Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Biol ; 19(11): e3001447, 2021 11.
Article in English | MEDLINE | ID: mdl-34758018

ABSTRACT

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Subject(s)
Glycogen Synthase/metabolism , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Myocardium/enzymology , Animals , Animals, Newborn , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Diet, High-Fat , Enzyme Activation , Feeding Behavior , Female , Gene Deletion , Glucose Intolerance/enzymology , Glycogen/metabolism , Glycogen Synthase Kinase 3/metabolism , Insulin Resistance , Lipid Metabolism , MAP Kinase Signaling System , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Organ Specificity , Phosphorylation
2.
Cell ; 137(5): 793-5, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19490884

ABSTRACT

By consolidating scientific societies into a confederation, Spanish scientists have developed a powerful platform from which to promote the importance of scientific research and science education to both the general public and government.


Subject(s)
Societies, Scientific/organization & administration , Government , Science/education , Spain
3.
J Neurochem ; 2023 Jul 04.
Article in English | MEDLINE | ID: mdl-37401737

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons in the spinal cord. Glial cells, including astrocytes and microglia, have been shown to contribute to neurodegeneration in ALS, and metabolic dysfunction plays an important role in the progression of the disease. Glycogen is a soluble polymer of glucose found at low levels in the central nervous system that plays an important role in memory formation, synaptic plasticity, and the prevention of seizures. However, its accumulation in astrocytes and/or neurons is associated with pathological conditions and aging. Importantly, glycogen accumulation has been reported in the spinal cord of human ALS patients and mouse models. In the present work, using the SOD1G93A mouse model of ALS, we show that glycogen accumulates in the spinal cord and brainstem during symptomatic and end stages of the disease and that the accumulated glycogen is associated with reactive astrocytes. To study the contribution of glycogen to ALS progression, we generated SOD1G93A mice with reduced glycogen synthesis (SOD1G93A GShet mice). SOD1G93A GShet mice had a significantly longer life span than SOD1G93A mice and showed lower levels of the astrocytic pro-inflammatory cytokine Cxcl10, suggesting that the accumulation of glycogen is associated with an inflammatory response. Supporting this, inducing an increase in glycogen synthesis reduced life span in SOD1G93A mice. Altogether, these results suggest that glycogen in reactive astrocytes contributes to neurotoxicity and disease progression in ALS.

4.
Hum Mol Genet ; 29(21): 3554-3565, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33219378

ABSTRACT

The glycogenin knockout mouse is a model of Glycogen Storage Disease type XV. These animals show high perinatal mortality (90%) due to respiratory failure. The lungs of glycogenin-deficient embryos and P0 mice have a lower glycogen content than that of wild-type counterparts. Embryonic lungs were found to have decreased levels of mature surfactant proteins SP-B and SP-C, together with incomplete processing of precursors. Furthermore, non-surviving pups showed collapsed sacculi, which may be linked to a significantly reduced amount of surfactant proteins. A similar pattern was observed in glycogen synthase1-deficient mice, which are devoid of glycogen in the lungs and are also affected by high perinatal mortality due to atelectasis. These results indicate that glycogen availability is a key factor for the burst of surfactant production required to ensure correct lung expansion at the establishment of air breathing. Our findings confirm that glycogen deficiency in lungs can cause respiratory distress syndrome and suggest that mutations in glycogenin and glycogen synthase 1 genes may underlie cases of idiopathic neonatal death.


