Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37502859

ABSTRACT

Obesity-related type II diabetes (diabesity) has increased global morbidity and mortality dramatically. Previously, the ancient drug salicylate demonstrated promise for the treatment of type II diabetes, but its clinical use was precluded due to high dose requirements. In this study, we present a nitroalkene derivative of salicylate, 5-(2-nitroethenyl)salicylic acid (SANA), a molecule with unprecedented beneficial effects in diet-induced obesity (DIO). SANA reduces DIO, liver steatosis and insulin resistance at doses up to 40 times lower than salicylate. Mechanistically, SANA stimulated mitochondrial respiration and increased creatine-dependent energy expenditure in adipose tissue. Indeed, depletion of creatine resulted in the loss of SANA action. Moreover, we found that SANA binds to creatine kinases CKMT1/2, and downregulation CKMT1 interferes with the effect of SANA in vivo. Together, these data demonstrate that SANA is a first-in-class activator of creatine-dependent energy expenditure and thermogenesis in adipose tissue and emerges as a candidate for the treatment of diabesity.

2.
Mol Med ; 26(1): 115, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33238880

ABSTRACT

BACKGROUND: Circulating high-mobility group box 1 (HMGB1) plays important roles in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Intracellular HMGB1 is critical for the biology of hepatocytes. However, the intracellular role of HMGB1 in hepatocellular steatosis is unknown. Therefore, we aimed to investigate the role of hepatocyte-specific HMGB1 (HC-HMGB1) in development of hepatic steatosis. METHODS: Wild type (WT) C57BL/6 and HC-HMGB1-/- mice were fed high-fat diet (HFD) or low-fat diet (LFD) for up to 16 weeks. RESULTS: As expected, HMGB1 translocated from nuclear into cytoplasm and released into circulation after HFD treatment. HC-HMGB1 deficiency significantly reduced circulating HMGB1, suggesting that hepatocyte is a major source of circulating HMGB1 during NAFLD. Unexpectedly, HC-HMGB1 deficiency promoted rapid weight gain with enhanced hepatic fat deposition compared with WT at as early as 4 weeks after HFD treatment. Furthermore, there was no difference between WT and HC-HMGB1-/- mice in glucose tolerance, energy expenditure, liver damage or systemic inflammation. Interestingly, hepatic gene expression related to free fatty acid (FFA) ß-oxidation was significantly down-regulated in HC-HMGB1-/- mice compared with WT, and endoplasmic reticulum (ER) stress markers were significantly higher in livers of HC-HMGB1-/- mice. In vitro experiments using primary mouse hepatocytes showed absence of HMGB1 increased FFA-induced intracellular lipid accumulation, accompanied by increased ER-stress, significant downregulation of FFA ß-oxidation, and reduced oxidative phosphorylation. CONCLUSIONS: Our findings suggest that hepatocyte HMGB1 protects against dysregulated lipid metabolism via maintenance of ß-oxidation and prevention of ER stress. This represents a novel mechanism for HMGB1-regulation of hepatocellular steatosis, and suggests that stabilizing HMGB1 in hepatocytes may be effective strategies for prevention and treatment of NAFLD.


Subject(s)
Diet, High-Fat , Fatty Liver/etiology , Fatty Liver/metabolism , HMGB1 Protein/genetics , Hepatocytes/metabolism , Stress, Physiological , Animals , Biopsy , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/pathology , HMGB1 Protein/blood , HMGB1 Protein/metabolism , Lipid Metabolism , Male , Mice , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction
3.
PLoS One ; 14(5): e0203101, 2019.
Article in English | MEDLINE | ID: mdl-31136580

ABSTRACT

Petite Integration Factor 1 (PIF1) is a multifunctional helicase present in nuclei and mitochondria. PIF1 knock out (KO) mice exhibit accelerated weight gain and decreased wheel running on a normal chow diet. In the current study, we investigated whether Pif1 ablation alters whole body metabolism in response to weight gain. PIF1 KO and wild type (WT) C57BL/6J mice were fed a Western diet (WD) rich in fat and carbohydrates before evaluation of their metabolic phenotype. Compared with weight gain-resistant WT female mice, WD-fed PIF1 KO females, but not males, showed accelerated adipose deposition, decreased locomotor activity, and reduced whole-body energy expenditure without increased dietary intake. Surprisingly, PIF1 KO females did not show obesity-induced alterations in fasting blood glucose and glucose clearance. WD-fed PIF1 KO females developed mild hepatic steatosis and associated changes in liver gene expression that were absent in weight-matched, WD-fed female controls, linking hepatic steatosis to Pif1 ablation rather than increased body weight. WD-fed PIF1 KO females also showed decreased expression of inflammation-associated genes in adipose tissue. Collectively, these data separated weight gain from inflammation and impaired glucose homeostasis. They also support a role for Pif1 in weight gain resistance and liver metabolic dysregulation during nutrient stress.


Subject(s)
DNA Helicases/deficiency , Diet, Western , Glucose/metabolism , Inflammation Mediators/metabolism , Weight Gain/genetics , Adipose Tissue/metabolism , Animals , Body Composition , Cholesterol/metabolism , Cytokines/metabolism , Energy Metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Glucose Tolerance Test , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Motor Activity
4.
Physiol Rep ; 7(8): e14043, 2019 04.
Article in English | MEDLINE | ID: mdl-31004398

ABSTRACT

Adropin is a liver- and brain-secreted peptide hormone with striking effects on fuel metabolism regulation in a number of tissues. Previous studies demonstrated that adropin secretion is decreased in obese mice subjected to a long-term high-fat diet (HFD), and that whole-body loss of adropin expression resulted in systemic insulin resistance. Treatment of obese mice with adropin improves glucose tolerance, which has been linked to increased glucose oxidation and inhibition of fatty acid utilization in isolated skeletal muscle homogenates. In this study, we used in vivo physiological measurements to determine how treatment of obese mice with adropin affects whole-body glucose metabolism. Treatment with adropin reduced fasting blood glucose and, as shown previously, increased glucose tolerance in HFD mice during standard glucose tolerance tests. Under hyperinsulinemic-euglycemic clamp conditions, adropin treatment led to a nonsignificant increase in whole-body insulin sensitivity, and a significant reduction in whole-body glucose uptake. Finally, we show that adropin treatment suppressed hepatic glucose production and improved hepatic insulin sensitivity. This correlated with reduced expression of fatty acid import proteins and gluconeogenic regulatory enzymes in the liver, suggesting that adropin treatment may impact the pathways that drive vital aspects of hepatic glucose metabolism.


Subject(s)
Anti-Obesity Agents/pharmacology , Blood Glucose/metabolism , Gluconeogenesis , Intercellular Signaling Peptides and Proteins/pharmacology , Liver/metabolism , Animals , Anti-Obesity Agents/therapeutic use , Diet, High-Fat/adverse effects , Insulin Resistance , Intercellular Signaling Peptides and Proteins/therapeutic use , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology
5.
Diabetes ; 68(6): 1221-1229, 2019 06.
Article in English | MEDLINE | ID: mdl-30936145

ABSTRACT

Systemic hyperuricemia (HyUA) in obesity/type 2 diabetes facilitated by elevated activity of xanthine oxidoreductase (XOR), which is the sole source of uric acid (UA) in mammals, has been proposed to contribute to the pathogenesis of insulin resistance/dyslipidemia in obesity. Here, the effects of hepatocyte-specific ablation of Xdh, the gene encoding XOR (HXO), and whole-body pharmacologic inhibition of XOR (febuxostat) on obesity-induced insulin resistance/dyslipidemia were assessed. Deletion of hepatocyte Xdh substantially lowered liver and plasma UA concentration. When exposed to an obesogenic diet, HXO and control floxed (FLX) mice became equally obese, but systemic HyUA was absent in HXO mice. Despite this, obese HXO mice became as insulin resistant and dyslipidemic as obese FLX mice. Similarly, febuxostat dramatically lowered plasma and tissue UA and XOR activity in obese wild-type mice without altering obesity-associated insulin resistance/dyslipidemia. These data demonstrate that hepatocyte XOR activity is a critical determinant of systemic UA homeostasis, that deletion of hepatocyte Xdh is sufficient to prevent systemic HyUA of obesity, and that neither prevention nor correction of HyUA improves insulin resistance/dyslipidemia in obesity. Thus, systemic HyUA, although clearly a biomarker of the metabolic abnormalities of obesity, does not appear to be causative.


Subject(s)
Glucose/metabolism , Hepatocytes/metabolism , Hyperuricemia/genetics , Lipid Metabolism , Obesity/metabolism , Uric Acid/metabolism , Xanthine Dehydrogenase/genetics , Animals , Diet, High-Fat , Fatty Acids, Nonesterified/metabolism , Febuxostat/pharmacology , Glucose Tolerance Test , Hepatocytes/drug effects , Hyperuricemia/metabolism , Mice , Triglycerides/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors
6.
J Biol Chem ; 293(46): 17676-17684, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30323061

ABSTRACT

Sirtuin 3 (SIRT3) deacetylates and activates several mitochondrial fatty acid oxidation enzymes in the liver. Here, we investigated whether the protein acetylase GCN5 general control of amino acid synthesis 5-like 1 (GCN5L1), previously shown to oppose SIRT3 activity, is involved in the regulation of hepatic fatty acid oxidation. We show that GCN5L1 abundance is significantly up-regulated in response to an acute high-fat diet (HFD). Transgenic GCN5L1 overexpression in the mouse liver increased protein acetylation levels, and proteomic detection of specific lysine residues identified numerous sites that are co-regulated by GCN5L1 and SIRT3. We analyzed several fatty acid oxidation proteins identified by the proteomic screen and found that hyperacetylation of hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit α (HADHA) correlates with increased GCN5L1 levels. Stable GCN5L1 knockdown in HepG2 cells reduced HADHA acetylation and increased activities of fatty acid oxidation enzymes. Mice with a liver-specific deletion of GCN5L1 were protected from hepatic lipid accumulation following a chronic HFD and did not exhibit hyperacetylation of HADHA compared with WT controls. Finally, we found that GCN5L1-knockout mice lack HADHA that is hyperacetylated at three specific lysine residues (Lys-350, Lys-383, and Lys-406) and that acetylation at these sites is significantly associated with increased HADHA activity. We conclude that GCN5L1-mediated regulation of mitochondrial protein acetylation plays a role in hepatic metabolic homeostasis.


Subject(s)
Fatty Acids/metabolism , Nerve Tissue Proteins/metabolism , Acetylation , Animals , Diet, High-Fat/adverse effects , Fatty Liver/prevention & control , Hep G2 Cells , Humans , Lysine/chemistry , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Nerve Tissue Proteins/genetics , Oxidation-Reduction , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Proteomics , Sirtuin 3/genetics
7.
Physiol Rep ; 6(18): e13836, 2018 09.
Article in English | MEDLINE | ID: mdl-30251338

ABSTRACT

Mice with a deletion of the p50 subunit of the proinflammatory nuclear factor kappa B pathway (NF-κB p50) have reduced weight compared to wild-type control mice. However, the physiological underpinning of this phenotype remains unknown. This study addressed this issue. Compared to littermate controls, lean male p50 null mice (p50-/- ) had an increased metabolic rate (~20%) that was associated with increased skeletal muscle (SkM, ~35%), but not liver, oxidative metabolism. These metabolic alterations were accompanied by decreases in adiposity, and tissue and plasma triglyceride levels (all ~30%). Notably, there was a marked decrease in skeletal muscle, but not liver, DGAT2 gene expression (~70%), but a surprising reduction in muscle PPARα and CPT1 (both ~20%) gene expression. Exposure to a high-fat diet accentuated the diminished adiposity of p50-/- mice despite elevated caloric intake, whereas plasma triglycerides and free fatty acids (both ~30%), and liver (~40%) and SkM (~50%) triglyceride accumulation were again reduced compared to WT. Although SkM cytokine expression (IL-6 and TNFα, each ~100%) were increased in p50-/- mice, neither cytokine acutely increased SkM oxidative metabolism. We conclude that the reduced susceptibility to diet-induced obesity and dyslipidemia in p50-/- mice results from an increase in metabolic rate, which is associated with elevated skeletal muscle oxidative metabolism and decreased DGAT2 expression.


Subject(s)
Basal Metabolism/physiology , Inflammation Mediators/metabolism , Muscle, Skeletal/metabolism , NF-kappa B p50 Subunit/deficiency , Obesity/metabolism , Oxidative Stress/physiology , Animals , Diet, High-Fat/adverse effects , Diet, High-Fat/trends , Energy Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/prevention & control
8.
Am J Physiol Endocrinol Metab ; 315(5): E758-E770, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30086648

ABSTRACT

Accumulation of myeloid cells in the liver, notably dendritic cells (DCs) and monocytes/macrophages (MCs), is a major component of the metainflammation of obesity. However, the mechanism(s) stimulating hepatic DC/MC infiltration remain ill defined. Herein, we addressed the hypothesis that adipose tissue (AT) free fatty acids (FFAs) play a central role in the initiation of hepatic DC/MC accumulation, using a number of mouse models of altered FFA supply to the liver. In two models of acute FFA elevation (lipid infusion and fasting) hepatic DC/MC and triglycerides (TGs) but not AT DC/MC were increased without altering plasma cytokines (PCs; TNFα and monocyte chemoattractant protein 1) and with variable effects on oxidative stress (OxS) markers. However, fasting in mice with profoundly reduced AT lipolysis (AT-specific deletion of adipose TG lipase; AAKO) failed to elevate liver DC/MC, TG, or PC, but liver OxS increased. Livers of obese AAKO mice that are known to be resistant to steatosis were similarly protected from inflammation. In high-fat feeding studies of 1, 3, 6, or 20-wk duration, liver DC/MC accumulation dissociated from PC and OxS but tracked with liver TGs. Furthermore, decreasing OxS by ~80% in obese mice failed to decrease liver DC/MC. Therefore, FFA and more specifically AT-derived FFA stimulate hepatic DC/MC accumulation, thus recapitulating the pathology of the obese liver. In a number of cases the effects of FFA can be dissociated from OxS and PC but match well with liver TG, a marker of FFA oversupply.


Subject(s)
Adipose Tissue/metabolism , Fasting/metabolism , Fatty Acids, Nonesterified/metabolism , Liver/metabolism , Myeloid Cells/metabolism , Animals , Cytokines/blood , Diet, High-Fat , Fatty Acids, Nonesterified/pharmacology , Lipase/genetics , Lipase/metabolism , Lipolysis/physiology , Liver/drug effects , Mice , Mice, Knockout , Obesity/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Triglycerides/metabolism
9.
Hepatology ; 68(4): 1347-1360, 2018 10.
Article in English | MEDLINE | ID: mdl-29631332

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a progressive, inflammatory form of fatty liver disease. It is the most rapidly rising risk factor for the development of hepatocellular carcinoma (HCC), which can arise in NASH with or without cirrhosis. The inflammatory signals promoting the progression of NASH to HCC remain largely unknown. The propensity of neutrophils to expel decondensed chromatin embedded with inflammatory proteins, known as neutrophil extracellular traps (NETs), has been shown to be important in chronic inflammatory conditions and in cancer progression. In this study, we asked whether NET formation occurs in NASH and contributes to the progression of HCC. We found elevated levels of a NET marker in serum of patients with NASH. In livers from STAM mice (NASH induced by neonatal streptozotocin and high-fat diet), early neutrophil infiltration and NET formation were seen, followed by an influx of monocyte-derived macrophages, production of inflammatory cytokines, and progression of HCC. Inhibiting NET formation, through treatment with deoxyribonuclease (DNase) or using mice knocked out for peptidyl arginine deaminase type IV (PAD4-/- ), did not affect the development of a fatty liver but altered the consequent pattern of liver inflammation, which ultimately resulted in decreased tumor growth. Mechanistically, we found that commonly elevated free fatty acids stimulate NET formation in vitro. CONCLUSION: Our findings implicate NETs in the protumorigenic inflammatory environment in NASH, suggesting that their elimination may reduce the progression of liver cancer in NASH. (Hepatology 2018).


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Disease Progression , Extracellular Traps/metabolism , Neutrophils/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biomarkers/metabolism , Biopsy, Needle , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Prognosis , Random Allocation , Risk Assessment
10.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515034

ABSTRACT

Insulin resistance is associated with increased incidence and enhanced progression of cancers. However, little is known about strategies that can effectively ameliorate insulin resistance and consequently halt cancer progression. Herein, we propose that the transcription factor Nrf2 (also known as Nfe2l2) may be such a target, given its central role in disease prevention. To this end, we developed a mouse that overexpresses the Notch intracellular domain in adipocytes (AdNICD), leading to lipodystrophy-induced severe insulin resistance and subsequent development of sarcomas, as a model reflecting that Notch signaling is deregulated in cancers and shows positive associations with insulin resistance and fatty liver disease in humans. Nrf2 pathway activation was achieved by knocking down Keap1, a repressor of Nrf2, in the AdNICD background. Constitutively enhanced Nrf2 signaling in this setting led to prevention of hepatic steatosis, dyslipidemia, and insulin resistance by repressing hepatic lipogenic pathways and restoration of the hepatic fatty acid profile to control levels. This protective effect of Nrf2 against diabetes extended to significant reduction and delay in sarcoma incidence and latency. Our study highlights that the Nrf2 pathway, which has been induced by small molecules in clinical trials, is a potential therapeutic target against insulin resistance and subsequent risk of cancer.


Subject(s)
Carcinogenesis/genetics , Insulin Resistance/genetics , NF-E2-Related Factor 2/metabolism , Receptors, Notch/metabolism , Sarcoma/genetics , Animals , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lipodystrophy/complications , Lipodystrophy/genetics , Lipodystrophy/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Protein Domains/genetics , Receptors, Notch/genetics , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction/genetics
11.
Am J Physiol Endocrinol Metab ; 315(2): E180-E195, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29486138

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a canonical regulator of cytoprotective gene expression, but evidence of its cross talk with other pathways, including metabolic ones, is ever increasing. Pharmacologic or systemic genetic activation of the Nrf2 pathway partially protects from obesity in mice and ameliorates fasting hyperglycemia in mice and humans. However, systemic Nrf2 deletion also protects from diet-induced obesity and insulin resistance in mice. To further investigate the effect of the disruption of Nrf2 on obesity in a tissue-specific manner, we focused on adipocytes and hepatocytes with targeted deletion of Nrf2. To this end, mice with cell-specific deletion of Nrf2 in adipocytes (ANKO) or hepatocytes (HeNKO) were fed a high-fat diet (HFD) for 6 mo and showed similar increases in body weight and body fat content. ANKO mice showed a partially deteriorated glucose tolerance, higher fasting glucose levels, and higher levels of cholesterol and nonesterified fatty acids compared with their Control counterparts. The HeNKO mice, though, had lower insulin levels and trended toward improved insulin sensitivity without having any difference in liver triglyceride accumulation. This study compared for the first time two conditional Nrf2 knockout models in adipocytes and in hepatocytes during HFD-induced obesity. None of these models could completely recapitulate the unexpected protection against obesity observed in the whole body Nrf2 knockout mice, but this study points out the differential roles that Nrf2 may play, beyond cytoprotection, in different target tissues and rather suggests systemic activation of the Nrf2 pathway as an effective means of prevention and treatment of obesity and type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Diet, High-Fat/adverse effects , Hepatocytes/metabolism , NF-E2-Related Factor 2/metabolism , Obesity/genetics , Obesity/metabolism , Adiposity/genetics , Animals , Blood Glucose/metabolism , Body Composition/genetics , Body Weight/genetics , Glucose Intolerance/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Triglycerides/blood
12.
Nat Commun ; 9(1): 708, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29453396

ABSTRACT

Obesity, a prevalent condition in adults and children, impairs bone marrow (BM) function. However, the underlying mechanisms are unclear. Here, we show that obese mice exhibit poor emergency immune responses in a toll-like receptor 4 (TLR4)-dependent manner. Canonical myeloid genes (Csf1r, Spi1, Runx1) are enhanced, and lymphoid genes (Flt3, Tcf3, Ebf1) are reduced. Using adoptive transfer and mixed BM chimera approaches we demonstrate that myeloid>lymphoid bias arises after 6 weeks of high-fat diet and depends on precursor cell-autonomous TLR4. Further, lean mice exposed to the TLR4 ligand lipopolysaccharide (LPS) at doses similar to that detectable in obese serum recapitulates BM lympho-myeloid alterations. Together, these results establish a mechanistic contribution of BM cell-intrinsic TLR4 to obesity-driven BM malfunction and demonstrate the importance of LPS. Our findings raises important questions about the impact of maternal obesity and endotoxemia to fetal hematopoiesis, as fetal immune precursors are also sensitive to TLR4 signals.


Subject(s)
Bone Marrow/physiopathology , Obesity/immunology , Toll-Like Receptor 4/physiology , Adoptive Transfer , Animals , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/physiopathology
13.
Am J Physiol Heart Circ Physiol ; 313(2): H265-H274, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28526709

ABSTRACT

Lysine acetylation is a reversible posttranslational modification and is particularly important in the regulation of mitochondrial metabolic enzymes. Acetylation uses acetyl-CoA derived from fuel metabolism as a cofactor, thereby linking nutrition to metabolic activity. In the present study, we investigated how mitochondrial acetylation status in the heart is controlled by food intake and how these changes affect mitochondrial metabolism. We found that there was a significant increase in cardiac mitochondrial protein acetylation in mice fed a long-term high-fat diet and that this change correlated with an increase in the abundance of the mitochondrial acetyltransferase-related protein GCN5L1. We showed that the acetylation status of several mitochondrial fatty acid oxidation enzymes (long-chain acyl-CoA dehydrogenase, short-chain acyl-CoA dehydrogenase, and hydroxyacyl-CoA dehydrogenase) and a pyruvate oxidation enzyme (pyruvate dehydrogenase) was significantly upregulated in high-fat diet-fed mice and that the increase in long-chain and short-chain acyl-CoA dehydrogenase acetylation correlated with increased enzymatic activity. Finally, we demonstrated that the acetylation of mitochondrial fatty acid oxidation proteins was decreased after GCN5L1 knockdown and that the reduced acetylation led to diminished fatty acid oxidation in cultured H9C2 cells. These data indicate that lysine acetylation promotes fatty acid oxidation in the heart and that this modification is regulated in part by the activity of GCN5L1.NEW & NOTEWORTHY Recent research has shown that acetylation of mitochondrial fatty acid oxidation enzymes has greatly contrasting effects on their activity in different tissues. Here, we provide new evidence that acetylation of cardiac mitochondrial fatty acid oxidation enzymes by GCN5L1 significantly upregulates their activity in diet-induced obese mice.


Subject(s)
Acetyltransferases/metabolism , Energy Metabolism , Fatty Acids/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/enzymology , Nerve Tissue Proteins/metabolism , Obesity/enzymology , Protein Processing, Post-Translational , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetylation , Acetyltransferases/genetics , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Animals , Cell Line , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Lysine , Male , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Obesity/genetics , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , RNA Interference , Rats , Sirtuin 3/genetics , Sirtuin 3/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Time Factors , Transfection
14.
Mol Metab ; 6(4): 327-339, 2017 04.
Article in English | MEDLINE | ID: mdl-28377872

ABSTRACT

OBJECTIVE: Overnutrition can alter gene expression patterns through epigenetic mechanisms that may persist through generations. However, it is less clear if overnutrition, for example a high fat diet, modifies epigenetic control of gene expression in adults, or by what molecular mechanisms, or if such mechanisms contribute to the pathology of the metabolic syndrome. Here we test the hypothesis that a high fat diet alters hepatic DNA methylation, transcription and gene expression patterns, and explore the contribution of such changes to the pathophysiology of obesity. METHODS: RNA-seq and targeted high-throughput bisulfite DNA sequencing were used to undertake a systematic analysis of the hepatic response to a high fat diet. RT-PCR, chromatin immunoprecipitation and in vivo knockdown of an identified driver gene, Phlda1, were used to validate the results. RESULTS: A high fat diet resulted in the hypermethylation and decreased transcription and expression of Phlda1 and several other genes. A subnetwork of genes associated with Phlda1 was identified from an existing Bayesian gene network that contained numerous hepatic regulatory genes involved in lipid and body weight homeostasis. Hepatic-specific depletion of Phlda1 in mice decreased expression of the genes in the subnetwork, and led to increased oil droplet size in standard chow-fed mice, an early indicator of steatosis, validating the contribution of this gene to the phenotype. CONCLUSIONS: We conclude that a high fat diet alters the epigenetics and transcriptional activity of key hepatic genes controlling lipid homeostasis, contributing to the pathophysiology of obesity.


Subject(s)
DNA Methylation , Diet, High-Fat/adverse effects , Epigenesis, Genetic , Obesity/etiology , Animals , Cells, Cultured , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
15.
Exp Gerontol ; 90: 1-13, 2017 04.
Article in English | MEDLINE | ID: mdl-28108329

ABSTRACT

Exercise is known to have numerous beneficial effects. Recent studies indicate that exercise improves mitochondrial energetics not only in skeletal muscle but also in other tissues. While exercise elicits positive effects on memory, neurogenesis, and synaptic plasticity, the effects of exercise on brain mitochondrial energetics remain relatively unknown. Herein, we studied the effects of exercise training in old and young mice on brain mitochondrial energetics, in comparison to known effects on peripheral tissues that utilize fatty acid oxidation. Exercise improved the capacity for muscle and liver to oxidize palmitate in old mice, but not young mice. In the brain, exercise increased rates of respiration and reactive oxygen species (ROS) production in the old group only while utilizing complex I substrates, effects that were not seen in the young group. Coupled complex I to III enzymatic activity was significantly increased in old trained versus untrained mice with no effect on coupled II to III enzymatic activity. Mitochondrial protein content and markers of mitochondrial biogenesis (PGC-1α and TFAM) were not affected by exercise training in the brain, in contrast to the skeletal muscle of old mice. Brain levels of the autophagy marker LC3-II and protein levels of other signaling proteins that regulate metabolism or transport (BDNF, HSP60, phosphorylated mTOR, FNDC5, SIRT3) were not significantly altered. Old exercised mice showed a significant increase in DRP1 protein levels in the brain without changes in phosphorylation, while MFN2 and OPA1 protein levels were unchanged. Our results suggest that exercise training in old mice can improve brain mitochondrial function through effects on electron transport chain function and mitochondrial dynamics without increasing mitochondrial biogenesis.


Subject(s)
Aging/metabolism , Dynamins/metabolism , Electron Transport Complex I/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Physical Conditioning, Animal , Animals , Cerebellar Cortex/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Dynamics , Organelle Biogenesis , Reactive Oxygen Species/metabolism , Signal Transduction
16.
Am J Physiol Endocrinol Metab ; 312(1): E11-E18, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27827807

ABSTRACT

Leptin has potent effects on lipid metabolism in a number of peripheral tissues. In liver, an acute leptin infusion (~120 min) stimulates hepatic fatty acid oxidation (~30%) and reduces triglycerides (TG, ~40%), effects that are dependent on phosphoinositol-3-kinase (PI3K) activity. In the current study we addressed the hypothesis that leptin actions on liver-resident immune cells are required for these metabolic effects. Myeloid cell-specific deletion of the leptin receptor (ObR) in mice or depletion of liver Kupffer cells (KC) in rats in vivo prevented the acute effects of leptin on liver lipid metabolism, while the metabolic effects of leptin were maintained in mice lacking ObR in hepatocytes. Notably, liver TG were elevated in both lean and obese myeloid cell ObR, but the degree of obesity and insulin resistance induced by a high-fat diet was similar to control mice. In isolated primary hepatocytes (HEP), leptin had no effects on HEP lipid metabolism and only weakly stimulated PI3K. However, the coculture of KC with HEP restored leptin action on HEP fatty acid metabolism and stimulation of HEP PI3K. Notably, leptin stimulated the release from KC of a number of cytokines. However, the exposure of HEP to these cytokines individually [granulocyte macrophage colony-stimulating factor, IL-1α, IL-1ß, IL-6, IL-10, and IL-18] or in combination had no effects on HEP lipid metabolism. Together, these data demonstrate a role for liver mononuclear cells in the regulation of liver lipid metabolism by leptin.


Subject(s)
Hepatocytes/metabolism , Kupffer Cells/physiology , Leptin/metabolism , Lipid Metabolism , Liver/metabolism , Triglycerides/metabolism , Animals , Cytokines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-10/immunology , Interleukin-18/immunology , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Myeloid Cells/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Rats , Rats, Wistar , Receptors, Leptin/genetics
17.
Am J Physiol Endocrinol Metab ; 311(1): E105-16, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27166280

ABSTRACT

Mitochondrial dysfunction is associated with many human diseases and results from mismatch of damage and repair over the life of the organelle. PARK2 is a ubiquitin E3 ligase that regulates mitophagy, a repair mechanism that selectively degrades damaged mitochondria. Deletion of PARK2 in multiple in vivo models results in susceptibility to stress-induced mitochondrial and cellular dysfunction. Surprisingly, Park2 knockout (KO) mice are protected from nutritional stress and do not develop obesity, hepatic steatosis or insulin resistance when fed a high-fat diet (HFD). However, these phenomena are casually related and the physiological basis for this phenotype is unknown. We therefore undertook a series of acute HFD studies to more completely understand the physiology of Park2 KO during nutritional stress. We find that intestinal lipid absorption is impaired in Park2 KO mice as evidenced by increased fecal lipids and reduced plasma triglycerides after intragastric fat challenge. Park2 KO mice developed hepatic steatosis in response to intravenous lipid infusion as well as during incubation of primary hepatocytes with fatty acids, suggesting that hepatic protection from nutritional stress was secondary to changes in energy balance due to altered intestinal triglyceride absorption. Park2 KO mice showed reduced adiposity after 1-wk HFD, as well as improved hepatic and peripheral insulin sensitivity. These studies suggest that changes in intestinal lipid absorption may play a primary role in protection from nutritional stress in Park2 KO mice by preventing HFD-induced weight gain and highlight the need for tissue-specific models to address the role of PARK2 during metabolic stress.


Subject(s)
Body Weight/genetics , Diet, High-Fat , Insulin Resistance/genetics , Intestinal Absorption/genetics , Lipid Metabolism/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Energy Metabolism , Fatty Acids/pharmacology , Fatty Liver/genetics , Feces/chemistry , Infusions, Intravenous , Intestinal Mucosa/metabolism , Lipids/analysis , Lipids/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , Triglycerides/blood , Weight Gain/genetics
18.
Physiol Rep ; 4(6)2016 Mar.
Article in English | MEDLINE | ID: mdl-27033445

ABSTRACT

In obesity, adipose tissue (AT) and liver are infiltrated with Th-1 polarized immune cells, which are proposed to play an important role in the pathogenesis of the metabolic abnormalities of obesity. Aging is also associated with increased adiposity, but the effects of this increase on inflammation and associated metabolic dysfunction are poorly understood. To address this issue, we assessed insulin resistance (IR) andATand liver immunophenotype in aged, lean (AL) and aged, obese (AO) mice, all of whom were maintained on a standard chow diet (11% fat diet) throughout their lives. For comparison, these variables were also assessed in young, lean (YL) and young diet-induced obese mice (41% fat diet,YO). Despite similar body weight and fat accumulation,YOmice were substantially moreIRand had greater liver steatosis compared toAOmice.YOalso had elevated infiltration of macrophages/dendritic cells inATand liver, but these increases were absent inAO Furthermore, liver immune cells ofYOwere more Th-1 polarized thenAO Notably, aging was associated with accumulation of T cells, but this occurred independent of obesity. Together, the data suggest that reduced inflammation inAOunderlies the improved insulin sensitivity and lowered steatosis compared toYO.


Subject(s)
Adipose Tissue , Aging , Diet, High-Fat , Fatty Liver/etiology , Insulin Resistance , Liver , Obesity/etiology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adiposity , Age Factors , Aging/blood , Aging/immunology , Animals , Dendritic Cells/immunology , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/immunology , Immunophenotyping , Liver/immunology , Liver/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , Obesity/blood , Obesity/immunology , Th1 Cells/immunology , Time Factors
19.
Oncotarget ; 7(21): 30379-95, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27105497

ABSTRACT

Establishing c-Myc's (Myc) role in liver regeneration has proven difficult particularly since the traditional model of partial hepatectomy may provoke an insufficiently demanding proliferative stress. We used a model of hereditary tyrosinemia whereby the affected parenchyma can be gradually replaced by transplanted hepatocytes, which replicate 50-100-fold, over several months. Prior to transplantation, livers from myc-/- (KO) mice were smaller in young animals and larger in older animals relative to myc+/+ (WT) counterparts. KO mice also consumed more oxygen, produced more CO2 and generated more heat. Although WT and KO hepatocytes showed few mitochondrial structural differences, the latter demonstrated defective electron transport chain function. RNAseq revealed differences in transcripts encoding ribosomal subunits, cytochrome p450 members and enzymes for triglyceride and sterol biosynthesis. KO hepatocytes also accumulated neutral lipids. WT and KO hepatocytes repopulated recipient tyrosinemic livers equally well although the latter were associated with a pro-inflammatory hepatic environment that correlated with worsening lipid accumulation, its extracellular deposition and parenchymal oxidative damage. Our results show Myc to be dispensable for sustained in vivo hepatocyte proliferation but necessary for maintaining normal lipid homeostasis. myc-/- livers resemble those encountered in non-alcoholic fatty liver disease and, under sustained proliferative stress, gradually acquire the features of non-alcoholic steatohepatitis.


Subject(s)
Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver Regeneration , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Proliferation , Cell Size , Cells, Cultured , Gene Expression Profiling/methods , Hepatocytes/cytology , Hepatocytes/transplantation , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Triglycerides/metabolism
20.
Br J Pharmacol ; 173(2): 267-78, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26447327

ABSTRACT

BACKGROUND AND PURPOSE: Thiazolidinediones (TZD) are known to ameliorate fatty liver in type 2 diabetes. To date, the underlying mechanisms of their hepatic actions remain unclear. EXPERIMENTAL APPROACH: Hepatic triglyceride content and export rates were assessed in 2 week high-sucrose-fed Wistar rats treated with troglitazone and compared with untreated high-sucrose rodent controls. Fractional de novo lipogenesis (DNL) contributions to hepatic triglyceride were quantified by analysis of triglyceride enrichment from deuterated water. Hepatic insulin clearance and NO status during a meal tolerance test were also evaluated. KEY RESULTS: TZD significantly reduced hepatic triglyceride (P < 0.01) by 48%, decreased DNL contribution to hepatic triglyceride (P < 0.01) and increased postprandial non-esterified fatty acids clearance rates (P < 0.01) in comparison with the high-sucrose rodent control group. During a meal tolerance test, plasma insulin AUC was significantly lower (P < 0.01), while blood glucose and plasma C-peptide levels were not different. Insulin clearance was increased (P < 0.001) by 24% and was associated with a 22% augmentation of hepatic insulin-degrading enzyme activity (P < 0.05). Finally, hepatic NO was decreased by 24% (P < 0.05). CONCLUSIONS: Overall, TZD show direct actions on liver by reducing hepatic DNL and increasing hepatic insulin clearance. The alterations in hepatic insulin clearance were associated with changes in insulin-degrading enzyme activity, with possible modulation of NO levels.


Subject(s)
Chromans/therapeutic use , Fatty Liver/chemically induced , Fatty Liver/prevention & control , Hyperinsulinism/chemically induced , Hyperinsulinism/prevention & control , Sucrose/toxicity , Thiazolidinediones/therapeutic use , Animals , Fatty Liver/blood , Hyperinsulinism/blood , Hypoglycemic Agents/therapeutic use , Male , Protective Agents/therapeutic use , Rats , Rats, Wistar , Sucrose/administration & dosage , Troglitazone
SELECTION OF CITATIONS
SEARCH DETAIL
...