Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Viruses ; 13(2)2021 02 08.
Article in English | MEDLINE | ID: mdl-33567674

ABSTRACT

Nucleolin is an essential cellular receptor to human respiratory syncytial virus (RSV). Pharmacological targeting of the nucleolin RNA binding domain RBD1,2 can inhibit RSV infections in vitro and in vivo; however, the site(s) on RBD1,2 which interact with RSV are not known. We undertook a series of experiments designed to: document RSV-nucleolin co-localization on the surface of polarized MDCK cells using immunogold electron microscopy, to identify domains on nucleolin that physically interact with RSV using biochemical methods and determine their biological effects on RSV infection in vitro, and to carry out structural analysis toward informing future RSV drug development. Results of immunogold transmission and scanning electron microscopy showed RSV-nucleolin co-localization on the cell surface, as would be expected for a viral receptor. RSV, through its fusion protein (RSV-F), physically interacts with RBD1,2 and these interactions can be competitively inhibited by treatment with Palivizumab or recombinant RBD1,2. Treatment with synthetic peptides derived from two 12-mer domains of RBD1,2 inhibited RSV infection in vitro, with structural analysis suggesting these domains are potentially feasible for targeting in drug development. In conclusion, the identification and characterization of domains of nucleolin that interact with RSV provide the essential groundwork toward informing design of novel nucleolin-targeting compounds in RSV drug development.


Subject(s)
Phosphoproteins/metabolism , Protein Interaction Domains and Motifs/physiology , RNA-Binding Proteins/metabolism , Receptors, Virus/metabolism , Respiratory Syncytial Viruses/metabolism , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Humans , Immunohistochemistry , Madin Darby Canine Kidney Cells , Microscopy, Electron , Palivizumab/pharmacology , Nucleolin
2.
Epileptic Disord ; 18(S2): 38-62, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27702709

ABSTRACT

Lafora disease (LD) is an autosomal recessive progressive myoclonus epilepsy due to mutations in the EPM2A (laforin) and EPM2B (malin) genes, with no substantial genotype-phenotype differences between the two. Founder effects and recurrent mutations are common, and mostly isolated to specific ethnic groups and/or geographical locations. Pathologically, LD is characterized by distinctive polyglucosans, which are formations of abnormal glycogen. Polyglucosans, or Lafora bodies (LB) are typically found in the brain, periportal hepatocytes of the liver, skeletal and cardiac myocytes, and in the eccrine duct and apocrine myoepithelial cells of sweat glands. Mouse models of the disease and other naturally occurring animal models have similar pathology and phenotype. Hypotheses of LB formation remain controversial, with compelling evidence and caveats for each hypothesis. However, it is clear that the laforin and malin functions regulating glycogen structure are key. With the exception of a few missense mutations LD is clinically homogeneous, with onset in adolescence. Symptoms begin with seizures, and neurological decline follows soon after. The disease course is progressive and fatal, with death occurring within 10 years of onset. Antiepileptic drugs are mostly non-effective, with none having a major influence on the progression of cognitive and behavioral symptoms. Diagnosis and genetic counseling are important aspects of LD, and social support is essential in disease management. Future therapeutics for LD will revolve around the pathogenesics of the disease. Currently, efforts at identifying compounds or approaches to reduce brain glycogen synthesis appear to be highly promising.


Subject(s)
Lafora Disease , Animals , Disease Models, Animal , Genetic Counseling , Glycogen/metabolism , Humans , Lafora Disease/drug therapy , Lafora Disease/genetics , Lafora Disease/metabolism , Lafora Disease/physiopathology , Mice , Protein Tyrosine Phosphatases, Non-Receptor/genetics
4.
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
5.
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
6.
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
7.
Acta Neuropathol ; 125(3): 439-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23315026

ABSTRACT

X-linked Myopathy with Excessive Autophagy (XMEA) is a childhood onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p, VMA21 is an essential assembly chaperone of the vacuolar ATPase (V-ATPase), the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids which leads to downregulation of the mTORC1 pathway, and consequent increased macroautophagy resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge, and vacuolate the cell. Our results uncover a novel mechanism of disease, namely macroautophagic overcompensation leading to cell vacuolation and tissue atrophy.


Subject(s)
Adenosine Triphosphatases/metabolism , Autophagy/genetics , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/prevention & control , Muscular Diseases/genetics , Muscular Diseases/prevention & control , Vacuolar Proton-Translocating ATPases/deficiency , Vacuolar Proton-Translocating ATPases/genetics , Animals , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Leucine/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Lysosomes/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Muscular Diseases/pathology , Mutation/genetics , RNA Interference/physiology , RNA, Messenger/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Subcellular Fractions/metabolism , Subcellular Fractions/pathology , Time Factors , Vacuoles/metabolism
8.
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
9.
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
11.
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
12.
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
13.
Neuron ; 67(1): 100-15, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20624595

ABSTRACT

Neurotransmitter release depends critically on close spatial coupling of Ca(2+) entry to synaptic vesicles at the nerve terminal; however, the molecular substrates determining their physical proximity are unknown. Using the calyx of Held synapse, where "microdomain" coupling predominates at immature stages and developmentally switches to "nanodomain" coupling, we demonstrate that deletion of the filamentous protein Septin 5 imparts immature synapses with striking morphological and functional features reminiscent of mature synapses. This includes synaptic vesicles tightly localized to active zones, resistance to the slow Ca(2+) buffer EGTA and a reduced number of Ca(2+) channels required to trigger single fusion events. Disrupting Septin 5 organization acutely transforms microdomain to nanodomain coupling and potentiates quantal output in immature wild-type terminals. These observations suggest that Septin 5 is a core molecular substrate that differentiates distinct release modalities at the central synapse.


Subject(s)
Calcium/metabolism , Membrane Microdomains/metabolism , Neurotransmitter Agents/metabolism , Selenoproteins/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Age Factors , Animals , Animals, Newborn , Antibodies/pharmacology , Brain Stem/cytology , Brain Stem/growth & development , CHO Cells , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Chelating Agents/pharmacology , Cricetinae , Cricetulus , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GTP-Binding Protein Regulators/genetics , In Vitro Techniques , Membrane Microdomains/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Models, Biological , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Selenoproteins/deficiency , Selenoproteins/immunology , Synapses/ultrastructure , Synaptic Transmission/genetics , Vesicular Glutamate Transport Protein 1/metabolism
14.
Ann Neurol ; 67(6): 802-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20517942

ABSTRACT

OBJECTIVE: Individuals with cystic fibrosis (CF) have exercise intolerance and skeletal muscle weakness not solely attributable to physical inactivity or pulmonary function abnormalities. CF transmembrane conductance regulator (CFTR) has been demonstrated in human bronchial smooth and cardiac muscle. Using (31)P-magnetic resonance spectroscopy of skeletal muscle, we showed CF patients to have lower resting muscle adenosine triphosphate and delayed phosphocreatine recovery times after high-intensity exercise, suggesting abnormal muscle aerobic metabolism; and higher end-exercise pH values, suggesting altered bicarbonate transport. Our objective was to study CFTR expression in human skeletal muscle. METHODS AND RESULTS: We studied CFTR expression in human skeletal muscle by Western blot with anti-CFTR antibody (Ab) L12B4 and demonstrated a single band with expected molecular weight of 168kDa. We isolated the cDNA by reverse transcription polymerase chain reaction and directly sequenced a 975bp segment (c. 3,600-4,575) that was identical to the human CFTR sequence. We showed punctate staining of CFTR in sarcoplasm and sarcolemma by immunofluorescence microscopy with L12B4 Ab and secondary Alexa 488-labeled Ab. We confirmed CFTR expression in the sarcotubular network and sarcolemma by electron microscopy, using immunogold-labeled anti-CFTR Ab. We observed activation of CFTR Cl(-) channels with iodide efflux, on addition of forskolin, 3-isobutyl-1-methyl-xanthine, and 8-chlorphenylthio-cyclic adenosine monophosphate, in wild-type C57BL/6J isolated muscle fibers in contrast to no efflux from mutant F508del-CFTR muscle. INTERPRETATION: We speculate that a defect in sarcoplasmic reticulum CFTR Cl(-) channels could alter the electrochemical gradient, causing dysregulation of Ca(2+) homeostasis, for example, ryanodine receptor or sarco(endo)plasmic reticulum Ca(2+) adenosine triphosphatases essential to excitation-contraction coupling leading to exercise intolerance and muscle weakness in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis/physiopathology , Exercise/physiology , Muscle, Skeletal/metabolism , Animals , Colforsin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Immunoelectron/methods , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Mutation/genetics , Phosphodiesterase Inhibitors/pharmacology , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Subcellular Fractions/metabolism
15.
Brain Pathol ; 19(4): 727-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19744044

ABSTRACT

A 22-year-old girl presented with convulsive status epilepticus and a previous history of recurrent seizures, myoclonus, ataxia and impaired cognitive functions. Neurological examination revealed rest and action-induced myoclonus, pyramidal signs and opposition hypertonia. Testing revealed severe metabolic acidosis, elevated transaminases and creatine kinase, and respiratory insufficiency. After intubation and ventilation, thiopental was introduced but the patient's condition worsened dramatically with death in a few hours. Autopsy showed profuse periodic acid-Schiff (PAS) positive intracellular inclusions in the CNS (Lafora bodies), most abundant in thalamus, cerebellum, and brainstem, as well as in other organs. Genetic testing revealed a homozygous missense mutation (c.205C > G, P69A) in the EPM2B (NHLRC1) gene, confirming the diagnosis of progressive myoclonic epilepsy Lafora-type.


Subject(s)
Brain/pathology , Lafora Disease/diagnosis , Lafora Disease/pathology , Seizures/pathology , Status Epilepticus/pathology , Adolescent , Atrophy , Carrier Proteins/genetics , Dendrites/pathology , Diagnosis, Differential , Fatal Outcome , Female , Humans , Inclusion Bodies/pathology , Lafora Disease/genetics , Microscopy, Electron , Mutation , Seizures/genetics , Staining and Labeling , Status Epilepticus/genetics , Ubiquitin-Protein Ligases , Young Adult
16.
Proc Natl Acad Sci U S A ; 106(33): 14085-90, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19666602

ABSTRACT

In a mouse mutagenesis screen, we isolated a mutant, Myshkin (Myk), with autosomal dominant complex partial and secondarily generalized seizures, a greatly reduced threshold for hippocampal seizures in vitro, posttetanic hyperexcitability of the CA3-CA1 hippocampal pathway, and neuronal degeneration in the hippocampus. Positional cloning and functional analysis revealed that Myk/+ mice carry a mutation (I810N) which renders the normally expressed Na(+),K(+)-ATPase alpha3 isoform inactive. Total Na(+),K(+)-ATPase activity was reduced by 42% in Myk/+ brain. The epilepsy in Myk/+ mice and in vitro hyperexcitability could be prevented by delivery of additional copies of wild-type Na(+),K(+)-ATPase alpha3 by transgenesis, which also rescued Na(+),K(+)-ATPase activity. Our findings reveal the functional significance of the Na(+),K(+)-ATPase alpha3 isoform in the control of epileptiform activity and seizure behavior.


Subject(s)
Central Nervous System/metabolism , Mutation , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Seizures/genetics , Seizures/pathology , Sequence Homology, Nucleic Acid , Sodium-Potassium-Exchanging ATPase/genetics
17.
Cell ; 137(2): 235-46, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19379691

ABSTRACT

X-linked myopathy with excessive autophagy (XMEA) is a childhood-onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p it is an essential assembly chaperone of the V-ATPase, the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH, which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids, which upregulates the mTOR pathway and mTOR-dependent macroautophagy, resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge together, and vacuolate the cell. Our results uncover macroautophagic overcompensation leading to cell vacuolation and tissue atrophy as a mechanism of disease.


Subject(s)
Genes, X-Linked , Muscular Diseases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Autophagy , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Vacuolar Proton-Translocating ATPases/genetics
18.
Dis Model Mech ; 1(4-5): 229-40, 2008.
Article in English | MEDLINE | ID: mdl-19093029

ABSTRACT

Demyelination in the central nervous system is the hallmark feature in multiple sclerosis (MS). The mechanism resulting in destabilization of myelin is a complex multi-faceted process, part of which involves deimination of myelin basic protein (MBP). Deimination, the conversion of protein-bound arginine to citrulline, is mediated by the peptidylarginine deiminase (PAD) family of enzymes, of which the PAD2 and PAD4 isoforms are present in myelin. To test the hypothesis that PAD contributes to destabilization of myelin in MS, we developed a transgenic mouse line (PD2) containing multiple copies of the cDNA encoding PAD2, under the control of the MBP promoter. Using previously established criteria, clinical signs were more severe in PD2 mice than in their normal littermates. The increase in PAD2 expression and activity in white matter was demonstrated by immunohistochemistry, reverse transcriptase-PCR, enzyme activity assays, and increased deimination of MBP. Light and electron microscopy revealed more severe focal demyelination and thinner myelin in the PD2 homozygous mice compared with heterozygous PD2 mice. Quantitation of the disease-associated molecules GFAP and CD68, as measured by immunoslot blots, were indicative of astrocytosis and macrophage activation. Concurrently, elevated levels of the pro-inflammatory cytokine TNF-alpha and nuclear histone deimination support initiation of demyelination by increased PAD activity. These data support the hypothesis that elevated PAD levels in white matter represents an early change that precedes demyelination.


Subject(s)
Central Nervous System/pathology , Hydrolases/metabolism , Myelin Sheath/pathology , Animals , Hydrolases/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Phenotype , Protein-Arginine Deiminases , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Nat Clin Pract Neurol ; 4(2): 106-11, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18256682

ABSTRACT

BACKGROUND: A 20-year-old woman presented to a specialist epilepsy center with a 3-year history of drug-resistant epileptic seizures, progressive myoclonus, ataxia, and cognitive decline. INVESTIGATIONS: Neurological examination, neuropsychological testing, electrophysiological studies, skin biopsy, MRI, genetic testing, and autopsy. DIAGNOSIS: Lafora disease (EPM2), resulting from a homozygous missense mutation in EPM2B (NHLRC1; c205C>G; Pro69Ala). MANAGEMENT: Symptomatic treatment with conventional antiepileptic and antimyoclonic drugs.


Subject(s)
Lafora Disease/pathology , Lafora Disease/physiopathology , Adult , Disease Progression , Electroencephalography , Fatal Outcome , Female , Humans , Lafora Disease/genetics
20.
J Child Neurol ; 23(2): 240-2, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18263761

ABSTRACT

Lafora epilepsy is characterized by starch formation in brain and skin and is diagnosed by skin biopsy or mutation detection. It has variable ages of onset (6-19 years) and death (18-32 years) even with the same mutation, likely due to extramutational factors. The authors identified 14 Lafora epilepsy patients in the genetic isolate of tribal Oman. The authors show that in this homogeneous environment and gene pool, the same mutation, EPM2B-c.468-469delAG, results in highly uniform ages of onset (14 years) and death (21 years). Biopsy, on the other hand, was not homogeneous (positive in 4/5 patients) and is, therefore, less sensitive than mutation testing.


Subject(s)
Carrier Proteins/genetics , Lafora Disease/genetics , Population Groups/genetics , Adolescent , Adult , Age of Onset , Carrier Proteins/metabolism , Consanguinity , DNA Mutational Analysis , Death , Humans , Lafora Disease/ethnology , Lafora Disease/mortality , Lafora Disease/physiopathology , Oman , Skin/metabolism , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...