Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
J Biol Chem ; 288(48): 34627-37, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24142699

ABSTRACT

Glycogen synthesis is a major component of the insulin response, and defective glycogen synthesis is a major portion of insulin resistance. Insulin regulates glycogen synthase (GS) through incompletely defined pathways that activate the enzyme through dephosphorylation and, more potently, allosteric activation. We identify Epm2aip1 as a GS-associated protein. We show that the absence of Epm2aip1 in mice impairs allosteric activation of GS by glucose 6-phosphate, decreases hepatic glycogen synthesis, increases liver fat, causes hepatic insulin resistance, and protects against age-related obesity. Our work identifies a novel GS-associated GS activity-modulating component of insulin resistance.


Subject(s)
Dual-Specificity Phosphatases/genetics , Glycogen Synthase/metabolism , Glycogen/biosynthesis , Insulin Resistance/genetics , Obesity/pathology , Aging/genetics , Animals , Dual-Specificity Phosphatases/metabolism , Glucose-6-Phosphate/metabolism , Glycogen/genetics , Glycogen Synthase/genetics , Humans , Insulin/genetics , Insulin/metabolism , Liver/enzymology , Liver/metabolism , Liver/pathology , Mice , Obesity/etiology , Obesity/genetics , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor
3.
Ann Neurol ; 74(2): 297-300, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23913475

ABSTRACT

Lafora disease (LD) is a fatal progressive myoclonus epilepsy characterized neuropathologically by aggregates of abnormally structured glycogen and proteins (Lafora bodies [LBs]), and neurodegeneration. Whether LBs could be prevented by inhibiting glycogen synthesis and whether they are pathogenic remain uncertain. We genetically eliminated brain glycogen synthesis in LD mice. This resulted in long-term prevention of LB formation, neurodegeneration, and seizure susceptibility. This study establishes that glycogen synthesis is requisite for LB formation and that LBs are pathogenic. It opens a therapeutic window for potential treatments in LD with known and future small molecule inhibitors of glycogen synthesis.


Subject(s)
Glycogen/antagonists & inhibitors , Glycogen/biosynthesis , Lafora Disease/prevention & control , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Gene Knockout Techniques , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Lafora Disease/pathology , Lafora Disease/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatases, Non-Receptor
4.
Brain ; 135(Pt 9): 2684-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22961547

ABSTRACT

The most common progressive myoclonus epilepsies are the late infantile and late infantile-variant neuronal ceroid lipofuscinoses (onset before the age of 6 years), Unverricht-Lundborg disease (onset after the age of 6 years) and Lafora disease. Lafora disease is a distinct disorder with uniform course: onset in teenage years, followed by progressively worsening myoclonus, seizures, visual hallucinations and cognitive decline, leading to a vegetative state in status myoclonicus and death within 10 years. Biopsy reveals Lafora bodies, which are pathognomonic and not seen with any other progressive myoclonus epilepsies. Lafora bodies are aggregates of polyglucosans, poorly constructed glycogen molecules with inordinately long strands that render them insoluble. Lafora disease is caused by mutations in the EPM2A or EPM2B genes, encoding the laforin phosphatase and the malin ubiquitin ligase, respectively, two cytoplasmically active enzymes that regulate glycogen construction, ensuring symmetric expansion into a spherical shape, essential to its solubility. In this work, we report a new progressive myoclonus epilepsy associated with Lafora bodies, early-onset Lafora body disease, map its locus to chromosome 4q21.21, identify its gene and mutation and characterize the relationship of its gene product with laforin and malin. Early-onset Lafora body disease presents early, at 5 years, with dysarthria, myoclonus and ataxia. The combination of early-onset and early dysarthria strongly suggests late infantile-variant neuronal ceroid lipofuscinosis, not Lafora disease. Pathology reveals no ceroid lipofuscinosis, but Lafora bodies. The subsequent course is a typical progressive myoclonus epilepsy, though much more protracted than any infantile neuronal ceroid lipofuscinosis, or Lafora disease, patients living into the fourth decade. The mutation, c.781T>C (Phe261Leu), is in a gene of unknown function, PRDM8. We show that the PRDM8 protein interacts with laforin and malin and causes translocation of the two proteins to the nucleus. We find that Phe261Leu-PRDM8 results in excessive sequestration of laforin and malin in the nucleus and that it therefore likely represents a gain-of-function mutation that leads to an effective deficiency of cytoplasmic laforin and malin. We have identified a new progressive myoclonus epilepsy with Lafora bodies, early-onset Lafora body disease, 101 years after Lafora disease was first described. The results to date suggest that PRDM8, the early-onset Lafora body disease protein, regulates the cytoplasmic quantities of the Lafora disease enzymes.


Subject(s)
Brain/pathology , Carrier Proteins/genetics , Lafora Disease/genetics , Muscle, Skeletal/pathology , Nuclear Proteins/genetics , Adolescent , Adult , Age of Onset , Atrophy , Child , Child, Preschool , Chromosomes, Human, Pair 4 , DNA-Binding Proteins , Disease Progression , Female , Histone Methyltransferases , Humans , Lafora Disease/pathology , Lod Score , Male , Mutation , Skin/pathology
5.
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
6.
Am J Med Genet B Neuropsychiatr Genet ; 159B(2): 210-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22213695

ABSTRACT

The overwhelming majority of Rett syndrome cases are caused by mutations in the gene MECP2. MECP2 has two isoforms, termed MECP2_e1 and MECP2_e2, which differ in their N-terminal amino acid sequences. A growing body of evidence has indicated that MECP2_e1 may be the etiologically relevant isoform in Rett Syndrome based on its expression profile in the brain and because, strikingly, no mutations have been discovered that affect MECP2_e2 exclusively. In this study we sought to characterize four classical Rett patients with mutations that putatively affect only the MECP2_e1 isoform. Our hypothesis was that the classical Rett phenotype seen here is the result of disrupted MECP2_e1 expression, but with MECP2_e2 expression unaltered. We used quantitative reverse transcriptase PCR to assay mRNA expression for each isoform independently, and used cytospinning methods to assay total MECP2 in peripheral blood lymphocytes (PBL). In the two Rett patients with identical 11 bp deletions within the coding portion of exon 1, MECP2_e2 levels were unaffected, whilst a significant reduction of MECP2_e1 levels was detected. In two Rett patients harboring mutations in the exon 1 start codon, MECP2_e1 and MECP2_e2 mRNA amounts were unaffected. In summary, we have shown that patients with exon 1 mutations transcribe normal levels of MECP2_e2 mRNA, and most PBL are positive for MeCP2 protein, despite them theoretically being unable to produce the MECP2_e1 isoform, and yet still exhibit the classical RTT phenotype. Altogether, our work further supports our hypothesis that MECP2_e1 is the predominant isoform involved in the neuropathology of Rett syndrome.


Subject(s)
Exons/genetics , Methyl-CpG-Binding Protein 2/genetics , Mutation/genetics , Rett Syndrome/genetics , Transcription, Genetic , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Phenotype , Prognosis , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , X Chromosome Inactivation , Young Adult
7.
PLoS Genet ; 7(4): e1002037, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21552327

ABSTRACT

Lafora disease is the most common teenage-onset neurodegenerative disease, the main teenage-onset form of progressive myoclonus epilepsy (PME), and one of the severest epilepsies. Pathologically, a starch-like compound, polyglucosan, accumulates in neuronal cell bodies and overtakes neuronal small processes, mainly dendrites. Polyglucosan formation is catalyzed by glycogen synthase, which is activated through dephosphorylation by glycogen-associated protein phosphatase-1 (PP1). Here we remove PTG, one of the proteins that target PP1 to glycogen, from mice with Lafora disease. This results in near-complete disappearance of polyglucosans and in resolution of neurodegeneration and myoclonic epilepsy. This work discloses an entryway to treating this fatal epilepsy and potentially other glycogen storage diseases.


Subject(s)
Glucans/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lafora Disease/physiopathology , Animals , Brain/physiopathology , Disease Models, Animal , Glucans/analysis , Glycogen Synthase/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lafora Disease/genetics , Mice , Mice, Knockout
8.
Ann Neurol ; 68(6): 925-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21077101

ABSTRACT

OBJECTIVE: Glycogen, the largest cytosolic macromolecule, acquires solubility, essential to its function, through extreme branching. Lafora bodies are aggregates of polyglucosan, a long, linear, poorly branched, and insoluble form of glycogen. Lafora bodies occupy vast numbers of neuronal dendrites and perikarya in Lafora disease in time-dependent fashion, leading to intractable and fatal progressive myoclonus epilepsy. Lafora disease is caused by deficiency of either the laforin glycogen phosphatase or the malin E3 ubiquitin ligase. The 2 leading hypotheses of Lafora body formation are: (1) increased glycogen synthase activity extends glycogen strands too rapidly to allow adequate branching, resulting in polyglucosans; and (2) increased glycogen phosphate leads to glycogen conformational change, unfolding, precipitation, and conversion to polyglucosan. Recently, it was shown that in the laforin phosphatase-deficient form of Lafora disease, there is no increase in glycogen synthase, but there is a dramatic increase in glycogen phosphate, with subsequent conversion of glycogen to polyglucosan. Here, we determine whether Lafora bodies in the malin ubiquitin ligase-deficient form of the disease are due to increased glycogen synthase or increased glycogen phosphate. METHODS: We generated malin-deficient mice and tested the 2 hypotheses. RESULTS: Malin-deficient mice precisely replicate the pathology of Lafora disease with Lafora body formation in skeletal muscle, liver, and brain, and in the latter in the pathognomonic perikaryal and dendritic locations. Glycogen synthase quantity and activity are unchanged. There is a highly significant increase in glycogen phosphate. INTERPRETATION: We identify a single common modification, glycogen hyperphosphorylation, as the root cause of Lafora body pathogenesis.


Subject(s)
Glycogen/metabolism , Hyperphosphatemia/etiology , Inclusion Bodies/metabolism , Lafora Disease/complications , Lafora Disease/pathology , Muscle, Skeletal/pathology , Animals , Brain/metabolism , Cerebellar Cortex/pathology , Cerebellar Cortex/ultrastructure , Disease Models, Animal , Dual-Specificity Phosphatases/metabolism , Gene Expression Regulation/genetics , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/ultrastructure , Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...