Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Hepatol ; 72(4): 627-635, 2020 04.
Article in English | MEDLINE | ID: mdl-31760070

ABSTRACT

BACKGROUND & AIMS: In non-alcoholic fatty liver disease (NAFLD), hepatocytes can undergo necroptosis: a regulated form of necrotic cell death mediated by the receptor-interacting protein kinase (RIPK) 1. Herein, we assessed the potential for RIPK1 and its downstream effector mixed lineage kinase domain-like protein (MLKL) to act as therapeutic targets and markers of activity in NAFLD. METHODS: C57/BL6J-mice were fed a normal chow diet or a high-fat diet (HFD). The effect of RIPA-56, a highly specific inhibitor of RIPK1, was evaluated in HFD-fed mice and in primary human steatotic hepatocytes. RIPK1 and MLKL concentrations were measured in the serum of patients with NAFLD. RESULTS: When used as either a prophylactic or curative treatment for HFD-fed mice, RIPA-56 caused a downregulation of MLKL and a reduction of liver injury, inflammation and fibrosis, characteristic of non-alcoholic steatohepatitis (NASH), as well as of steatosis. This latter effect was reproduced by treating primary human steatotic hepatocytes with RIPA-56 or necrosulfonamide, a specific inhibitor of human MLKL, and by knockout (KO) of Mlkl in fat-loaded AML-12 mouse hepatocytes. Mlkl-KO led to activation of mitochondrial respiration and an increase in ß-oxidation in steatotic hepatocytes. Along with decreased MLKL activation, Ripk3-KO mice exhibited increased activities of the liver mitochondrial respiratory chain complexes in experimental NASH. In patients with NAFLD, serum concentrations of RIPK1 and MLKL increased in correlation with activity. CONCLUSION: The inhibition of RIPK1 improves NASH features in HFD-fed mice and reverses steatosis via an MLKL-dependent mechanism that, at least partly, involves an increase in mitochondrial respiration. RIPK1 and MLKL are potential serum markers of activity and promising therapeutic targets in NAFLD. LAY SUMMARY: There are currently no pharmacological treatment options for non-alcoholic fatty liver disease (NAFLD), which is now the most frequent liver disease. Necroptosis is a regulated process of cell death that can occur in hepatocytes during NAFLD. Herein, we show that RIPK1, a gatekeeper of the necroptosis pathway that is activated in NAFLD, can be inhibited by RIPA-56 to reduce not only liver injury, inflammation and fibrosis, but also steatosis in experimental models. These results highlight the potential of RIPK1 as a therapeutic target in NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Protein Kinase Inhibitors/administration & dosage , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/blood , Acrylamides/pharmacology , Aged , Animals , Diet, High-Fat , Disease Models, Animal , Female , Gene Knockout Techniques , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Necroptosis/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Protein Kinases/blood , Protein Kinases/deficiency , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , Treatment Outcome
2.
Nature ; 501(7467): 412-5, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23965628

ABSTRACT

Ageing is due to an accumulation of various types of damage, and mitochondrial dysfunction has long been considered to be important in this process. There is substantial sequence variation in mammalian mitochondrial DNA (mtDNA), and the high mutation rate is counteracted by different mechanisms that decrease maternal transmission of mutated mtDNA. Despite these protective mechanisms, it is becoming increasingly clear that low-level mtDNA heteroplasmy is quite common and often inherited in humans. We designed a series of mouse mutants to investigate the extent to which inherited mtDNA mutations can contribute to ageing. Here we report that maternally transmitted mtDNA mutations can induce mild ageing phenotypes in mice with a wild-type nuclear genome. Furthermore, maternally transmitted mtDNA mutations lead to anticipation of reduced fertility in mice that are heterozygous for the mtDNA mutator allele (PolgA(wt/mut)) and aggravate premature ageing phenotypes in mtDNA mutator mice (PolgA(mut/mut)). Unexpectedly, a combination of maternally transmitted and somatic mtDNA mutations also leads to stochastic brain malformations. Our findings show that a pre-existing mutation load will not only allow somatic mutagenesis to create a critically high total mtDNA mutation load sooner but will also increase clonal expansion of mtDNA mutations to enhance the normally occurring mosaic respiratory chain deficiency in ageing tissues. Our findings suggest that maternally transmitted mtDNA mutations may have a similar role in aggravating aspects of normal human ageing.


Subject(s)
Aging/genetics , Brain/abnormalities , Brain/metabolism , DNA, Mitochondrial/genetics , Extrachromosomal Inheritance/genetics , Mitochondria/genetics , Mutation/genetics , Aging/pathology , Alleles , Animals , Brain/growth & development , Cell Nucleus/genetics , Female , Genome/genetics , Heterozygote , Litter Size , Male , Mice , Mice, Inbred C57BL , Mutagenesis/genetics , Phenotype , Reproduction/genetics , Reproduction/physiology , Stochastic Processes
3.
PLoS Genet ; 11(8): e1005423, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26247782

ABSTRACT

We have studied the in vivo role of SLIRP in regulation of mitochondrial DNA (mtDNA) gene expression and show here that it stabilizes its interacting partner protein LRPPRC by protecting it from degradation. Although SLIRP is completely dependent on LRPPRC for its stability, reduced levels of LRPPRC persist in the absence of SLIRP in vivo. Surprisingly, Slirp knockout mice are apparently healthy and only display a minor weight loss, despite a 50-70% reduction in the steady-state levels of mtDNA-encoded mRNAs. In contrast to LRPPRC, SLIRP is dispensable for polyadenylation of mtDNA-encoded mRNAs. Instead, deep RNA sequencing (RNAseq) of mitochondrial ribosomal fractions and additional molecular analyses show that SLIRP is required for proper association of mRNAs to the mitochondrial ribosome and efficient translation. Our findings thus establish distinct functions for SLIRP and LRPPRC within the LRPPRC-SLIRP complex, with a novel role for SLIRP in mitochondrial translation. Very surprisingly, our results also demonstrate that mammalian mitochondria have a great excess of transcripts under basal physiological conditions in vivo.


Subject(s)
Mitochondrial Proteins/biosynthesis , Neoplasm Proteins/metabolism , RNA-Binding Proteins/physiology , Animals , Cells, Cultured , Female , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Polyadenylation , Protein Biosynthesis , Proteolysis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
4.
PLoS Genet ; 9(1): e1003178, 2013.
Article in English | MEDLINE | ID: mdl-23300484

ABSTRACT

Regulation of mitochondrial DNA (mtDNA) expression is critical for the control of oxidative phosphorylation in response to physiological demand, and this regulation is often impaired in disease and aging. We have previously shown that mitochondrial transcription termination factor 3 (MTERF3) is a key regulator that represses mtDNA transcription in the mouse, but its molecular mode of action has remained elusive. Based on the hypothesis that key regulatory mechanisms for mtDNA expression are conserved in metazoans, we analyzed Mterf3 knockout and knockdown flies. We demonstrate here that decreased expression of MTERF3 not only leads to activation of mtDNA transcription, but also impairs assembly of the large mitochondrial ribosomal subunit. This novel function of MTERF3 in mitochondrial ribosomal biogenesis is conserved in the mouse, thus we identify a novel and unexpected role for MTERF3 in coordinating the crosstalk between transcription and translation for the regulation of mammalian mtDNA gene expression.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/genetics , Mitochondria , Mitochondrial Proteins , Ribosomes , Animals , DNA, Mitochondrial/genetics , Drosophila Proteins/genetics , Gene Expression Regulation , Invertebrates/genetics , Invertebrates/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidative Phosphorylation , Ribosomes/genetics , Ribosomes/metabolism , Transcription, Genetic
5.
Nature ; 458(7241): 1056-60, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19262508

ABSTRACT

AMP-activated protein kinase (AMPK) is a metabolic fuel gauge conserved along the evolutionary scale in eukaryotes that senses changes in the intracellular AMP/ATP ratio. Recent evidence indicated an important role for AMPK in the therapeutic benefits of metformin, thiazolidinediones and exercise, which form the cornerstones of the clinical management of type 2 diabetes and associated metabolic disorders. In general, activation of AMPK acts to maintain cellular energy stores, switching on catabolic pathways that produce ATP, mostly by enhancing oxidative metabolism and mitochondrial biogenesis, while switching off anabolic pathways that consume ATP. This regulation can take place acutely, through the regulation of fast post-translational events, but also by transcriptionally reprogramming the cell to meet energetic needs. Here we demonstrate that AMPK controls the expression of genes involved in energy metabolism in mouse skeletal muscle by acting in coordination with another metabolic sensor, the NAD+-dependent type III deacetylase SIRT1. AMPK enhances SIRT1 activity by increasing cellular NAD+ levels, resulting in the deacetylation and modulation of the activity of downstream SIRT1 targets that include the peroxisome proliferator-activated receptor-gamma coactivator 1alpha and the forkhead box O1 (FOXO1) and O3 (FOXO3a) transcription factors. The AMPK-induced SIRT1-mediated deacetylation of these targets explains many of the convergent biological effects of AMPK and SIRT1 on energy metabolism.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , NAD/metabolism , Sirtuins/metabolism , Acetylation , Aminoimidazole Carboxamide/analogs & derivatives , Animals , Cell Line , Energy Metabolism/genetics , Enzyme Activation , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genes, Mitochondrial/genetics , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Mutation , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Ribonucleotides , Sirtuin 1 , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic
6.
Proc Natl Acad Sci U S A ; 105(44): 17187-92, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18957541

ABSTRACT

Transcriptional control of metabolic circuits requires coordination between specific transcription factors and coregulators and is often deregulated in metabolic diseases. We characterized here the mechanisms through which the coactivator SRC-3 controls energy homeostasis. SRC-3 knock-out mice present a more favorable metabolic profile relative to their wild-type littermates. This metabolic improvement in SRC-3(-/-) mice is caused by an increase in mitochondrial function and in energy expenditure as a consequence of activation of PGC-1alpha. By controlling the expression of the only characterized PGC-1alpha acetyltransferase GCN5, SRC-3 induces PGC-1alpha acetylation and consequently inhibits its activity. Interestingly, SRC-3 expression is induced by caloric excess, resulting in the inhibition of PGC-1alpha activity and energy expenditure, whereas caloric restriction reduces SRC-3 levels leading to enhanced PGC-1alpha activity and energy expenditure. Collectively, these data suggest that SRC-3 is a critical link in a cofactor network that uses PGC-1alpha as an effector to control mitochondrial function and energy homeostasis.


Subject(s)
Histone Acetyltransferases/genetics , Obesity/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Acetylation , Animals , Caloric Restriction , Energy Metabolism , Histone Acetyltransferases/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nuclear Receptor Coactivator 3 , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors , p300-CBP Transcription Factors/metabolism
7.
Med Sci (Paris) ; 23(10): 840-4, 2007 Oct.
Article in French | MEDLINE | ID: mdl-17937892

ABSTRACT

Neurodegenerative diseases are more and more prevalent in our aging societies. A rapid overview of the etiology of many neurodegenerative diseases like Alzheimer, Parkinson, Huntington disease and amyotrophic lateral sclerosis suggests a tight link with mitochondrial dysfunction. Since it has been recently demonstrated that activation of the SIRT1/PGC-1 pathway, in a metabolic context promotes mitochondrial function, we performed a detailed literature review on the implication of this pathway in neurodegeneration. Interestingly, transgenic mice with impaired PGC-1 expression have neurodegenerative lesions and show behavioural abnormalities. As evidenced from independent investigations, enhanced SIRT1 activity has been demonstrated to protect against axonal degeneration and to decrease the accumulation of amyloid beta peptides, the hallmark of Alzheimer disease, in cultured murine embryonic neurons. In addition, several studies suggest that resveratrol, a specific activator of SIRT1, could have protective effects in animal models of neurodegenerative diseases. Taken together, these results strongly suggest that the modulation of the SIRT1/PGC-1 pathway, which has not been well documented in the central nervous system, could become the cornerstone for new therapeutical approaches to combat neurodegeneration.


Subject(s)
Mitochondria/physiology , Neurodegenerative Diseases/physiopathology , Sirtuins/physiology , Transcription Factors/physiology , Acetylation , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured/drug effects , Humans , Mice , Mice, Knockout , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein Processing, Post-Translational/drug effects , Reactive Oxygen Species , Resveratrol , Sirtuin 1 , Sirtuins/genetics , Stilbenes/pharmacology , Stilbenes/therapeutic use , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation
8.
Nat Rev Endocrinol ; 12(12): 710-722, 2016 12.
Article in English | MEDLINE | ID: mdl-27516341

ABSTRACT

Hepatic steatosis, the first step in the progression of nonalcoholic fatty liver disease, is characterized by triglyceride accumulation in hepatocytes and is highly prevalent in people with obesity. Although initially asymptomatic, hepatic steatosis is an important risk factor for the development of hepatic insulin resistance and type 2 diabetes mellitus and can also progress to more severe pathologies such as nonalcoholic steatohepatitis, liver fibrosis and cirrhosis; hepatic steatosis has, therefore, received considerable research interest in the past 20 years. The lipid accumulation that defines hepatic steatosis disturbs the function of the endoplasmic reticulum (ER) in hepatocytes, thereby generating chronic ER stress that interferes with normal cellular function. Although ubiquitous stress response mechanisms (namely, ER-associated degradation, unfolded protein response and autophagy) are the main processes for restoring cellular proteostasis, these mechanisms are unable to alleviate ER stress in the context of the fatty liver. Furthermore, ER stress and ER stress responses can promote lipid accumulation in hepatocytes in a counter-productive manner and could, therefore, be the origin of a vicious pathological cycle.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Endoplasmic Reticulum-Associated Degradation , Fatty Liver/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Triglycerides/metabolism , Unfolded Protein Response , Diabetes Mellitus, Type 2 , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Insulin Resistance , Liver/metabolism , Obesity , Organelles/metabolism , Proteins/metabolism
9.
J Cell Biol ; 208(4): 429-42, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25688136

ABSTRACT

Mitochondria form a dynamic network within the cell as a result of balanced fusion and fission. Despite the established role of mitofusins (MFN1 and MFN2) in mitochondrial fusion, only MFN2 has been associated with metabolic and neurodegenerative diseases, which suggests that MFN2 is needed to maintain mitochondrial energy metabolism. The molecular basis for the mitochondrial dysfunction encountered in the absence of MFN2 is not understood. Here we show that loss of MFN2 leads to impaired mitochondrial respiration and reduced ATP production, and that this defective oxidative phosphorylation process unexpectedly originates from a depletion of the mitochondrial coenzyme Q pool. Our study unravels an unexpected and novel role for MFN2 in maintenance of the terpenoid biosynthesis pathway, which is necessary for mitochondrial coenzyme Q biosynthesis. The reduced respiratory chain function in cells lacking MFN2 can be partially rescued by coenzyme Q10 supplementation, which suggests a possible therapeutic strategy for patients with diseases caused by mutations in the Mfn2 gene.


Subject(s)
Electron Transport/genetics , GTP Phosphohydrolases/physiology , Mitochondria/enzymology , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/biosynthesis , Animals , Cells, Cultured , Dynamins/genetics , Energy Metabolism/genetics , Energy Metabolism/physiology , GTP Phosphohydrolases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Dynamics/physiology , Oxidative Phosphorylation , RNA Interference , RNA, Small Interfering , Terpenes/metabolism , Ubiquinone/biosynthesis
10.
Biomaterials ; 44: 143-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25617134

ABSTRACT

Human neural stem cells (hNSCs) hold great promise for the treatment of neurological diseases. Considerable progress has been made to induce neural differentiation in the cell culture in vitro and upon transplantation in vivo [2] in order to explore restoration of damaged neuronal circuits. However, in vivo conventional strategies are limited to post mortem analysis. Here, we apply our developed first fate mapping platform to monitor neuronal differentiation in vivo by magnetic resonance imaging, bioluminescence imaging, and fluorescence imaging. Ferritin, Luciferase and GFP under neuronal-specific promoters for immature and mature neurons, respectively, were used to generate transgenic hNSCs. Differentiation-linked imaging reporter expression was validated in vitro. The time profile of spontaneous neuronal maturation after transplantation into mouse brain cortex demonstrated early neuronal differentiation within 6 weeks. Fully mature neurons expressing synaptogenesis were observed only after three months or longer. Our trimodal fate mapping strategy represents a unique non-invasive tool to monitor the time course of neuronal differentiation of transplanted stem cells in vivo.


Subject(s)
Brain/cytology , Cell Differentiation , Neural Stem Cells/transplantation , Neurons/cytology , Animals , Cell Lineage , Computer Systems , Electrophysiological Phenomena , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Imaging, Three-Dimensional , Male , Mice , Neural Stem Cells/cytology , Promoter Regions, Genetic/genetics , Stem Cell Transplantation , Time Factors
11.
Cell Metab ; 11(3): 213-9, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20197054

ABSTRACT

During fasting and after exercise, skeletal muscle efficiently switches from carbohydrate to lipid as the main energy source to preserve glycogen stores and blood glucose levels for glucose-dependent tissues. Skeletal muscle cells sense this limitation in glucose availability and transform this information into transcriptional and metabolic adaptations. Here we demonstrate that AMPK acts as the prime initial sensor that translates this information into SIRT1-dependent deacetylation of the transcriptional regulators PGC-1alpha and FOXO1, culminating in the transcriptional modulation of mitochondrial and lipid utilization genes. Deficient AMPK activity compromises SIRT1-dependent responses to exercise and fasting, resulting in impaired PGC-1alpha deacetylation and blunted induction of mitochondrial gene expression. Thus, we conclude that AMPK acts as the primordial trigger for fasting- and exercise-induced adaptations in skeletal muscle and that activation of SIRT1 and its downstream signaling pathways are improperly triggered in AMPK-deficient states.


Subject(s)
AMP-Activated Protein Kinases/genetics , Fasting/metabolism , Forkhead Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Sirtuin 1/metabolism , Trans-Activators/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Energy Metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Genes, Mitochondrial/genetics , Glucose/metabolism , Lipids/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Sirtuin 1/genetics , Trans-Activators/genetics , Transcription Factors , Up-Regulation/genetics
12.
Diabetes ; 59(4): 829-35, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20107110

ABSTRACT

OBJECTIVE: Sirtuin 1 (SIRT1) is implicated in the regulation of mitochondrial function, energy metabolism, and insulin sensitivity in rodents. No studies are available in humans to demonstrate that SIRT1 expression in insulin-sensitive tissues is associated with energy expenditure and insulin sensitivity. RESEARCH DESIGN AND METHODS: Energy expenditure (EE), insulin sensitivity, and SIRT1 mRNA adipose tissue expression (n = 81) were measured by indirect calorimetry, hyperinsulinemic-euglycemic clamp, and quantitative RT-PCR in 247 nondiabetic offspring of type 2 diabetic patients. RESULTS: High EE during the clamp (r = 0.375, P = 2.8 x 10(-9)) and high DeltaEE (EE during the clamp - EE in the fasting state) (r = 0.602, P = 2.5 x 10(-24)) were associated with high insulin sensitivity. Adipose tissue SIRT1 mRNA expression was significantly associated with EE (r = 0.289, P = 0.010) and with insulin sensitivity (r = 0.334, P = 0.002) during hyperinsulinemic-euglycemic clamp. Furthermore, SIRT1 mRNA expression correlated significantly with the expression of several genes regulating mitochondrial function and energy metabolism (e.g., peroxisome proliferator-activated receptor gamma coactivator-1beta, estrogen-related receptor alpha, nuclear respiratory factor-1, and mitochondrial transcription factor A), and with several genes of the respiratory chain (e.g., including NADH dehydrogenase [ubiquinone] 1alpha subcomplex 2, cytochrome c, cytochrome c oxidase subunit IV, and ATP synthase). CONCLUSIONS: Impaired stimulation of EE by insulin and low SIRT1 expression in insulin-sensitive tissues is likely to reflect impaired regulation of mitochondrial function associated with insulin resistance in humans.


Subject(s)
Diabetes Mellitus, Type 2/genetics , RNA, Messenger/genetics , Sirtuin 1/genetics , Adult , Animals , Body Mass Index , Crosses, Genetic , Diabetes Mellitus, Type 2/physiopathology , Energy Metabolism/genetics , Female , Gene Expression Regulation , Glucose Intolerance/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Reference Values
13.
Curr Protoc Mol Biol ; Chapter 29: Unit 29B.5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18972384

ABSTRACT

The maintenance of metabolic homeostasis relies on the balanced intake of nutrients from food. Consequently, diet composition strongly impacts whole-body physiology. Dietary formulations with strong nutrient imbalances can lead to metabolic disorders, with lipids and simple sugars playing a prominent role. This unit describes how diet formulation can be modified to generate mouse models of human metabolic pathologies, and it details methodological procedures linked to dietary manipulations, including caloric restriction and introduction of a test compound.


Subject(s)
Diet , Animal Feed/analysis , Animals , Caloric Restriction , Diet/adverse effects , Disease Models, Animal , Humans , Metabolic Diseases/etiology , Mice , Molecular Biology/methods
14.
Cell Metab ; 8(5): 347-58, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19046567

ABSTRACT

The NAD(+)-dependent deacetylase SIRT1 controls metabolic processes in response to low nutrient availability. We report the metabolic phenotype of mice treated with SRT1720, a specific and potent synthetic activator of SIRT1 that is devoid of direct action on AMPK. SRT1720 administration robustly enhances endurance running performance and strongly protects from diet-induced obesity and insulin resistance by enhancing oxidative metabolism in skeletal muscle, liver, and brown adipose tissue. These metabolic effects of SRT1720 are mediated by the induction of a genetic network controlling fatty acid oxidation through a multifaceted mechanism that involves the direct deacetylation of PGC-1alpha, FOXO1, and p53 and the indirect stimulation of AMPK signaling through a global metabolic adaptation mimicking low energy levels. Combined with our previous work on resveratrol, the current study further validates SIRT1 as a target for the treatment of metabolic disorders and characterizes the mechanisms underlying the therapeutic potential of SIRT1 activation.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Lipid Metabolism/drug effects , Metabolic Diseases/metabolism , Obesity/metabolism , Sirtuins/metabolism , Acetylation , Animals , Diet , Dietary Fats/administration & dosage , Energy Metabolism , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Insulin Resistance/physiology , Male , Metabolic Diseases/drug therapy , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Obesity/drug therapy , Oxidation-Reduction , Physical Conditioning, Animal , Sirtuin 1 , Sirtuins/agonists , Sirtuins/genetics
15.
Ann Med ; 39(5): 335-45, 2007.
Article in English | MEDLINE | ID: mdl-17701476

ABSTRACT

The sirtuin family of histone deacetylases (HDACs) was named after their homology to the Saccharomyces cerevisiae gene silent information regulator 2 (Sir2). In the yeast, Sir2 has been shown to mediate the effects of calorie restriction on the extension of life span and high levels of Sir2 activity promote longevity. Like their yeast homologs, the mammalian sirtuins (SIRT1-7) are class III HDACs and require NAD(+) as a cofactor to deacetylate substrates ranging from histones to transcriptional regulators. Through this activity, sirtuins are shown to regulate important biological processes ranging from apoptosis, adipocyte and muscle differentiation, and energy expenditure to gluconeogenesis. We review here the current knowledge regarding the role of sirtuins in metabolism, longevity, and discuss the possible therapeutic applications that could result from the understanding of their function in different organs and pathologies.


Subject(s)
Histone Deacetylases , Longevity , NAD/metabolism , Sirtuins/physiology , Animals , Caenorhabditis elegans , Caloric Restriction , Drosophila melanogaster , Energy Metabolism , Histone Deacetylases/genetics , Histone Deacetylases/physiology , Humans , Mammals , Neurodegenerative Diseases/genetics , Saccharomyces cerevisiae , Sirtuins/classification , Sirtuins/genetics
16.
Cell ; 127(6): 1109-22, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17112576

ABSTRACT

Diminished mitochondrial oxidative phosphorylation and aerobic capacity are associated with reduced longevity. We tested whether resveratrol (RSV), which is known to extend lifespan, impacts mitochondrial function and metabolic homeostasis. Treatment of mice with RSV significantly increased their aerobic capacity, as evidenced by their increased running time and consumption of oxygen in muscle fibers. RSV's effects were associated with an induction of genes for oxidative phosphorylation and mitochondrial biogenesis and were largely explained by an RSV-mediated decrease in PGC-1alpha acetylation and an increase in PGC-1alpha activity. This mechanism is consistent with RSV being a known activator of the protein deacetylase, SIRT1, and by the lack of effect of RSV in SIRT1(-/-) MEFs. Importantly, RSV treatment protected mice against diet-induced-obesity and insulin resistance. These pharmacological effects of RSV combined with the association of three Sirt1 SNPs and energy homeostasis in Finnish subjects implicates SIRT1 as a key regulator of energy and metabolic homeostasis.


Subject(s)
Metabolic Diseases/prevention & control , Mitochondria, Muscle/physiology , Sirtuins/genetics , Sirtuins/metabolism , Stilbenes/pharmacology , Trans-Activators/metabolism , Acetylation , Adult , Animals , Dietary Fats/administration & dosage , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Motor Activity/drug effects , Muscle Fibers, Skeletal/physiology , Obesity/prevention & control , Oxidative Phosphorylation , Oxygen Consumption/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polymorphism, Single Nucleotide , Resveratrol , Sirtuin 1 , Specific Pathogen-Free Organisms , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL