Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Epileptic Disord ; 18(S2): 38-62, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27702709

RESUMO

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.


Assuntos
Doença de Lafora , Animais , Modelos Animais de Doenças , Aconselhamento Genético , Glicogênio/metabolismo , Humanos , Doença de Lafora/tratamento farmacológico , Doença de Lafora/genética , Doença de Lafora/metabolismo , Doença de Lafora/fisiopatologia , Camundongos , Proteínas Tirosina Fosfatases não Receptoras/genética
2.
J Biol Chem ; 288(48): 34627-37, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24142699

RESUMO

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.


Assuntos
Fosfatases de Especificidade Dupla/genética , Glicogênio Sintase/metabolismo , Glicogênio/biossíntese , Resistência à Insulina/genética , Obesidade/patologia , Envelhecimento/genética , Animais , Fosfatases de Especificidade Dupla/metabolismo , Glucose-6-Fosfato/metabolismo , Glicogênio/genética , Glicogênio Sintase/genética , Humanos , Insulina/genética , Insulina/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Obesidade/etiologia , Obesidade/genética , Fosforilação , Proteínas Tirosina Fosfatases não Receptoras
3.
Brain ; 135(Pt 9): 2684-98, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22961547

RESUMO

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.


Assuntos
Encéfalo/patologia , Proteínas de Transporte/genética , Doença de Lafora/genética , Músculo Esquelético/patologia , Proteínas Nucleares/genética , Adolescente , Adulto , Idade de Início , Atrofia , Criança , Pré-Escolar , Cromossomos Humanos Par 4 , Proteínas de Ligação a DNA , Progressão da Doença , Feminino , Histona Metiltransferases , Humanos , Doença de Lafora/patologia , Escore Lod , Masculino , Mutação , Pele/patologia
4.
J Biol Chem ; 287(30): 25650-9, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22669944

RESUMO

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.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Glicogênio/metabolismo , Doença de Lafora/enzimologia , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Adolescente , Animais , Células Cultivadas , Fosfatases de Especificidade Dupla/genética , Feminino , Glicogênio/genética , Humanos , Doença de Lafora/genética , Doença de Lafora/patologia , Masculino , Camundongos , Camundongos Knockout , Fosforilação/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica/genética , Proteínas Tirosina Fosfatases não Receptoras , Ubiquitina-Proteína Ligases/genética
5.
PLoS Genet ; 7(4): e1002037, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21552327

RESUMO

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.


Assuntos
Glucanos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Doença de Lafora/fisiopatologia , Animais , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Glucanos/análise , Glicogênio Sintase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Doença de Lafora/genética , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...