Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Metab ; 6(3): 567-577, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38378996

ABSTRACT

Uptake of circulating succinate by brown adipose tissue (BAT) and beige fat elevates whole-body energy expenditure, counteracts obesity and antagonizes systemic tissue inflammation in mice. The plasma membrane transporters that facilitate succinate uptake in these adipocytes remain undefined. Here we elucidate a mechanism underlying succinate import into BAT via monocarboxylate transporters (MCTs). We show that succinate transport is strongly dependent on the proportion that is present in the monocarboxylate form. MCTs facilitate monocarboxylate succinate uptake, which is promoted by alkalinization of the cytosol driven by adrenoreceptor stimulation. In brown adipocytes, we show that MCT1 primarily facilitates succinate import. In male mice, we show that both acute pharmacological inhibition of MCT1 and congenital depletion of MCT1 decrease succinate uptake into BAT and consequent catabolism. In sum, we define a mechanism of succinate uptake in BAT that underlies its protective activity in mouse models of metabolic disease.


Subject(s)
Adipocytes, Brown , Succinic Acid , Male , Mice , Animals , Adipocytes, Brown/metabolism , Succinic Acid/metabolism , Adipose Tissue, Brown/metabolism , Biological Transport , Membrane Transport Proteins/metabolism
2.
Mol Cell ; 83(11): 1903-1920.e12, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267907

ABSTRACT

Exercise benefits the human body in many ways. Irisin is secreted by muscle, increased with exercise, and conveys physiological benefits, including improved cognition and resistance to neurodegeneration. Irisin acts via αV integrins; however, a mechanistic understanding of how small polypeptides like irisin can signal through integrins is poorly understood. Using mass spectrometry and cryo-EM, we demonstrate that the extracellular heat shock protein 90α (eHsp90α) is secreted by muscle with exercise and activates integrin αVß5. This allows for high-affinity irisin binding and signaling through an Hsp90α/αV/ß5 complex. By including hydrogen/deuterium exchange data, we generate and experimentally validate a 2.98 Å RMSD irisin/αVß5 complex docking model. Irisin binds very tightly to an alternative interface on αVß5 distinct from that used by known ligands. These data elucidate a non-canonical mechanism by which a small polypeptide hormone like irisin can function through an integrin receptor.


Subject(s)
Cell Communication , Fibronectins , Humans , Fibronectins/metabolism , Signal Transduction
3.
Cell Metab ; 35(3): 535-549.e7, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36681077

ABSTRACT

Proteins are secreted from cells to send information to neighboring cells or distant tissues. Because of the highly integrated nature of energy balance systems, there has been particular interest in myokines and adipokines. These are challenging to study through proteomics because serum or plasma contains highly abundant proteins that limit the detection of proteins with lower abundance. We show here that extracellular fluid (EF) from muscle and fat tissues of mice shows a different protein composition than either serum or tissues. Mass spectrometry analyses of EFs from mice with physiological perturbations, like exercise or cold exposure, allowed the quantification of many potentially novel myokines and adipokines. Using this approach, we identify prosaposin as a secreted product of muscle and fat. Prosaposin expression stimulates thermogenic gene expression and induces mitochondrial respiration in primary fat cells. These studies together illustrate the utility of EF isolation as a discovery tool for adipokines and myokines.


Subject(s)
Extracellular Fluid , Saposins , Mice , Animals , Extracellular Fluid/metabolism , Saposins/metabolism , Muscles/metabolism , Adipose Tissue/metabolism , Adipokines
4.
Cell ; 185(24): 4654-4673.e28, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36334589

ABSTRACT

Brown adipose tissue (BAT) regulates metabolic physiology. However, nearly all mechanistic studies of BAT protein function occur in a single inbred mouse strain, which has limited the understanding of generalizable mechanisms of BAT regulation over physiology. Here, we perform deep quantitative proteomics of BAT across a cohort of 163 genetically defined diversity outbred mice, a model that parallels the genetic and phenotypic variation found in humans. We leverage this diversity to define the functional architecture of the outbred BAT proteome, comprising 10,479 proteins. We assign co-operative functions to 2,578 proteins, enabling systematic discovery of regulators of BAT. We also identify 638 proteins that correlate with protection from, or sensitivity to, at least one parameter of metabolic disease. We use these findings to uncover SFXN5, LETMD1, and ATP1A2 as modulators of BAT thermogenesis or adiposity, and provide OPABAT as a resource for understanding the conserved mechanisms of BAT regulation over metabolic physiology.


Subject(s)
Adipose Tissue, Brown , Proteome , Humans , Mice , Animals , Adipose Tissue, Brown/metabolism , Proteome/metabolism , Thermogenesis/physiology , Adiposity , Obesity/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism
5.
Cell Metab ; 34(1): 140-157.e8, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34861155

ABSTRACT

Uncoupling protein 1 (UCP1) is a major regulator of brown and beige adipocyte energy expenditure and metabolic homeostasis. However, the widely employed UCP1 loss-of-function model has recently been shown to have a severe deficiency in the entire electron transport chain of thermogenic fat. As such, the role of UCP1 in metabolic regulation in vivo remains unclear. We recently identified cysteine-253 as a regulatory site on UCP1 that elevates protein activity upon covalent modification. Here, we examine the physiological importance of this site through the generation of a UCP1 cysteine-253-null (UCP1 C253A) mouse, a precise genetic model for selective disruption of UCP1 in vivo. UCP1 C253A mice exhibit significantly compromised thermogenic responses in both males and females but display no measurable effect on fat accumulation in an obesogenic environment. Unexpectedly, we find that a lack of C253 results in adipose tissue redox stress, which drives substantial immune cell infiltration and systemic inflammatory pathology in adipose tissues and liver of male, but not female, mice. Elevation of systemic estrogen reverses this male-specific pathology, providing a basis for protection from inflammation due to loss of UCP1 C253 in females. Together, our results establish the UCP1 C253 activation site as a regulator of acute thermogenesis and sex-dependent tissue inflammation.


Subject(s)
Adipose Tissue, Brown , Cysteine , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Animals , Cysteine/metabolism , Energy Metabolism , Female , Inflammation/metabolism , Male , Mice , Thermogenesis/physiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
6.
Nature ; 593(7860): 580-585, 2021 05.
Article in English | MEDLINE | ID: mdl-33981039

ABSTRACT

Adaptive thermogenesis has attracted much attention because of its ability to increase systemic energy expenditure and to counter obesity and diabetes1-3. Recent data have indicated that thermogenic fat cells use creatine to stimulate futile substrate cycling, dissipating chemical energy as heat4,5. This model was based on the super-stoichiometric relationship between the amount of creatine added to mitochondria and the quantity of oxygen consumed. Here we provide direct evidence for the molecular basis of this futile creatine cycling activity in mice. Thermogenic fat cells have robust phosphocreatine phosphatase activity, which is attributed to tissue-nonspecific alkaline phosphatase (TNAP). TNAP hydrolyses phosphocreatine to initiate a futile cycle of creatine dephosphorylation and phosphorylation. Unlike in other cells, TNAP in thermogenic fat cells is localized to the mitochondria, where futile creatine cycling occurs. TNAP expression is powerfully induced when mice are exposed to cold conditions, and its inhibition in isolated mitochondria leads to a loss of futile creatine cycling. In addition, genetic ablation of TNAP in adipocytes reduces whole-body energy expenditure and leads to rapid-onset obesity in mice, with no change in movement or feeding behaviour. These data illustrate the critical role of TNAP as a phosphocreatine phosphatase in the futile creatine cycle.


Subject(s)
Alkaline Phosphatase/metabolism , Mitochondria/enzymology , Phosphocreatine/metabolism , Thermogenesis , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Cold Temperature , Energy Metabolism , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Obesity/metabolism
7.
Cell ; 183(1): 62-75.e17, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32946811

ABSTRACT

In response to skeletal muscle contraction during exercise, paracrine factors coordinate tissue remodeling, which underlies this healthy adaptation. Here we describe a pH-sensing metabolite signal that initiates muscle remodeling upon exercise. In mice and humans, exercising skeletal muscle releases the mitochondrial metabolite succinate into the local interstitium and circulation. Selective secretion of succinate is facilitated by its transient protonation, which occurs upon muscle cell acidification. In the protonated monocarboxylic form, succinate is rendered a transport substrate for monocarboxylate transporter 1, which facilitates pH-gated release. Upon secretion, succinate signals via its cognate receptor SUCNR1 in non-myofibrillar cells in muscle tissue to control muscle-remodeling transcriptional programs. This succinate-SUCNR1 signaling is required for paracrine regulation of muscle innervation, muscle matrix remodeling, and muscle strength in response to exercise training. In sum, we define a bioenergetic sensor in muscle that utilizes intracellular pH and succinate to coordinate tissue adaptation to exercise.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Succinic Acid/metabolism , Animals , Humans , Hydrogen-Ion Concentration , Inflammation/metabolism , Mice , Mitochondria/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Contraction , Receptors, G-Protein-Coupled/physiology , Signal Transduction , Succinates/metabolism , Symporters/metabolism
8.
Proc Natl Acad Sci U S A ; 117(20): 10789-10796, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32358195

ABSTRACT

Oxidation of cysteine thiols by physiological reactive oxygen species (ROS) initiates thermogenesis in brown and beige adipose tissues. Cellular selenocysteines, where sulfur is replaced with selenium, exhibit enhanced reactivity with ROS. Despite their critical roles in physiology, methods for broad and direct detection of proteogenic selenocysteines are limited. Here we developed a mass spectrometric method to interrogate incorporation of selenium into proteins. Unexpectedly, this approach revealed facultative incorporation of selenium as selenocysteine or selenomethionine into proteins that lack canonical encoding for selenocysteine. Selenium was selectively incorporated into regulatory sites on key metabolic proteins, including as selenocysteine-replacing cysteine at position 253 in uncoupling protein 1 (UCP1). This facultative utilization of selenium was initiated by increasing cellular levels of organic, but not inorganic, forms of selenium. Remarkably, dietary selenium supplementation elevated facultative incorporation into UCP1, elevated energy expenditure through thermogenic adipose tissue, and protected against obesity. Together, these findings reveal the existence of facultative protein selenation, which correlates with impacts on thermogenic adipocyte function and presumably other biological processes as well.


Subject(s)
Adipose Tissue/metabolism , Cysteine/metabolism , Obesity/metabolism , Selenium/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism , Adipose Tissue/physiology , Animals , Cells, Cultured , Male , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
9.
Cell Metab ; 30(1): 190-200.e6, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31105043

ABSTRACT

Mitochondrial abundance and function are tightly controlled during metabolic adaptation but dysregulated in pathological states such as diabetes, neurodegeneration, cancer, and kidney disease. We show here that translation of PGC1α, a key governor of mitochondrial biogenesis and oxidative metabolism, is negatively regulated by an upstream open reading frame (uORF) in the 5' untranslated region of its gene (PPARGC1A). We find that uORF-mediated translational repression is a feature of PPARGC1A orthologs from human to fly. Strikingly, whereas multiple inhibitory uORFs are broadly present in fish PPARGC1A orthologs, they are completely absent in the Atlantic bluefin tuna, an animal with exceptionally high mitochondrial content. In mice, an engineered mutation disrupting the PPARGC1A uORF increases PGC1α protein levels and oxidative metabolism and confers protection from acute kidney injury. These studies identify a translational regulatory element governing oxidative metabolism and highlight its potential contribution to the evolution of organismal mitochondrial function.


Subject(s)
5' Untranslated Regions/genetics , Open Reading Frames/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Diptera , Female , HEK293 Cells , Humans , Immunoprecipitation , Male , Mice , Mutation/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phylogeny , Protein Processing, Post-Translational/genetics , Tuna , Zebrafish
10.
Proc Natl Acad Sci U S A ; 115(3): 561-566, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29295932

ABSTRACT

The peroxisome-proliferator receptor-γ (PPARγ) is expressed in multiple cancer types. Recently, our group has shown that PPARγ is phosphorylated on serine 273 (S273), which selectively modulates the transcriptional program controlled by this protein. PPARγ ligands, including thiazolidinediones (TZDs), block S273 phosphorylation. This activity is chemically separable from the canonical activation of the receptor by agonist ligands and, importantly, these noncanonical agonist ligands do not cause some of the known side effects of TZDs. Here, we show that phosphorylation of S273 of PPARγ occurs in cancer cells on exposure to DNA damaging agents. Blocking this phosphorylation genetically or pharmacologically increases accumulation of DNA damage, resulting in apoptotic cell death. A genetic signature of PPARγ phosphorylation is associated with worse outcomes in response to chemotherapy in human patients. Noncanonical agonist ligands sensitize lung cancer xenografts and genetically induced lung tumors to carboplatin therapy. Moreover, inhibition of this phosphorylation results in deregulation of p53 signaling, and biochemical studies show that PPARγ physically interacts with p53 in a manner dependent on S273 phosphorylation. These data implicate a role for PPARγ in modifying the p53 response to cytotoxic therapy, which can be modulated for therapeutic gain using these compounds.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA Damage , Lung Neoplasms/drug therapy , PPAR gamma/metabolism , Thiazolidinediones/administration & dosage , Amino Acid Motifs , Animals , Apoptosis/drug effects , Carboplatin/administration & dosage , Cell Line, Tumor , DNA Damage/drug effects , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , PPAR gamma/agonists , PPAR gamma/chemistry , PPAR gamma/genetics , Phosphorylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Cell Metab ; 26(4): 660-671.e3, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28844881

ABSTRACT

Diet-induced thermogenesis is an important homeostatic mechanism that limits weight gain in response to caloric excess and contributes to the relative stability of body weight in most individuals. We previously demonstrated that creatine enhances energy expenditure through stimulation of mitochondrial ATP turnover, but the physiological role and importance of creatine energetics in adipose tissue have not been explored. Here, we have inactivated the first and rate-limiting enzyme of creatine biosynthesis, glycine amidinotransferase (GATM), selectively in fat (Adipo-Gatm KO). Adipo-Gatm KO mice are prone to diet-induced obesity due to the suppression of elevated energy expenditure that occurs in response to high-calorie feeding. This is paralleled by a blunted capacity for ß3-adrenergic activation of metabolic rate, which is rescued by dietary creatine supplementation. These results provide strong in vivo genetic support for a role of GATM and creatine metabolism in energy expenditure, diet-induced thermogenesis, and defense against diet-induced obesity.


Subject(s)
Adipocytes/metabolism , Amidinotransferases/metabolism , Creatine/metabolism , Diet, High-Fat/adverse effects , Obesity/etiology , Obesity/metabolism , Thermogenesis , Adipocytes/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiopathology , Amidinotransferases/genetics , Animals , Basal Metabolism , Creatine/genetics , Energy Metabolism , Mice , Mice, Knockout , Obesity/genetics , Obesity/physiopathology
14.
Nature ; 532(7597): 112-6, 2016 04 07.
Article in English | MEDLINE | ID: mdl-27027295

ABSTRACT

Brown and beige adipose tissues can dissipate chemical energy as heat through thermogenic respiration, which requires uncoupling protein 1 (UCP1). Thermogenesis from these adipocytes can combat obesity and diabetes, encouraging investigation of factors that control UCP1-dependent respiration in vivo. Here we show that acutely activated thermogenesis in brown adipose tissue is defined by a substantial increase in levels of mitochondrial reactive oxygen species (ROS). Remarkably, this process supports in vivo thermogenesis, as pharmacological depletion of mitochondrial ROS results in hypothermia upon cold exposure, and inhibits UCP1-dependent increases in whole-body energy expenditure. We further establish that thermogenic ROS alter the redox status of cysteine thiols in brown adipose tissue to drive increased respiration, and that Cys253 of UCP1 is a key target. UCP1 Cys253 is sulfenylated during thermogenesis, while mutation of this site desensitizes the purine-nucleotide-inhibited state of the carrier to adrenergic activation and uncoupling. These studies identify mitochondrial ROS induction in brown adipose tissue as a mechanism that supports UCP1-dependent thermogenesis and whole-body energy expenditure, which opens the way to improved therapeutic strategies for combating metabolic disorders.


Subject(s)
Cysteine/chemistry , Energy Metabolism , Ion Channels/chemistry , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Thermogenesis , Adipose Tissue, Brown/chemistry , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Cell Respiration , Cysteine/genetics , Cysteine/metabolism , Energy Metabolism/drug effects , Female , Humans , Ion Channels/deficiency , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oxidation-Reduction , Sulfhydryl Compounds/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1
15.
Cell ; 163(3): 643-55, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496606

ABSTRACT

Thermogenic brown and beige adipose tissues dissipate chemical energy as heat, and their thermogenic activities can combat obesity and diabetes. Herein the functional adaptations to cold of brown and beige adipose depots are examined using quantitative mitochondrial proteomics. We identify arginine/creatine metabolism as a beige adipose signature and demonstrate that creatine enhances respiration in beige-fat mitochondria when ADP is limiting. In murine beige fat, cold exposure stimulates mitochondrial creatine kinase activity and induces coordinated expression of genes associated with creatine metabolism. Pharmacological reduction of creatine levels decreases whole-body energy expenditure after administration of a ß3-agonist and reduces beige and brown adipose metabolic rate. Genes of creatine metabolism are compensatorily induced when UCP1-dependent thermogenesis is ablated, and creatine reduction in Ucp1-deficient mice reduces core body temperature. These findings link a futile cycle of creatine metabolism to adipose tissue energy expenditure and thermal homeostasis. PAPERCLIP.


Subject(s)
Adipose Tissue, Brown/metabolism , Creatine/metabolism , Thermogenesis , Adenosine Diphosphate/metabolism , Adipose Tissue/metabolism , Animals , Energy Metabolism , Homeostasis , Humans , Ion Channels/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Uncoupling Protein 1
16.
Nature ; 517(7534): 391-5, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25409143

ABSTRACT

Obesity-linked insulin resistance is a major precursor to the development of type 2 diabetes. Previous work has shown that phosphorylation of PPARγ (peroxisome proliferator-activated receptor γ) at serine 273 by cyclin-dependent kinase 5 (Cdk5) stimulates diabetogenic gene expression in adipose tissues. Inhibition of this modification is a key therapeutic mechanism for anti-diabetic drugs that bind PPARγ, such as the thiazolidinediones and PPARγ partial agonists or non-agonists. For a better understanding of the importance of this obesity-linked PPARγ phosphorylation, we created mice that ablated Cdk5 specifically in adipose tissues. These mice have both a paradoxical increase in PPARγ phosphorylation at serine 273 and worsened insulin resistance. Unbiased proteomic studies show that extracellular signal-regulated kinase (ERK) kinases are activated in these knockout animals. Here we show that ERK directly phosphorylates serine 273 of PPARγ in a robust manner and that Cdk5 suppresses ERKs through direct action on a novel site in MAP kinase/ERK kinase (MEK). Importantly, pharmacological inhibition of MEK and ERK markedly improves insulin resistance in both obese wild-type and ob/ob mice, and also completely reverses the deleterious effects of the Cdk5 ablation. These data show that an ERK/Cdk5 axis controls PPARγ function and suggest that MEK/ERK inhibitors may hold promise for the treatment of type 2 diabetes.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Diabetes Mellitus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , PPAR gamma/metabolism , Adipocytes/enzymology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Diet, High-Fat , Humans , Insulin Resistance , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Obese , PPAR gamma/chemistry , Phosphorylation
17.
Am J Physiol Endocrinol Metab ; 306(2): E157-67, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24280126

ABSTRACT

Diabetes risk increases significantly with age and correlates with lower oxidative capacity in muscle. Decreased expression of peroxisome proliferator-activated receptor-γ coactivator-1α (Pgc-1α) and target gene pathways involved in mitochondrial oxidative phosphorylation are associated with muscle insulin resistance, but a causative role has not been established. We sought to determine whether a decline in Pgc-1α and oxidative gene expression occurs during aging and potentiates the development of age-associated insulin resistance. Muscle-specific Pgc-1α knockout (MKO) mice and wild-type littermate controls were aged for 2 yr. Genetic signatures of skeletal muscle (microarray and mRNA expression) and metabolic profiles (glucose homeostasis, mitochondrial metabolism, body composition, lipids, and indirect calorimetry) of mice were compared at 3, 12, and 24 mo of age. Microarray and gene set enrichment analysis highlighted decreased function of the electron transport chain as characteristic of both aging muscle and loss of Pgc-1α expression. Despite significant reductions in oxidative gene expression and succinate dehydrogenase activity, young mice lacking Pgc-1α in muscle had lower fasting glucose and insulin. Consistent with loss of oxidative capacity during aging, Pgc-1α and Pgc-1ß expression were reduced in aged wild-type mouse muscle. Interestingly, the combination of age and loss of muscle Pgc-1α expression impaired glucose tolerance and led to increased fat mass, insulin resistance, and inflammatory markers in white adipose and liver tissues. Therefore, loss of Pgc-1α expression and decreased mitochondrial oxidative capacity contribute to worsening glucose tolerance and chronic systemic inflammation associated with aging.


Subject(s)
Aging/physiology , Glucose Intolerance/genetics , Inflammation/genetics , Muscle, Skeletal/metabolism , Transcription Factors/genetics , Animals , Gene Deletion , Gene Expression Profiling , Glucose Intolerance/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Mitochondria, Muscle/metabolism , Muscle, Skeletal/pathology , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
18.
Cell Metab ; 18(5): 649-59, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24120943

ABSTRACT

Exercise can improve cognitive function and has been linked to the increased expression of brain-derived neurotrophic factor (BDNF). However, the underlying molecular mechanisms driving the elevation of this neurotrophin remain unknown. Here we show that FNDC5, a previously identified muscle protein that is induced in exercise and is cleaved and secreted as irisin, is also elevated by endurance exercise in the hippocampus of mice. Neuronal Fndc5 gene expression is regulated by PGC-1α, and Pgc1a(-/-) mice show reduced Fndc5 expression in the brain. Forced expression of FNDC5 in primary cortical neurons increases Bdnf expression, whereas RNAi-mediated knockdown of FNDC5 reduces Bdnf. Importantly, peripheral delivery of FNDC5 to the liver via adenoviral vectors, resulting in elevated blood irisin, induces expression of Bdnf and other neuroprotective genes in the hippocampus. Taken together, our findings link endurance exercise and the important metabolic mediators, PGC-1α and FNDC5, with BDNF expression in the brain.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Fibronectins/metabolism , Hippocampus/metabolism , Physical Conditioning, Animal , Signal Transduction , Transcription Factors/metabolism , Adenoviridae/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Feedback, Physiological/drug effects , Fibronectins/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Genetic Vectors/metabolism , Hippocampus/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Binding/drug effects , Receptors, Estrogen/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , ERRalpha Estrogen-Related Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...