Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
JCI Insight ; 9(12)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38912588

RESUMO

BackgroundGlycogen storage disease type IV (GSD IV) is an ultrarare autosomal recessive disorder that causes deficiency of functional glycogen branching enzyme and formation of abnormally structured glycogen termed polyglucosan. GSD IV has traditionally been categorized based on primary hepatic or neuromuscular involvement, with hepatic GSD IV subclassified as discrete subtypes: classic (progressive) and nonprogressive.MethodsTo better understand the progression of liver disease in GSD IV, we present clinical and histopathology data from 23 patients from around the world and characterized the liver involvement in the Gbe1ys/ys knockin mouse model.ResultsWe propose an alternative to the established subtype-based terminology for characterizing liver disease in GSD IV and recognize 3 tiers of disease severity: (i) "severe progressive" liver disease, (ii) "intermediate progressive" liver disease, and (iii) "attenuated" liver disease. Analysis of liver pathology revealed that risk for liver failure cannot be predicted from liver biopsy findings alone in individuals affected by GSD IV. Moreover, analysis of postmortem liver pathology from an individual who died over 40 years after being diagnosed with nonprogressive hepatic GSD IV in childhood verified that liver fibrosis did not regress. Last, characterization of the liver involvement in a mouse model known to recapitulate the adult-onset neurodegenerative form of GSD IV (Gbe1ys/ys mouse model) demonstrated hepatic disease.ConclusionOur findings challenge the established subtype-based view of GSD IV and suggest that liver disease severity among patients with GSD IV represents a disease continuum.Trial registrationClinicalTrials.gov NCT02683512FundingNone.


Assuntos
Modelos Animais de Doenças , Doença de Depósito de Glicogênio Tipo IV , Fígado , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem , Progressão da Doença , Sistema da Enzima Desramificadora do Glicogênio/genética , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/patologia , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Fígado/patologia , Fígado/metabolismo , Hepatopatias/patologia , Hepatopatias/metabolismo
2.
Arch Biochem Biophys ; 695: 108626, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33049291

RESUMO

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


Assuntos
Envelhecimento/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/deficiência , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Fígado/enzimologia , Estresse Oxidativo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Glicogênio/genética , Glicogênio/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/patologia , Fígado/patologia , Camundongos , Camundongos Knockout , Carbonilação Proteica/genética
3.
Virchows Arch ; 475(6): 671-686, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31363843

RESUMO

An abnormal structural form of glycogen (with less branching points or amylopectin-like polysaccharide) called polyglucosan (PG) may accumulate in various tissues such as striated and smooth muscles, brain, nerve, liver and skin, and cause a group of nine different genetic disorders manifesting with a variety of clinical phenotypes that affect mainly the nervous system (Lafora disease, adult PG body disease), the heart (glycogen storage disease type XV, hypertrophic cardiomyopathy type 6, PG body myopathy type 1) and the skeletal muscle (glycogen storage disease type IV, glycogen storage disease type VII, PG body myopathy type 2), depending on the organs which are mostly affected by the PG aggregates. The pathological feature of PG storage in tissues is a hallmark of these disorders. Whole-genome sequencing has allowed to obtain a diagnosis in a large number of patients with a previously unrecognized disorder. We describe the clinical, pathological and molecular features of these genetic disorders, for many of which the pathological mechanisms underlying the corresponding mutant gene have been investigated and, at least in part, understood.


Assuntos
Glucanos/metabolismo , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio/metabolismo , Doenças do Sistema Nervoso/metabolismo , Polissacarídeos/metabolismo , Animais , Humanos , Fígado/metabolismo , Fígado/patologia , Músculo Esquelético/patologia
4.
Pediatr Dev Pathol ; 21(4): 423-427, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28497716

RESUMO

The fatal infantile neuromuscular type is the most severe form of glycogen storage disease type IV (GSD IV). We report a case of a 22-day-old female neonate born at 34 weeks gestation with polyhyramnios, fetal hydrops, and severe hypotonia. Placental examination revealed numerous periodic acid schiff-positive diastase-resistant polyglucosan bodies in the cytoplasm of extravillous trophoblast predominantly in the placental basal plate. Muscle biopsy and autopsy findings supported a diagnosis of neuromuscular-type glycogen storage disease type IV with extensive involvement of skeletal muscle, heart, and liver. The diagnosis was confirmed by molecular genetic testing. We could only find 1 prior report in the English literature that describes placental pathological changes. Our findings suggest that placental examination can be a useful adjunct for early diagnosis, as placentas are often received for pathological examination shortly after birth and usually before a diagnostic muscle biopsy can be performed. Pathologists need to be aware of characteristic placental features.


Assuntos
Glucanos/metabolismo , Doença de Depósito de Glicogênio Tipo IV/diagnóstico , Placenta/patologia , Trofoblastos/patologia , Biomarcadores/metabolismo , Evolução Fatal , Feminino , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/patologia , Humanos , Recém-Nascido , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo
5.
Hum Mol Genet ; 24(23): 6801-10, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26385640

RESUMO

Glycogen storage disease type IV (GSD IV) is a rare autosomal recessive disorder caused by deficiency of the glycogen-branching enzyme (GBE). The diagnostic hallmark of the disease is the accumulation of a poorly branched form of glycogen known as polyglucosan (PG). The disease is clinically heterogeneous, with variable tissue involvement and age at onset. Complete loss of enzyme activity is lethal in utero or in infancy and affects primarily the muscle and the liver. However, residual enzyme activity as low as 5-20% leads to juvenile or adult onset of a disorder that primarily affects the central and peripheral nervous system and muscles and in the latter is termed adult polyglucosan body disease (APBD). Here, we describe a mouse model of GSD IV that reflects this spectrum of disease. Homologous recombination was used to knock in the most common GBE1 mutation p.Y329S c.986A > C found in APBD patients of Ashkenazi Jewish decent. Mice homozygous for this allele (Gbe1(ys/ys)) exhibit a phenotype similar to APBD, with widespread accumulation of PG. Adult mice exhibit progressive neuromuscular dysfunction and die prematurely. While the onset of symptoms is limited to adult mice, PG accumulates in tissues of newborn mice but is initially absent from the cerebral cortex and heart muscle. Thus, PG is well tolerated in most tissues, but the eventual accumulation in neurons and their axons causes neuropathy that leads to hind limb spasticity and premature death. This mouse model mimics the pathology and pathophysiologic features of human adult-onset branching enzyme deficiency.


Assuntos
Modelos Animais de Doenças , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Mutação , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Técnicas de Introdução de Genes , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/fisiopatologia , Camundongos , Músculo Estriado/metabolismo , Músculo Estriado/fisiopatologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/fisiopatologia , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/fisiopatologia , Fenótipo
6.
Hum Mol Genet ; 24(2): 471-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25205110

RESUMO

Andersen's syndrome (AS) is a rare and dominantly inherited pathology, linked to the inwardly rectifying potassium channel Kir2.1. AS patients exhibit a triad of symptoms that include periodic paralysis, cardiac dysrhythmia and bone malformations. Some progress has been made in understanding the contribution of the Kir2.1 channel to skeletal and cardiac muscle dysfunctions, but its role in bone morphogenesis remains unclear. We isolated myoblast precursors from muscle biopsies of healthy individuals and typical AS patients with dysmorphic features. Myoblast cultures underwent osteogenic differentiation that led to extracellular matrix mineralization. Osteoblastogenesis was monitored through the activity of alkaline phosphatase, and through the hydroxyapatite formation using Alizarin Red and Von Kossa staining techniques. Patch-clamp recordings revealed the presence of an inwardly rectifying current in healthy cells that was absent in AS osteoblasts, showing the dominant-negative effect of the Kir2.1 mutant allele in osteoblasts. We also found that while control cells actively synthesize hydroxyapatite, AS osteoblasts are unable to efficiently form any extracellular matrix. To further demonstrate the role of the Kir2.1 channels during the osteogenesis, we inhibited Kir2.1 channel activity in healthy patient cells by applying extracellular Ba(2+) or using adenoviruses carrying mutant Kir2.1 channels. In both cases, cells were no longer able to produce extracellular matrixes. Moreover, osteogenic activity of AS osteoblasts was restored by rescue experiments, via wild-type Kir2.1 channel overexpression. These observations provide a proof that normal Kir2.1 channel function is essential during osteoblastogenesis.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/metabolismo , Mioblastos/metabolismo , Osteogênese , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/fisiopatologia , Humanos , Mioblastos/citologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética
7.
Fortschr Neurol Psychiatr ; 79(10): 598-605; quiz 606, 2011 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-21989512
8.
Hum Mol Genet ; 20(22): 4430-9, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21856731

RESUMO

Glycogen storage disease type IV (GSD IV) is a rare autosomal recessive disorder caused by deficiency of the glycogen branching enzyme (GBE). The diagnostic feature of the disease is the accumulation of a poorly branched form of glycogen known as polyglucosan (PG). The disease is clinically heterogeneous, with variable tissue involvement and age of disease onset. Absence of enzyme activity is lethal in utero or in infancy affecting primarily muscle and liver. However, residual enzyme activity (5-20%) leads to juvenile or adult onset of a disorder that primarily affects muscle as well as central and peripheral nervous system. Here, we describe two mouse models of GSD IV that reflect this spectrum of disease. Homologous recombination was used to insert flippase recognition target recombination sites around exon 7 of the Gbe1 gene and a phosphoglycerate kinase-Neomycin cassette within intron 7, leading to a reduced synthesis of GBE. Mice bearing this mutation (Gbe1(neo/neo)) exhibit a phenotype similar to juvenile onset GSD IV, with wide spread accumulation of PG. Meanwhile, FLPe-mediated homozygous deletion of exon 7 completely eliminated GBE activity (Gbe1(-/-)), leading to a phenotype of lethal early onset GSD IV, with significant in utero accumulation of PG. Adult mice with residual GBE exhibit progressive neuromuscular dysfunction and die prematurely. Differently from muscle, PG in liver is a degradable source of glucose and readily depleted by fasting, emphasizing that there are structural and regulatory differences in glycogen metabolism among tissues. Both mouse models recapitulate typical histological and physiological features of two human variants of branching enzyme deficiency.


Assuntos
Enzima Ramificadora de 1,4-alfa-Glucana/genética , Doença de Depósito de Glicogênio Tipo IV/genética , Animais , Modelos Animais de Doenças , Glucanos , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Camundongos
9.
Hum Mol Genet ; 20(3): 455-65, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21075835

RESUMO

Glycogen storage disease type IV (GSD-IV) is an autosomal recessive disease caused by a deficiency in glycogen-branching enzyme (GBE1) activity that results in the accumulation of amylopectin-like polysaccharide, which presumably leads to osmotic swelling and cell death. This disease is extremely heterogeneous in terms of tissue involvement, age of onset and clinical manifestation. The most severe fetal form presents as hydrops fetalis; however, its pathogenetic mechanisms are largely unknown. In this study, mice carrying a stop codon mutation (E609X) in the Gbe1 gene were generated using a gene-driven mutagenesis approach. Homozygous mutants (Gbe(-/-) mice) recapitulated the clinical features of hydrops fetalis and the embryonic lethality of the severe fetal form of GSD-IV. However, contrary to conventional expectations, little amylopectin accumulation and no cell degeneration were found in Gbe(-/-) embryonic tissues. Glycogen accumulation was reduced in developing hearts of Gbe(-/-)embryos, and abnormal cardiac development, including hypertrabeculation and noncompaction of the ventricular wall, was observed. Further, Gbe1 ablation led to poor ventricular function in late gestation and ultimately caused heart failure, fetal hydrops and embryonic lethality. We also found that the cell-cycle regulators cyclin D1 and c-Myc were highly expressed in cardiomyocytes and likely contributed to cardiomyocyte proliferation and trabeculation/compaction of the ventricular wall. Our results reveal that early molecular events associated with Gbe1 deficiency contribute to abnormal cardiac development and fetal hydrops in the fetal form of GSD-IV.


Assuntos
Enzima Ramificadora de 1,4-alfa-Glucana/deficiência , Enzima Ramificadora de 1,4-alfa-Glucana/genética , Doença de Depósito de Glicogênio Tipo IV/genética , Glicogênio/metabolismo , Cardiopatias Congênitas/genética , Coração/embriologia , Amilopectina/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células , Códon de Terminação , Ciclina D1/genética , Perda do Embrião , Imunofluorescência , Genes myc , Doença de Depósito de Glicogênio Tipo IV/embriologia , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Cardiopatias Congênitas/metabolismo , Insuficiência Cardíaca , Frequência Cardíaca , Hidropisia Fetal , Camundongos , Miócitos Cardíacos , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Função Ventricular
10.
Folia Neuropathol ; 46(3): 165-75, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18825592