Subject(s)
Glucosyltransferases/physiology , Glycogen Synthase/physiology , Glycoproteins/physiology , Pulmonary Surfactants/metabolism , Respiratory Distress Syndrome/pathology , Animals , Animals, Newborn , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism
5.
Int J Mol Sci ; 24(3)2023 Jan 29.
Article in English | MEDLINE | ID: mdl-36768897

ABSTRACT

Many lines of evidence demonstrate a correlation between liver glycogen content and food intake. We previously demonstrated that mice overexpressing protein targeting to glycogen (PTG) specifically in the liver-which have increased glycogen content in this organ-are protected from high-fat diet (HFD)-induced obesity by reduced food intake. However, the use of PTG to increase liver glycogen implies certain limitations. PTG stimulates glycogen synthesis but also inhibits the enzyme responsible for glycogen degradation. Furthermore, as PTG is a regulatory subunit of protein phosphatase 1 (PP1), which regulates many cellular functions, its overexpression could have side effects beyond the regulation of glycogen metabolism. Therefore, it is necessary to determine whether the direct activation of glycogen synthesis, without affecting its degradation or other cellular functions, has the same effects. To this end, we generated mice overexpressing a non-inactivatable form of glycogen synthase (GS) specifically in the liver (9A-MGSAlb mice). Control and 9a-MGSAlb mice were fed a standard diet (SD) or HFD for 16 weeks. Glucose tolerance and feeding behavior were analyzed. 9A-MGSAlb mice showed an increase in hepatic glycogen in fed and fasting conditions. When fed an HFD, these animals preserved their hepatic energy state, had a reduced food intake, and presented a lower body weight and fat mass than control animals, without changes in energy expenditure. Furthermore, 9A-MGSAlb animals showed improved glucose tolerance when fed an SD or HFD. Moreover, liver triacylglycerol levels that were increased after HFD feeding were lower in these mice. These results confirm that increased liver glycogen stores contribute to decreased appetite and improve glucose tolerance in mice fed an HFD. On the basis of our findings, strategies to preserve hepatic glycogen stores emerge as potential treatments for obesity and hyperglycemia.


Subject(s)
Glucose Intolerance , Liver Glycogen , Animals , Mice , Body Weight , Diet, High-Fat , Eating/physiology , Glucose/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/prevention & control , Glucose Intolerance/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Liver/metabolism , Mice, Inbred C57BL , Obesity/etiology , Obesity/prevention & control , Obesity/metabolism
6.
J Biol Chem ; 297(2): 100976, 2021 08.
Article in English | MEDLINE | ID: mdl-34284060

ABSTRACT

Muscle glycogen depletion has been proposed as one of the main causes of fatigue during exercise. However, few studies have addressed the contribution of liver glycogen to exercise performance. Using a low-intensity running protocol, here, we analyzed exercise capacity in mice overexpressing protein targeting to glycogen (PTG) specifically in the liver (PTGOE mice), which show a high concentration of glycogen in this organ. PTGOE mice showed improved exercise capacity, as determined by the distance covered and time ran in an extenuating endurance exercise, compared with control mice. Moreover, fasting decreased exercise capacity in control mice but not in PTGOE mice. After exercise, liver glycogen stores were totally depleted in control mice, but PTGOE mice maintained significant glycogen levels even in fasting conditions. In addition, PTGOE mice displayed an increased hepatic energy state after exercise compared with control mice. Exercise caused a reduction in the blood glucose concentration in control mice that was less pronounced in PTGOE mice. No changes were found in the levels of blood lactate, plasma free fatty acids, or ß-hydroxybutyrate. Plasma glucagon was elevated after exercise in control mice, but not in PTGOE mice. Exercise-induced changes in skeletal muscle were similar in both genotypes. These results identify hepatic glycogen as a key regulator of endurance capacity in mice, an effect that may be exerted through the maintenance of blood glucose levels.


Subject(s)
Blood Glucose/metabolism , Exercise Tolerance/physiology , Fatty Acids, Nonesterified/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Glycogen/metabolism , Muscle, Skeletal/metabolism , Animals , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
J Biol Chem ; 296: 100498, 2021.
Article in English | MEDLINE | ID: mdl-33667544

ABSTRACT

Hepatic glycogen metabolism is impaired in diabetes. We previously demonstrated that strategies to increase liver glycogen content in a high-fat-diet mouse model of obesity and insulin resistance led to a reduction in food intake and ameliorated obesity and glucose tolerance. These effects were accompanied by a decrease in insulin levels, but whether this decrease contributed to the phenotype observed in this animal was unclear. Here we sought to evaluate this aspect directly, by examining the long-term effects of increasing liver glycogen in an animal model of insulin-deficient and monogenic diabetes, namely the Akita mouse, which is characterized by reduced insulin production. We crossed Akita mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate Akita mice with increased liver glycogen content (Akita-PTGOE). Akita-PTGOE animals showed lower glycemia, lower food intake, and decreased water consumption and urine output compared with Akita mice. Furthermore, Akita-PTGOE mice showed a restoration of the hepatic energy state and a normalization of gluconeogenesis and glycolysis back to nondiabetic levels. Moreover, hepatic lipogenesis, which is reduced in Akita mice, was reverted in Akita-PTGOE animals. These results demonstrate that strategies to increase liver glycogen content lead to the long-term reduction of the diabetic phenotype, independently of circulating insulin.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Glycogen/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Disease Models, Animal , Female , Gluconeogenesis , Glycolysis , Male , Mice , Mice, Inbred C57BL , Phenotype
8.
Gastroenterology ; 160(5): 1725-1740.e2, 2021 04.
Article in English | MEDLINE | ID: mdl-33309778

ABSTRACT

BACKGROUND & AIMS: We recently showed that alcoholic hepatitis (AH) is characterized by dedifferentiation of hepatocytes and loss of mature functions. Glucose metabolism is tightly regulated in healthy hepatocytes. We hypothesize that AH may lead to metabolic reprogramming of the liver, including dysregulation of glucose metabolism. METHODS: We performed integrated metabolomic and transcriptomic analyses of liver tissue from patients with AH or alcoholic cirrhosis or normal liver tissue from hepatic resection. Focused analyses of chromatin immunoprecipitation coupled to DNA sequencing was performed. Functional in vitro studies were performed in primary rat and human hepatocytes and HepG2 cells. RESULTS: Patients with AH exhibited specific changes in the levels of intermediates of glycolysis/gluconeogenesis, the tricarboxylic acid cycle, and monosaccharide and disaccharide metabolism. Integrated analysis of the transcriptome and metabolome showed the used of alternate energetic pathways, metabolite sinks and bottlenecks, and dysregulated glucose storage in patients with AH. Among genes involved in glucose metabolism, hexokinase domain containing 1 (HKDC1) was identified as the most up-regulated kinase in patients with AH. Histone active promoter and enhancer markers were increased in the HKDC1 genomic region. High HKDC1 levels were associated with the development of acute kidney injury and decreased survival. Increased HKDC1 activity contributed to the accumulation of glucose-6-P and glycogen in primary rat hepatocytes. CONCLUSIONS: Altered metabolite levels and messenger RNA expression of metabolic enzymes suggest the existence of extensive reprogramming of glucose metabolism in AH. Increased HKDC1 expression may contribute to dysregulated glucose metabolism and represents a novel biomarker and therapeutic target for AH.


Subject(s)
Cell Dedifferentiation , Energy Metabolism , Gene Expression Profiling , Glucose/metabolism , Hepatitis, Alcoholic/enzymology , Hepatocytes/enzymology , Hexokinase/metabolism , Liver/enzymology , Metabolomics , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Adaptation, Physiological , Animals , Europe , Female , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Hep G2 Cells , Hepatitis, Alcoholic/genetics , Hepatitis, Alcoholic/pathology , Hepatocytes/pathology , Hexokinase/genetics , Humans , Liver/pathology , Male , Metabolome , Middle Aged , Rats, Wistar , Transcriptome , United States
9.
Brain ; 144(8): 2349-2360, 2021 09 04.
Article in English | MEDLINE | ID: mdl-33822008

ABSTRACT

The hallmark of Lafora disease, a fatal neurodegenerative disorder, is the accumulation of intracellular glycogen aggregates called Lafora bodies. Until recently, it was widely believed that brain Lafora bodies were present exclusively in neurons and thus that Lafora disease pathology derived from their accumulation in this cell population. However, recent evidence indicates that Lafora bodies are also present in astrocytes. To define the role of astrocytic Lafora bodies in Lafora disease pathology, we deleted glycogen synthase specifically from astrocytes in a mouse model of the disease (malinKO). Strikingly, blocking glycogen synthesis in astrocytes-thus impeding Lafora bodies accumulation in this cell type-prevented the increase in neurodegeneration markers, autophagy impairment, and metabolic changes characteristic of the malinKO model. Conversely, mice that over-accumulate glycogen in astrocytes showed an increase in these markers. These results unveil the deleterious consequences of the deregulation of glycogen metabolism in astrocytes and change the perspective that Lafora disease is caused solely by alterations in neurons.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glycogen/metabolism , Lafora Disease/metabolism , Nerve Degeneration/metabolism , Animals , Astrocytes/pathology , Brain/pathology , Disease Models, Animal , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Lafora Disease/genetics , Lafora Disease/pathology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Neurobiol Dis ; 147: 105173, 2021 01.
Article in English | MEDLINE | ID: mdl-33171226

ABSTRACT

Lafora disease (LD) is a fatal adolescence-onset neurodegenerative condition. The hallmark of LD is the accumulation of aberrant glycogen aggregates called Lafora bodies (LBs) in the brain and other tissues. Impeding glycogen synthesis from early embryonic stages by genetic suppression of glycogen synthase (MGS) in an animal model of LD prevents LB formation and ultimately the pathological manifestations of LD thereby indicating that LBs are responsible for the pathophysiology of the disease. However, it is not clear whether eliminating glycogen synthesis in an adult animal after LBs have already formed would halt or reverse the progression of LD. Herein we generated a mouse model of LD with inducible MGS suppression. We evaluated the effect of MGS suppression at different time points on LB accumulation as well as on the appearance of neuroinflammation, a pathologic trait of LD models. In the skeletal muscle, MGS suppression in adult LD mice blocked the formation of new LBs and reduced the number of glycogen aggregates. In the brain, early but not late MGS suppression halted the accumulation of LBs. However, the neuroinflammatory response was still present, as shown by the levels of reactive astrocytes, microglia and inflammatory cytokines. Our results confirm that MGS as a promising therapeutic target for LD and highlight the importance of an early diagnosis for effective treatment of the disease.


Subject(s)
Brain/pathology , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Lafora Disease/pathology , Muscle, Skeletal/pathology , Animals , Disease Models, Animal , Glycogen/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Arch Biochem Biophys ; 695: 108626, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33049291

ABSTRACT

Glycogen branching enzyme (GBE1) introduces branching points in the glycogen molecule during its synthesis. Pathogenic GBE1 gene mutations lead to glycogen storage disease type IV (GSD IV), which is characterized by excessive intracellular accumulation of abnormal, poorly branched glycogen in affected tissues and organs, mostly in the liver. Using heterozygous Gbe1 knock-out mice (Gbe1+/-), we analyzed the effects of moderate GBE1 deficiency on oxidative stress in the liver. The livers of aged Gbe1+/- mice (22 months old) had decreased GBE1 protein levels, which caused a mild decrease in the degree of glycogen branching, but did not affect the tissue glycogen content. GBE1 deficiency was accompanied by increased protein carbonylation and elevated oxidation of the glutathione pool, indicating the existence of oxidative stress. Furthermore, we have observed increased levels of glutathione peroxidase and decreased activity of respiratory complex I in Gbe1+/- livers. Our data indicate that even mild changes in the degree of glycogen branching, which did not lead to excessive glycogen accumulation, may have broader effects on cellular bioenergetics and redox homeostasis. In young animals cellular homeostatic mechanisms are able to counteract those changes, while in aged tissues the changes may lead to increased oxidative stress.


Subject(s)
Aging/metabolism , Glycogen Debranching Enzyme System/deficiency , Glycogen Storage Disease Type IV/metabolism , Liver/enzymology , Oxidative Stress , Aging/genetics , Aging/pathology , Animals , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glycogen/genetics , Glycogen/metabolism , Glycogen Debranching Enzyme System/metabolism , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/pathology , Liver/pathology , Mice , Mice, Knockout , Protein Carbonylation/genetics
12.
Epilepsy Behav ; 103(Pt A): 106839, 2020 02.
Article in English | MEDLINE | ID: mdl-31932179

ABSTRACT

Lafora disease (LD) is both a fatal childhood epilepsy and a glycogen storage disease caused by recessive mutations in either the Epilepsy progressive myoclonus 2A (EPM2A) or EPM2B genes. Hallmarks of LD are aberrant, cytoplasmic carbohydrate aggregates called Lafora bodies (LBs) that are a disease driver. The 5th International Lafora Epilepsy Workshop was recently held in Alcala de Henares, Spain. The workshop brought together nearly 100 clinicians, academic and industry scientists, trainees, National Institutes of Health (NIH) representation, and friends and family members of patients with LD. The workshop covered aspects of LD ranging from defining basic scientific mechanisms to elucidating a LD therapy or cure and a recently launched LD natural history study.


Subject(s)
Congresses as Topic/trends , Education/trends , Internationality , Lafora Disease/therapy , Animals , Humans , Lafora Disease/epidemiology , Lafora Disease/genetics , Mutation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Spain/epidemiology
13.
J Biol Chem ; 293(19): 7117-7125, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29483193

ABSTRACT

Lafora disease (LD) is a fatal, autosomal recessive, glycogen-storage disorder that manifests as severe epilepsy. LD results from mutations in the gene encoding either the glycogen phosphatase laforin or the E3 ubiquitin ligase malin. Individuals with LD develop cytoplasmic, aberrant glycogen inclusions in nearly all tissues that more closely resemble plant starch than human glycogen. This Minireview discusses the unique window into glycogen metabolism that LD research offers. It also highlights recent discoveries, including that glycogen contains covalently bound phosphate and that neurons synthesize glycogen and express both glycogen synthase and glycogen phosphorylase.


Subject(s)
Glycogen/metabolism , Lafora Disease/metabolism , Neurons/metabolism , Animals , Carbohydrate Conformation , Carrier Proteins/genetics , Disease Models, Animal , Glycogen/biosynthesis , Glycogen/chemistry , Glycogen Phosphorylase/genetics , Humans , Lafora Disease/genetics , Lafora Disease/pathology , Lafora Disease/therapy , Phosphates/metabolism , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases/genetics
14.
Glia ; 66(10): 2094-2107, 2018 10.
Article in English | MEDLINE | ID: mdl-30152044

ABSTRACT

Lafora disease (LD), the most devastating adolescence-onset epilepsy, is caused by mutations in the EPM2A or EPM2B genes, which encode the proteins laforin and malin, respectively. Loss of function of one of these proteins, which are involved in the regulation of glycogen synthesis, induces the accumulation of polyglucosan bodies (PGBs)-known as Lafora bodies (LBs) and associated with neurons-in the brain. Ageing and some neurodegenerative conditions lead to the appearance of another type of PGB called corpora amylacea, which are associated with astrocytes and contain neo-epitopes that can be recognized by natural antibodies. Here we studied the PGBs in the cerebral cortex and hippocampus of malin knockout mice, a mouse model of LD. These animals presented not only LBs associated with neurons but also a significant number of PGBs associated with astrocytes. These astrocytic PGBs were also increased in mice from senescence-accelerated mouse-prone 8 (SAMP8) strain and mice with overexpression of Protein Targeting to Glycogen (PTGOE ), indicating that they are not exclusive of LD. The astrocytic PGBs, but not neuronal LBs, contained neo-epitopes that are recognized by natural antibodies. The astrocytic PGBs appeared predominantly in the hippocampus but were also present in some cortical brain regions, while neuronal LBs were found mainly in the brain cortex and the pyramidal layer of hippocampal regions CA2 and CA3. Our results indicate that astrocytes, contrary to current belief, are involved in the etiopathogenesis of LD.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Glucans/metabolism , Inclusion Bodies/metabolism , Lafora Disease/metabolism , Neurons/metabolism , Animals , Astrocytes/pathology , Cerebral Cortex/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Inclusion Bodies/pathology , Lafora Disease/pathology , Mice, Transgenic , Neurons/pathology
15.
Diabetologia ; 60(6): 1076-1083, 2017 06.
Article in English | MEDLINE | ID: mdl-28299379

ABSTRACT

AIMS/HYPOTHESIS: Liver glycogen plays a key role in regulating food intake and blood glucose. Mice that accumulate large amounts of this polysaccharide in the liver are protected from high-fat diet (HFD)-induced obesity by reduced food intake. Furthermore, these animals show reversal of the glucose intolerance and hyperinsulinaemia caused by the HFD. The aim of this study was to examine the involvement of the hepatic branch of the vagus nerve in regulating food intake and glucose homeostasis in this model. METHODS: We performed hepatic branch vagotomy (HBV) or a sham operation on mice overexpressing protein targeting to glycogen (Ptg OE). Starting 1 week after surgery, mice were fed an HFD for 10 weeks. RESULTS: HBV did not alter liver glycogen or ATP levels, thereby indicating that this procedure does not interfere with hepatic energy balance. However, HBV reversed the effect of glycogen accumulation on food intake. In wild-type mice, HBV led to a significant reduction in body weight without a change in food intake. Consistent with their body weight reduction, these animals had decreased fat deposition, adipocyte size, and insulin and leptin levels, together with increased energy expenditure. Ptg OE mice showed an increase in energy expenditure and glucose oxidation, and these differences were abolished by HBV. Moreover, Ptg OE mice showed an improvement in HFD-induced glucose intolerance, which was suppressed by HBV. CONCLUSIONS/INTERPRETATION: Our results demonstrate that the regulation of food intake and glucose homeostasis by liver glycogen is dependent on the hepatic branch of the vagus nerve.


Subject(s)
Blood Glucose/physiology , Eating/physiology , Liver Glycogen/metabolism , Vagus Nerve/metabolism , Vagus Nerve/physiology , Animals , Diet, High-Fat/adverse effects , Glucose/metabolism , Homeostasis , Liver/metabolism , Mice , Obesity/etiology , Obesity/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Polymerase Chain Reaction
16.
PLoS Comput Biol ; 12(4): e1004899, 2016 04.
Article in English | MEDLINE | ID: mdl-27124774

ABSTRACT

The liver performs many essential metabolic functions, which can be studied using computational models of hepatocytes. Here we present HepatoDyn, a highly detailed dynamic model of hepatocyte metabolism. HepatoDyn includes a large metabolic network, highly detailed kinetic laws, and is capable of dynamically simulating the redox and energy metabolism of hepatocytes. Furthermore, the model was coupled to the module for isotopic label propagation of the software package IsoDyn, allowing HepatoDyn to integrate data derived from 13C based experiments. As an example of dynamical simulations applied to hepatocytes, we studied the effects of high fructose concentrations on hepatocyte metabolism by integrating data from experiments in which rat hepatocytes were incubated with 20 mM glucose supplemented with either 3 mM or 20 mM fructose. These experiments showed that glycogen accumulation was significantly lower in hepatocytes incubated with medium supplemented with 20 mM fructose than in hepatocytes incubated with medium supplemented with 3 mM fructose. Through the integration of extracellular fluxes and 13C enrichment measurements, HepatoDyn predicted that this phenomenon can be attributed to a depletion of cytosolic ATP and phosphate induced by high fructose concentrations in the medium.


Subject(s)
Hepatocytes/metabolism , Models, Biological , Animals , Carbon Isotopes , Computational Biology , Computer Simulation , Fructose/metabolism , Glucose/metabolism , In Vitro Techniques , Kinetics , Male , Metabolic Networks and Pathways , Rats , Rats, Wistar
17.
Proc Natl Acad Sci U S A ; 111(28): E2831-40, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982189

ABSTRACT

Glycogen is a primary form of energy storage in eukaryotes that is essential for glucose homeostasis. The glycogen polymer is synthesized from glucose through the cooperative action of glycogen synthase (GS), glycogenin (GN), and glycogen branching enzyme and forms particles that range in size from 10 to 290 nm. GS is regulated by allosteric activation upon glucose-6-phosphate binding and inactivation by phosphorylation on its N- and C-terminal regulatory tails. GS alone is incapable of starting synthesis of a glycogen particle de novo, but instead it extends preexisting chains initiated by glycogenin. The molecular determinants by which GS recognizes self-glucosylated GN, the first step in glycogenesis, are unknown. We describe the crystal structure of Caenorhabditis elegans GS in complex with a minimal GS targeting sequence in GN and show that a 34-residue region of GN binds to a conserved surface on GS that is distinct from previously characterized allosteric and binding surfaces on the enzyme. The interaction identified in the GS-GN costructure is required for GS-GN interaction and for glycogen synthesis in a cell-free system and in intact cells. The interaction of full-length GS-GN proteins is enhanced by an avidity effect imparted by a dimeric state of GN and a tetrameric state of GS. Finally, the structure of the N- and C-terminal regulatory tails of GS provide a basis for understanding phosphoregulation of glycogen synthesis. These results uncover a central molecular mechanism that governs glycogen metabolism.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans/enzymology , Glucosyltransferases , Glycogen Synthase , Glycoproteins , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell-Free System , Cells, Cultured , Crystallography, X-Ray , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycogen/biosynthesis , Glycogen/chemistry , Glycogen/genetics , Glycogen Synthase/chemistry , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Mice , Mice, Knockout , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Structure-Activity Relationship
18.
Diabetologia ; 59(5): 1012-20, 2016 May.
Article in English | MEDLINE | ID: mdl-26825527

ABSTRACT

AIMS/HYPOTHESIS: Glycogen accumulation occurs in beta cells of diabetic patients and has been proposed to partly mediate glucotoxicity-induced beta cell dysfunction. However, the role of glycogen metabolism in beta cell function and its contribution to diabetes pathophysiology remain poorly understood. We investigated the function of beta cell glycogen by studying glucose homeostasis in mice with (1) defective glycogen synthesis in the pancreas; and (2) excessive glycogen accumulation in beta cells. METHODS: Conditional deletion of the Gys1 gene and overexpression of protein targeting to glycogen (PTG) was accomplished by Cre-lox recombination using pancreas-specific Cre lines. Glucose homeostasis was assessed by determining fasting glycaemia, insulinaemia and glucose tolerance. Beta cell mass was determined by morphometry. Glycogen was detected histologically by periodic acid-Schiff's reagent staining. Isolated islets were used for the determination of glycogen and insulin content, insulin secretion, immunoblots and gene expression assays. RESULTS: Gys1 knockout (Gys1 (KO)) mice did not exhibit differences in glucose tolerance or basal glycaemia and insulinaemia relative to controls. Insulin secretion and gene expression in isolated islets was also indistinguishable between Gys1 (KO) and controls. Conversely, despite effective glycogen overaccumulation in islets, mice with PTG overexpression (PTG(OE)) presented similar glucose tolerance to controls. However, under fasting conditions they exhibited lower glycaemia and higher insulinaemia. Importantly, neither young nor aged PTG(OE) mice showed differences in beta cell mass relative to age-matched controls. Finally, a high-fat diet did not reveal a beta cell-autonomous phenotype in either model. CONCLUSIONS/INTERPRETATION: Glycogen metabolism is not required for the maintenance of beta cell function. Glycogen accumulation in beta cells alone is not sufficient to trigger the dysfunction or loss of these cells, or progression to diabetes.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Insulin-Secreting Cells/metabolism , Animals , Female , Glycogen/physiology , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Homeostasis , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Knockout
19.
J Cell Biochem ; 117(11): 2597-607, 2016 11.
Article in English | MEDLINE | ID: mdl-27017955

ABSTRACT

Sertoli cell metabolism actively maintains the nutritional needs of germ cells. It has been described that after glucose incorporation in Sertoli cells, less than 1% is converted to glycogen suggesting low levels of glycogen synthase activity. Phosphorylation of muscle glycogen synthase (MGS) at serine 640 (pS640MGS) decreases its activity, and this form of the enzyme was discovered as a non-ribosomal protein that modulates the translation of a subset of transcripts in HeLa cells. The aim of our study was to functionally characterize MGS in cultured Sertoli cells, as well as to explore this new feature related to RNA molecules. We detected MGS in the cytoplasm of Sertoli cells as well as in the nuclei. The activity rates of the enzyme were extremely low indicating that MGS is expressed but almost inactive. Protein targeting to glycogen (PTG) overexpression was performed to activate MGS by dephosphorylation. PTG induced glycogen synthesis massively, confirming that this enzyme is present but inactive. This finding correlates with high levels of pS640MGS, which were assayed by phosphatase treatment. To explore a putative new function for MGS in Sertoli cells, we performed RNA immunoprecipitation coupled to microarray studies. The results revealed that MGS co-immunoprecipitated with the several mRNAs and also rRNAs. These findings indicate that MGS is expressed Sertoli cells but in an inactive form, and also support a possibly novel feature of this metabolic enzyme associated with RNA-related molecules. J. Cell. Biochem. 117: 2597-2607, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Glycogen Synthase/metabolism , Glycogen/biosynthesis , Muscle, Skeletal/enzymology , RNA/metabolism , Sertoli Cells/enzymology , Animals , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Glucose/metabolism , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL
20.
Hum Mol Genet ; 23(12): 3147-56, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24452334

ABSTRACT

Lafora disease is a fatal neurodegenerative condition characterized by the accumulation of abnormal glycogen inclusions known as Lafora bodies. It is an autosomal recessive disorder caused by mutations in either the laforin or malin gene. To study whether glycogen is primarily responsible for the neurodegeneration in Lafora disease, we generated malin knockout mice with impaired (totally or partially) glycogen synthesis. These animals did not show the increase in markers of neurodegeneration, the impairments in electrophysiological properties of hippocampal synapses, nor the susceptibility to kainate-induced epilepsy seen in the malin knockout model. Interestingly, the autophagy impairment that has been described in malin knockout animals was also rescued in this double knockout model. Conversely, two other mouse models in which glycogen is over-accumulated in the brain independently of the lack of malin showed impairment in autophagy. Our findings reveal that glycogen accumulation accounts for the neurodegeneration and functional consequences seen in the malin knockout model, as well as the impaired autophagy. These results identify the regulation of glycogen synthesis as a key target for the treatment of Lafora disease.


Subject(s)
Autophagy , Dual-Specificity Phosphatases/metabolism , Glycogen Synthase/genetics , Glycogen/metabolism , Lafora Disease/physiopathology , Ubiquitin-Protein Ligases/genetics , Animals , Biomarkers/metabolism , Disease Models, Animal , Electrical Synapses/metabolism , Epilepsy/chemically induced , Epilepsy/pathology , Glycogen Synthase/metabolism , Hippocampus/physiology , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Kainic Acid/pharmacology , Lafora Disease/metabolism , Lafora Disease/pathology , Mice , Mice, Knockout , Mutation , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL