuclear respiratory factor 1 increases glucose transport capacity. FASEB J 17: 1666 1673, 2003. 5. Baar K, Wende AR, Jones TE, Marison M, Nolte LA, Chen M, Kelly DP, and Holloszy JO. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC-1. FASEB J 16: 1879 1886, 2002. 6. Barbera MJ, Schluter A, Pedraza N, Iglesias R, Villarroya F, and Giralt M. Peroxisome proliferator-activated receptor alpha activates transcription of the brown fat uncoupling protein-1 gene. A link between regulation of the thermogenic and lipid oxidation pathways in the brown fat cell. J Biol Chem 276: 1486 1493, 2001. 7. Burgess SC, Leone TC, Wende AR, Croce MA, Chen Z, Sherry AD, Malloy CR, and Finck BN. Diminished hepatic gluconeogenesis via defects in tricarboxylic acid cycle ux in peroxisome proliferator-activated receptor coactivator-1 (PGC-1 )-decient mice. J Biol Chem 281: 19000 19008, 2006. 8. Cannon B, Houstek J, and Nedergaard J. Brown adipose tissue. More than an effector of thermogenesis? Ann NY Acad Sci 856: 171187, 1998. 9. Cassard-Doulcier AM, Larose M, Matamala JC, Champigny O, Bouillaud F, and Ricquier D. In vitro interactions between nuclear

10.

11. 12.

13.

14. 15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27. 28.

proteins and uncoupling protein gene promoter reveal several putative transactivating factors including Ets1, retinoid X receptor, thyroid hormone receptor, and a CACCC box-binding protein. J Biol Chem 269: 2433524342, 1994. Czubryt MP, McAnally J, Fishman GI, and Olson EN. Regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha) and mitochondrial function by MEF2 and HDAC5. Proc Natl Acad Sci USA 100: 17111716, 2003. Dinneen S, Gerich J, and Rizza R. Carbohydrate metabolism in noninsulin-dependent diabetes mellitus. N Engl J Med 327: 707713, 1992. Finck BN and Kelly DP. PGC-1 coactivators: inducible regulators of energy metabolism in health and disease. J Clin Invest 116: 615 622, 2006. Garcia-Roves PM, Huss J, and Holloszy JO. Role of calcineurin in exercise-induced mitochondrial biogenesis. Am J Physiol Endocrinol Metab 290: E1172E1179, 2006. Garesse R and Vallejo CG. Animal mitochondrial biogenesis and function: a regulatory cross-talk between two genomes. Gene 263: 116, 2001. Goto M, Terada S, Kato M, Katoh M, Yokozeki T, Tabata I, and Shimokawa T. cDNA Cloning and mRNA analysis of PGC-1 in epitrochlearis muscle in swimming-exercised rats. Biochem Biophys Res Commun 274: 350 354, 2000. Guan HP, Ishizuka T, Chui PC, Lehrke M, and Lazar MA. Corepressors selectively control the transcriptional activity of PPARgamma in adipocytes. Genes Dev 19: 453 461, 2005. Handschin C, Rhee J, Lin J, Tarr PT, and Spiegelman BM. An autoregulatory loop controls peroxisome proliferator-activated receptor gamma coactivator 1alpha expression in muscle. Proc Natl Acad Sci USA 100: 71117116, 2003. Hara K, Tobe K, Okada T, Kadowaki H, Akanuma Y, Ito C, Kimura S, and Kadowaki T. A genetic variation in the PGC-1 gene could confer insulin resistance and susceptibility to Type II diabetes. Diabetologia 45: 740 743, 2002. Herzig S, Long F, Jhala US, Hedrick S, Quinn R, Bauer A, Rudolph D, Schutz G, Yoon C, Puigserver P, Spiegelman B, and Montminy M. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature 413: 179 183, 2001. Huss JM, Kopp RP, and Kelly DP. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identication of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem 277: 40265 40274, 2002. 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. Koo SH, Satoh H, Herzig S, Lee CH, Hedrick S, Kulkarni R, Evans RM, Olefsky J, and Montminy M. PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB-3. Nat Med 10: 530 534, 2004. Kressler D, Schreiber SN, Knutti D, and Kralli A. The PGC-1-related protein PERC is a selective coactivator of estrogen receptor alpha. J Biol Chem 277: 13918 13925, 2002. Larsson NG, Wang J, Wilhelmsson H, Oldfors A, Rustin P, Lewandoski M, Barsh GS, and Clayton DA. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat Genet 18: 231236, 1998. Lehman JJ, Barger PM, Kovacs A, Saftz JE, Medeiros DM, and Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. J Clin Invest 106: 847 856, 2000. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina S, Courtois M, Wozniak DF, Sambandam N, Bernal-Mizrachi C, Chen Z, Holloszy JO, Medeiros DM, Schmidt RE, Saftz JE, Abel ED, Semenkovich CF, and Kelly DP. PGC-1alpha deciency causes multisystem energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol 3: e101, 2005. Lin J, Handschin C, and Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab 1: 361370, 2005. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel-Duby R, and Spiegelman BM. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle bres. Nature 418: 797 801, 2002.
30 DECEMBER 2006

Downloaded from ajpadvan.physiology.org on January 28, 2010

Advances in Physiology Education VOL

Staying Current
150 PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM 46. Puigserver P, Adelmant G, Wu Z, Fan M, Xu J, OMalley B, and Spiegelman BM. Activation of PPARgamma coactivator-1 through transcription factor docking. Science 286: 1368 1371, 1999. 47. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente F, Kitamura Y, Altomonte J, Dong H, Accili D, and Spiegelman BM. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature 423: 550 555, 2003. 48. Puigserver P and Spiegelman BM. Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator. Endocr Rev 24: 78 90, 2003. 49. Puigserver P, Wu Z, Park CW, Graves R, Wright M, and Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92: 829 839, 1998. 50. Rangwala SM and Lazar MA. Peroxisome proliferator-activated receptor gamma in diabetes and metabolism. Trends Pharmacol Sci 25: 331 336, 2004. 51. Rothwell NJ and Stock MJ. A role for brown adipose tissue in dietinduced thermogenesis. Nature 281: 3135, 1979. 52. Russell AP, Feilchenfeldt J, Schreiber S, Praz M, Crettenand A, Gobelet C, Meier CA, Bell DR, Kralli A, Giacobino JP, and Deriaz O. Endurance training in humans leads to ber type-specic increases in levels of peroxisome proliferator-activated receptor-gamma coactivator-1 and peroxisome proliferator-activated receptor-alpha in skeletal muscle. Diabetes 52: 2874 2881, 2003. 53. Russell LK, Manseld CM, Lehman JJ, Kovacs A, Courtois M, Saftz JE, Medeiros DM, Valencik ML, McDonald JA, and Kelly DP. Cardiac-specic induction of the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha promotes mitochondrial biogenesis and reversible cardiomyopathy in a developmental stage-dependent manner. Circ Res 94: 525533, 2004. 54. Sack MN, Disch DL, Rockman HA, and Kelly DP. A role for Sp and nuclear receptor transcription factors in a cardiac hypertrophic growth program. Proc Natl Acad Sci USA 94: 6438 6443, 1997. 55. Sano M, Wang SC, Shirai M, Scaglia F, Xie M, Sakai S, Tanaka T, Kulkarni PA, Barger PM, Youker KA, Taffet GE, Hamamori Y, Michael LH, Craigen WJ, and Schneider MD. Activation of cardiac Cdk9 represses PGC-1 and confers a predisposition to heart failure. EMBO J 23: 3559 3569, 2004. 56. Scarpulla RC. Nuclear activators and coactivators in mammalian mitochondrial biogenesis. Biochim Biophys Acta 1576: 114, 2002. 57. Scarpulla RC. Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene 286: 81 89, 2002. 58. Sears IB, MacGinnitie MA, Kovacs LG, and Graves RA. Differentiation-dependent expression of the brown adipocyte uncoupling protein gene: regulation by peroxisome proliferator-activated receptor gamma. Mol Cell Biol 16: 3410 3419, 1996. 59. Spiegelman BM. PPAR-gamma: adipogenic regulator and thiazolidinedione receptor. Diabetes 47: 507514, 1998. 60. Unger RH. Lipotoxicity in the pathogenesis of obesity-dependent NIDDM. Genetic and clinical implications. Diabetes 44: 863 870, 1995. 61. van Raalte DH, Li M, Pritchard PH, and Wasan KM. Peroxisome proliferator-activated receptor (PPAR)-alpha: a pharmacological target with a promising future. Pharm Res 21: 15311538, 2004. 62. Vega RB, Huss JM, and Kelly DP. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol 20: 1868 1876, 2000. 63. Vondra K, Rath R, Bass A, Slabochova Z, Teisinger J, and Vitek V. Enzyme activities in quadriceps femoris muscle of obese diabetic male patients. Diabetologia 13: 527529, 1977. 64. Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, and Evans RM. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 113: 159 170, 2003. 65. Wende AR, Huss JM, Schaeffer PJ, Giguere V, and Kelly DP. PGC-1alpha coactivates PDK4 gene expression via the orphan nuclear receptor ERRalpha: a mechanism for transcriptional control of muscle glucose metabolism. Mol Cell Biol 25: 10684 10694, 2005. 66. Wilson-Fritch L, Nicoloro S, Chouinard M, Lazar MA, Chui PC, Leszyk J, Straubhaar J, Czech MP, and Corvera S. Mitochondrial remodeling in adipose tissue associated with obesity and treatment with rosiglitazone. J Clin Invest 114: 12811289, 2004.
30 DECEMBER 2006

29. Lin J, Wu PH, Tarr PT, Lindenberg KS, St Pierre J, Zhang CY, Mootha VK, Jager S, Vianna CR, Reznick RM, Cui L, Manieri M, Donovan MX, Wu Z, Cooper MP, Fan MC, Rohas LM, Zavacki AM, Cinti S, Shulman GI, Lowell BB, Krainc D, and Spiegelman BM. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell 119: 121135, 2004. 30. Lin J, Yang R, Tarr PT, Wu PH, Handschin C, Li S, Yang W, Pei L, Uldry M, Tontonoz P, Newgard CB, and Spiegelman BM. Hyperlipidemic effects of dietary saturated fats mediated through PGC-1beta coactivation of SREBP. Cell 120: 261273, 2005. 31. Lowell BB. PPARgamma: an essential regulator of adipogenesis and modulator of fat cell function. Cell 99: 239 242, 1999. 32. Lowell BB and Shulman GI. Mitochondrial dysfunction and type 2 diabetes. Science 307: 384 387, 2005. 33. Michael LF, Wu Z, Cheatham RB, Puigserver P, Adelmant G, Lehman JJ, Kelly DP, and Spiegelman BM. Restoration of insulin-sensitive glucose transporter (GLUT4) gene expression in muscle cells by the transcriptional coactivator PGC-1. Proc Natl Acad Sci USA 98: 3820 3825, 2001. 34. Miura S, Kai Y, Ono M, and Ezaki O. Overexpression of peroxisome proliferator-activated receptor gamma coactivator-1alpha down-regulates GLUT4 mRNA in skeletal muscles. J Biol Chem 278: 3138531390, 2003. 35. 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. Err 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. 36. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstrale M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, and Groop LC. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 34: 267 273, 2003. 37. Mora S and Pessin JE. The MEF2A isoform is required for striated muscle-specic expression of the insulin-responsive GLUT4 glucose transporter. J Biol Chem 275: 1632316328, 2000. 38. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes N, Neschen S, White MF, Bilz S, Sono S, Pypaert M, and Shulman GI. Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest 115: 35873593, 2005. 39. Mortensen OH, Frandsen L, Schjerling P, Nishimura E, and Grunnet N. PGC-1 and PGC-1 have both similar and distinct effects upon myober switching towards an oxidative phenotype. Am J Physiol Endocrinol Metab 291: E807E816, 2006. 40. Muller YL, Bogardus C, Pedersen O, and Baier L. A Gly482Ser missense mutation in the peroxisome proliferator-activated receptor gamma coactivator-1 is associated with altered lipid oxidation and early insulin secretion in Pima Indians. Diabetes 52: 895 898, 2003. 41. Norrbom J, Sundberg CJ, Ameln H, Kraus WE, Jansson E, and Gustafsson T. PGC-1 mRNA expression is inuenced by metabolic perturbation in exercising human skeletal muscle. J Appl Physiol 96: 189 194, 2004. 42. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldne AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, and Mandarino LJ. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc Natl Acad Sci USA 100: 8466 8471, 2003. 43. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, DiPietro L, Cline GW, and Shulman GI. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science 300: 1140 1142, 2003. 44. Petersen KF, Dufour S, Befroy D, Garcia R, and Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med 350: 664 671, 2004. 45. Puigserver P. Tissue-specic regulation of metabolic pathways through the transcriptional coactivator PGC1-alpha. Int J Obes (Lond) 29, Suppl 1: S5S9, 2005.

Downloaded from ajpadvan.physiology.org on January 28, 2010

Advances in Physiology Education VOL

Staying Current
PGC-1 : A KEY REGULATOR OF ENERGY METABOLISM 67. Wu H, Kanatous SB, Thurmond FA, Gallardo T, Isotani E, BasselDuby R, and Williams RS. Regulation of mitochondrial biogenesis in skeletal muscle by CaMK. Science 296: 349 352, 2002. 68. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, and Spiegelman BM. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 98: 115124, 1999. 69. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, Newgard CB, and Spiegelman BM. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413: 131138, 2001. 70. Yoon JC, Xu G, Deeney JT, Yang SN, Rhee J, Puigserver P, Levens AR, Yang R, Zhang CY, Lowell BB, Berggren PO, Newgard CB, Bonner-Weir S, Weir G, and Spiegelman BM. Suppression of beta cell 151

energy metabolism and insulin release by PGC-1alpha. Dev Cell 5: 73 83, 2003. 71. Zhang Y, Castellani LW, Sinal CJ, Gonzalez FJ, and Edwards PA. Peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha) regulates triglyceride metabolism by activation of the nuclear receptor FXR. Genes Dev 18: 157169, 2004. 72. Zhang Y, Ma K, Song S, Elam MB, Cook GA, and Park EA. Peroxisomal proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1 alpha) enhances the thyroid hormone induction of carnitine palmitoyltransferase I (CPT-I alpha). J Biol Chem 279: 5396353971, 2004. 73. Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ, and Shulman GI. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proc Natl Acad Sci USA 99: 1598315987, 2002.

Downloaded from ajpadvan.physiology.org on January 28, 2010

Advances in Physiology Education VOL

30 DECEMBER 2006

You might also like