Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Biochem J ; 473(3): 335-45, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26578817

ABSTRACT

Laforin is a human dual-specificity phosphatase (DSP) involved in glycogen metabolism regulation containing a carbohydrate-binding module (CBM). Mutations in the gene coding for laforin are responsible for the development of Lafora disease, a progressive fatal myoclonus epilepsy with early onset, characterized by the intracellular deposition of abnormally branched, hyperphosphorylated insoluble glycogen-like polymers, called Lafora bodies. Despite the known importance of the CBM domain of laforin in the regulation of glycogen metabolism, the molecular mechanism of laforin-glycogen interaction is still poorly understood. Recently, the structure of laforin with bound maltohexaose was determined and despite the importance of such breakthrough, some molecular interaction details remained missing. We herein report a thorough biophysical characterization of laforin-carbohydrate interaction using soluble glycans. We demonstrated an increased preference of laforin for the interaction with glycans with higher order of polymerization and confirmed the importance of tryptophan residues for glycan interaction. Moreover, and in line with what has been described for other CBMs and lectins, our results confirmed that laforin-glycan interactions occur with a favourable enthalpic contribution counter-balanced by an unfavourable entropic contribution. The analysis of laforin-glycan interaction through the glycan side by saturation transfer difference (STD)-NMR has shown that the CBM-binding site can accommodate between 5 and 6 sugar units, which is in line with the recently obtained crystal structure of laforin. Overall, the work in the present study complements the structural characterization of laforin and sheds light on the molecular mechanism of laforin-glycan interaction, which is a pivotal requisite to understand the physiological and pathological roles of laforin.


Subject(s)
Lafora Disease/enzymology , Polysaccharides/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Binding Sites , Glycogen/chemistry , Glycogen/metabolism , Humans , Lafora Disease/genetics , Lafora Disease/metabolism , Polysaccharides/chemistry , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Substrate Specificity
2.
Ann Neurol ; 75(3): 442-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24419970

ABSTRACT

Ubiquitin ligases regulate quantities and activities of target proteins, often pleiotropically. The malin ubiquitin E3 ligase is reported to regulate autophagy, the misfolded protein response, microRNA silencing, Wnt signaling, neuronatin-mediated endoplasmic reticulum stress, and the laforin glycogen phosphatase. Malin deficiency causes Lafora disease, pathologically characterized by neurodegeneration and accumulations of malformed glycogen (Lafora bodies). We show that reducing glycogen production in malin-deficient mice by genetically removing PTG, a glycogen synthesis activator protein, nearly completely eliminates Lafora bodies and rescues the neurodegeneration, myoclonus, seizure susceptibility, and behavioral abnormality. Glycogen synthesis downregulation is a potential therapy for the fatal adolescence onset epilepsy Lafora disease.


Subject(s)
Intracellular Signaling Peptides and Proteins/therapeutic use , Lafora Disease/enzymology , Lafora Disease/therapy , Ubiquitin-Protein Ligases/deficiency , Animals , Brain/metabolism , Brain/pathology , Conditioning, Psychological , Down-Regulation , Fear/psychology , Glycogen/metabolism , Glycogen Synthase/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lafora Disease/psychology , Mice , Mice, Knockout , Myoclonus/enzymology , Myoclonus/genetics , Myoclonus/therapy , Neuroprotective Agents/metabolism , Plaque, Amyloid , Seizures/enzymology , Seizures/genetics , Seizures/therapy
3.
Brain ; 137(Pt 3): 806-18, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24430976

