Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Mol Cell Cardiol. Author manuscript; available in PMC 2012 Oct 1.
Published in final edited form as:
PMCID: PMC3042046
NIHMSID: NIHMS249263
PMID: 20888832

The PGC-1 Cascade as a Therapeutic Target for Heart Failure

Abstract

The PPARγ coactivator-1 (PGC-1) family of transcriptional coactivators, together with estrogen related receptors (ERRs), play a key role in regulating genes involved in myocardial fuel metabolism and cardiac function. Increasing evidence implicates dysregulation of this transcriptional regulatory circuit in the metabolic and functional disturbances that presage heart failure due to common diseases such as hypertension and diabetes. Accordingly, the PGC-1/ERR axis is a plausible candidate therapeutic target for novel therapeutics aimed at reversing the energy metabolic disturbances that contribute to heart failure. This review describes the biologic actions of the PGC-1 and ERR cascade and summarizes the evidence that dysregulation of this transcriptional regulatory circuit contributes to heart failure. Potential strategies to modulate this target pathway are reviewed.

The constant generation of ATP is vital for the heart to perform its contractile function. Cardiac myocytes must also be able to dynamically re-program fuel and energy metabolic capacity in response to environmental and physiological cues to ensure that energy supply meets demands. One important mechanism by which this is achieved is at the level of metabolic gene expression. The transcriptional coactivators, PPARγ coactivator-1 (PGC-1) α and β, and their target nuclear receptors (NRs), estrogen-related receptors α and γ (ERRα and γ), comprise a key gene regulatory network controlling the expression of cardiac genes involved in multiple mitochondrial energy transduction and ATP-generating pathways. The purpose of this article is to review the function of the PGC-1/ERR axis in the normal heart and the evidence that dysregulation of this circuit contributes to the metabolic and functional disturbances that lead to heart failure. The PGC-1/ERR gene regulatory pathway as a novel therapeutic candidate for metabolic modulation in cardiovascular disease will also be discussed.

PGC-1/ERR gene regulatory axis

PGC-1 Coactivators

PGC-1α was originally described as a coactivator of peroxisome proliferator-activated receptor γ (PPARγ), a brown adipose enriched NR, where it was shown to regulate adaptive thermogenesis and mitochondrial function [1]. Subsequently, a structurally-related protein, PGC-1β, was found to be capable of regulating many of the target pathways known to be controlled by PGC-1α [2]. It is now well-established that PGC-1α and β are inducible, transcriptional coregulators that play a vital role in the control cellular ATP-producing capacity and mitochondrial function under basal conditions and in response to physiological stressors. The PGC-1 coactivators exert their biologic effects by directly binding to, and enhancing, the transcriptional activity of NR and non-NR transcription factors [3]. Known targets of PGC-1 coactivators expressed in heart include NR (ERRα, ERRγ, PPARα, PPARβ/δ) and non-NR (NRF-1 and MEF2) transcription factors [4]. Together, the PGC-1 network serves to coordinately regulate the expression of numerous genes involved in mitochondrial pathways such as fatty acid oxidation (FAO), oxidative phosphorylation, and ATP synthesis along with the regulatory machinery involved in mitochondrial biogenesis (Figure 1) [5].

An external file that holds a picture, illustration, etc.
Object name is nihms249263f1.jpg
PGC-1α and β: Inducible transcriptional coregulators of cardiac myocyte fuel and energy metabolism

The PGC-1 coactivators and their ERR nuclear receptor targets are shown schematically. A variety of upstream signaling pathways can upregulate (black arrows) or downregulate (red lines) the expression and/or activity of the PGC-1 coactivators (described in text). Together via ERRα or γ, PGC-1α and β influence the expression of a number of genes involved in mitochondrial biogenesis, respiratory function, and mitochondrial fatty acid oxidation.

PGC-1α and β are enriched in tissues with high oxidative capacity such as heart, brown adipose, kidney, and slow-twitch skeletal muscle [1, 2, 6]. The expression of PGC-1α coactivators is highly inducible at the transcriptional level via the action of a variety of upstream signaling pathways (Figure 1). For example, PGC-1α gene expression is induced by cold exposure, fasting, and exercise, all of which demand increased mitochondrial oxidative flux for ATP production or thermogenesis [1, 7, 8]. Transcription of the PGC-1α gene can be influenced by CaM kinase, calcineurin, β-adrenergic receptor (β-AR)/cAMP, nitric oxide (NO) and AMP-activated protein kinase (AMPK) [913]. Transcription factors that transduce upstream signals to the control of PGC-1α gene expression include cAMP response element-binding protein (CREB) and MEF2 [9, 14]. The PGC-1α promoter can also be suppressed by the action of class II histone deacetylases (HDACs) via inhibition of MEF2 activity [14]. In contrast to PGC-1α, less is known about the transcriptional regulation of PGC-1β; however, recent investigations have shown that both interleukin-4 (IL-4) and interferon (IFN)-γ activate the PGC-1β promoter via STAT6 or STAT1, respectively [15, 16]. Relevant to these findings, cytokine-mediated induction of PGC-1β is important for the oxidative burst and microbicidal function of activated macrophages [15].

PGC-1α activity is also modulated at the post-translational level via acetylation and phosphorylation [17, 18]. Acetylation of PGC-1α can occur at multiple sites and serves to decrease its transcriptional activity. Recent evidence indicates that PGC-1α acetylation status is regulated in large part by the balance between the deacetylase silent information regulator (SIRT)1 and acetyltransferase GCN5 [17]. When cellular energy stores are reduced, the level of NAD+ increases and this activates SIRT1. Subsequently, SIRT1 deacetylates PGC-1α thereby boosting the generation of ATP and reducing equivalents via mitochondrial substrate oxidation [17]. In addition, AMPK and p38 MAPK activate PGC-1α via direct phosphorylation [18, 19]. AMPK can also facilitate activation of SIRT1, further increasing PGC-1α transcriptional activity [20]. Similar to PGC-1α, PGC-1β was recently shown to be negatively regulated by acetylation [21].

PGC-1α is also produced in variant forms via alternative splicing. One of the better characterized alternatively spliced forms of PGC-1α contains only the amino terminal portion of the protein (termed NT-PGC-1α), but is still capable of coactivating many of the known downstream targets [22]. Interestingly, in contrast to full-length PGC-1α which resides in the nucleus, NT-PGC-1α cycles between the nucleus and cytoplasm and in many conditions is predominantly cytoplasmic. Protein kinase A (PKA), and possibly other kinases can phosphorylate NT-PGC1α, trapping it in the nucleus leading to enhanced transcription of PGC-1α target genes [23].

ERRα/γ

ERRα and related family members, ERRβ and ERRγ, were originally discovered through a screen to identify steroid hormone receptors related to the estrogen receptor (ER) [24]. However, in contrast to ER, the ERRs do not have known ligands (thus termed “orphan” nuclear receptors). ERRα was also identified in a screen for novel PGC-1α binding partners [25]. It is now well-accepted that ERRα and γ serve key roles in the gene regulatory control of cardiac mitochondrial energetic pathways. Interestingly, current evidence indicates that ERR family members must interact with PGC-1 coactivators to be transcriptionally active, leading to the notion that PGC-1α and β should be considered “protein ligands” for these NRs [26]. Conversely, ERRα is required for many PGC-1-dependent effects on metabolism and mitochondrial function, suggesting that this co-activator/NR pairing is intimately intertwined in a feed-forward cycle [27].

The PGC-1/ERR axis in cardiac metabolism and physiology

Significant insight into the role of PGC-1α and β in the control of cardiac metabolism has been gained from studies of genetically-modified mouse models (Table 1). During the early postnatal period, the heart undergoes a dramatic shift in fuel utilization from glucose and lactate oxidation during the fetal period, to a high reliance on mitochondrial FAO after birth. This metabolic shift is accompanied by a surge in the expression of PGC-1α and β mRNA and a burst of mitochondrial biogenesis [7, 28]. The importance of PGC-1 coactivators in controlling perinatal mitochondrial biogenesis is supported by the observation that PGC-1α overexpression in cardiac myocytes in vitro and in vivo in transgenic mice is sufficient to trigger mitochondrial proliferation [7]. Conversely, in mice with combined PGC-1α/β gene disruption, perinatal mitochondrial biogenesis is arrested and the mice die within days after birth from heart failure [28]. Together, these results indicate that the PGC-1 coactivators are necessary and sufficient for postnatal metabolic maturation and cardiac function.

Table 1

Cardiac phenotypes of genetically modified PGC-1/ERR animal models.

Genetic ModelCommentsCardiac PhenotypeReferences
PGC-1α KO1) Disruption of PGC-1 by insertion of additional exon 3 between exons 5-6.1) Mild bradycardia, blunted heart rate response to dobutamine, reduced myocardial FAO capacity, slightly reduced expression of mitochondrial and FAO genes, reduced ATP generation, increased expression of PGC-1β.31, 32
2) Disruption of PGC-1α gene by deletion of exons 3-5.2) Blunted contractile response to dobutamine infusion in Langendorff ex vivo heart, reduced cardiac function with aging or pressure overload, reduced ATP generation slightly reduced expression of mitochondrial and FAO genes.29, 30, 48

PGC-1β KO4 independent KO/KD mouse models generatedMild bradycardia, blunted heart response to dobutamine, reduced ATP generation, slightly reduced expression of mitochondrial genes in heart and skeletal muscle, increased expression of PGC- 1α.28, 3335

PGC-1α/β DKOPGC-1α global KO mice crossed with a cardiac- specific KO of PGC-1βArrest in cardiac perinatal mitochondrial biogenesis, marked reduction in cardiac mitochondria, death from heart failure in first day of life.28

ERRα KOGlobal knockoutIncreased expression of PGC-1α and ERRγ, heart failure after aortic banding; reduction of cardiac ATP/energy reserves.39, 43

ERRγ KOGlobal knockoutIncreased expression of PGC-1α and ERRα, absent mitochondrial biogenesis after birth, QT prolongation on ECG, early mortality38

PGC-1α OEPGC-1α expression driven by MHC promoterUncontrolled mitochondrial proliferation, severe cardiomyopathy and death from heart failure8

PGC-1α COETetracycline inducible double transgenic mouse using MHC-rtTA and TRE- PGC-1αIncreased expression of mitochondrial genes, modest mitochondrial biogenesis, dilated cardiomyopathy within 2 weeks of PGC-1α induction (reversible)36

KO (knockout), KD (knockdown), DKO (double knockout), OE (overexpression), COE (conditional overexpression), TAC (transaortic constriction), rtTA (tetracycline transactivator), TRE (tetracycline response element)

PGC-1α and β also serve important functions in the adult heart. PGC-1α-deficient mice are developmentally normal, but have reduced FAO capacity, lower exercise tolerance, and reduced cardiac reserve with dobutamine challenge or aging [28-32]. In addition, cardiac mitochondrial and FAO gene expression is modestly decreased in the absence of PGC-1α. Of note, PGC-1β expression is increased in PGC-1α-deficient mice and this appears to partially compensate for the loss of PGC-1α [28]. Similar to PGC-1α-deficient mice, animals lacking PGC-1β have a modest downregulation in FAO and mitochondrial gene expression and display reduced contractile reserve with dobutamine treatment [28, 3335]. The effects of PGC-1α overexpression in the adult heart have also been investigated. Forced cardiac expression via a tetracycline-inducible, myocyte-specific transgenic system, demonstrated that PGC-1α drives a modest mitochondrial biogenic response [36]. However, chronic overexpression of PGC-1α in heart leads to significant mitochondrial structural abnormalities and a severe, but reversible cardiomyopathy [36]. Thus, PGC-1α and β are potent regulators of mitochondrial proliferation/function and fuel metabolism in the adult heart.

In addition to the regulation of mitochondrial function and energy metabolism, PGC-1α and β can also regulate the expression of genes encoding reactive oxygen species (ROS) scavengers. The production of ROS has been implicated in the pathogenesis of many diseases including heart failure, atherosclerosis, diabetes, and neurodegeneration. In cell culture, oxidant stress induced by hydrogen peroxide results in the upregulation of PGC-1α and β and the ROS scavengers superoxide dismutase (SOD)1, SOD2, catalase, and glutathione peroxidase 1 (GPX1) [37]. Interestingly, the upregulation of ROS defense molecules is dependent on the PGC-1 coactivators [37]. The in vivo relevance of this response is supported by the observation that PGC-1α KO mice have reduced expression of antioxidant molecules and may be more susceptible to neurodegeneration [37].

ERRα and ERRγ mediate a number of the biologic functions of the PGC-1 coactivators. Genetically-modified mice have helped provide insight into the function of the ERRs in heart (Table 1). Several lines of evidence support the importance of ERRα and γ in the cardiac PGC-1 gene regulatory circuit. First, ERRα and γ expression in heart increases dramatically after birth in parallel with the expression pattern of PGC-1 coactivators and their target genes [25, 38]. Second, ERRγ KO mice fail to make the postnatal transition to oxidative FA metabolism in the heart, similar to PGC-1α/β double deficient mice [28, 38]. Third, overexpression of ERRα leads to induction of the expression of mitochondrial and FAO genes in a profile that mimics PGC-1 overexpression [39]. Finally, PGC-1α overexpression fails to induce the expression of genes involved in mitochondrial respiration and oxidative stress protection in cells lacking ERRα [27]. Thus, the PGC-1 coactivators and ERR family members are central partners in the regulation of cardiac metabolism and mitochondrial oxidative flux.

The PGC-1/ERR axis in cardiac pathophysiology

Fuel and energy metabolic disturbances are a well-described signature of cardiac hypertrophy and heart failure [40]. The development of pathologic cardiac hypertrophy due to pressure overload is associated with a relative increase in glucose utilization and reduction in FAO, a shift towards a fetal metabolic profile [41, 42]. This metabolic shift is related to the downregulation of expression and activity of PGC-1α, ERRα and downstream target genes (Figure 2) [43]. Although this fuel shift may initially serve an adaptive function to divert flux away from mitochondrial pathways [44], over the long-term this response is likely maladaptive leading to energy starvation and heart failure. Indeed, studies in animal models and humans have shown high energy phosphate reserves are reduced in the hypertrophied heart [4547]. In line with the observed energetic deficiency, PGC-1α and ERRα null mice subjected to the stress of aortic constriction develop more severe heart failure compared to WT animals [43, 48]. These results suggest that chronic progressive deactivation of the PGC-1/ERR axis and consequent reduction in capacity for ATP production may play a causative role in the transition from compensated hypertrophy to heart failure.

An external file that holds a picture, illustration, etc.
Object name is nihms249263f2.jpg
Dysregulation of the PGC-1/ERR axis in hypertensive and diabetic forms of cardiomyopathy

Hypertension (HTN) and diabetes (DM) lead to distinct early myocardial energy metabolic derangements that likely contribute to the pathogenesis of heart failure. HTN elicits LV hypertrophy and downregulation of PGC-1α and ERRα related pathways. Subsequently, a decline in mitochondrial function leads to reduced capacity to generate ATP (“energy starvation”), and may contribute to the development of heart failure. In DM, myocyte lipid overload leads to an early activation of PGC-1/ERR as an adaptive response to increase capacity to burn fats in the mitochondrion. However, as the disease progresses, PGC-1 becomes “deactivated” via unknown mechanisms setting the stage for the development of lipotoxicity, energy starvation, and subsequent cardiomyopathy.

Similar to what has been observed in cardiac hypertrophy, cardiomyopathic hearts also exhibit a reduction in oxidative fuel metabolism [49]. The extent to which this metabolic shift occurs appears to be dependent on the severity and etiology of the LV dysfunction. Positron emission tomography (PET) has shown reduced rates of FAO in patients with idiopathic dilated cardiomyopathy [49]. Animal models and human tissue have provided additional evidence that the expression and activity of PGC-1α and ERRα is diminished in heart failure [5052]. Although it remains unclear as to the extent that changes in PGC-1 expression are causal for heart failure, the phenotype of PGC-1 loss-of-function mice demonstrates that reduced PGC-1/ERR activity can lead to cardiac dysfunction [28, 43, 48]. Similarly, inhibiting PGC-1 expression via cardiac-specific overexpression of HDAC5 also produces a cardiomyopathy that is associated with mitochondrial abnormalities and myocardial lipid accumulation [14]. The upstream signals that deactivate the PGC-1/ERR circuit in heart failure are unknown, but is an area of active investigation.

In contrast to cardiac hypertrophy and heart failure, obesity-related insulin resistance and diabetes leads to increased activation of PGC-1/ERR dependent pathways, at least in the early stages (Figure 2). Obesity and insulin resistance are also associated with myocardial triglyceride (TAG) accumulation which has led to the term cardiac “lipotoxicity”. Both animal and human studies have shown that myocardial FAO capacity is increased in response to this fat load [5355]. Moreover, insulin resistant hearts undergo a mitochondrial biogenic response that is mediated by PGC-1α [56]. The upregulation of PGC-1-related pathways appears to be an early compensatory mechanism to handle the excess lipid delivered to the heart in obese and diabetic states. However, in later stages of diabetes the expression of PGC-1α and PPARα is diminished [5557]. Once this occurs, it is speculated that the excess fatty acids can no longer be effectively oxidized in the mitochondrion, perhaps leading to the generation of toxic lipids, accumulation of reactive oxygen species, and increased risk of heart failure [58].

Manipulating the PGC-1/ERR axis as a candidate therapeutic target

As summarized above, the PGC-1/ERR transcriptional regulatory circuit is deactivated during the development of heart failure. This observation, coupled with the known potential for PGC-1α/β and ERRα/γ to stimulate energy production and reduce oxidative stress suggests that this transcriptional regulatory circuit could be an attractive target for prevention and treatment of heart failure and metabolic diseases. Two major proof-of-concept questions must be addressed prior to pursuing this strategy with vigor. First, how can the PGC-1/ERR cascade be activated in a way that harnesses the powerful regulatory effects without producing toxicity? In this regard, the delivery strategy is an important concern given that chronic, persistent PGC-1 overexpression in the hearts of mice produces a cardiomyopathy [7, 36]. However, acute induction of PGC-1α in skeletal muscle has been shown to increase mitochondrial respiratory capacity and augment skeletal muscle refueling after exercise without evidence of significant toxicity [59]. Thus, it may be possible to therapeutically augment PGC-1 activity using intermittent dosing regimens. Secondly, does reactivation of the PGC-1/ERR circuit prevent or ameliorate heart failure in pre-clinical models?

Assuming that proof-of-concept studies suggest that the PGC-1/ERR axis is a viable therapeutic target, it will be important to define optimal strategies to modulate this pathway. Conceptually, this could be achieved through approaches that increase either the expression or activity of one or several of these molecules. Given that PGC-1α/β are master regulators of ERRα/γ expression and mitochondrial function, targeting the coactivators is an obvious approach. Several mechanisms to augment PGC-1 expression could be considered based on the known regulators of the PGC-1 promoter. For example, molecules that increase the activity of PKA or calcium signaling, or inhibit HDAC activity, could increase PGC-1 gene expression via CREB and/or MEF2. However, augmenting PKA activation via β-AR/cAMP signaling is counter-intuitive for the treatment of heart failure given the poor outcomes of heart failure patients treated chronically with β-adrenergic agonists [60]. Moreover, interfering with calcium signaling in heart failure would have the potential for significant off-target effects and arrhythmias. Targeting AMPK-mediated activation of PGC-1α could also prove effective [19, 61]. Interestingly, AMPK is activated by metformin, an FDA-approved drug for the treatment of diabetes. Recent evidence based on human and animal studies suggests that metformin may improve cardiac function and outcomes in patients with diabetes and heart failure [62, 63]. Intriguingly, a recent report demonstrated that metformin-mediated activation of AMPK improved survival and cardiac function after myocardial infarction in animals, and that this was associated with increased levels of PGC-1α mRNA in the heart [64]. Whether the benefit of metformin and AMPK activation is dependent on PGC-1α is not known. Moreover, given the potential risk of lactic acidosis with the use of metformin, larger clinical studies will be necessary to determine the value and safety of metformin for the treatment of heart failure in diabetic and non-diabetic patients.

PGC-1α and β protein activity is negatively regulated by acetylation. Thus, strategies to reduce PGC-1 acetylation represent another approach to modulate these coactivators. Published data suggests that the sirtuin SIRT1 is the predominant deacetylase for PGC-1α/β [17, 21]. The compound resveratrol activates SIRT1 enzyme activity [65], which subsequently deacetylates the PGC-1 coactivators and amplifies PGC-1 activity. Interestingly, resveratrol has been shown to be cardioprotective in a variety of heart failure models [6668]. Whether these effects are mediated through modulating PGC-1 activity is not known. As an alternative approach, inhibiting the acetyltransferase GCN5 could reduce PGC-1 acetylation. However, both SIRT1 and GCN5 have other targets besides the PGC-1 coactivators and, thus, the potential for off-target effects is significant. Further investigation into manipulating PGC-1 acetylation status for therapeutic gain is warranted.

One potential negative aspect of activating PGC-1 activity is that it coactivates many targets, setting the stage for effects beyond the mitochondrial targets. Accordingly, targeting ERR or the PGC-1/ERR interface would activate the pathways known to be deactivated in the hypertrophied and failing heart. Agonists of ERRα have not been described. However, a recent study described a compound that activates ERRγ and its downstream mitochondrial targets in cells [69]. Moreover, it has been shown that additional coactivators, such as Bcl3, can physically interact with and modulate the transcriptional activity of PGC-1α/ERRα [70]. This raises the possibility that targeting the PGC-1/ERR interface, rather than ERR, could have therapeutic application. The use of chemical biology screens to identify new small molecules that alter the ERR/PGC-1 transcriptional complex would be one approach to explore this concept.

Summary

The PGC-1/ERR axis is a central regulator of energy metabolism and mitochondrial function in heart. Together, these molecules orchestrate coordinated shifts in cardiac fuel and energy metabolism during cardiac development and in response to physiological stressors. Given that the activity of PGC-1 coactivators and ERRs are suppressed in heart failure caused by hypertension or late stages of diabetes, the PGC-1/ERR axis represents a novel potential therapeutic target for cardiovascular diseases. Further proof-of-concept studies investigating the consequences of PGC-1/ERR activation in animal models of heart failure are needed to address the potential benefit of this approach. If such studies prove promising, many strategies aimed at modulating this transcriptional regulatory axis can be considered.

Acknowledgments

This work was supported by NIH grants DK45416, HL058493 and KO8 22-3040-53603. Special thanks to Genevieve DeMaria for assistance with manuscript preparation and Teresa Leone for critique of the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998 Mar 20;92(6):829–39. [PubMed] [Google Scholar]
2. Lin J, Puigserver P, Donovan J, Tarr P, Spiegelman BM. Peroxisome proliferator-activated receptor gamma coactivator 1beta (PGC-1beta ), a novel PGC-1-related transcription coactivator associated with host cell factor. The Journal of Biological Chemistry. 2002 Jan 18;277(3):1645–8. [PubMed] [Google Scholar]
3. Huss JM, Kelly DP. Nuclear receptor signaling and cardiac energetics. Circulation Research. 2004 Sep 17;95(6):568–78. [PubMed] [Google Scholar]
4. Finck BN, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) regulatory cascade in cardiac physiology and disease. Circulation. 2007 May 15;115(19):2540–8. [PubMed] [Google Scholar]
5. Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metabolism. 2005 Jun;1(6):361–70. [PubMed] [Google Scholar]
6. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 1999 Jul 9;98(1):115–24. [PubMed] [Google Scholar]
7. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. The Journal of Clinical Investigation. 2000 Oct;106(7):847–56. [PMC free article] [PubMed] [Google Scholar]
8. Goto M, Terada S, Kato M, Katoh M, Yokozeki T, Tabata I, et al. cDNA Cloning and mRNA analysis of PGC-1 in epitrochlearis muscle in swimming-exercised rats. Biochemical and Biophysical Research Communications. 2000 Aug 2;274(2):350–4. [PubMed] [Google Scholar]
9. Handschin C, Rhee J, Lin J, Tarr PT, Spiegelman BM. An autoregulatory loop controls peroxisome proliferator-activated receptor gamma coactivator 1alpha expression in muscle. Proceedings of the National Academy of Sciences of the United States of America. 2003 Jun 10;100(12):7111–6. [PMC free article] [PubMed] [Google Scholar]
10. Schaeffer PJ, Wende AR, Magee CJ, Neilson JR, Leone TC, Chen F, et al. Calcineurin and calcium/calmodulin-dependent protein kinase activate distinct metabolic gene regulatory programs in cardiac muscle. The Journal of Biological Chemistry. 2004 Sep 17;279(38):39593–603. [PubMed] [Google Scholar]
11. Rohas LM, St-Pierre J, Uldry M, Jager S, Handschin C, Spiegelman BM. A fundamental system of cellular energy homeostasis regulated by PGC-1alpha. Proceedings of the National Academy of Sciences of the United States of America. 2007 May 8;104(19):7933–8. [PMC free article] [PubMed] [Google Scholar]
12. Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ, et al. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proceedings of the National Academy of Sciences of the United States of America. 2002 Dec 10;99(25):15983–7. [PMC free article] [PubMed] [Google Scholar]
13. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, et al. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science. 2003 Feb 7;299(5608):896–9. [PubMed] [Google Scholar]
14. Czubryt MP, McAnally J, Fishman GI, Olson EN. Regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha ) and mitochondrial function by MEF2 and HDAC5. Proceedings of the National Academy of Sciences of the United States of America. 2003 Feb 18;100(4):1711–6. [PMC free article] [PubMed] [Google Scholar]
15. Sonoda J, Laganiere J, Mehl IR, Barish GD, Chong LW, Li X, et al. Nuclear receptor ERR alpha and coactivator PGC-1 beta are effectors of IFN-gamma-induced host defense. Genes & Development. 2007 Aug 1;21(15):1909–20. [PMC free article] [PubMed] [Google Scholar]
16. Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metabolism. 2006 Jul;4(1):13–24. [PMC free article] [PubMed] [Google Scholar]
17. Canto C, Auwerx J. PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Current Opinion in Lipidology. 2009 Apr;20(2):98–105. [PMC free article] [PubMed] [Google Scholar]
18. Puigserver P, Rhee J, Lin J, Wu Z, Yoon JC, Zhang CY, et al. Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator-1. Molecular Cell. 2001 Nov;8(5):971–82. [PubMed] [Google Scholar]
19. Jager S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proceedings of the National Academy of Sciences of the United States of America. 2007 Jul 17;104(29):12017–22. [PMC free article] [PubMed] [Google Scholar]
20. Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009 Apr 23;458(7241):1056–60. [PMC free article] [PubMed] [Google Scholar]
21. Kelly TJ, Lerin C, Haas W, Gygi SP, Puigserver P. GCN5-mediated transcriptional control of the metabolic coactivator PGC-1beta through lysine acetylation. The Journal of Biological Chemistry. 2009 Jul 24;284(30):19945–52. [PMC free article] [PubMed] [Google Scholar]
22. Zhang Y, Huypens P, Adamson AW, Chang JS, Henagan TM, Boudreau A, et al. Alternative mRNA splicing produces a novel biologically active short isoform of PGC-1alpha. The Journal of Biological Chemistry. 2009 Nov 20;284(47):32813–26. [PMC free article] [PubMed] [Google Scholar]
23. Chang JS, Huypens P, Zhang Y, Black C, Kralli A, Gettys TW. Regulation of NT-PGC-1alpha subcellular localization and function by protein kinase A-dependent modulation of nuclear export by CRM1. The Journal of Biological Chemistry. Jun 4;285(23):18039–50. [PMC free article] [PubMed] [Google Scholar]
24. Giguere V, Yang N, Segui P, Evans RM. Identification of a new class of steroid hormone receptors. Nature. 1988 Jan 7;331(6151):91–4. [PubMed] [Google Scholar]
25. Huss JM, Kopp RP, Kelly DP. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. The Journal of Biological Chemistry. 2002 Oct 25;277(43):40265–74. [PubMed] [Google Scholar]
26. Kamei Y, Ohizumi H, Fujitani Y, Nemoto T, Tanaka T, Takahashi N, et al. PPARgamma coactivator 1beta/ERR ligand 1 is an ERR protein ligand, whose expression induces a high-energy expenditure and antagonizes obesity. Proceedings of the National Academy of Sciences of the United States of America. 2003 Oct 14;100(21):12378–83. [PMC free article] [PubMed] [Google Scholar]
27. Rangwala SM, Li X, Lindsley L, Wang X, Shaughnessy S, Daniels TG, et al. Estrogen-related receptor alpha is essential for the expression of antioxidant protection genes and mitochondrial function. Biochemical and Biophysical Research Communications. 2007 May 25;357(1):231–6. [PubMed] [Google Scholar]
28. Lai L, Leone TC, Zechner C, Schaeffer PJ, Kelly SM, Flanagan DP, et al. Transcriptional coactivators PGC-1alpha and PGC-lbeta control overlapping programs required for perinatal maturation of the heart. Genes & Development. 2008 Jul 15;22(14):1948–61. [PMC free article] [PubMed] [Google Scholar]
29. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang CY, et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell. 2004 Oct 1;119(1):121–35. [PubMed] [Google Scholar]
30. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina S, et al. PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biology. 2005 Apr;3(4):e101. [PMC free article] [PubMed] [Google Scholar]
31. Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, et al. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metabolism. 2005 Apr;1(4):259–71. [PubMed] [Google Scholar]
32. Lehman JJ, Boudina S, Banke NH, Sambandam N, Han X, Young DM, et al. The transcriptional coactivator PGC-1alpha is essential for maximal and efficient cardiac mitochondrial fatty acid oxidation and lipid homeostasis. Am J Physiol Heart Circ Physiol. 2008 Jul;295(1):H185–96. [PMC free article] [PubMed] [Google Scholar]
33. Lelliott CJ, Medina-Gomez G, Petrovic N, Kis A, Feldmann HM, Bjursell M, et al. Ablation of PGC-1beta results in defective mitochondrial activity, thermogenesis, hepatic function, and cardiac performance. PLoS Biology. 2006 Nov;4(11):e369. [PMC free article] [PubMed] [Google Scholar]
34. Vianna CR, Huntgeburth M, Coppari R, Choi CS, Lin J, Krauss S, et al. Hypomorphic mutation of PGC-1beta causes mitochondrial dysfunction and liver insulin resistance. Cell Metabolism. 2006 Dec;4(6):453–64. [PMC free article] [PubMed] [Google Scholar]
35. Sonoda J, Mehl IR, Chong LW, Nofsinger RR, Evans RM. PGC-1beta controls mitochondrial metabolism to modulate circadian activity, adaptive thermogenesis, and hepatic steatosis. Proceedings of the National Academy of Sciences of the United States of America. 2007 Mar 20;104(12):5223–8. [PMC free article] [PubMed] [Google Scholar]
36. Russell LK, Mansfield CM, Lehman JJ, Kovacs A, Courtois M, Saffitz JE, et al. Cardiac-specific induction of the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha promotes mitochondrial biogenesis and reversible cardiomyopathy in a developmental stage-dependent manner. Circulation Research. 2004 Mar 5;94(4):525–33. [PubMed] [Google Scholar]
37. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager S, et al. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell. 2006 Oct 20;127(2):397–408. [PubMed] [Google Scholar]
38. Alaynick WA, Kondo RP, Xie W, He W, Dufour CR, Downes M, et al. ERRgamma directs and maintains the transition to oxidative metabolism in the postnatal heart. Cell Metabolism. 2007 Jul;6(1):13–24. [PubMed] [Google Scholar]
39. Huss JM, Torra IP, Staels B, Giguere V, Kelly DP. Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Molecular and Cellular Biology. 2004 Oct;24(20):9079–91. [PMC free article] [PubMed] [Google Scholar]
40. Taegtmeyer H, Golfman L, Sharma S, Razeghi P, van Arsdall M. Linking gene expression to function: metabolic flexibility in the normal and diseased heart. Ann N Y Acad Sci. 2004 May;1015:202–13. [PubMed] [Google Scholar]
41. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabolism in the normal and failing heart. Physiological Reviews. 2005 Jul;85(3):1093–129. [PubMed] [Google Scholar]
42. de las Fuentes L, Herrero P, Peterson LR, Kelly DP, Gropler RJ, Davila-Roman VG. Myocardial fatty acid metabolism: independent predictor of left ventricular mass in hypertensive heart disease. Hypertension. 2003 Jan;41(1):83–7. [PubMed] [Google Scholar]
43. Huss JM, Imahashi K, Dufour CR, Weinheimer CJ, Courtois M, Kovacs A, et al. The nuclear receptor ERRalpha is required for the bioenergetic and functional adaptation to cardiac pressure overload. Cell Metabolism. 2007 Jul;6(1):25–37. [PubMed] [Google Scholar]
44. Young ME, Laws FA, Goodwin GW, Taegtmeyer H. Reactivation of peroxisome proliferator-activated receptor alpha is associated with contractile dysfunction in hypertrophied rat heart. The Journal of Biological Chemistry. 2001 Nov 30;276(48):44390–5. [PubMed] [Google Scholar]
45. Ingwall JS, Kramer MF, Fifer MA, Lorell BH, Shemin R, Grossman W, et al. The Creatine Kinase System in Normal and Diseased Human Myocardium. New England Journal of Medicine. 1985;313(17):1050–4. [PubMed] [Google Scholar]
46. Bottomley PA, Wu KC, Gerstenblith G, Schulman SP, Steinberg A, Weiss RG. Reduced Myocardial Creatine Kinase Flux in Human Myocardial Infarction: An In Vivo Phosphorus Magnetic Resonance Spectroscopy Study. Circulation. 2009 April 14;119(14):1918–24. [PMC free article] [PubMed] [Google Scholar]
47. Gupta A, Chacko VP, Weiss RG. Abnormal energetics and ATP depletion in pressure-overload mouse hearts: in vivo high-energy phosphate concentration measures by noninvasive magnetic resonance. Am J Physiol Heart Circ Physiol. 2009 July 1;297(1):H59–64. [PMC free article] [PubMed] [Google Scholar]
48. Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, Spiegelman BM. Transverse aortic constriction leads to accelerated heart failure in mice lacking PPAR-gamma coactivator 1alpha. Proceedings of the National Academy of Sciences of the United States of America. 2006 Jun 27;103(26):10086–91. [PMC free article] [PubMed] [Google Scholar]
49. Davila-Roman VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG, Kelly DP, et al. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. Journal of the American College of Cardiology. 2002 Jul 17;40(2):271–7. [PubMed] [Google Scholar]
50. Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP. Fatty acid oxidation enzyme gene expression is downregulated in the failing heart. Circulation. 1996 Dec 1;94(11):2837–42. [PubMed] [Google Scholar]
51. Sihag S, Cresci S, Li AY, Sucharov CC, Lehman JJ. PGC-1alpha and ERRalpha target gene downregulation is a signature of the failing human heart. Journal of Molecular and Cellular Cardiology. 2009 Feb;46(2):201–12. [PMC free article] [PubMed] [Google Scholar]
52. Koitabashi N, Bedja D, Zaiman AL, Pinto YM, Zhang M, Gabrielson KL, et al. Avoidance of transient cardiomyopathy in cardiomyocyte-targeted tamoxifen-induced MerCreMer gene deletion models. Circulation Research. 2009 Jul 2;105(1):12–5. [PMC free article] [PubMed] [Google Scholar]
53. Peterson LR, Herrero P, Schechtman KB, Racette SB, Waggoner AD, Kisrieva-Ware Z, et al. Effect of obesity and insulin resistance on myocardial substrate metabolism and efficiency in young women. Circulation. 2004 May 11;109(18):2191–6. [PubMed] [Google Scholar]
54. Herrero P, Peterson LR, McGill JB, Matthew S, Lesniak D, Dence C, et al. Increased myocardial fatty acid metabolism in patients with type 1 diabetes mellitus. Journal of the American College of Cardiology. 2006 Feb 7;47(3):598–604. [PubMed] [Google Scholar]
55. Buchanan J, Mazumder PK, Hu P, Chakrabarti G, Roberts MW, Yun UJ, et al. Reduced cardiac efficiency and altered substrate metabolism precedes the onset of hyperglycemia and contractile dysfunction in two mouse models of insulin resistance and obesity. Endocrinology. 2005 Dec;146(12):5341–9. [PubMed] [Google Scholar]
56. Duncan JG, Fong JL, Medeiros DM, Finck BN, Kelly DP. Insulin-resistant heart exhibits a mitochondrial biogenic response driven by the peroxisome proliferator-activated receptor-alpha/PGC-1alpha gene regulatory pathway. Circulation. 2007 Feb 20;115(7):909–17. [PMC free article] [PubMed] [Google Scholar]
57. Sharma S, Adrogue JV, Golfman L, Uray I, Lemm J, Youker K, et al. Intramyocardial lipid accumulation in the failing human heart resembles the lipotoxic rat heart. Faseb J. 2004 Nov;18(14):1692–700. [PubMed] [Google Scholar]
58. Brookheart RT, Michel CI, Schaffer JE. As a matter of fat. Cell Metabolism. 2009 Jul;10(1):9–12. [PMC free article] [PubMed] [Google Scholar]
59. Wende AR, Schaeffer PJ, Parker GJ, Zechner C, Han DH, Chen MM, et al. A role for the transcriptional coactivator PGC-1alpha in muscle refueling. The Journal of Biological Chemistry. 2007 Dec 14;282(50):36642–51. [PubMed] [Google Scholar]
60. Elkayam U, Tasissa G, Binanay C, Stevenson LW, Gheorghiade M, Warnica JW, et al. Use and impact of inotropes and vasodilator therapy in hospitalized patients with severe heart failure. American Heart Journal. 2007 Jan;153(1):98–104. [PubMed] [Google Scholar]
61. Lee WJ, Kim M, Park HS, Kim HS, Jeon MJ, Oh KS, et al. AMPK activation increases fatty acid oxidation in skeletal muscle by activating PPARalpha and PGC-1. Biochemical and Biophysical Research Communications. 2006 Feb 3;340(1):291–5. [PubMed] [Google Scholar]
62. Eurich DT, McAlister FA, Blackburn DF, Majumdar SR, Tsuyuki RT, Varney J, et al. Benefits and harms of antidiabetic agents in patients with diabetes and heart failure: systematic review. BMJ (Clinical research ed) 2007 Sep 8;335(7618):497. [PMC free article] [PubMed] [Google Scholar]
63. Sasaki H, Asanuma H, Fujita M, Takahama H, Wakeno M, Ito S, et al. Metformin prevents progression of heart failure in dogs: role of AMP-activated protein kinase. Circulation. 2009 May 19;119(19):2568–77. [PubMed] [Google Scholar]
64. Gundewar S, Calvert JW, Jha S, Toedt-Pingel I, Ji SY, Nunez D, et al. Activation of AMP-activated protein kinase by metformin improves left ventricular function and survival in heart failure. Circulation Research. 2009 Feb 13;104(3):403–11. [PMC free article] [PubMed] [Google Scholar]
65. Elliott PJ, Jirousek M. Sirtuins: novel targets for metabolic disease. Curr Opin Investig Drugs. 2008 Apr;9(4):371–8. [PubMed] [Google Scholar]
66. Chen CJ, Yu W, Fu YC, Wang X, Li JL, Wang W. Resveratrol protects cardiomyocytes from hypoxia-induced apoptosis through the SIRT1-FoxO1 pathway. Biochemical and Biophysical Research Communications. 2009 Jan 16;378(3):389–93. [PubMed] [Google Scholar]
67. Lin JF, Lin SM, Chih CL, Nien MW, Su HH, Hu BR, et al. Resveratrol reduces infarct size and improves ventricular function after myocardial ischemia in rats. Life Sciences. 2008 Aug 29;83(9–10):313–7. [PubMed] [Google Scholar]
68. Thirunavukkarasu M, Penumathsa SV, Koneru S, Juhasz B, Zhan L, Otani H, et al. Resveratrol alleviates cardiac dysfunction in streptozotocin-induced diabetes: Role of nitric oxide, thioredoxin, and heme oxygenase. Free Radical Biology & Medicine. 2007 Sep 1;43(5):720–9. [PMC free article] [PubMed] [Google Scholar]
69. Rangwala SM, Wang X, Calvo JA, Lindsley L, Zhang Y, Deyneko G, et al. Estrogen-related Receptor Is a Key Regulator of Muscle Mitochondrial Activity and Oxidative Capacity. Journal of Biological Chemistry. 2010 July 16;285(29):22619–29. [PMC free article] [PubMed] [Google Scholar]
70. Yang J, Williams RS, Kelly DP. Bcl3 interacts cooperatively with peroxisome proliferator-activated receptor gamma (PPARgamma) coactivator 1alpha to coactivate nuclear receptors estrogen-related receptor alpha and PPARalpha. Molecular and Cellular Biology. 2009 Aug;29(15):4091–102. [PMC free article] [PubMed] [Google Scholar]
-