Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 02.
Article in English | MEDLINE | ID: mdl-38746425

ABSTRACT

Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are novel, potent heart failure medications with an unknown mechanism of action. We sought to determine if the beneficial actions of SGLT2i in heart failure were on- or off-target, and related to metabolic reprogramming, including increased lipolysis and ketogenesis. The phenotype of mice treated with empagliflozin and genetically engineered mice constitutively lacking SGLT2 mirrored metabolic changes seen in human clinical trials (including reduced blood glucose, increased ketogenesis, and profound glucosuria). In a mouse heart failure model, SGLT2i treatment, but not generalized SGLT2 knockout, resulted in improved systolic function and reduced pathologic cardiac remodeling. SGLT2i treatment of the SGLT2 knockout mice sustained the cardiac benefits, demonstrating an off-target role for these drugs. This benefit is independent of metabolic changes, including ketosis. The mechanism of action and target of SGLT2i in HF remain elusive.

2.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36927960

ABSTRACT

During the development of heart failure (HF), the capacity for cardiomyocyte (CM) fatty acid oxidation (FAO) and ATP production is progressively diminished, contributing to pathologic cardiac hypertrophy and contractile dysfunction. Receptor-interacting protein 140 (RIP140, encoded by Nrip1) has been shown to function as a transcriptional corepressor of oxidative metabolism. We found that mice with striated muscle deficiency of RIP140 (strNrip1-/-) exhibited increased expression of a broad array of genes involved in mitochondrial energy metabolism and contractile function in heart and skeletal muscle. strNrip1-/- mice were resistant to the development of pressure overload-induced cardiac hypertrophy, and CM-specific RIP140-deficient (csNrip1-/-) mice were protected against the development of HF caused by pressure overload combined with myocardial infarction. Genomic enhancers activated by RIP140 deficiency in CMs were enriched in binding motifs for transcriptional regulators of mitochondrial function (estrogen-related receptor) and cardiac contractile proteins (myocyte enhancer factor 2). Consistent with a role in the control of cardiac fatty acid oxidation, loss of RIP140 in heart resulted in augmented triacylglyceride turnover and fatty acid utilization. We conclude that RIP140 functions as a suppressor of a transcriptional regulatory network that controls cardiac fuel metabolism and contractile function, representing a potential therapeutic target for the treatment of HF.


Subject(s)
Heart Failure , Nuclear Receptor Interacting Protein 1 , Animals , Mice , Cardiomegaly/metabolism , Energy Metabolism/genetics , Fatty Acids/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Nuclear Receptor Interacting Protein 1/genetics , Nuclear Receptor Interacting Protein 1/metabolism
3.
Circ Heart Fail ; 14(1): e007684, 2021 01.
Article in English | MEDLINE | ID: mdl-33356362

ABSTRACT

BACKGROUND: Accumulating evidence suggests that the failing heart reprograms fuel metabolism toward increased utilization of ketone bodies and that increasing cardiac ketone delivery ameliorates cardiac dysfunction. As an initial step toward development of ketone therapies, we investigated the effect of chronic oral ketone ester (KE) supplementation as a prevention or treatment strategy in rodent heart failure models. METHODS: Two independent rodent heart failure models were used for the studies: transverse aortic constriction/myocardial infarction (MI) in mice and post-MI remodeling in rats. Seventy-five mice underwent a prevention treatment strategy with a KE comprised of hexanoyl-hexyl-3-hydroxybutyrate KE (KE-1) diet, and 77 rats were treated in either a prevention or treatment regimen using a commercially available ß-hydroxybutyrate-(R)-1,3-butanediol monoester (DeltaG; KE-2) diet. RESULTS: The KE-1 diet in mice elevated ß-hydroxybutyrate levels during nocturnal feeding, whereas the KE-2 diet in rats induced ketonemia throughout a 24-hour period. The KE-1 diet preventive strategy attenuated development of left ventricular dysfunction and remodeling post-transverse aortic constriction/MI (left ventricular ejection fraction±SD, 36±8 in vehicle versus 45±11 in KE-1; P=0.016). The KE-2 diet therapeutic approach also attenuated left ventricular dysfunction and remodeling post-MI (left ventricular ejection fraction, 41±11 in MI-vehicle versus 61±7 in MI-KE-2; P<0.001). In addition, ventricular weight, cardiomyocyte cross-sectional area, and the expression of ANP (atrial natriuretic peptide) were significantly attenuated in the KE-2-treated MI group. However, treatment with KE-2 did not influence cardiac fibrosis post-MI. The myocardial expression of the ketone transporter and 2 ketolytic enzymes was significantly increased in rats fed KE-2 diet along with normalization of myocardial ATP levels to sham values. CONCLUSIONS: Chronic oral supplementation with KE was effective in both prevention and treatment of heart failure in 2 preclinical animal models. In addition, our results indicate that treatment with KE reprogrammed the expression of genes involved in ketone body utilization and normalized myocardial ATP production following MI, consistent with provision of an auxiliary fuel. These findings provide rationale for the assessment of KEs as a treatment for patients with heart failure.


Subject(s)
Dietary Supplements , Heart Failure/physiopathology , Hydroxybutyrates , Myocardial Infarction/physiopathology , Myocardium/metabolism , Stroke Volume/physiology , Ventricular Dysfunction, Left/physiopathology , Adenosine Triphosphate/metabolism , Animals , Aorta/surgery , Atrial Natriuretic Factor/metabolism , Constriction, Pathologic , Fibrosis , Heart Failure/metabolism , Heart Failure/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Size , Rats , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Function, Left
4.
Circ Res ; 126(2): 197-199, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31944916
5.
Am J Physiol Endocrinol Metab ; 312(5): E394-E406, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28270443

ABSTRACT

Peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α and -1ß serve as master transcriptional regulators of muscle mitochondrial functional capacity and are capable of enhancing muscle endurance when overexpressed in mice. We sought to determine whether muscle-specific transgenic overexpression of PGC-1ß affects the detraining response following endurance training. First, we established and validated a mouse exercise-training-detraining protocol. Second, using multiple physiological and gene expression end points, we found that PGC-1ß overexpression in skeletal muscle of sedentary mice fully recapitulated the training response. Lastly, PGC-1ß overexpression during the detraining period resulted in partial prevention of the detraining response. Specifically, an increase in the plateau at which O2 uptake (V̇o2) did not change from baseline with increasing treadmill speed [peak V̇o2 (ΔV̇o2max)] was maintained in trained mice with PGC-1ß overexpression in muscle 6 wk after cessation of training. However, other detraining responses, including changes in running performance and in situ half relaxation time (a measure of contractility), were not affected by PGC-1ß overexpression. We conclude that while activation of muscle PGC-1ß is sufficient to drive the complete endurance phenotype in sedentary mice, it only partially prevents the detraining response following exercise training, suggesting that the process of endurance detraining involves mechanisms beyond the reversal of muscle autonomous mechanisms involved in endurance fitness. In addition, the protocol described here should be useful for assessing early-stage proof-of-concept interventions in preclinical models of muscle disuse atrophy.


Subject(s)
Muscle, Skeletal/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal/methods , Physical Endurance/physiology , Physical Fitness/physiology , Running/physiology , Animals , Male , Mice , Mice, Transgenic , Muscular Disorders, Atrophic/physiopathology , Muscular Disorders, Atrophic/prevention & control , Phenotype
6.
J Clin Invest ; 126(9): 3567-79, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27500491

ABSTRACT

Intramuscular lipid accumulation is a common manifestation of chronic caloric excess and obesity that is strongly associated with insulin resistance. The mechanistic links between lipid accumulation in myocytes and insulin resistance are not completely understood. In this work, we used a high-throughput chemical biology screen to identify a small-molecule probe, SBI-477, that coordinately inhibited triacylglyceride (TAG) synthesis and enhanced basal glucose uptake in human skeletal myocytes. We then determined that SBI-477 stimulated insulin signaling by deactivating the transcription factor MondoA, leading to reduced expression of the insulin pathway suppressors thioredoxin-interacting protein (TXNIP) and arrestin domain-containing 4 (ARRDC4). Depleting MondoA in myocytes reproduced the effects of SBI-477 on glucose uptake and myocyte lipid accumulation. Furthermore, an analog of SBI-477 suppressed TXNIP expression, reduced muscle and liver TAG levels, enhanced insulin signaling, and improved glucose tolerance in mice fed a high-fat diet. These results identify a key role for MondoA-directed programs in the coordinated control of myocyte lipid balance and insulin signaling and suggest that this pathway may have potential as a therapeutic target for insulin resistance and lipotoxicity.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Insulin/metabolism , Lipid Metabolism , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Animals , Arrestins/metabolism , Biopsy , Carrier Proteins/metabolism , Cell Cycle Proteins , Diet, High-Fat , Glucose/metabolism , Homeostasis , Insulin Resistance/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Signal Transduction , Thioredoxins/metabolism , Transfection , Triglycerides/metabolism
7.
JCI Insight ; 2(1)2016 Feb.
Article in English | MEDLINE | ID: mdl-26998524

ABSTRACT

Myocardial fuel and energy metabolic derangements contribute to the pathogenesis of heart failure. Recent evidence implicates posttranslational mechanisms in the energy metabolic disturbances that contribute to the pathogenesis of heart failure. We hypothesized that accumulation of metabolite intermediates of fuel oxidation pathways drives posttranslational modifications of mitochondrial proteins during the development of heart failure. Myocardial acetylproteomics demonstrated extensive mitochondrial protein lysine hyperacetylation in the early stages of heart failure in well-defined mouse models and the in end-stage failing human heart. To determine the functional impact of increased mitochondrial protein acetylation, we focused on succinate dehydrogenase A (SDHA), a critical component of both the tricarboxylic acid (TCA) cycle and respiratory complex II. An acetyl-mimetic mutation targeting an SDHA lysine residue shown to be hyperacetylated in the failing human heart reduced catalytic function and reduced complex II-driven respiration. These results identify alterations in mitochondrial acetyl-CoA homeostasis as a potential driver of the development of energy metabolic derangements that contribute to heart failure.

8.
Circulation ; 133(8): 698-705, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26819376

ABSTRACT

BACKGROUND: Significant evidence indicates that the failing heart is energy starved. During the development of heart failure, the capacity of the heart to utilize fatty acids, the chief fuel, is diminished. Identification of alternate pathways for myocardial fuel oxidation could unveil novel strategies to treat heart failure. METHODS AND RESULTS: Quantitative mitochondrial proteomics was used to identify energy metabolic derangements that occur during the development of cardiac hypertrophy and heart failure in well-defined mouse models. As expected, the amounts of proteins involved in fatty acid utilization were downregulated in myocardial samples from the failing heart. Conversely, expression of ß-hydroxybutyrate dehydrogenase 1, a key enzyme in the ketone oxidation pathway, was increased in the heart failure samples. Studies of relative oxidation in an isolated heart preparation using ex vivo nuclear magnetic resonance combined with targeted quantitative myocardial metabolomic profiling using mass spectrometry revealed that the hypertrophied and failing heart shifts to oxidizing ketone bodies as a fuel source in the context of reduced capacity to oxidize fatty acids. Distinct myocardial metabolomic signatures of ketone oxidation were identified. CONCLUSIONS: These results indicate that the hypertrophied and failing heart shifts to ketone bodies as a significant fuel source for oxidative ATP production. Specific metabolite biosignatures of in vivo cardiac ketone utilization were identified. Future studies aimed at determining whether this fuel shift is adaptive or maladaptive could unveil new therapeutic strategies for heart failure.


Subject(s)
Diet, Ketogenic/methods , Fatty Acids/metabolism , Heart Failure/metabolism , Heart Failure/pathology , Ketone Bodies/metabolism , Animals , Female , Gene Expression Profiling/methods , Heart Failure/diet therapy , Mice , Mice, Inbred C57BL
9.
J Clin Invest ; 125(9): 3461-76, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26241060

ABSTRACT

Mitochondrial homeostasis is critical for tissue health, and mitochondrial dysfunction contributes to numerous diseases, including heart failure. Here, we have shown that the transcription factor Kruppel-like factor 4 (KLF4) governs mitochondrial biogenesis, metabolic function, dynamics, and autophagic clearance. Adult mice with cardiac-specific Klf4 deficiency developed cardiac dysfunction with aging or in response to pressure overload that was characterized by reduced myocardial ATP levels, elevated ROS, and marked alterations in mitochondrial shape, size, ultrastructure, and alignment. Evaluation of mitochondria isolated from KLF4-deficient hearts revealed a reduced respiration rate that is likely due to defects in electron transport chain complex I. Further, cardiac-specific, embryonic Klf4 deletion resulted in postnatal premature mortality, impaired mitochondrial biogenesis, and altered mitochondrial maturation. We determined that KLF4 binds to, cooperates with, and is requisite for optimal function of the estrogen-related receptor/PPARγ coactivator 1 (ERR/PGC-1) transcriptional regulatory module on metabolic and mitochondrial targets. Finally, we found that KLF4 regulates autophagy flux through transcriptional regulation of a broad array of autophagy genes in cardiomyocytes. Collectively, these findings identify KLF4 as a nodal transcriptional regulator of mitochondrial homeostasis.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Transcription, Genetic , Animals , Autophagy/genetics , HEK293 Cells , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Oxygen Consumption/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Acta Neuropathol Commun ; 3: 16, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25853296

ABSTRACT

INTRODUCTION: Mitochondrial dysfunction and oxidative stress are critical factors in the pathogenesis of age-dependent neurodegenerative diseases. PGC-1α, a master regulator of mitochondrial biogenesis and cellular antioxidant defense, has emerged as a possible therapeutic target for Parkinson's disease, with important roles in the function and survival of dopaminergic neurons in the substantia nigra. The objective of this study is to determine if the loss of PGC-1α activity contributes to α-synuclein-induced degeneration. RESULTS: We explore the vulnerability of PGC-1α null mice to the accumulation of human α-synuclein in nigral neurons, and assess the neuroprotective effect of AAV-mediated PGC-1α expression in this experimental model. Using neuronal cultures derived from these mice, mitochondrial respiration and production of reactive oxygen species are assessed in conditions of human α-synuclein overexpression. We find ultrastructural evidence for abnormal mitochondria and fragmented endoplasmic reticulum in the nigral dopaminergic neurons of PGC-1α null mice. Furthermore, PGC-1α null nigral neurons are more prone to degenerate following overexpression of human α-synuclein, an effect more apparent in male mice. PGC-1α overexpression restores mitochondrial morphology, oxidative stress detoxification and basal respiration, which is consistent with the observed neuroprotection against α-synuclein toxicity in male PGC-1α null mice. CONCLUSIONS: Altogether, our results highlight an important role for PGC-1α in controlling the mitochondrial function of nigral neurons accumulating α-synuclein, which may be critical for gender-dependent vulnerability to Parkinson's disease.


Subject(s)
Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Substantia Nigra/cytology , Transcription Factors/metabolism , alpha-Synuclein/metabolism , Animals , Cell Culture Techniques , Dopaminergic Neurons/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Oxidative Stress/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Sex Factors , Transcription Factors/deficiency , Transcription Factors/genetics
11.
Circ Heart Fail ; 7(6): 1022-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25236884

ABSTRACT

BACKGROUND: An unbiased systems approach was used to define energy metabolic events that occur during the pathological cardiac remodeling en route to heart failure (HF). METHODS AND RESULTS: Combined myocardial transcriptomic and metabolomic profiling were conducted in a well-defined mouse model of HF that allows comparative assessment of compensated and decompensated (HF) forms of cardiac hypertrophy because of pressure overload. The pressure overload data sets were also compared with the myocardial transcriptome and metabolome for an adaptive (physiological) form of cardiac hypertrophy because of endurance exercise training. Comparative analysis of the data sets led to the following conclusions: (1) expression of most genes involved in mitochondrial energy transduction were not significantly changed in the hypertrophied or failing heart, with the notable exception of a progressive downregulation of transcripts encoding proteins and enzymes involved in myocyte fatty acid transport and oxidation during the development of HF; (2) tissue metabolite profiles were more broadly regulated than corresponding metabolic gene regulatory changes, suggesting significant regulation at the post-transcriptional level; (3) metabolomic signatures distinguished pathological and physiological forms of cardiac hypertrophy and served as robust markers for the onset of HF; and (4) the pattern of metabolite derangements in the failing heart suggests bottlenecks of carbon substrate flux into the Krebs cycle. CONCLUSIONS: Mitochondrial energy metabolic derangements that occur during the early development of pressure overload-induced HF involve both transcriptional and post-transcriptional events. A subset of the myocardial metabolomic profile robustly distinguished pathological and physiological cardiac remodeling.


Subject(s)
Cardiomegaly/physiopathology , Energy Metabolism/physiology , Heart Failure/physiopathology , Ventricular Remodeling/physiology , Amino Acids/metabolism , Animals , Disease Models, Animal , Down-Regulation/physiology , Energy Metabolism/genetics , Gene Expression Profiling , Heart Failure/metabolism , Lipid Metabolism/genetics , Metabolome , Mice , Up-Regulation/physiology
12.
J Biol Chem ; 289(4): 2250-9, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24337569

ABSTRACT

The energy demands of the adult mammalian heart are met largely by ATP generated via oxidation of fatty acids in a high capacity mitochondrial system. Peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1)-α and -ß serve as inducible transcriptional coregulators of genes involved in mitochondrial biogenesis and metabolism. Whether PGC-1 plays a role in the regulation of mitochondrial structure is unknown. In this study, mice with combined deficiency of PGC-1α and PGC-1ß (PGC-1αß(-/-)) in adult heart were analyzed. PGC-1αß(-/-) hearts exhibited a distinctive mitochondrial cristae-stacking abnormality suggestive of a phospholipid abnormality as has been described in humans with genetic defects in cardiolipin (CL) synthesis (Barth syndrome). A subset of molecular species, containing n-3 polyunsaturated species in the CL, phosphatidylcholine, and phosphatidylethanolamine profiles, was reduced in PGC-1αß-deficient hearts. Gene expression profiling of PGC-1αß(-/-) hearts revealed reduced expression of the gene encoding CDP-diacylglycerol synthase 1 (Cds1), an enzyme that catalyzes the proximal step in CL biosynthesis. Cds1 gene promoter-reporter cotransfection experiments and chromatin immunoprecipitation studies demonstrated that PGC-1α coregulates estrogen-related receptors to activate the transcription of the Cds1 gene. We conclude that the PGC-1/estrogen-related receptor axis coordinately regulates metabolic and membrane structural programs relevant to the maintenance of high capacity mitochondrial function in heart.


Subject(s)
Diacylglycerol Cholinephosphotransferase/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Phosphatidylcholines/biosynthesis , Phosphatidylethanolamines/biosynthesis , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , Barth Syndrome/genetics , Barth Syndrome/metabolism , Barth Syndrome/pathology , Cell Line , Diacylglycerol Cholinephosphotransferase/genetics , Female , Humans , Mice , Mice, Knockout , Mitochondria, Heart , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphatidylcholines/genetics , Phosphatidylethanolamines/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transcription Factors/genetics
13.
Circ Res ; 114(4): 626-36, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24366168

ABSTRACT

RATIONALE: Increasing evidence has shown that proper control of mitochondrial dynamics (fusion and fission) is required for high-capacity ATP production in the heart. Transcriptional coactivators, peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1) α and PGC-1ß, have been shown to regulate mitochondrial biogenesis in the heart at the time of birth. The function of PGC-1 coactivators in the heart after birth has been incompletely understood. OBJECTIVE: Our aim was to assess the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts in mice. METHODS AND RESULTS: Conditional gene targeting was used in mice to explore the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts. Marked mitochondrial structural derangements were observed in hearts of PGC-1α/ß-deficient mice during postnatal growth, including fragmentation and elongation, associated with the development of a lethal cardiomyopathy. The expression of genes involved in mitochondrial fusion (Mfn1, Opa1) and fission (Drp1, Fis1) was altered in the hearts of PGC-1α/ß-deficient mice. PGC-lα was shown to directly regulate Mfn1 gene transcription by coactivating the estrogen-related receptor α on a conserved DNA element. Surprisingly, PGC-1α/ß deficiency in the adult heart did not result in evidence of abnormal mitochondrial dynamics or heart failure. However, transcriptional profiling demonstrated that PGC-1 coactivators are required for high-level expression of nuclear- and mitochondrial-encoded genes involved in mitochondrial dynamics and energy transduction in the adult heart. CONCLUSIONS: These results reveal distinct developmental stage-specific programs involved in cardiac mitochondrial dynamics.


Subject(s)
Cardiomyopathies/metabolism , Heart/growth & development , Mitochondria, Heart/metabolism , Transcription Factors/metabolism , Age Factors , Animals , Cardiomyopathies/genetics , Disease Progression , Energy Metabolism/physiology , Estrogen Receptor alpha/metabolism , Female , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics
14.
Mol Metab ; 2(3): 194-204, 2013.
Article in English | MEDLINE | ID: mdl-24049734

ABSTRACT

Peroxisome proliferator-activated receptorγ coactivators (PGC-1α and PGC-1ß) play important roles in the transcriptional regulation of intermediary metabolism. To evaluate the effects of overexpressing PGC-1α or PGC-1ß at physiologic levels in liver, we generated transgenic mice with inducible overexpression of PGC-1α or PGC-1ß. Gene expression array profiling revealed that whereas both PGC-1 family proteins induced mitochondrial oxidative enzymes, the expression of several genes involved in converting glucose to fatty acid was induced by PGC-1ß, but not PGC-1α. The increased expression of enzymes involved in carbohydrate utilization and de novo lipogenesis by PGC-1ß required carbohydrate response element binding protein (ChREBP). The interaction between PGC-1ß and ChREBP, as well as PGC-1ß occupancy of the liver-type pyruvate kinase promoter, was influenced by glucose concentration and liver-specific PGC-1ß(-/-) hepatocytes were refractory to the lipogenic response to high glucose conditions. These data suggest that PGC-1ß-mediated coactivation of ChREBP is involved in the lipogenic response to hyperglycemia.

15.
J Clin Invest ; 123(6): 2564-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676496

ABSTRACT

The mechanisms involved in the coordinate regulation of the metabolic and structural programs controlling muscle fitness and endurance are unknown. Recently, the nuclear receptor PPARß/δ was shown to activate muscle endurance programs in transgenic mice. In contrast, muscle-specific transgenic overexpression of the related nuclear receptor, PPARα, results in reduced capacity for endurance exercise. We took advantage of the divergent actions of PPARß/δ and PPARα to explore the downstream regulatory circuitry that orchestrates the programs linking muscle fiber type with energy metabolism. Our results indicate that, in addition to the well-established role in transcriptional control of muscle metabolic genes, PPARß/δ and PPARα participate in programs that exert opposing actions upon the type I fiber program through a distinct muscle microRNA (miRNA) network, dependent on the actions of another nuclear receptor, estrogen-related receptor γ (ERRγ). Gain-of-function and loss-of-function strategies in mice, together with assessment of muscle biopsies from humans, demonstrated that type I muscle fiber proportion is increased via the stimulatory actions of ERRγ on the expression of miR-499 and miR-208b. This nuclear receptor/miRNA regulatory circuit shows promise for the identification of therapeutic targets aimed at maintaining muscle fitness in a variety of chronic disease states, such as obesity, skeletal myopathies, and heart failure.


Subject(s)
Energy Metabolism , MicroRNAs/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Animals , Cell Line , Gene Expression , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , MicroRNAs/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , PPAR alpha/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal-To-Noise Ratio
16.
PLoS One ; 8(1): e54054, 2013.
Article in English | MEDLINE | ID: mdl-23342071

ABSTRACT

BACKGROUND: Inflammatory cytokines play an important role in the pathogenesis of heart failure. We have recently found the cytokine TWEAK (tumor necrosis factor (TNF)-like weak inducer of apoptosis), a member of the TNF superfamily, to be increased in patients with cardiomyopathy and result in the development of heart failure when overexpressed in mice. The molecular mechanisms underlying TWEAK-induced cardiac pathology, however, remain unknown. METHODOLOGY AND CRITICAL FINDING: Using mouse models of elevated circulating TWEAK levels, established through intravenous injection of adenovirus expressing TWEAK or recombinant TWEAK protein, we find that TWEAK induces a progressive dilated cardiomyopathy with impaired contractile function in mice. Moreover, TWEAK treatment is associated with decreased expression of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC1α) and genes required for mitochondrial oxidative phosphorylation, which precede the onset of cardiac dysfunction. TWEAK-induced downregulation of PGC1α requires expression of its cell surface receptor, fibroblast growth factor-inducible 14 (Fn14). We further find that TWEAK downregulates PGC1α gene expression via the TNF receptor-associated factor 2 (TRAF2) and NFκB signaling pathways. Maintaining PGC1α levels through adenoviral-mediated gene expression is sufficient to protect against TWEAK-induced cardiomyocyte dysfunction. CONCLUSION: Collectively, our data suggest that TWEAK induces cardiac dysfunction via downregulation of PGC1α, through FN14-TRAF2-NFκB-dependent signaling. Selective targeting of the FN14-TRAF2-NFκB-dependent signaling pathway or augmenting PGC1α levels may serve as novel therapeutic strategies for cardiomyopathy and heart failure.


Subject(s)
Tumor Necrosis Factors/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokine TWEAK , Echocardiography , Fibroblast Growth Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 2/metabolism , Trans-Activators/metabolism , Transcription Factors , Tumor Necrosis Factors/genetics
17.
Nature ; 494(7435): 105-10, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23354045

ABSTRACT

Cellular reprogramming of somatic cells to patient-specific induced pluripotent stem cells (iPSCs) enables in vitro modelling of human genetic disorders for pathogenic investigations and therapeutic screens. However, using iPSC-derived cardiomyocytes (iPSC-CMs) to model an adult-onset heart disease remains challenging owing to the uncertainty regarding the ability of relatively immature iPSC-CMs to fully recapitulate adult disease phenotypes. Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is an inherited heart disease characterized by pathological fatty infiltration and cardiomyocyte loss predominantly in the right ventricle, which is associated with life-threatening ventricular arrhythmias. Over 50% of affected individuals have desmosome gene mutations, most commonly in PKP2, encoding plakophilin-2 (ref. 9). The median age at presentation of ARVD/C is 26 years. We used previously published methods to generate iPSC lines from fibroblasts of two patients with ARVD/C and PKP2 mutations. Mutant PKP2 iPSC-CMs demonstrate abnormal plakoglobin nuclear translocation and decreased ß-catenin activity in cardiogenic conditions; yet, these abnormal features are insufficient to reproduce the pathological phenotypes of ARVD/C in standard cardiogenic conditions. Here we show that induction of adult-like metabolic energetics from an embryonic/glycolytic state and abnormal peroxisome proliferator-activated receptor gamma (PPAR-γ) activation underlie the pathogenesis of ARVD/C. By co-activating normal PPAR-alpha-dependent metabolism and abnormal PPAR-γ pathway in beating embryoid bodies (EBs) with defined media, we established an efficient ARVD/C in vitro model within 2 months. This model manifests exaggerated lipogenesis and apoptosis in mutant PKP2 iPSC-CMs. iPSC-CMs with a homozygous PKP2 mutation also had calcium-handling deficits. Our study is the first to demonstrate that induction of adult-like metabolism has a critical role in establishing an adult-onset disease model using patient-specific iPSCs. Using this model, we revealed crucial pathogenic insights that metabolic derangement in adult-like metabolic milieu underlies ARVD/C pathologies, enabling us to propose novel disease-modifying therapeutic strategies.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/metabolism , Arrhythmogenic Right Ventricular Dysplasia/pathology , Induced Pluripotent Stem Cells/pathology , Models, Biological , Active Transport, Cell Nucleus , Age of Onset , Apoptosis/genetics , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Cellular Reprogramming , Culture Media/pharmacology , Embryoid Bodies/drug effects , Embryoid Bodies/physiology , Energy Metabolism/genetics , Fatty Acids/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Glucose/metabolism , Glycolysis , Humans , Induced Pluripotent Stem Cells/metabolism , Lipogenesis/genetics , Myocardial Contraction/drug effects , Myocytes, Cardiac/pathology , PPAR alpha/metabolism , PPAR gamma/metabolism , Phenotype , Plakophilins/genetics , Time Factors , beta Catenin/metabolism
18.
PLoS One ; 7(12): e52645, 2012.
Article in English | MEDLINE | ID: mdl-23285128

ABSTRACT

PGC-1ß plays pleiotropic roles in regulating intermediary metabolism and has been shown to regulate both catabolic and anabolic processes in liver. We sought to evaluate the effects of PGC-1ß on liver energy metabolism by generating mice with postnatal, liver-specific deletion of PGC-1ß (LS-PGC-1ß(-/-) mice). LS-PGC-1ß(-/-) mice were outwardly normal, but exhibited a significant increase in hepatic triglyceride content at 6 weeks of age. Hepatic steatosis was due, at least in part, to impaired capacity for fatty acid oxidation and marked mitochondrial dysfunction. Mitochondrial DNA content and the expression of genes encoding multiple steps in mitochondrial fatty acid oxidation and oxidative phosphorylation pathways were significantly diminished in LS-PGC-1ß(-/-) mice. Liquid chromatography mass spectrometry-based analyses also revealed that acetylcarnitine and butyrylcarnitine levels were depleted whereas palmitoylcarnitine content was increased in LS-PGC-1ß(-/-) liver, which is consistent with attenuated rates of fatty acid oxidation. Interestingly, loss of PGC-1ß also significantly impaired inducible expression of glycolytic and lipogenic enzymes that occurs with high carbohydrate diet refeeding after a prolonged fast. These results suggest that PGC-1ß plays dual roles in regulating hepatic fatty acid metabolism by controlling the expression of programs of genes involved in both fatty acid oxidation and de novo fatty acid synthesis.


Subject(s)
Fasting/metabolism , Lipid Metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Trans-Activators/deficiency , Animals , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Energy Metabolism , Female , Gene Deletion , Gene Expression , Glycolysis , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/genetics , Transcription Factors , Triglycerides/metabolism
19.
Genes Dev ; 25(24): 2619-30, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22135324

ABSTRACT

To identify new gene regulatory pathways controlling skeletal muscle energy metabolism, comparative studies were conducted on muscle-specific transgenic mouse lines expressing the nuclear receptors peroxisome proliferator-activated receptor α (PPARα; muscle creatine kinase [MCK]-PPARα) or PPARß/δ (MCK-PPARß/δ). MCK-PPARß/δ mice are known to have enhanced exercise performance, whereas MCK-PPARα mice perform at low levels. Transcriptional profiling revealed that the lactate dehydrogenase b (Ldhb)/Ldha gene expression ratio is increased in MCK-PPARß/δ muscle, an isoenzyme shift that diverts pyruvate into the mitochondrion for the final steps of glucose oxidation. PPARß/δ gain- and loss-of-function studies in skeletal myotubes demonstrated that PPARß/δ, but not PPARα, interacts with the exercise-inducible kinase AMP-activated protein kinase (AMPK) to synergistically activate Ldhb gene transcription by cooperating with myocyte enhancer factor 2A (MEF2A) in a PPARß/δ ligand-independent manner. MCK-PPARß/δ muscle was shown to have high glycogen stores, increased levels of GLUT4, and augmented capacity for mitochondrial pyruvate oxidation, suggesting a broad reprogramming of glucose utilization pathways. Lastly, exercise studies demonstrated that MCK-PPARß/δ mice persistently oxidized glucose compared with nontransgenic controls, while exhibiting supranormal performance. These results identify a transcriptional regulatory mechanism that increases capacity for muscle glucose utilization in a pattern that resembles the effects of exercise training.


Subject(s)
Glucose/metabolism , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , PPAR delta/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cells, Cultured , Female , Lactate Dehydrogenases/genetics , Lactate Dehydrogenases/metabolism , Male , Mice , Muscle, Skeletal/enzymology , Oxidation-Reduction , PPAR alpha/metabolism , Physical Conditioning, Animal , Transcriptional Activation
20.
Circ Heart Fail ; 4(4): 474-82, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558447

ABSTRACT

BACKGROUND: Currently, there are no specific therapies available to treat cardiac dysfunction caused by sepsis and other chronic inflammatory conditions. Activation of toll-like receptor 4 (TLR4) by lipopolysaccharide (LPS) is an early event in Gram-negative bacterial sepsis, triggering a robust inflammatory response and changes in metabolism. Peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) α and ß serve as critical physiological regulators of energy metabolic gene expression in heart. METHODS AND RESULTS: Injection of mice with LPS triggered a myocardial fuel switch similar to that of the failing heart: reduced mitochondrial substrate flux and myocyte lipid accumulation. The LPS-induced metabolic changes were associated with diminished ventricular function and suppression of the genes encoding PGC-1α and ß, known transcriptional regulators of mitochondrial function. This cascade of events required TLR4 and nuclear factor-κB activation. Restoration of PGC-1ß expression in cardiac myocytes in culture and in vivo in mice reversed the gene regulatory, metabolic, and functional derangements triggered by LPS. Interestingly, the effects of PGC-1ß overexpression were independent of the upstream inflammatory response, highlighting the potential utility of modulating downstream metabolic derangements in cardiac myocytes as a novel strategy to prevent or treat sepsis-induced heart failure. CONCLUSIONS: LPS triggers cardiac energy metabolic reprogramming through suppression of PGC-1 coactivators in the cardiac myocyte. Reactivation of PGC-1ß expression can reverse the metabolic and functional derangements caused by LPS-TLR4 activation, identifying the PGC-1 axis as a candidate therapeutic target for sepsis-induced heart failure.


Subject(s)
Energy Metabolism/physiology , Heart Failure/metabolism , Myocardium/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Transcription Factors/antagonists & inhibitors , Animals , Cells, Cultured , Disease Models, Animal , Fatty Acids/metabolism , Heart Failure/chemically induced , Heart Failure/physiopathology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...