ABSTRACT

Lafora progressive myoclonus epilepsy (Lafora disease) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. The vast majority of patients carry mutations in either the EPM2A or EPM2B genes, encoding laforin, a glucan phosphatase, and malin, an E3 ubiquitin ligase, respectively. Although the precise physiological role of these proteins is not fully understood, work in past years has established a link between glycogen synthesis, Lafora bodies formation and Lafora disease development. To determine the role of the phosphatase activity of laforin in disease development we generated two Epm2a(-/-) mouse lines expressing either wild-type laforin or a mutant (C265S) laforin lacking only the phosphatase activity. Our results demonstrate that expression of either transgene blocks formation of Lafora bodies and restores the impairment in macroautophagy, preventing the development of Lafora bodies in Epm2a(-/-) mice. These data indicate that the critical pathogenic process is the control of abnormal glycogen accumulation through intracellular proteolytic systems by the laforin-malin complex, and not glycogen dephosphorylation by laforin. Understanding which is the essential process leading to Lafora disease pathogenesis represents a critical conceptual advance that should facilitate development of appropriate therapeutics.


Subject(s)
Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/metabolism , Lafora Disease/metabolism , Animals , Autophagy/genetics , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Female , Lafora Disease/enzymology , Lafora Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Phosphorylation/genetics , Protein Tyrosine Phosphatases, Non-Receptor
4.
J Biol Chem ; 287(30): 25650-9, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22669944

ABSTRACT

The solubility of glycogen, essential to its metabolism, is a property of its shape, a sphere generated through extensive branching during synthesis. Lafora disease (LD) is a severe teenage-onset neurodegenerative epilepsy and results from multiorgan accumulations, termed Lafora bodies (LB), of abnormally structured aggregation-prone and digestion-resistant glycogen. LD is caused by loss-of-function mutations in the EPM2A or EPM2B gene, encoding the interacting laforin phosphatase and malin E3 ubiquitin ligase enzymes, respectively. The substrate and function of malin are unknown; an early counterintuitive observation in cell culture experiments that it targets laforin to proteasomal degradation was not pursued until now. The substrate and function of laforin have recently been elucidated. Laforin dephosphorylates glycogen during synthesis, without which phosphate ions interfere with and distort glycogen construction, leading to LB. We hypothesized that laforin in excess or not removed following its action on glycogen also interferes with glycogen formation. We show in malin-deficient mice that the absence of malin results in massively increased laforin preceding the appearance of LB and that laforin gradually accumulates in glycogen, which corresponds to progressive LB generation. We show that increasing the amounts of laforin in cell culture causes LB formation and that this occurs only with glycogen binding-competent laforin. In summary, malin deficiency causes increased laforin, increased laforin binding to glycogen, and LB formation. Furthermore, increased levels of laforin, when it can bind glycogen, causes LB. We conclude that malin functions to regulate laforin and that malin deficiency at least in part causes LB and LD through increased laforin binding to glycogen.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Glycogen/metabolism , Lafora Disease/enzymology , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Adolescent , Animals , Cells, Cultured , Dual-Specificity Phosphatases/genetics , Female , Glycogen/genetics , Humans , Lafora Disease/genetics , Lafora Disease/pathology , Male , Mice , Mice, Knockout , Phosphorylation/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding/genetics , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases/genetics
5.
Hum Mol Genet ; 20(13): 2571-84, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21493628

ABSTRACT

Laforin is a dual specificity protein phosphatase involved in Lafora disease (LD), a fatal form of progressive myoclonus epilepsy characterized by neurodegeneration and the presence of intracellular polyglucosan inclusions (Lafora bodies) in different tissues. In this work, we describe that mice lacking laforin (epm2a-/-) have enhanced insulin response leading to altered whole-body energy balance. This enhanced insulin response overactivates the Akt pathway which increases glucose uptake in the heart, resulting in increased glycogen levels and the formation of polyglucosan inclusions. In addition, enhanced insulin response resulted in increased liver lipid biosynthesis, resulting in hepatic steatosis. On the contrary, overexpression in rat hepatoma FTO2B cells of native laforin but not of a form lacking phosphatase activity (C266S) resulted in attenuation of insulin signaling. These results define laforin as a new regulator of insulin sensitivity, which provides novel insights into LD pathogenesis and identifies this phosphatase as a potential novel component of the insulin signaling cascade.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Energy Metabolism , Insulin/metabolism , Lafora Disease/enzymology , Animals , Cell Line, Tumor , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Energy Metabolism/genetics , Female , Glucose/metabolism , Lafora Disease/genetics , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Knockout , Motor Activity/genetics , Myocardium/metabolism , Protein Tyrosine Phosphatases, Non-Receptor , Rats , Signal Transduction/genetics
6.
Biochem J ; 439(2): 265-75, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21728993

ABSTRACT

Lafora progressive myoclonus epilepsy [LD (Lafora disease)] is a fatal autosomal recessive neurodegenerative disorder caused by loss-of-function mutations in either the EPM2A gene, encoding the dual-specificity phosphatase laforin, or the EPM2B gene, encoding the E3-ubiquitin ligase malin. Previously, we and others showed that laforin and malin form a functional complex that regulates multiple aspects of glycogen metabolism, and that the interaction between laforin and malin is enhanced by conditions activating AMPK (AMP-activated protein kinase). In the present study, we demonstrate that laforin is a phosphoprotein, as indicated by two-dimensional electrophoresis, and we identify Ser(25) as the residue involved in this modification. We also show that Ser(25) is phosphorylated both in vitro and in vivo by AMPK. Lastly, we demonstrate that this residue plays a critical role for both the phosphatase activity and the ability of laforin to interact with itself and with previously established binding partners. The results of the present study suggest that phosphorylation of laforin-Ser(25) by AMPK provides a mechanism to modulate the interaction between laforin and malin. Regulation of this complex is necessary to maintain normal glycogen metabolism. Importantly, Ser(25) is mutated in some LD patients (S25P), and our results begin to elucidate the mechanism of disease in these patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lafora Disease/enzymology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Serine/metabolism , Cell Line , Electrophoresis, Gel, Two-Dimensional , Humans , Models, Molecular , Phosphorylation , Protein Conformation , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Two-Hybrid System Techniques
7.
BMC Evol Biol ; 11: 225, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798009

ABSTRACT

BACKGROUND: Malin is an E3-ubiquitin ligase that is mutated in Lafora disease, a fatal form of progressive myoclonus epilepsy. In order to perform its function, malin forms a functional complex with laforin, a glucan phosphatase that facilitates targeting of malin to its corresponding substrates. While laforin phylogeny has been studied, there are no data on the evolutionary lineage of malin. RESULTS: After an extensive search for malin orthologs, we found that malin is present in all vertebrate species and a cephalochordate, in contrast with the broader species distribution previously reported for laforin. These data suggest that in addition to forming a functional complex, laforin and perhaps malin may also have independent functions. In addition, we found that malin shares significant identity with the E3-ubiquitin ligase TRIM32, which belongs to the tripartite-motif containing family of proteins. We present experimental evidence that both malin and TRIM32 share some substrates for ubiquitination, although they produce ubiquitin chains with different topologies. However, TRIM32-specific substrates were not reciprocally ubiquitinated by the laforin-malin complex. CONCLUSIONS: We found that malin and laforin are not conserved in the same genomes. In addition, we found that malin shares significant identity with the E3-ubiquitin ligase TRIM32. The latter result suggests a common origin for malin and TRIM32 and provides insights into possible functional relationships between both proteins.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Evolution, Molecular , Lafora Disease/enzymology , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Humans , Lafora Disease/genetics , Molecular Sequence Data , Phylogeny , Sequence Alignment , Transcription Factors/chemistry , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitination , Vertebrates/classification , Vertebrates/genetics
8.
Neurobiol Dis ; 44(1): 133-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21742036

ABSTRACT

Lafora disease (LD) is the inherited progressive myoclonus epilepsy caused by mutations in either EPM2A gene, encoding the protein phosphatase laforin or the NHLRC1 gene, encoding the ubiquitin ligase malin. Since malin is an ubiquitin ligase and its mutations cause LD, it is hypothesized that improper clearance of its substrates might lead to LD pathogenesis. Here, we demonstrate for the first time that neuronatin is a novel substrate of malin. Malin interacts with neuronatin and enhances its degradation through proteasome. Interestingly, neuronatin is an aggregate prone protein, forms aggresome upon inhibition of cellular proteasome function and malin recruited to those aggresomes. Neuronatin is found to stimulate the glycogen synthesis through the activation of glycogen synthase and malin prevents neuronatin-induced glycogen synthesis. Several LD-associated mutants of malin are ineffective in the degradation of neuronatin and suppression of neuronatin-induced glycogen synthesis. Finally, we demonstrate the increased levels of neuronatin in the skin biopsy sample of LD patients. Overall, our results indicate that malin negatively regulates neuronatin and its loss of function in LD results in increased accumulation of neuronatin, which might be implicated in the formation of Lafora body or other aspect of disease pathogenesis.


Subject(s)
Carrier Proteins/pharmacology , Glycogen/biosynthesis , Lafora Disease/enzymology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteasome Endopeptidase Complex/drug effects , Animals , Blotting, Western , Carrier Proteins/genetics , DNA, Complementary/genetics , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Lafora Disease/genetics , Lafora Disease/metabolism , PC12 Cells , Rats , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Skin/pathology , Transfection , Ubiquitin-Protein Ligases , Ubiquitination/drug effects
9.
Clin Biochem ; 93: 80-89, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33831386

ABSTRACT

OBJECTIVES: Mutations in the gene encoding the glycogen phosphatase laforin result in the fatal childhood dementia Lafora disease (LD). A cellular hallmark of LD is cytoplasmic, hyper-phosphorylated, glycogen-like aggregates called Lafora bodies (LBs) that form in nearly all tissues and drive disease progression. Additional tools are needed to define the cellular function of laforin, understand the pathological role of laforin in LD, and determine the role of glycogen phosphate in glycogen metabolism. In this work, we present the generation and characterization of laforin nanobodies, with one being a laforin inhibitor. DESIGN AND METHODS: We identify multiple classes of specific laforin-binding nanobodies and determine their binding epitopes using hydrogen deuterium exchange (HDX) mass spectrometry. Using para-nitrophenyl phosphate (pNPP) and a malachite gold-based assay specific for glucan phosphatase activity, we assess the inhibitory effect of one nanobody on laforin's catalytic activity. RESULTS: Six families of laforin nanobodies are characterized and their epitopes mapped. One nanobody is identified and characterized that serves as an inhibitor of laforin's phosphatase activity. CONCLUSIONS: The six generated and characterized laforin nanobodies, with one being a laforin inhibitor, are an important set of tools that open new avenues to define unresolved glycogen metabolism questions.


Subject(s)
Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/chemistry , Animals , Biological Assay , Camelids, New World , Chromatography, Gel , Enzyme Inhibitors/pharmacology , Epitope Mapping , Glycogen/metabolism , Gold/chemistry , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Lafora Disease/enzymology , Models, Molecular , Nitrophenols/chemistry , Organometallic Compounds/chemistry , Organophosphorus Compounds/chemistry , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Single-Domain Antibodies/isolation & purification
10.
Proc Natl Acad Sci U S A ; 104(49): 19262-6, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18040046

ABSTRACT

Lafora disease is a progressive myoclonus epilepsy with onset typically in the second decade of life and death within 10 years. Lafora bodies, deposits of abnormally branched, insoluble glycogen-like polymers, form in neurons, muscle, liver, and other tissues. Approximately half of the cases of Lafora disease result from mutations in the EPM2A gene, which encodes laforin, a member of the dual-specificity protein phosphatase family that additionally contains a glycogen binding domain. The molecular basis for the formation of Lafora bodies is completely unknown. Glycogen, a branched polymer of glucose, contains a small amount of covalently linked phosphate whose origin and function are obscure. We report here that recombinant laforin is able to release this phosphate in vitro, in a time-dependent reaction with an apparent K(m) for glycogen of 4.5 mg/ml. Mutations of laforin that disable the glycogen binding domain also eliminate its ability to dephosphorylate glycogen. We have also analyzed glycogen from a mouse model of Lafora disease, Epm2a(-/-) mice, which develop Lafora bodies in several tissues. Glycogen isolated from these mice had a 40% increase in the covalent phosphate content in liver and a 4-fold elevation in muscle. We propose that excessive phosphorylation of glycogen leads to aberrant branching and Lafora body formation. This study provides a molecular link between an observed biochemical property of laforin and the phenotype of a mouse model of Lafora disease. The results also have important implications for glycogen metabolism generally.


Subject(s)
Dual-Specificity Phosphatases/deficiency , Glycogen/metabolism , Lafora Disease/enzymology , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Glycogen Synthase/analysis , Glycogen Synthase/metabolism , Male , Mice , Mice, Knockout , Mutation , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor , Rabbits , Recombinant Proteins/pharmacology
11.
Hum Mutat ; 30(5): 715-23, 2009 May.
Article in English | MEDLINE | ID: mdl-19267391

ABSTRACT

Lafora disease (LD) is an autosomal recessive and fatal form of progressive myoclonus epilepsy. LD patients manifest myoclonus and tonic-clonic seizures, visual hallucinations, and progressive neurologic deterioration beginning at 12 to 15 years of age. The two genes known to be associated with LD are EPM2A and NHLRC1. Mutations in at least one other as yet unknown gene also cause LD. The EMP2A encodes a protein phosphatase and NHLRC1 encodes an ubiquitin ligase. These two proteins interact with each other and, as a complex, are thought to regulate critical neuronal functions. Nearly 100 distinct mutations have been discovered in the two genes in over 200 independent LD families. Nearly half of them are missense mutations, and the deletion mutations account for one-quarter. Several reports have provided functional data for the mutant proteins and a few also provide genotype-phenotype correlations. In this review we provide an update on the spectrum of EPM2A and NHLRC1 mutations, and discuss their distribution in the patient population, genotype-phenotype correlations, and on the possible effect of disease mutations on the cellular functions of LD proteins.


Subject(s)
Carrier Proteins/genetics , Lafora Disease/enzymology , Lafora Disease/genetics , Mutation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Adolescent , Carrier Proteins/chemistry , Genotype , Humans , Lafora Disease/diagnosis , Phenotype , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Ubiquitin-Protein Ligases
12.
Biochimie ; 88(12): 1961-71, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17010495

ABSTRACT

Lafora disease is a progressive myoclonus epilepsy with an early fatal issue. Two genes were identified thus far, the mutations of which cause the disease. The first one, EPM2A, encodes the consensus sequence of a protein tyrosine phosphatase. Its product, laforin, is the object of the present work. We analysed in detail the amino acid sequence of this protein. This suggested, as also observed by others, that it could present two domains, a carbohydrate-binding domain (CBM20, known as a starch-binding domain) and the catalytic domain of a dual-specificity protein phosphatase. We produced the enzyme as two different GST-fused proteins and as an N-terminally His-tagged protein. Differences in solubility were observed between the constructs. Moreover, the N-terminal carbohydrate-binding domain contains a thrombin cleavage site, which is hidden in the simplest GST-fusion protein we produced, but was accessible after introducing a five-residue linker between the engineered cleavage site and the enzyme N-terminus. The two types of constructs hydrolyse pNPP and OMFP with kinetic parameters consistent with those of a dual-specificity phosphatase. We show in addition that the protein not only binds glycogen, but also starch, amylose and cyclodextrin. Neither binding of glycogen nor of beta-cyclodextrin appreciably affects the phosphatase activity. These results suggest that the role of the N-terminal domain is rather that of targeting the protein in the cell, probably to glycogen and the protein complexes attached to it, rather than that of directly modulating the catalytic activity.


Subject(s)
Lafora Disease/enzymology , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Blotting, Western , Glycogen/metabolism , Humans , Kinetics , Lafora Disease/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
13.
Methods Mol Biol ; 1447: 107-19, 2016.
Article in English | MEDLINE | ID: mdl-27514803

ABSTRACT

Glucan phosphatases are a recently discovered family of enzymes that dephosphorylate either starch or glycogen and are essential for proper starch metabolism in plants and glycogen metabolism in humans. Mutations in the gene encoding the only human glucan phosphatase, laforin, result in the fatal, neurodegenerative, epilepsy known as Lafora disease. Here, we describe phosphatase assays to assess both generic laforin phosphatase activity and laforin's unique glycogen phosphatase activity.


Subject(s)
Enzyme Assays/methods , Glycogen/metabolism , Lafora Disease/enzymology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Fluoresceins/metabolism , Humans , Lafora Disease/metabolism , Nitrophenols/metabolism , Organophosphorus Compounds/metabolism , Substrate Specificity
14.
Epilepsy Res ; 40(1): 33-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10771256

ABSTRACT

Progressive myoclonic epilepsies (EPM) are difficult to treat and refractory to most antiepileptic drugs. Besides epilepsy, EPMs also involve continuous neurological deterioration. Oxidative stress is thought to be an important factor in this process. We therefore analyzed a series of antioxidant enzymes in the blood of patients and compared with healthy age matched controls. In addition patients were given high doses of N-acetylcysteine (NAC), a glutathione percursor to determine if symptoms of EPM would improve. Five patients, four with EPM 1 (Unverricht-Lundborg disease) and one patient with EPM2 (Lafora body disease) were treated with 6 g/day of NAC. Before treatment, plasma samples were analyzed for glutathione peroxidase activity, catalase activity, extracellular superoxide dismutase (SOD) and CuZn-SOD and compared with the controls. Erythrocyte CuZn-SOD was significantly lower in the EPM patients compared to controls. NAC improved markedly and stabilized the neurological symptoms in patients with EPM 1 but had a doubtful effect in the patient with EPM 2.


Subject(s)
Epilepsies, Myoclonic/enzymology , Glutathione Peroxidase/metabolism , Superoxide Dismutase/metabolism , Acetylcysteine/therapeutic use , Adult , Epilepsies, Myoclonic/blood , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/physiopathology , Erythrocytes/enzymology , Female , Humans , Lafora Disease/blood , Lafora Disease/drug therapy , Lafora Disease/enzymology , Lafora Disease/physiopathology , Male , Reference Values , Unverricht-Lundborg Syndrome/drug therapy , Unverricht-Lundborg Syndrome/metabolism , Unverricht-Lundborg Syndrome/physiopathology
15.
Cell Metab ; 17(5): 756-67, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23663739

ABSTRACT

Laforin or malin deficiency causes Lafora disease, characterized by altered glycogen metabolism and teenage-onset neurodegeneration with intractable and invariably fatal epilepsy. Plant starches possess small amounts of metabolically essential monophosphate esters. Glycogen contains similar phosphate amounts, which are thought to originate from a glycogen synthase error side reaction and therefore lack any specific function. Glycogen is also believed to lack monophosphates at glucosyl carbon C6, an essential phosphorylation site in plant starch metabolism. We now show that glycogen phosphorylation is not due to a glycogen synthase side reaction, that C6 is a major glycogen phosphorylation site, and that C6 monophosphates predominate near centers of glycogen molecules and positively correlate with glycogen chain lengths. Laforin or malin deficiency causes C6 hyperphosphorylation, which results in malformed long-chained glycogen that accumulates in many tissues, causing neurodegeneration in brain. Our work advances the understanding of Lafora disease pathogenesis and suggests that glycogen phosphorylation has important metabolic function.


Subject(s)
Glycogen/metabolism , Lafora Disease/metabolism , Animals , Brain/enzymology , Brain/metabolism , Carbon/metabolism , Glycogen Synthase/metabolism , Lafora Disease/enzymology , Male , Mice , Phosphorylation , Rabbits
16.
Int J Biochem Cell Biol ; 45(7): 1479-88, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23624058

ABSTRACT

Protein phosphatase type 1 (PP1) plays a major role in the regulation of glycogen biosynthesis. PP1 is recruited to sites of glycogen formation by its binding to specific targeting subunits. There, it dephosphorylates different enzymes involved in glycogen homeostasis leading to an activation of glycogen biosynthesis. Regulation of these targeting subunits is crucial, as excess of them leads to an enhancement of the action of PP1, which results in glycogen accumulation. In this work we present evidence that PPP1R3D (R6), one of the PP1 glycogenic targeting subunits, interacts physically with laforin, a glucan phosphatase involved in Lafora disease, a fatal type of progressive myoclonus epilepsy. Binding of R6 to laforin allows the ubiquitination of R6 by the E3-ubiquitin ligase malin, what targets R6 for autophagic degradation. As a result of the action of the laforin-malin complex on R6, its glycogenic activity is downregulated. Since R6 is expressed in brain, our results suggest that the laforin-malin complex downregulates the glycogenic activity of R6 present in neuron cells to prevent glycogen accumulation.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Lafora Disease/metabolism , Protein Phosphatase 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagy , Carrier Proteins , Cell Line, Tumor , Down-Regulation , Glycogen/metabolism , HEK293 Cells , Humans , Lafora Disease/enzymology , Mice , Neurons/metabolism , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitination
17.
Mol Neurobiol ; 48(1): 49-61, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23546741

ABSTRACT

Glycogen, the largest cytosolic macromolecule, is soluble because of intricate construction generating perfect hydrophilic-surfaced spheres. Little is known about neuronal glycogen function and metabolism, though progress is accruing through the neurodegenerative epilepsy Lafora disease (LD) proteins laforin and malin. Neurons in LD exhibit Lafora bodies (LBs), large accumulations of malconstructed insoluble glycogen (polyglucosans). We demonstrated that the laforin-malin complex reduces LBs and protects neuronal cells against endoplasmic reticulum stress-induced apoptosis. We now show that stress induces polyglucosan formation in normal neurons in culture and in the brain. This is mediated by increased glucose-6-phosphate allosterically hyperactivating muscle glycogen synthase (GS1) and is followed by activation of the glycogen digesting enzyme glycogen phosphorylase. In the absence of laforin, stress-induced polyglucosans are undigested and accumulate into massive LBs, and in laforin-deficient mice, stress drastically accelerates LB accumulation and LD. The mechanism through which laforin-malin mediates polyglucosan degradation remains unclear but involves GS1 dephosphorylation by laforin. Our work uncovers the presence of rapid polyglucosan metabolism as part of the normal physiology of neuroprotection. We propose that deficiency in the degradative phase of this metabolism, leading to LB accumulation and resultant seizure predisposition and neurodegeneration, underlies LD.


Subject(s)
Disease Progression , Dual-Specificity Phosphatases/metabolism , Endoplasmic Reticulum Stress , Glucans/metabolism , Inclusion Bodies/metabolism , Lafora Disease/pathology , Neurons/pathology , Allosteric Regulation , Animals , Apoptosis , Enzyme Activation , Gene Knockdown Techniques , Glycogen/biosynthesis , Glycogen Phosphorylase/metabolism , Glycogen Synthase/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Hydrolysis , Lafora Disease/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Neurons/enzymology , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor , alpha-Amylases/metabolism
18.
Cell Metab ; 13(3): 274-82, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21356517

ABSTRACT

Glycogen is a branched polymer of glucose that serves as an energy store. Phosphate, a trace constituent of glycogen, has profound effects on glycogen structure, and phosphate hyperaccumulation is linked to Lafora disease, a fatal progressive myoclonus epilepsy that can be caused by mutations of laforin, a glycogen phosphatase. However, little is known about the metabolism of glycogen phosphate. We demonstrate here that the biosynthetic enzyme glycogen synthase, which normally adds glucose residues to glycogen, is capable of incorporating the ß-phosphate of its substrate UDP-glucose at a rate of one phosphate per approximately 10,000 glucoses, in what may be considered a catalytic error. We show that the phosphate in glycogen is present as C2 and C3 phosphomonoesters. Since hyperphosphorylation of glycogen causes Lafora disease, phosphate removal by laforin may thus be considered a repair or damage control mechanism.