RESUMO

Glycogenosis type IV is caused by a deficiency of glycogen branching enzyme (alpha-1,4 glucan 6-transglucosylase). Adult polyglucosan body disease (APBD) may represent a neuropathological hallmark of the adult form of this storage disease of the central nervous system. We analysed a case of a 45-year-old unconscious woman who died three days after admission to the hospital. Neuropathological examination revealed massive accumulation of polyglucosan bodies (PBs) in the cortex and white matter of the whole brain. PBs were located in the processes of neurons, astrocytes and microglial cells. The storage material in the cytoplasm of neurons and glial cells was visible as fine granules. Ultrastructurally, PBs consisted of non-membrane-bound deposits of branched and densely packed filaments, measuring about 7-10 nm in diameter, typical of polyglucosan bodies. APBD patients develop upper and lower neuron disease and dementia, probably secondary to the disruption of neuron and astrocyte functions.


Assuntos
Encéfalo/metabolismo , Encéfalo/ultraestrutura , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/patologia , Feminino , Glucanos/metabolismo , Humanos , Imuno-Histoquímica , Corpos de Inclusão/metabolismo , Corpos de Inclusão/ultraestrutura , Pessoa de Meia-Idade
11.
Mol Genet Metab ; 95(1-2): 52-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18691923

RESUMO

We describe a 15-year-old boy and his 19-year-old sister with progressive dilated cardiomyopathy and mild non-progressive proximal lower limb myopathy, secondary to the accumulation of amylopectin-like fibrillar glycogen, (polyglucosan) bodies, in heart and skeletal muscle. Evidence of idiopathic amylopectinosis or polysaccharidosis was demonstrated in heart and skeletal muscle tissue by histology, electron microscopy, biochemical, and genetic analysis. In both siblings the heart muscle stored PAS-positive, proteinase-k resistant and partly diastase resistant granulo-filamentous material, simulating polyglucosan bodies. Glycogen branching enzyme activity, and phosphofructokinase enzyme activity, measured in skeletal muscle tissue and explanted heart tissue were all within the normal limits, however glycogen content was elevated. Furthermore, GBE1, PRKAG2, desmin, alphabeta-crystallin, ZASP, myotilin, and LAMP-2 gene sequencing revealed no mutation, excluding e.g. glycogen storage disease type 4 and desmin-related myofibrillar cardiomyopathies. In both patients the diagnosis of an idiopathic polysaccharidosis with progressive dilated cardiomyopathy was made, requiring heart transplantation at age 13 and 14, respectively. Both patients belong to an autosomal recessive group of biochemically and genetically unclassified severe vacuolar glycogen storage disease of the heart and skeletal muscle. Up to now unidentified glycogen synthesis or glycogen degradation pathways are supposed to contribute to this idiopathic glycogen storage disease.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/diagnóstico , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Adolescente , Adulto , Feminino , Alemanha , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/enzimologia , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/patologia , Humanos , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Miocárdio/enzimologia , Miocárdio/patologia , Linhagem , Irmãos
12.
Muscle Nerve ; 37(4): 530-6, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17994551

RESUMO

Adult polyglucosan body disease (APBD) is characterized by the accumulation of insoluble glucose polymers within the central and peripheral nervous systems. A common missense mutation in the glycogen branching enzyme (GBE1) gene has been identified in Ashkenazi patients with APBD. We report on a non-Jewish patient with APBD on whom we performed proton magnetic resonance spectroscopic imaging of the brain. GBE activity in fibroblasts was markedly reduced, and a novel heterozygous mutation was identified in the GBE1 gene. Our findings widen the spectrum of APBD genotypes, underline the importance of performing GBE analysis in all APBD patients, and suggest that brain white matter degeneration in APBD may result from tissue damage involving axons and myelin.


Assuntos
Enzima Ramificadora de 1,4-alfa-Glucana/genética , Glucanos/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Espectroscopia de Ressonância Magnética , Doenças do Sistema Nervoso/genética , Adulto , Encéfalo/metabolismo , Encéfalo/patologia , Eletromiografia , Saúde da Família , Feminino , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/patologia , Humanos , Mutação de Sentido Incorreto , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Prótons
13.
Mol Genet Metab ; 90(4): 383-92, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17257876

RESUMO

Deficiency of glycogen branching enzyme (GBE) activity causes glycogen storage disease type IV (GSD IV), an autosomal recessive error of metabolism. Abnormal glycogen accumulates in myocytes, hepatocytes, and neurons, causing variably progressive, benign to lethal organ dysfunctions. A naturally occurring orthologue of human GSD IV was described previously in Norwegian forest cats (NFC). Here, we report that while most affected kittens die at or soon after birth, presumably due to hypoglycemia, survivors of the perinatal period appear clinically normal until onset of progressive neuromuscular degeneration at 5 months of age. Molecular investigation of affected cats revealed abnormally spliced GBE1 mRNA products and lack of GBE cross-reactive material in liver and muscle. Affected cats are homozygous for a complex rearrangement of genomic DNA in GBE1, constituted by a 334 bp insertion at the site of a 6.2 kb deletion that extends from intron 11 to intron 12 (g. IVS11+1552_IVS12-1339 del6.2kb ins334 bp), removing exon 12. An allele-specific, PCR-based test demonstrates that the rearrangement segregates with the disease in the GSD IV kindred and is not found in unrelated normal cats. Screening of 402 privately owned NFC revealed 58 carriers and 4 affected cats. The molecular characterization of feline GSD IV will enhance further studies of GSD IV pathophysiology and development of novel therapies in this unique animal model.


Assuntos
Enzima Ramificadora de 1,4-alfa-Glucana/genética , Processamento Alternativo , Doença de Depósito de Glicogênio Tipo IV/genética , Animais , Sequência de Bases , Cruzamento , Gatos , Éxons , Feminino , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/fisiopatologia , Hipoglicemia/genética , Hipoglicemia/metabolismo , Hipoglicemia/fisiopatologia , Fígado/metabolismo , Masculino , Dados de Sequência Molecular , Mutação , Doenças Neuromusculares/genética , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/fisiopatologia , Linhagem
14.
Curr Opin Lipidol ; 15(2): 191-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15017362

RESUMO

PURPOSE OF REVIEW: The transport of lipoproteins through the secretory pathways of enterocytes and hepatocytes is pivotal for whole-body lipid homeostasis. This review focuses on the assembly and structural evolution of COPII (coat protein) transport carriers that are essential for the transport of chylomicrons from the endoplasmic reticulum to the Golgi apparatus. RECENT FINDINGS: The assembly of endoplasmic reticulum to Golgi transport carriers commences with the coating of specific areas of the endoplasmic reticulum membrane with Sar1-GTP and the Sec23/24 heterodimer. An important advance has been the crystallographic analysis of the Sar1-Sec23/24 complex. The proteins form a bow-tie shaped structure, with a concave face that seems to match the curvature of transport carriers. Mammalian cells produce two isoforms of Sar1, designated Sar1a and Sar1b, both of which are expressed in enterocytes. Sar1b is defective in chylomicron retention disease and Anderson disease, two rare recessive disorders characterized by severe fat malabsorption and a failure to thrive in infancy. Patients with chylomicron retention disease and Anderson disease selectively retain chylomicron-like particles within membrane-bound compartments. By analogy with procollagen, chylomicrons may drive the formation of endoplasmic reticulum to Golgi transport carriers from endoplasmic reticulum sites close to, but separate from, domains of the endoplasmic reticulum coated with Sar1-Sec23/24. The COPII machinery also mediates the transport of VLDL to the Golgi. SUMMARY: New insights into the role of the COPII machinery in the intracellular transport of cargo, including chylomicrons and VLDL, may suggest new drug targets for ameliorating the lipid abnormalities of the metabolic syndrome.


Assuntos
Quilomícrons/metabolismo , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Sequência de Aminoácidos , Animais , Transporte Biológico Ativo , Proteínas de Transporte , Retículo Endoplasmático/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Humanos , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/química , Proteínas Monoméricas de Ligação ao GTP/genética , Mutação , Fosfoproteínas , Proteínas de Saccharomyces cerevisiae , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/metabolismo , Proteínas de Transporte Vesicular
15.
Neuromuscul Disord ; 14(4): 253-60, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15019703

RESUMO

Glycogen storage disease type IV or Andersen disease is an autosomal recessive disorder due to deficiency of glycogen branching enzyme. Typically, glycogen storage disease type IV presents with rapidly progressive liver cirrhosis and death in childhood. Variants include a cardiopathic form of childhood, a relatively benign myopathic form of young adults, and a late-onset neurodegenerative disorder (adult polyglucosan body disease). A severe neuromuscular variant resembling Werdnig-Hoffmann disease has also been described in two patients. The objective was to describe two additional infants with the neuromuscular variant and novel mutations in the GBE1 gene. Branching enzyme assay, Western blot, RT-PCR and sequencing were performed in muscle biopsies from both patients. The cDNA of patient 1 was subcloned and sequenced to define the mutations. Muscle biopsies showed accumulation of periodic acid Schiff-positive, diastase-resistant storage material in both patients and increased lysosomal enzyme activity in patient 1. Branching enzyme activity in muscle was negligible in both patients, and Western blot showed decreased branching enzyme protein. Patient 1 had two single base pair deletions, one in exon 10 (1238delT) and the other in exon 12 (1467delC), and each parent was heterozygous for one of the deletions. Patient 2 had a large homozygous deletion that spanned 627 bp and included exons 8-12. Patient 1, who died at 41 days, had neurophysiological and neuropathological features of Spinal Muscular Atrophy. Patient 2, who died at 5(1/2) weeks, had a predominantly myopathic process. The infantile neuromuscular form of glycogen storage disease type IV is considered extremely rare, but our encountering two patients in close succession suggests that the disease may be underdiagnosed.


Assuntos
Doença de Depósito de Glicogênio Tipo IV , Músculo Esquelético/patologia , Doenças Neuromusculares , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Ácido Aminossalicílico/metabolismo , Biópsia/métodos , Western Blotting/métodos , Tronco Encefálico/enzimologia , Tronco Encefálico/patologia , Análise Mutacional de DNA/métodos , Éxons , Feminino , Doença de Depósito de Glicogênio Tipo IV/complicações , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Humanos , Lactente , Oxirredutases Intramoleculares , Lisossomos/enzimologia , Microscopia Eletrônica/métodos , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mutação , Doenças Neuromusculares/enzimologia , Doenças Neuromusculares/patologia , Prostaglandina-E Sintases , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Atrofias Musculares Espinais da Infância/complicações , Atrofias Musculares Espinais da Infância/patologia , Transativadores/genética
16.
J Inherit Metab Dis ; 27(5): 609-19, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15669676

RESUMO

Glycogenosis type IV is an autosomal recessive disease, exceptionally diagnosed at birth: only very few reports of the fatal perinatal neuromuscular form have been described. We report on two sibling male newborns who died at 10 and 4 weeks of age with clinical signs of a systemic storage disease. Prenatal history included polyhydramnios, reduced fetal movements and fetal hydrops, and Caesarean section was performed at 36 weeks of gestational age because of fetal distress. At birth, both babies showed severe hypotonia, hyporeflexia and no spontaneous breathing activity. They never showed active movements, sucking and swallowing and were respirator-dependent until death. A muscle biopsy revealed, in both patients, the presence of PAS-positive and partially diastase-resistant cytoplasmic inclusions containing granular and filamentous amylopectin-like material. This suggested that the stored material consisted of abnormal glycogen. At autopsy, ultrastructural examination of cardiac and skeletal muscle, liver, kidney and brain showed PAS-positive diastase-resistant eosinophilic cytoplasmic inclusions. Determination of branching enzyme activity, in cultured fibroblasts from the second patient, showed markedly reduced enzyme activity, confirming diagnosis of glycogenosis type IV. Our patients showed the full spectrum of both prenatal signs (hydrops, polyhydramnios) and postnatal signs (hypotonia, hyporeflexia, absence of active movements, cardiomegaly), which have been reported previously. They suffered from a very severe form of glycogenosis type IV with clinical and histological involvement of many tissues and organs. Diagnosis was accomplished on the second baby and required several biochemical and histological studies, in order to rule out both neuromuscular disorders and the most common storage diseases with neonatal onset. In our experience, the correct interpretation of the histological findings was essential in the search for the diagnosis.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/diagnóstico , Doença de Depósito de Glicogênio Tipo IV/genética , Idade de Início , Autopsia , Sistema Nervoso Central/metabolismo , Citoplasma/metabolismo , Saúde da Família , Evolução Fatal , Genes Recessivos , Glicogênio/sangue , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Músculo Esquelético/patologia , Distribuição Tecidual
17.
J Mol Cell Cardiol ; 35(4): 409-15, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12689820

RESUMO

Andersen's syndrome (AS) (which is characterized by periodic paralysis, cardiac arrhythmias and dysmorphic features), a hereditary disease, and missense mutations of KCNJ2 (which encodes an inward rectifying potassium channel) have been reported recently. We performed clinical and molecular analyses of a patient with AS, and found a novel mutation (G215D) of KCNJ2. Twelve-lead electrocardiography revealed a long QT interval and frequent premature ventricular contractions, and polymorphic ventricular tachycardia was induced by programmed electrical stimulation. Use of a conventional whole-cell patch-clamp system with COS7 cells demonstrated that the G215D mutant was non-functional, and that co-expression of wild type (WT)- and mutant-KCNJ2 shows a dominant negative effect on both inward and outward currents. We performed confocal laser scanning microscopy to assess the cellular trafficking of WT- and mutant-KCNJ2 subunits tagged with yellow fluorescent protein (YFP) and cyan fluorescent protein (CFP), respectively. Tagging with the YFP did not affect the channel function of WT-KCNJ2 and both proteins showed similar plasma membrane fluorescence patterns. Furthermore, the result of fluorescence resonance energy transfer (FRET) studies at the plasma membrane region suggested that both YFP-tagged WT- and CFP-tagged mutant-KCNJ2 combine to construct a hetero-multimer of the potassium channel. In conclusion, the G215D mutant of KCNJ2 is distributed normally in the plasma membrane, but exhibits a dominant-negative effect and reduces the Kir2.1 current, presumably due to hetero-multimer construction.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adulto , Sequência de Aminoácidos , Animais , Células COS , Membrana Celular/metabolismo , Análise Mutacional de DNA , Feminino , Transferência Ressonante de Energia de Fluorescência , Técnicas de Transferência de Genes , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Humanos , Mutação de Sentido Incorreto , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
18.
Neuron ; 34(6): 933-44, 2002 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12086641

RESUMO

Inwardly rectifying K(+) (Kir) channels are important regulators of resting membrane potential and cell excitability. The activity of Kir channels is critically dependent on the integrity of channel interactions with phosphatidylinositol 4,5-bisphosphate (PIP(2)). Here we identify and characterize channel-PIP(2) interactions that are conserved among Kir family members. We find basic residues that interact with PIP(2), two of which have been associated with Andersen's and Bartter's syndromes. We show that several naturally occurring mutants decrease channel-PIP(2) interactions, leading to disease.


Assuntos
Sequência Conservada/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Sequência de Aminoácidos/genética , Animais , Síndrome de Bartter/genética , Síndrome de Bartter/metabolismo , Feminino , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Ativação do Canal Iônico/genética , Potenciais da Membrana/genética , Dados de Sequência Molecular , Mutação/genética , Oócitos/metabolismo , Xenopus laevis
20.
Neurology ; 56(12): 1739-45, 2001 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-11425943

RESUMO

BACKGROUND: The authors previously reported the generation of a knockout mouse model of Pompe disease caused by the inherited deficiency of lysosomal acid alpha-glucosidase (GAA). The disorder in the knockout mice (GAA-/-) resembles the human disease closely, except that the clinical symptoms develop late relative to the lifespan of the animals. In an attempt to accelerate the course of the disease in the knockouts, the authors increased the level of cytoplasmic glycogen by overexpressing glycogen synthase (GSase) or GlutI glucose transporter. METHODS: GAA-/- mice were crossed to transgenic mice overexpressing GSase or GlutI in skeletal muscle. RESULTS: Both transgenics on a GAA knockout background (GS/GAA-/- and GlutI/GAA-/-) developed a severe muscle wasting disorder with an early age at onset. This finding, however, is not the major focus of the study. Unexpectedly, the mice bearing the GSase transgene, but not those bearing the GlutI transgene, accumulated structurally abnormal polysaccharide (polyglucosan) similar to that observed in patients with Lafora disease, glycogenosis type IV, and glycogenosis type VII. Ultrastructurally, the periodic acid-Schiff (PAS)-positive polysaccharide inclusions were composed of short, amorphous, irregular branching filaments indistinguishable from classic polyglucosan bodies. The authors show here that increased level of GSase in the presence of normal glycogen branching enzyme (GBE) activity leads to polyglucosan accumulation. The authors have further shown that inactivation of lysosomal acid alpha-glucosidase in the knockout mice does not contribute to the process of polyglucosan formation. CONCLUSIONS: An imbalance between GSase and GBE activities is proposed as the mechanism involved in the production of polyglucosan bodies. The authors may have inadvertently created a "muscle polyglucosan disease" by simulating the mechanism for polyglucosan formation.


Assuntos
Engenharia Genética , Glucanos/genética , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/patologia , Músculos/patologia , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Animais , Modelos Animais de Doenças , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Glicogênio Sintase/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica , Músculos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...