Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 576(7785): 138-142, 2019 12.
Article in English | MEDLINE | ID: mdl-31748741

ABSTRACT

Haem is an essential prosthetic group of numerous proteins and a central signalling molecule in many physiologic processes1,2. The chemical reactivity of haem means that a network of intracellular chaperone proteins is required to avert the cytotoxic effects of free haem, but the constituents of such trafficking pathways are unknown3,4. Haem synthesis is completed in mitochondria, with ferrochelatase adding iron to protoporphyrin IX. How this vital but highly reactive metabolite is delivered from mitochondria to haemoproteins throughout the cell remains poorly defined3,4. Here we show that progesterone receptor membrane component 2 (PGRMC2) is required for delivery of labile, or signalling haem, to the nucleus. Deletion of PGMRC2 in brown fat, which has a high demand for haem, reduced labile haem in the nucleus and increased stability of the haem-responsive transcriptional repressors Rev-Erbα and BACH1. Ensuing alterations in gene expression caused severe mitochondrial defects that rendered adipose-specific PGRMC2-null mice unable to activate adaptive thermogenesis and prone to greater metabolic deterioration when fed a high-fat diet. By contrast, obese-diabetic mice treated with a small-molecule PGRMC2 activator showed substantial improvement of diabetic features. These studies uncover a role for PGRMC2 in intracellular haem transport, reveal the influence of adipose tissue haem dynamics on physiology and suggest that modulation of PGRMC2 may revert obesity-linked defects in adipocytes.


Subject(s)
Adipocytes/metabolism , Heme/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Homeostasis , Humans , Intracellular Space/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Molecular Chaperones/metabolism , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Transcription, Genetic
2.
FASEB J ; 30(5): 1976-86, 2016 05.
Article in English | MEDLINE | ID: mdl-26849960

ABSTRACT

Ketone bodies (KBs) are crucial energy substrates during states of low carbohydrate availability. However, an aberrant regulation of KB homeostasis can lead to complications such as diabetic ketoacidosis. Exercise and diabetes affect systemic KB homeostasis, but the regulation of KB metabolism is still enigmatic. In our study in mice with either knockout or overexpression of the peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α in skeletal muscle, PGC-1α regulated ketolytic gene transcription in muscle. Furthermore, KB homeostasis of these mice was investigated during withholding of food, exercise, and ketogenic diet feeding, and after streptozotocin injection. In response to these ketogenic stimuli, modulation of PGC-1α levels in muscle affected systemic KB homeostasis. Moreover, the data demonstrate that skeletal muscle PGC-1α is necessary for the enhanced ketolytic capacity in response to exercise training and overexpression of PGC-1α in muscle enhances systemic ketolytic capacity and is sufficient to ameliorate diabetic hyperketonemia in mice. In cultured myotubes, the transcription factor estrogen-related receptor-α was a partner of PGC-1α in the regulation of ketolytic gene transcription. These results demonstrate a central role of skeletal muscle PGC-1α in the transcriptional regulation of systemic ketolytic capacity.-Svensson, K., Albert, V., Cardel, B., Salatino, S., Handschin, C. Skeletal muscle PGC-1α modulates systemic ketone body homeostasis and ameliorates diabetic hyperketonemia in mice.


Subject(s)
Diet, Ketogenic , Food Deprivation , Homeostasis/physiology , Ketone Bodies/blood , Motor Activity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Animals, Genetically Modified , Cell Line , Ketone Bodies/metabolism , Male , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
3.
Proc Natl Acad Sci U S A ; 111(32): 11592-9, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25082895