Subject(s)
Glycogen/biosynthesis , Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Glycogen Synthase/metabolism , Lafora Disease/enzymology , Phosphorylation , Rabbits , Uridine Diphosphate Glucose/metabolism
19.
PLoS One ; 6(8): e24040, 2011.
Article in English | MEDLINE | ID: mdl-21887368

ABSTRACT

Lafora Disease (LD) is a fatal neurodegenerative epileptic disorder that presents as a neurological deterioration with the accumulation of insoluble, intracellular, hyperphosphorylated carbohydrates called Lafora bodies (LBs). LD is caused by mutations in either the gene encoding laforin or malin. Laforin contains a dual specificity phosphatase domain and a carbohydrate-binding module, and is a member of the recently described family of glucan phosphatases. In the current study, we investigated the functional and physiological relevance of laforin dimerization. We purified recombinant human laforin and subjected the monomer and dimer fractions to denaturing gel electrophoresis, mass spectrometry, phosphatase assays, protein-protein interaction assays, and glucan binding assays. Our results demonstrate that laforin prevalently exists as a monomer with a small dimer fraction both in vitro and in vivo. Of mechanistic importance, laforin monomer and dimer possess equal phosphatase activity, and they both associate with malin and bind glucans to a similar extent. However, we found differences between the two states' ability to interact simultaneously with malin and carbohydrates. Furthermore, we tested other members of the glucan phosphatase family. Cumulatively, our data suggest that laforin monomer is the dominant form of the protein and that it contains phosphatase activity.


Subject(s)
Dual-Specificity Phosphatases/analysis , Lafora Disease/enzymology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Carbohydrates , Carrier Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Humans , Protein Binding , Protein Multimerization , Protein Tyrosine Phosphatases, Non-Receptor/analysis , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Ubiquitin-Protein Ligases
20.
EMBO Mol Med ; 3(11): 667-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21882344

ABSTRACT

Lafora disease (LD) is caused by mutations in either the laforin or malin gene. The hallmark of the disease is the accumulation of polyglucosan inclusions called Lafora Bodies (LBs). Malin knockout (KO) mice present polyglucosan accumulations in several brain areas, as do patients of LD. These structures are abundant in the cerebellum and hippocampus. Here, we report a large increase in glycogen synthase (GS) in these mice, in which the enzyme accumulates in LBs. Our study focused on the hippocampus where, under physiological conditions, astrocytes and parvalbumin-positive (PV(+)) interneurons expressed GS and malin. Although LBs have been described only in neurons, we found this polyglucosan accumulation in the astrocytes of the KO mice. They also had LBs in the soma and some processes of PV(+) interneurons. This phenomenon was accompanied by the progressive loss of these neuronal cells and, importantly, neurophysiological alterations potentially related to impairment of hippocampal function. Our results emphasize the relevance of the laforin-malin complex in the control of glycogen metabolism and highlight altered glycogen accumulation as a key contributor to neurodegeneration in LD.


Subject(s)
Disease Models, Animal , Glycogen Synthase/metabolism , Lafora Disease/enzymology , Lafora Disease/physiopathology , Mice , Nerve Degeneration/enzymology , Animals , Astrocytes/enzymology , Female , Glycogen/metabolism , Glycogen Synthase/genetics , Hippocampus/enzymology , Humans , Inclusion Bodies/enzymology , Inclusion Bodies/genetics , Lafora Disease/genetics , Lafora Disease/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL