Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Metab ; 82: 101912, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38458566

ABSTRACT

OBJECTIVE: Skeletal muscle plasticity and remodeling are critical for adapting tissue function to use, disuse, and regeneration. The aim of this study was to identify genes and molecular pathways that regulate the transition from atrophy to compensatory hypertrophy or recovery from injury. Here, we have used a mouse model of hindlimb unloading and reloading, which causes skeletal muscle atrophy, and compensatory regeneration and hypertrophy, respectively. METHODS: We analyzed mouse skeletal muscle at the transition from hindlimb unloading to reloading for changes in transcriptome and extracellular fluid proteome. We then used qRT-PCR, immunohistochemistry, and bulk and single-cell RNA sequencing data to determine Mustn1 gene and protein expression, including changes in gene expression in mouse and human skeletal muscle with different challenges such as exercise and muscle injury. We generated Mustn1-deficient genetic mouse models and characterized them in vivo and ex vivo with regard to muscle function and whole-body metabolism. We isolated smooth muscle cells and functionally characterized them, and performed transcriptomics and proteomics analysis of skeletal muscle and aorta of Mustn1-deficient mice. RESULTS: We show that Mustn1 (Musculoskeletal embryonic nuclear protein 1, also known as Mustang) is highly expressed in skeletal muscle during the early stages of hindlimb reloading. Mustn1 expression is transiently elevated in mouse and human skeletal muscle in response to intense exercise, resistance exercise, or injury. We find that Mustn1 expression is highest in smooth muscle-rich tissues, followed by skeletal muscle fibers. Muscle from heterozygous Mustn1-deficient mice exhibit differences in gene expression related to extracellular matrix and cell adhesion, compared to wild-type littermates. Mustn1-deficient mice have normal muscle and aorta function and whole-body glucose metabolism. We show that Mustn1 is secreted from smooth muscle cells, and that it is present in arterioles of the muscle microvasculature and in muscle extracellular fluid, particularly during the hindlimb reloading phase. Proteomics analysis of muscle from Mustn1-deficient mice confirms differences in extracellular matrix composition, and female mice display higher collagen content after chemically induced muscle injury compared to wild-type littermates. CONCLUSIONS: We show that, in addition to its previously reported intracellular localization, Mustn1 is a microprotein secreted from smooth muscle cells into the muscle extracellular space. We explore its role in muscle ECM deposition and remodeling in homeostasis and upon muscle injury. The role of Mustn1 in fibrosis and immune infiltration upon muscle injury and dystrophies remains to be investigated, as does its potential for therapeutic interventions.


Subject(s)
Micropeptides , Muscle, Skeletal , Animals , Female , Humans , Mice , Extracellular Matrix/metabolism , Hypertrophy/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myocytes, Smooth Muscle/metabolism
2.
bioRxiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37398033

ABSTRACT

Muscular atrophy is a mortality risk factor that happens with disuse, chronic disease, and aging. Recovery from atrophy requires changes in several cell types including muscle fibers, and satellite and immune cells. Here we show that Zfp697/ZNF697 is a damage-induced regulator of muscle regeneration, during which its expression is transiently elevated. Conversely, sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Myofiber-specific Zfp697 ablation hinders the inflammatory and regenerative response to muscle injury, compromising functional recovery. We uncover Zfp697 as an essential interferon gamma mediator in muscle cells, interacting primarily with ncRNAs such as the pro-regenerative miR-206. In sum, we identify Zfp697 as an integrator of cell-cell communication necessary for tissue regeneration.

3.
Proc Natl Acad Sci U S A ; 119(48): e2119824119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36409897

ABSTRACT

Fatty acids are vital for the survival of eukaryotes, but when present in excess can have deleterious consequences. The AMP-activated protein kinase (AMPK) is an important regulator of multiple branches of metabolism. Studies in purified enzyme preparations and cultured cells have shown that AMPK is allosterically activated by small molecules as well as fatty acyl-CoAs through a mechanism involving Ser108 within the regulatory AMPK ß1 isoform. However, the in vivo physiological significance of this residue has not been evaluated. In the current study, we generated mice with a targeted germline knock-in (KI) mutation of AMPKß1 Ser108 to Ala (S108A-KI), which renders the site phospho-deficient. S108A-KI mice had reduced AMPK activity (50 to 75%) in the liver but not in the skeletal muscle. On a chow diet, S108A-KI mice had impairments in exogenous lipid-induced fatty acid oxidation. Studies in mice fed a high-fat diet found that S108A-KI mice had a tendency for greater glucose intolerance and elevated liver triglycerides. Consistent with increased liver triglycerides, livers of S108A-KI mice had reductions in mitochondrial content and respiration that were accompanied by enlarged mitochondria, suggestive of impairments in mitophagy. Subsequent studies in primary hepatocytes found that S108A-KI mice had reductions in palmitate- stimulated Cpt1a and Ppargc1a mRNA, ULK1 phosphorylation and autophagic/mitophagic flux. These data demonstrate an important physiological role of AMPKß1 Ser108 phosphorylation in promoting fatty acid oxidation, mitochondrial biogenesis and autophagy under conditions of high lipid availability. As both ketogenic diets and intermittent fasting increase circulating free fatty acid levels, AMPK activity, mitochondrial biogenesis, and mitophagy, these data suggest a potential unifying mechanism which may be important in mediating these effects.


Subject(s)
AMP-Activated Protein Kinases , Fatty Acids , Mice , Animals , Phosphorylation , Fatty Acids/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Mitochondria/metabolism , Homeostasis , Autophagy , Triglycerides/metabolism
4.
Cell Metab ; 33(11): 2215-2230.e8, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34592133

ABSTRACT

Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.


Subject(s)
Motor Neurons , Neurturin , Animals , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurturin/genetics , Neurturin/metabolism , Neurturin/pharmacology , Oxidative Stress
5.
Cell Signal ; 57: 45-57, 2019 05.
Article in English | MEDLINE | ID: mdl-30772465

ABSTRACT

AMP-activated protein kinase (AMPK) is a key regulator of cellular energy homeostasis, acting as a sensor of energy and nutrient status. As such, AMPK is considered a promising drug target for treatment of medical conditions particularly associated with metabolic dysfunctions. To better understand the downstream effectors and physiological consequences of AMPK activation, we have employed a chemical genetic screen in mouse primary hepatocytes in an attempt to identify novel AMPK targets. Treatment of hepatocytes with a potent and specific AMPK activator 991 resulted in identification of 65 proteins phosphorylated upon AMPK activation, which are involved in a variety of cellular processes such as lipid/glycogen metabolism, vesicle trafficking, and cytoskeleton organisation. Further characterisation and validation using mass spectrometry followed by immunoblotting analysis with phosphorylation site-specific antibodies identified AMPK-dependent phosphorylation of Gapex-5 (also known as GTPase-activating protein and VPS9 domain-containing protein 1 (GAPVD1)) on Ser902 in hepatocytes and starch-binding domain 1 (STBD1) on Ser175 in multiple cells/tissues. As new promising roles of AMPK as a key metabolic regulator continue to emerge, the substrates we identified could provide new mechanistic and therapeutic insights into AMPK-activating drugs in the liver.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Animals , Hepatocytes/metabolism , Homeostasis/genetics , Homeostasis/physiology , Lipid Metabolism/genetics , Mass Spectrometry/methods , Mice, Knockout , Phosphorylation , Substrate Specificity
6.
Diabetologia ; 60(2): 336-345, 2017 02.
Article in English | MEDLINE | ID: mdl-27826658

ABSTRACT

AIMS/HYPOTHESIS: TBC1D1 (tre-2/USP6, BUB2, cdc16 domain family member 1) is a Rab GTPase-activating protein (RabGAP) that has been implicated in regulating GLUT4 trafficking. TBC1D1 can be phosphorylated by the AMP-activated protein kinase (AMPK) on Ser231, which consequently interacts with 14-3-3 proteins. Given the key role for AMPK in regulating insulin-independent muscle glucose uptake, we hypothesised that TBC1D1-Ser231 phosphorylation and/or 14-3-3 binding may mediate AMPK-governed glucose homeostasis. METHODS: Whole-body glucose homeostasis and muscle glucose uptake were assayed in mice bearing a Tbc1d1 Ser231Ala-knockin mutation or harbouring skeletal muscle-specific Ampkα1/α2 (also known as Prkaa1/2) double-knockout mutations in response to an AMPK-activating agent, 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR). Exercise-induced muscle glucose uptake and exercise capacity were also determined in the Tbc1d1 Ser231Ala-knockin mice. RESULTS: Skeletal muscle-specific deletion of Ampkα1/a2 in mice prevented AICAR-induced hypoglycaemia and muscle glucose uptake. The Tbc1d1 Ser231Ala-knockin mutation also attenuated the glucose-lowering effect of AICAR in mice. Glucose uptake and cell surface GLUT4 content were significantly lower in muscle isolated from the Tbc1d1 Ser231Ala-knockin mice upon stimulation with a submaximal dose of AICAR. However, this Tbc1d1 Ser231Ala-knockin mutation neither impaired exercise-induced muscle glucose uptake nor affected exercise capacity in mice. CONCLUSIONS/INTERPRETATION: TBC1D1-Ser231 phosphorylation and/or 14-3-3 binding partially mediates AMPK-governed glucose homeostasis and muscle glucose uptake in a context-dependent manner.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Exercise/physiology , GTPase-Activating Proteins/genetics , Glucose/metabolism , Ribonucleotides/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/metabolism , Animals , Biological Transport , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mice , Muscle, Skeletal/metabolism , Mutation/genetics , Phosphorylation , Ribonucleotides/genetics , Signal Transduction/genetics , Signal Transduction/physiology
7.
Skelet Muscle ; 6(1): 30, 2016.
Article in English | MEDLINE | ID: mdl-27597886

ABSTRACT

BACKGROUND: Adult skeletal muscles are composed of slow and fast myofiber subtypes which each express selective genes required for their specific contractile and metabolic activity. Six homeoproteins are transcription factors regulating muscle cell fate through activation of myogenic regulatory factors and driving fast-type gene expression during embryogenesis. RESULTS: We show here that Six1 protein accumulates more robustly in the nuclei of adult fast-type muscles than in adult slow-type muscles, this specific enrichment takes place during perinatal growth. Deletion of Six1 in soleus impaired fast-type myofiber specialization during perinatal development, resulting in a slow phenotype and a complete lack of Myosin heavy chain 2A (MyHCIIA) expression. Global transcriptomic analysis of wild-type and Six1 mutant myofibers identified the gene networks controlled by Six1 in adult soleus muscle. This analysis showed that Six1 is required for the expression of numerous genes encoding fast-type sarcomeric proteins, glycolytic enzymes and controlling intracellular calcium homeostasis. Parvalbumin, a key player of calcium buffering, in particular, is a direct target of Six1 in the adult myofiber. CONCLUSIONS: This analysis revealed that Six1 controls distinct aspects of adult muscle physiology in vivo, and acts as a main determinant of fast-fiber type acquisition and maintenance.


Subject(s)
Homeodomain Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Calcium/metabolism , Gene Deletion , Gene Regulatory Networks , Glycolysis , Homeodomain Proteins/genetics , Male , Mice , Muscle Fibers, Skeletal/cytology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Transcriptome
8.
Cell Metab ; 24(1): 118-29, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27411013

ABSTRACT

Brown (BAT) and white (WAT) adipose tissues play distinct roles in maintaining whole-body energy homeostasis, and their dysfunction can contribute to non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes. The AMP-activated protein kinase (AMPK) is a cellular energy sensor, but its role in regulating BAT and WAT metabolism is unclear. We generated an inducible model for deletion of the two AMPK ß subunits in adipocytes (iß1ß2AKO) and found that iß1ß2AKO mice were cold intolerant and resistant to ß-adrenergic activation of BAT and beiging of WAT. BAT from iß1ß2AKO mice had impairments in mitochondrial structure, function, and markers of mitophagy. In response to a high-fat diet, iß1ß2AKO mice more rapidly developed liver steatosis as well as glucose and insulin intolerance. Thus, AMPK in adipocytes is vital for maintaining mitochondrial integrity, responding to pharmacological agents and thermal stress, and protecting against nutrient-overload-induced NAFLD and insulin resistance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/enzymology , Adipose Tissue, Beige/enzymology , Adipose Tissue, Brown/enzymology , Fatty Liver/enzymology , Insulin Resistance , Adipocytes/drug effects , Adipose Tissue, Beige/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat , Enzyme Activation/drug effects , Fatty Liver/pathology , Gene Deletion , Homeostasis/drug effects , Lipolysis/drug effects , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Norepinephrine/pharmacology , Thermogenesis/drug effects
9.
Cell Signal ; 27(5): 978-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25683918

ABSTRACT

AMP-activated protein kinase (AMPK) is a key cellular energy sensor and regulator of metabolic homeostasis. Although it is best known for its effects on carbohydrate and lipid metabolism, AMPK is implicated in diverse cellular processes, including mitochondrial biogenesis, autophagy, and cell growth and proliferation. To further our understanding of energy homeostasis through AMPK-dependent processes, the design and application of approaches to identify and characterise novel AMPK substrates are invaluable. Here, we report an affinity proteomicstrategy for the discovery and validation of AMPK targets using an antibody to isolate proteins containing the phospho-AMPK substrate recognition motif from hepatocytes that had been treated with pharmacological AMPK activators. We identified 57 proteins that were uniquely enriched in the activator-treated hepatocytes, but were absent in hepatocytes lacking AMPK. We focused on two candidates, cingulin and mitochondrial fission factor (MFF), and further characterised/validated them as AMPK-dependent targets by immunoblotting with phosphorylation site-specific antibodies. A small-molecule AMPK activator caused transient phosphorylation of endogenous cingulin at S137 in intestinal Caco2 cells. Multiple splice-variants of MFF appear to express in hepatocytes and we identified a common AMPK-dependent phospho-site (S129) in all the 3 predominant variants spanning the mass range and a short variant-specific site (S146). Collectively, our proteomic-based approach using a phospho-AMPK substrate antibody in combination with genetic models and selective AMPK activators will provide a powerful and reliable platform for identifying novel AMPK-dependent cellular targets.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hepatocytes/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , AMP-Activated Protein Kinases/analysis , Amino Acid Sequence , Animals , Caco-2 Cells , Cells, Cultured , Humans , Male , Mass Spectrometry , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/analysis , Molecular Sequence Data , Phosphorylation , Proteomics
10.
Am J Physiol Endocrinol Metab ; 306(6): E688-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24425763

ABSTRACT

AMP-activated protein kinase (AMPK) is a key cellular energy sensor and regulator of metabolic homeostasis. Activation of AMPK provides beneficial outcomes in fighting against metabolic disorders such as insulin resistance and type 2 diabetes. Currently, there is no allosteric AMPK activator available for the treatment of metabolic diseases, and limited compounds are available to robustly stimulate cellular/tissue AMPK in a specific manner. Here we investigated whether simultaneous administration of two different pharmacological AMPK activators, which bind and act on different sites, would result in an additive or synergistic effect on AMPK and its downstream signaling and physiological events in intact cells. We observed that cotreating primary hepatocytes with the AMP mimetic 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR) and a low dose (1 µM) of the allosteric activator A769662 produced a synergistic effect on AMPK Thr172 phosphorylation and catalytic activity, which was associated with a more profound increase/decrease in phosphorylation of downstream AMPK targets and inhibition of hepatic lipogenesis compared with single-compound treatment. Mechanistically, we found that cotreatment does not stimulate LKB1, upstream kinase for AMPK, but it protects against dephosphorylation of Thr172 phosphorylation by protein phosphatase PP2Cα in an additive manner in a cell-free assay. Collectively, we demonstrate that AICAR sensitizes the effect of A769662 and promotes AMPK activity and its downstream events. The study demonstrates the feasibility of promoting AMPK activity by using two activators with distinct modes of action in order to achieve a greater activation of AMPK and downstream signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Enzyme Activators/pharmacology , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Myoblasts/drug effects , Pyrones/pharmacology , Ribonucleotides/pharmacology , Thiophenes/pharmacology , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Allosteric Regulation/drug effects , Aminoimidazole Carboxamide/pharmacology , Animals , Biphenyl Compounds , Cell Line , Cells, Cultured , Glucose/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Lipogenesis/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Subunits/agonists , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects
11.
Biochem J ; 449(2): 479-89, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23078342

ABSTRACT

AS160 (Akt substrate of 160 kDa) is a Rab GTPase-activating protein implicated in insulin control of GLUT4 (glucose transporter 4) trafficking. In humans, a truncation mutation (R363X) in one allele of AS160 decreased the expression of the protein and caused severe postprandial hyperinsulinaemia during puberty. To complement the limited studies possible in humans, we generated an AS160-knockout mouse. In wild-type mice, AS160 expression is relatively high in adipose tissue and soleus muscle, low in EDL (extensor digitorum longus) muscle and detectable in liver only after enrichment. Despite having lower blood glucose levels under both fasted and random-fed conditions, the AS160-knockout mice exhibited insulin resistance in both muscle and liver in a euglycaemic clamp study. Consistent with this paradoxical phenotype, basal glucose uptake was higher in AS160-knockout primary adipocytes and normal in isolated soleus muscle, but their insulin-stimulated glucose uptake and overall GLUT4 levels were markedly decreased. In contrast, insulin-stimulated glucose uptake and GLUT4 levels were normal in EDL muscle. The liver also contributes to the AS160-knockout phenotype via hepatic insulin resistance, elevated hepatic expression of phosphoenolpyruvate carboxykinase isoforms and pyruvate intolerance, which are indicative of increased gluconeogenesis. Overall, as well as its catalytic function, AS160 influences expression of other proteins, and its loss deregulates basal and insulin-regulated glucose homoeostasis, not only in tissues that normally express AS160, but also by influencing liver function.


Subject(s)
Adipose Tissue/metabolism , GTPase-Activating Proteins/genetics , Insulin Resistance/genetics , Muscle, Skeletal/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Animals , Blood Glucose/metabolism , Blotting, Western , Cells, Cultured , Female , GTPase-Activating Proteins/deficiency , Glucose/metabolism , Glucose/pharmacokinetics , Glucose Tolerance Test , Glucose Transporter Type 4/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Insulin/blood , Insulin/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Phosphorylation/drug effects
12.
Am J Physiol Endocrinol Metab ; 302(9): E1036-43, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22318952

ABSTRACT

AS160 and its closely related protein TBC1D1 have emerged as key mediators for both insulin- and contraction-stimulated muscle glucose uptake through regulating GLUT4 trafficking. Insulin increases AS160 phosphorylation at multiple Akt/PKB consensus sites, including Thr(649), and promotes its binding to 14-3-3 proteins through phospho-Thr(649). We recently provided genetic evidence that AS160-Thr(649) phosphorylation/14-3-3 binding plays a key role in mediating insulin-stimulated glucose uptake in muscle. Contraction has also been proposed to increase phosphorylation of AS160 and TBC1D1 via AMPK, which could be detected by a generic phospho-Akt substrate (PAS) antibody. Here, analysis of AS160 immunoprecipitates from muscle extracts with site-specific phospho-antibodies revealed that contraction and AICAR caused no increase but rather a slight decrease in phosphorylation of the major PAS recognition site AS160-Thr(649). In line with this, contraction failed to enhance 14-3-3 binding to AS160. Consistent with previous reports, we also observed that in situ contraction stimulated the signal intensity of PAS antibody immunoreactive protein of ∼150-160 kDa in muscle extracts. Using a TBC1D1 deletion mutant mouse, we showed that TBC1D1 protein accounted for the majority of the PAS antibody immunoreactive signals of ∼150-160 kDa in extracts of contracted muscles. Consistent with the proposed role of AS160-Thr(649) phosphorylation/14-3-3 binding in mediating glucose uptake, AS160-Thr(649)Ala knock-in mice displayed normal glucose uptake upon contraction and AICAR in isolated muscles. We conclude that the previously reported PAS antibody immunoreactive band ∼150-160 kDa, which were increased upon contraction, does not represent AS160 but TBC1D1, and that AS160-Thr(649)Ala substitution impairs insulin- but neither contraction- nor AICAR-stimulated glucose uptake in mouse skeletal muscle.


Subject(s)
GTPase-Activating Proteins/metabolism , Insulin/physiology , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , 14-3-3 Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Amino Acid Substitution , Aminoimidazole Carboxamide/analogs & derivatives , Analysis of Variance , Animals , Blood Glucose/metabolism , GTPase-Activating Proteins/genetics , Gene Knock-In Techniques , Glucose Transporter Type 4/metabolism , Mice , Mice, Transgenic , Mutation , Phosphorylation , Ribonucleotides/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...