ABSTRACT

The liver is a key metabolic organ that controls whole-body physiology in response to nutrient availability. Mammalian target of rapamycin (mTOR) is a nutrient-activated kinase and central controller of growth and metabolism that is negatively regulated by the tumor suppressor tuberous sclerosis complex 1 (TSC1). To investigate the role of hepatic mTOR complex 1 (mTORC1) in whole-body physiology, we generated liver-specific Tsc1 (L-Tsc1 KO) knockout mice. L-Tsc1 KO mice displayed reduced locomotor activity, body temperature, and hepatic triglyceride content in a rapamycin-sensitive manner. Ectopic activation of mTORC1 also caused depletion of hepatic and plasma glutamine, leading to peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α)-dependent fibroblast growth factor 21 (FGF21) expression in the liver. Injection of glutamine or knockdown of PGC-1α or FGF21 in the liver suppressed the behavioral and metabolic defects due to mTORC1 activation. Thus, mTORC1 in the liver controls whole-body physiology through PGC-1α and FGF21. Finally, mTORC1 signaling correlated with FGF21 expression in human liver tumors, suggesting that treatment of glutamine-addicted cancers with mTOR inhibitors might have beneficial effects at both the tumor and whole-body level.


Subject(s)
Body Temperature/physiology , Fibroblast Growth Factors/metabolism , Lipid Metabolism , Liver/metabolism , Motor Activity/physiology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Gene Knockdown Techniques , Glutamine/metabolism , Humans , Liver Neoplasms/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
J Biol Chem ; 290(26): 16059-76, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25987562

ABSTRACT

Resveratrol (RSV) and SRT1720 (SRT) elicit beneficial metabolic effects and are postulated to ameliorate obesity and related metabolic complications. The co-activator, peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), has emerged as a major downstream effector responsible for metabolic remodeling of muscle and other metabolic tissues in response to RSV or SRT treatment. However, the requirement of PGC-1α in skeletal muscle for the systemic metabolic effects of these compounds has so far not been demonstrated. Using muscle-specific PGC-1α knock-out mice, we show that PGC-1α is necessary for transcriptional induction of mitochondrial genes in muscle with both RSV and SRT treatment. Surprisingly, the beneficial effects of SRT on glucose homeostasis and of both compounds on energy expenditure occur even in the absence of muscle PGC-1α. Moreover, RSV and SRT treatment elicit differential transcriptional effects on genes involved in lipid metabolism and mitochondrial biogenesis in liver and adipose tissue. These findings indicate that RSV and SRT do not induce analogous metabolic effects in vivo. Our results provide important insights into the mechanism, effects, and organ specificity of the caloric restriction mimetics RSV and SRT. These findings are important for the design of future therapeutic interventions aimed at ameliorating obesity and obesity-related metabolic dysfunction.


Subject(s)
Adipose Tissue/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Liver/metabolism , Muscle, Skeletal/metabolism , Stilbenes/pharmacology , Transcription Factors/metabolism , Adipose Tissue/drug effects , Animals , Energy Metabolism/drug effects , Female , Glucose/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Male , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Resveratrol , Transcription Factors/genetics
5.
Biochem Biophys Res Commun ; 464(2): 480-6, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26159924

ABSTRACT

Orexigenic agouti-related protein/neuropeptide Y (Agrp/NPY) neurons and an orexigenic pro-opiomelanocortin (POMC) neurons of the hypothalamus regulate feeding behavior and energy homeostasis. An understanding of the molecular signaling pathways that regulate Agrp/NPY and POMC function could lead to novel treatments for metabolic disorders. Target of Rapamycin Complex 1 (TORC1) is a nutrient-activated protein kinase and central controller of growth and metabolism. We therefore investigated the role of mammalian TORC1 (mTORC1) in Agrp neurons. We generated and characterized Agrp neuron-specific raptor knockout (Agrp-raptor KO) mice. Agrp-raptor KO mice displayed reduced, non-circadian expression of Agrp and NPY but normal feeding behavior and energy homeostasis on both normal and high fat diet. Thus, mTORC1 in Agrp neurons controls circadian expression of orexigenic neuropeptides but is dispensable for the regulation of feeding behavior and energy metabolism.


Subject(s)
Agouti-Related Protein/metabolism , Circadian Rhythm , Feeding Behavior , Multiprotein Complexes/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Agouti-Related Protein/genetics , Animals , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Oxidative Stress , Regulatory-Associated Protein of mTOR , Signal Transduction
6.
Arterioscler Thromb Vasc Biol ; 33(9): 2105-11, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868942

ABSTRACT

OBJECTIVE: Perivascular adipose tissue (PVAT) wraps blood vessels and modulates vasoreactivity by secretion of vasoactive molecules. Mammalian target of rapamycin complex 2 (mTORC2) has been shown to control inflammation and is expressed in adipose tissue. In this study, we investigated whether adipose-specific deletion of rictor and thereby inactivation of mTORC2 in PVAT may modulate vascular function by increasing inflammation in PVAT. APPROACH AND RESULTS: Rictor, an essential mTORC2 component, was deleted specifically in mouse adipose tissue (rictor(ad-/-)). Phosphorylation of mTORC2 downstream target Akt at Serine 473 was reduced in PVAT from rictor(ad-/-) mice but unaffected in aortic tissue. Ex vivo functional analysis of thoracic aortae revealed increased contractions and impaired dilation in rings with PVAT from rictor(ad-/-) mice. Adipose rictor knockout increased gene expression and protein release of interleukin-6, macrophage inflammatory protein-1α, and tumor necrosis factor-α in PVAT as shown by quantitative real-time polymerase chain reaction and Bioplex analysis for the cytokines in the conditioned media, respectively. Moreover, gene and protein expression of inducible nitric oxide synthase was upregulated without affecting macrophage infiltration in PVAT from rictor(ad-/-) mice. Inhibition of inducible nitric oxide synthase normalized vascular reactivity in aortic rings from rictor(ad-/-) mice with no effect in rictor(fl/fl) mice. Interestingly, in perivascular and epididymal adipose depots, high-fat diet feeding induced downregulation of rictor gene expression. CONCLUSIONS: Here, we identify mTORC2 as a critical regulator of PVAT-directed protection of normal vascular tone. Modulation of mTORC2 activity in adipose tissue may be a potential therapeutic approach for inflammation-related vascular damage.


Subject(s)
Adipose Tissue/metabolism , Aorta, Thoracic/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Vasoconstriction , Vasodilation , 3T3-L1 Cells , Adipose Tissue/immunology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/immunology , Carrier Proteins/genetics , Chemokine CCL3/metabolism , Culture Media, Conditioned/metabolism , Cytokines/genetics , Diet, High-Fat , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inflammation/immunology , Inflammation/physiopathology , Interleukin-6/metabolism , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Real-Time Polymerase Chain Reaction , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
Proc Natl Acad Sci U S A ; 108(51): 20808-13, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143799

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is central to the control of cell, organ, and body size. Skeletal muscle-specific inactivation of mTORC1 in mice results in smaller muscle fibers, fewer mitochondria, increased glycogen stores, and a progressive myopathy that causes premature death. In mTORC1-deficient muscles, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), which regulates mitochondrial biogenesis and glucose homeostasis, is strongly down-regulated. Here we tested whether induction of mitochondrial biogenesis pharmacologically or by the overexpression of PGC-1α is sufficient to reverse the phenotype of mice deficient for mTORC1. We show that both approaches normalize mitochondrial function, such as oxidative capacity and expression of mitochondrial genes. However, they do not prevent or delay the progressive myopathy. In addition, we find that mTORC1 has a much stronger effect than PGC-1α on the glycogen content in muscle. This effect is based on the strong activation of PKB/Akt in mTORC1-deficient mice. We also show that activation of PKB/Akt not only affects glycogen synthesis but also diminishes glycogen degradation. Thus, our work provides strong functional evidence that mitochondrial dysfunction in mice with inactivated mTORC1 signaling is caused by the down-regulation of PGC-1α. However, our data also show that the impairment of mitochondria does not lead directly to the lethal myopathy.


Subject(s)
Bezafibrate/pharmacology , Gene Expression Regulation , Muscular Diseases/metabolism , Proteins/metabolism , Animals , Glycogen/chemistry , Glycogen/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Models, Genetic , Multiprotein Complexes , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , TOR Serine-Threonine Kinases , Trans-Activators/metabolism , Transcription Factors
9.
Nat Commun ; 13(1): 608, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105890

ABSTRACT

In obesity, signaling through the IRE1 arm of the unfolded protein response exerts both protective and harmful effects. Overexpression of the IRE1-regulated transcription factor XBP1s in liver or fat protects against obesity-linked metabolic deterioration. However, hyperactivation of IRE1 engages regulated IRE1-dependent decay (RIDD) and TRAF2/JNK pro-inflammatory signaling, which accelerate metabolic dysfunction. These pathologic IRE1-regulated processes have hindered efforts to pharmacologically harness the protective benefits of IRE1/XBP1s signaling in obesity-linked conditions. Here, we report the effects of a XBP1s-selective pharmacological IRE1 activator, IXA4, in diet-induced obese (DIO) mice. IXA4 transiently activates protective IRE1/XBP1s signaling in liver without inducing RIDD or TRAF2/JNK signaling. IXA4 treatment improves systemic glucose metabolism and liver insulin action through IRE1-dependent remodeling of the hepatic transcriptome that reduces glucose production and steatosis. IXA4-stimulated IRE1 activation also enhances pancreatic function. Our findings indicate that systemic, transient activation of IRE1/XBP1s signaling engenders multi-tissue benefits that integrate to mitigate obesity-driven metabolic dysfunction.


Subject(s)
Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , X-Box Binding Protein 1/metabolism , Animals , Fatty Liver/metabolism , Gene Expression Regulation , Glucose/metabolism , Homeostasis , Liver/metabolism , Membrane Proteins/genetics , Mice , Mice, Obese , Molecular Medicine , Obesity/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription Factors/metabolism , Unfolded Protein Response , X-Box Binding Protein 1/genetics
10.
iScience ; 2: 221-237, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29888756

ABSTRACT

Adrenergic stimulation of brown adipose tissue (BAT) induces acute and long-term responses. The acute adrenergic response activates thermogenesis by uncoupling oxidative phosphorylation and enabling increased substrate oxidation. Long-term, adrenergic signaling remodels BAT, inducing adaptive transcriptional changes that expand thermogenic capacity. Here, we show that the estrogen-related receptors alpha and gamma (ERRα, ERRγ) are collectively critical effectors of adrenergically stimulated transcriptional reprogramming of BAT. Mice lacking adipose ERRs (ERRαγAd-/-) have reduced oxidative and thermogenic capacity and rapidly become hypothermic when exposed to cold. ERRαγAd-/- mice treated long term with a ß3-adrenergic agonist fail to expand oxidative or thermogenic capacity and do not increase energy expenditure in response to norepinephrine (NE). Furthermore, ERRαγAd-/- mice fed a high-fat diet do not lose weight or show improved glucose tolerance when dosed with ß3-adrenergic agonists. The molecular basis of these defects is the finding that ERRs mediate the bulk of the transcriptional response to adrenergic stimulation.

11.
J Clin Invest ; 128(4): 1538-1550, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29528335

ABSTRACT

Obesity is a major risk factor for insulin resistance and type 2 diabetes. In adipose tissue, obesity-mediated insulin resistance correlates with the accumulation of proinflammatory macrophages and inflammation. However, the causal relationship of these events is unclear. Here, we report that obesity-induced insulin resistance in mice precedes macrophage accumulation and inflammation in adipose tissue. Using a mouse model that combines genetically induced, adipose-specific insulin resistance (mTORC2-knockout) and diet-induced obesity, we found that insulin resistance causes local accumulation of proinflammatory macrophages. Mechanistically, insulin resistance in adipocytes results in production of the chemokine monocyte chemoattractant protein 1 (MCP1), which recruits monocytes and activates proinflammatory macrophages. Finally, insulin resistance (high homeostatic model assessment of insulin resistance [HOMA-IR]) correlated with reduced insulin/mTORC2 signaling and elevated MCP1 production in visceral adipose tissue from obese human subjects. Our findings suggest that insulin resistance in adipose tissue leads to inflammation rather than vice versa.


Subject(s)
Insulin Resistance , Intra-Abdominal Fat/metabolism , Macrophages/metabolism , Obesity/metabolism , Panniculitis/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intra-Abdominal Fat/pathology , Macrophages/pathology , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Knockout , Obesity/genetics , Obesity/pathology , Panniculitis/genetics , Panniculitis/pathology
12.
Mol Metab ; 16: 76-87, 2018 10.
Article in English | MEDLINE | ID: mdl-30120064

ABSTRACT

OBJECTIVES: Extracts of the hops plant have been shown to reduce weight and insulin resistance in rodents and humans, but elucidation of the mechanisms responsible for these benefits has been hindered by the use of heterogeneous hops-derived mixtures. Because hop extracts are used as flavoring agents for their bitter properties, we hypothesized that bitter taste receptors (Tas2rs) could be mediating their beneficial effects in metabolic disease. Studies have shown that exposure of cultured enteroendocrine cells to bitter tastants can stimulate release of hormones, including glucagon-like peptide 1 (GLP-1). These findings have led to the suggestion that activation of Tas2rs may be of benefit in diabetes, but this tenet has not been tested. Here, we have assessed the ability of a pure derivative of a hops isohumulone with anti-diabetic properties, KDT501, to signal through Tas2rs. We have further used this compound as a tool to systematically assess the impact of bitter taste receptor activation in obesity-diabetes. METHODS: KDT501 was tested in a panel of bitter taste receptor signaling assays. Diet-induced obese mice (DIO) were dosed orally with KDT501 and acute effects on glucose homeostasis determined. A wide range of metabolic parameters were evaluated in DIO mice chronically treated with KDT501 to establish the full impact of activating gut bitter taste signaling. RESULTS: We show that KDT501 signals through Tas2r108, one of 35 mouse Tas2rs. In DIO mice, acute treatment stimulated GLP-1 secretion and enhanced glucose tolerance. Chronic treatment caused weight and fat mass loss, increased energy expenditure, enhanced glucose tolerance and insulin sensitivity, normalized plasma lipids, and induced broad suppression of inflammatory markers. Chronic KDT501 treatment altered enteroendocrine hormone levels and bile acid homeostasis and stimulated sustained GLP-1 release. Combined treatment with a dipeptidyl peptidase IV inhibitor amplified the incretin-based benefits of this pure isohumulone. CONCLUSIONS: Activation of Tas2r108 in the gut results in a remodeling of enteroendocrine hormone release and bile acid metabolism that ameliorates multiple features of metabolic syndrome. Targeting extraoral bitter taste receptors may be useful in metabolic disease.


Subject(s)
Cyclopentanes/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Body Weight/drug effects , Cyclopentanes/pharmacology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/drug effects , Glucagon-Like Peptide 1/metabolism , Humulus/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Intestinal Mucosa/metabolism , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/drug effects
13.
Cell Rep ; 25(10): 2904-2918.e8, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517875

ABSTRACT

Pancreatic ß cell physiology changes substantially throughout life, yet the mechanisms that drive these changes are poorly understood. Here, we performed comprehensive in vivo quantitative proteomic profiling of pancreatic islets from juvenile and 1-year-old mice. The analysis revealed striking differences in abundance of enzymes controlling glucose metabolism. We show that these changes in protein abundance are associated with higher activities of glucose metabolic enzymes involved in coupling factor generation as well as increased activity of the coupling factor-dependent amplifying pathway of insulin secretion. Nutrient tracing and targeted metabolomics demonstrated accelerated accumulation of glucose-derived metabolites and coupling factors in islets from 1-year-old mice, indicating that age-related changes in glucose metabolism contribute to improved glucose-stimulated insulin secretion with age. Together, our study provides an in-depth characterization of age-related changes in the islet proteome and establishes metabolic rewiring as an important mechanism for age-associated changes in ß cell function.


Subject(s)
Cellular Senescence , Insulin-Secreting Cells/metabolism , Metabolomics/methods , Proteomics/methods , Aging , Animals , Carbon/metabolism , Cell Respiration/drug effects , Citric Acid Cycle/drug effects , Female , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Insulin Secretion , Male , Mice, Inbred C57BL , Proteome/metabolism
14.
EMBO Mol Med ; 8(3): 232-46, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26772600

ABSTRACT

Activation of non-shivering thermogenesis (NST) in brown adipose tissue (BAT) has been proposed as an anti-obesity treatment. Moreover, cold-induced glucose uptake could normalize blood glucose levels in insulin-resistant patients. It is therefore important to identify novel regulators of NST and cold-induced glucose uptake. Mammalian target of rapamycin complex 2 (mTORC2) mediates insulin-stimulated glucose uptake in metabolic tissues, but its role in NST is unknown. We show that mTORC2 is activated in brown adipocytes upon ß-adrenergic stimulation. Furthermore, mice lacking mTORC2 specifically in adipose tissue (AdRiKO mice) are hypothermic, display increased sensitivity to cold, and show impaired cold-induced glucose uptake and glycolysis. Restoration of glucose uptake in BAT by overexpression of hexokinase II or activated Akt2 was sufficient to increase body temperature and improve cold tolerance in AdRiKO mice. Thus, mTORC2 in BAT mediates temperature homeostasis via regulation of cold-induced glucose uptake. Our findings demonstrate the importance of glucose metabolism in temperature regulation.


Subject(s)
Adipose Tissue, Brown/metabolism , Glucose/metabolism , Glycolysis , Multiprotein Complexes/metabolism , Oncogene Protein v-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Thermogenesis , Animals , Mechanistic Target of Rapamycin Complex 2 , Mice
15.
Curr Opin Cell Biol ; 33: 55-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25554914

ABSTRACT

Mammalian TOR (mTOR) signaling controls growth, metabolism and energy homeostasis in a cell autonomous manner. Recent findings indicate that mTOR signaling in one tissue can also affect other organs thereby affecting whole body metabolism and energy homeostasis in a non-cell autonomous manner. It is thus not surprising that mTOR signaling mediates aging and is often deregulated in metabolic disorders, such as obesity, diabetes and cancer. This review discusses the regulation of cellular and whole body energy metabolism by mTOR, with particular focus on the non-cell autonomous function of mTOR.


Subject(s)
Energy Metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Homeostasis , Humans , Hypothalamus/enzymology , Hypothalamus/metabolism , Liver/enzymology , Liver/metabolism , Metabolic Diseases/enzymology , Metabolic Diseases/metabolism , Mitochondria/enzymology , Mitochondria/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Nucleotides/biosynthesis , Protein Biosynthesis
16.
Curr Opin Genet Dev ; 23(1): 53-62, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23317514

ABSTRACT

The target of rapamycin (TOR) is a highly conserved serine/threonine kinase that is part of two structurally and functionally distinct complexes, TORC1 and TORC2. In multicellular organisms, TOR regulates cell growth and metabolism in response to nutrients, growth factors and cellular energy. Deregulation of TOR signaling alters whole body metabolism and causes age-related disease. This review describes the most recent advances in TOR signaling with a particular focus on mammalian TOR (mTOR) in metabolic tissues vis-a-vis aging, obesity, type 2 diabetes, and cancer.


Subject(s)
Aging/genetics , Neoplasms/genetics , TOR Serine-Threonine Kinases/genetics , Aging/metabolism , Animals , Cell Proliferation , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Deletion , Gene Expression Regulation , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Obesity/genetics , Obesity/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL