Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Traffic ; 9(7): 1157-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18410487

RESUMEN

The adaptor complexes AP-1 and AP-3 are localized to endosomes and/or the trans Golgi network (TGN). Because of limitations in analysing intracellular adaptor function directly, their site of function is a matter of ongoing uncertainty. To overcome this problem and to analyse adaptor sorting at the TGN, we reconstituted vesicle formation from Golgi/TGN-enriched membranes in a novel in vitro budding assay. Melanocytes were metabolically labelled followed by a 19 degrees C temperature block to accumulate newly synthesized proteins in Golgi membranes, which were then enriched by subcellular fractionation and used as donor membranes for vesicle formation in vitro. The incorporation of the melanosomal proteins tyrosinase and tyrosinase-related protein 1 (TRP-1) as well as Lamp-1 and 46 kDa mannose-6-phosphate receptor (MPR46) into Golgi/TGN-derived vesicles was temperature, nucleotide, cytosol, ADP ribosylation factor 1 and adaptor dependent. We show that sorting of TRP-1 and MPR46 was AP-1 dependent, while budding of tyrosinase and Lamp-1 required AP-3. Depletion of clathrin inhibited sorting of all four cargo proteins, suggesting that AP-1 and AP-3 are involved in the formation of distinct types of clathrin-coated vesicles, each of which is characterized by the incorporation of specific cargo membrane proteins.


Asunto(s)
Complejo 1 de Proteína Adaptadora/metabolismo , Complejo 3 de Proteína Adaptadora/metabolismo , Regulación de la Expresión Génica , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Animales , Membrana Celular/metabolismo , Clatrina/metabolismo , Fibroblastos/metabolismo , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Modelos Biológicos , Fracciones Subcelulares/metabolismo , Porcinos
2.
Biochim Biophys Acta ; 1793(4): 710-25, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19124046

RESUMEN

Multiple sulfatase deficiency (MSD), mucolipidosis (ML) II/III and Niemann-Pick type C1 (NPC1) disease are rare but fatal lysosomal storage disorders caused by the genetic defect of non-lysosomal proteins. The NPC1 protein mainly localizes to late endosomes and is essential for cholesterol redistribution from endocytosed LDL to cellular membranes. NPC1 deficiency leads to lysosomal accumulation of a broad range of lipids. The precise functional mechanism of this membrane protein, however, remains puzzling. ML II, also termed I cell disease, and the less severe ML III result from deficiencies of the Golgi enzyme N-acetylglucosamine 1-phosphotransferase leading to a global defect of lysosome biogenesis. In patient cells, newly synthesized lysosomal proteins are not equipped with the critical lysosomal trafficking marker mannose 6-phosphate, thus escaping from lysosomal sorting at the trans Golgi network. MSD affects the entire sulfatase family, at least seven members of which are lysosomal enzymes that are specifically involved in the degradation of sulfated glycosaminoglycans, sulfolipids or other sulfated molecules. The combined deficiencies of all sulfatases result from a defective post-translational modification by the ER-localized formylglycine-generating enzyme (FGE), which oxidizes a specific cysteine residue to formylglycine, the catalytic residue enabling a unique mechanism of sulfate ester hydrolysis. This review gives an update on the molecular bases of these enigmatic diseases, which have been challenging researchers since many decades and so far led to a number of surprising findings that give deeper insight into both the cell biology and the pathobiochemistry underlying these complex disorders. In case of MSD, considerable progress has been made in recent years towards an understanding of disease-causing FGE mutations. First approaches to link molecular parameters with clinical manifestation have been described and even therapeutical options have been addressed. Further, the discovery of FGE as an essential sulfatase activating enzyme has considerable impact on enzyme replacement or gene therapy of lysosomal storage disorders caused by single sulfatase deficiencies.


Asunto(s)
Mucolipidosis/patología , Enfermedad por Deficiencia de Múltiples Sulfatasas/patología , Enfermedad de Niemann-Pick Tipo C/patología , Proteínas/metabolismo , Transporte Biológico , Humanos , Mucolipidosis/clasificación , Enfermedad por Deficiencia de Múltiples Sulfatasas/enzimología , Enfermedad por Deficiencia de Múltiples Sulfatasas/genética , Enfermedad por Deficiencia de Múltiples Sulfatasas/terapia , Procesamiento Proteico-Postraduccional
3.
J Cell Mol Med ; 13(11-12): 4505-21, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20394677

RESUMEN

The extracellular sulfatases Sulf1 and Sulf2 remove specific 6-O-sulfate groups from heparan sulfate, thereby modulating numerous signalling pathways underlying development and homeostasis. In vitro data have suggested that the two enzymes show functional redundancy. To elucidate their in vivo functions and to further address the question of a putative redundancy, we have generated Sulf1- and Sulf2-deficient mice. Phenotypic analysis of these animals revealed higher embryonic lethality of Sulf2 knockout mice, which can be associated with neuroanatomical malformations during embryogenesis. Sulf1 seems not to be essential for developmental or postnatal viability, as mice deficient in this sulfatase show no overt phenotype. However, neurite outgrowth deficits were observed in hippocampal and cerebellar neurons of both mutant mouse lines, suggesting that not only Sulf2 but also Sulf1 function plays a role in the developing nervous system. Behavioural analysis revealed differential deficits with regard to cage activity and spatial learning for Sulf1- and Sulf2-deficient mouse lines. In addition, Sulf1-specific deficits were shown for synaptic plasticity in the CA1 region of the hippocampus, associated with a reduced spine density. These results reveal that Sulf1 and Sulf2 fulfil non-redundant functions in vivo in the development and maintenance of the murine nervous system.


Asunto(s)
Conducta Animal , Encéfalo/embriología , Encéfalo/enzimología , Plasticidad Neuronal , Neuronas/enzimología , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Animales Recién Nacidos , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Pérdida del Embrión/fisiopatología , Espacio Extracelular/enzimología , Hipocampo/enzimología , Hipocampo/patología , Hipocampo/fisiopatología , Hipocampo/ultraestructura , Hidrocefalia/complicaciones , Hidrocefalia/enzimología , Hidrocefalia/patología , Hidrocefalia/fisiopatología , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Malformaciones del Sistema Nervioso/complicaciones , Malformaciones del Sistema Nervioso/enzimología , Malformaciones del Sistema Nervioso/fisiopatología , Neuritas/enzimología , Neuritas/patología , Neuronas/patología , Fenotipo , Sulfatasas/deficiencia , Sulfotransferasas/deficiencia , Transmisión Sináptica/fisiología
4.
J Cell Biol ; 156(5): 791-5, 2002 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-11877457

RESUMEN

During receptor-mediated endocytosis, AP2 complexes act as a bridge between the cargo membrane proteins and the clathrin coat by binding to sorting signals via the mu 2 subunit and to clathrin via the beta subunit. Here we show that binding of AP2 to sorting signals in vitro is regulated by phosphorylation of the mu 2 subunit of AP2. Phosphorylation of mu 2 enhances the binding affinity of AP2 for sorting motifs as much as 25-fold compared with dephosphorylated AP2. The recognition of sorting signals was not affected by the phosphorylation status of the alpha or beta 2 subunit, suggesting that phosphorylation of mu 2 is critical for regulation of AP2 binding to sorting signals. Phosphorylation of mu 2 occurs at a single threonine residue (Thr-156) and is mediated by the newly discovered adaptor-associated kinase, AAK1, which copurifies with AP2. We propose that phosphorylation of the AP2 mu 2 subunit by AAK1 ensures high affinity binding of AP2 to sorting signals of cargo membrane proteins during the initial steps of receptor-mediated endocytosis.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Endocitosis/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/genética , Receptores de Superficie Celular/metabolismo , Complejo 2 de Proteína Adaptadora , Proteínas Adaptadoras del Transporte Vesicular , Animales , Sitios de Unión/fisiología , Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Porcinos
5.
Mol Cell Biol ; 26(15): 5615-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16847317

RESUMEN

Mutations in the cytosolic enzyme phosphomannomutase 2 (PMM2), which catalyzes the conversion of mannose-6-phosphate to mannose-1-phosphate, cause the most common form of congenital disorders of glycosylation, termed CDG-Ia. It is an inherited multisystemic disease with severe neurological impairment. To study the pathophysiology of CDG-Ia and to investigate possible therapeutic approaches, we generated a mouse model for CDG-Ia by targeted disruption of the Pmm2 gene. Heterozygous mutant mice appeared normal in development, gross anatomy, and fertility. In contrast, embryos homozygous for the Pmm2-null allele were recovered in embryonic development at days 2.5 to 3.5. These results indicate that Pmm2 is essential for early development of mice. Mating experiments of heterozygous mice with wild-type mice could further show that transmission of the female Pmm2-null allele is impaired.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/genética , Embrión de Mamíferos/fisiología , Isoenzimas/metabolismo , Fosfotransferasas (Fosfomutasas)/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Humanos , Isoenzimas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Sistemas de Lectura Abierta , Linaje , Fosfotransferasas (Fosfomutasas)/genética , Embarazo
6.
Hum Mutat ; 29(1): 205, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18157819

RESUMEN

Multiple Sulfatase Deficiency (MSD) is a rare inborn autosomal-recessive disorder, which mainly combines clinical features of metachromatic leukodystrophy, mucopolysaccharidosis and X-linked ichthyosis. The clinical course ranges from neonatal severe to mild juvenile cases. MSD is caused by mutations in the SUMF1 gene encoding the formylglycine-generating enzyme (FGE). FGE posttranslationally activates sulfatases by generating formylglycine in their catalytic sites. We analyzed the functional consequences of missense mutations p.A177P, p.W179S, p.A279V and p.R349W with regard to FGE's subcellular localization, enzymatic activity, protein stability, intracellular retention and resulting sulfatase activities. All four mutations did not affect localization of FGE in the endoplasmic reticulum of MSD fibroblasts. However, they decreased its specific enzymatic activity to less than 1% (p.A177P and p.R349W), 3% (p.W179S) or 23% (p.A279V). Protein stability was severely decreased for p.A279V and p.R349W, and almost comparable to wild type for p.A177P and p.W179S. The patient with the mildest clinical phenotype carries the mutation p.A279V leading to decreased FGE protein stability, but high residual enzymatic activity and only slightly reduced sulfatase activities. In contrast, the most severely affected patient carries the mutation p.R349W leading to drastically decreased protein stability, very low residual enzymatic activity and considerably reduced sulfatase activities. Our functional studies provide novel insight into the molecular defect underlying MSD and reveal that both residual enzyme activity and protein stability of FGE contribute to the clinical phenotype. The application of improved functional assays to determine these two molecular parameters of FGE mutants may enable the prediction of the clinical outcome in the future.


Asunto(s)
Enfermedad por Deficiencia de Múltiples Sulfatasas/diagnóstico , Enfermedad por Deficiencia de Múltiples Sulfatasas/genética , Mutación , Sulfatasas/genética , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Genotipo , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Fenotipo , Sulfatasas/análisis , Sulfatasas/metabolismo
7.
FEBS J ; 275(6): 1118-30, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18266766

RESUMEN

Formylglycine-generating enzyme (FGE) catalyzes in newly synthesized sulfatases the oxidation of a specific cysteine residue to formylglycine, which is the catalytic residue required for sulfate ester hydrolysis. This post-translational modification occurs in the endoplasmic reticulum (ER), and is an essential step in the biogenesis of this enzyme family. A paralog of FGE (pFGE) also localizes to the ER. It shares many properties with FGE, but lacks formylglycine-generating activity. There is evidence that FGE and pFGE act in concert, possibly by forming complexes with sulfatases and one another. Here we show that human pFGE, but not FGE, is retained in the ER through its C-terminal tetrapeptide PGEL, a noncanonical variant of the classic KDEL ER-retention signal. Surprisingly, PGEL, although having two nonconsensus residues (PG), confers efficient ER retention when fused to a secretory protein. Inducible coexpression of pFGE at different levels in FGE-expressing cells did not significantly influence the kinetics of FGE secretion, suggesting that pFGE is not a retention factor for FGE in vivo. PGEL is accessible at the surface of the pFGE structure. It is found in 21 mammalian species with available pFGE sequences. Other species carry either canonical signals (eight mammals and 26 nonmammals) or different noncanonical variants (six mammals and six nonmammals). Among the latter, SGEL was tested and found to also confer ER retention. Although evolutionarily conserved for mammalian pFGE, the PGEL signal is found only in one further human protein entering the ER. Its consequences for KDEL receptor-mediated ER retrieval and benefit for pFGE functionality remain to be fully resolved.


Asunto(s)
Retículo Endoplásmico/enzimología , Glicina/análogos & derivados , Señales de Clasificación de Proteína , Sulfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Biología Computacional , Secuencia Conservada , Evolución Molecular , Glicina/metabolismo , Humanos , Datos de Secuencia Molecular , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Conformación Proteica , Sulfatasas/análisis , Sulfatasas/química , Sulfatasas/genética
8.
J Clin Invest ; 111(11): 1733-45, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12782676

RESUMEN

Thyroid function depends on processing of the prohormone thyroglobulin by sequential proteolytic events. From in vitro analysis it is known that cysteine proteinases mediate proteolytic processing of thyroglobulin. Here, we have analyzed mice with deficiencies in cathepsins B, K, L, B and K, or K and L in order to investigate which of the cysteine proteinases is most important for proteolytic processing of thyroglobulin in vivo. Immunolabeling demonstrated a rearrangement of the endocytic system and a redistribution of extracellularly located enzymes in thyroids of cathepsin-deficient mice. Cathepsin L was upregulated in thyroids of cathepsin K(-/-) or B(-/-)/K(-/-) mice, suggesting a compensation of cathepsin L for cathepsin K deficiency. Impaired proteolysis resulted in the persistence of thyroglobulin in the thyroids of mice with deficiencies in cathepsin B or L. The typical multilayered appearance of extracellularly stored thyroglobulin was retained in cathepsin K(-/-) mice only. These results suggest that cathepsins B and L are involved in the solubilization of thyroglobulin from its covalently cross-linked storage form. Cathepsin K(-/-)/L(-/-) mice had significantly reduced levels of free thyroxine, indicating that utilization of luminal thyroglobulin for thyroxine liberation is mediated by a combinatory action of cathepsins K and L.


Asunto(s)
Catepsina B/metabolismo , Catepsinas/metabolismo , Glándula Tiroides/metabolismo , Animales , Catepsina B/genética , Catepsina K , Catepsina L , Catepsinas/genética , Cisteína Endopeptidasas , Endocitosis , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Modelos Biológicos , Mutación , Tiroglobulina/metabolismo , Tiroxina/sangre , Tiroxina/metabolismo , Factores de Tiempo , Regulación hacia Arriba
9.
J Clin Invest ; 109(6): 725-33, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11901181

RESUMEN

Deficiency of the Golgi enzyme UDP-Gal:N-acetylglucosamine beta-1,4-galactosyltransferase I (beta4GalT I) (E.C.2.4.1.38) causes a new congenital disorder of glycosylation (CDG), designated type IId (CDG-IId), a severe neurologic disease characterized by a hydrocephalus, myopathy, and blood-clotting defects. Analysis of oligosaccharides from serum transferrin by HPLC, mass spectrometry, and lectin binding revealed the loss of sialic acid and galactose residues. In skin fibroblasts and leukocytes, galactosyltransferase activity was reduced to 5% that of controls. In fibroblasts, a truncated polypeptide was detected that was about 12 kDa smaller in size than wild-type beta4GalT I and that failed to localize to the Golgi apparatus. Sequencing of the beta4GalT I cDNA and gene revealed an insertion of a single nucleotide (1031-1032insC) leading to premature translation stop and loss of the C-terminal 50 amino acids of the enzyme. The patient was homozygous and his parents heterozygous for this mutation. Expression of a corresponding mutant cDNA in COS-7 cells led to the synthesis of a truncated, inactive polypeptide, which localized to the endoplasmic reticulum.


Asunto(s)
Trastornos Congénitos de Glicosilación/enzimología , Glicoproteínas/sangre , Transferrina/análisis , Uridina Difosfato Galactosa/deficiencia , beta-N-Acetilglucosaminilglicopéptido beta-1,4-Galactosiltransferasa/deficiencia , Células Cultivadas , Preescolar , Cromatografía de Afinidad , Trastornos Congénitos de Glicosilación/etiología , Trastornos Congénitos de Glicosilación/metabolismo , Fibroblastos/citología , Fibroblastos/enzimología , Galactosa/metabolismo , Aparato de Golgi/metabolismo , Humanos , Inmunohistoquímica , Lactante , Leucocitos/enzimología , Masculino , Transporte de Proteínas/fisiología , Piel/citología , Piel/metabolismo , Uridina Difosfato Galactosa/metabolismo , beta-N-Acetilglucosaminilglicopéptido beta-1,4-Galactosiltransferasa/genética , beta-N-Acetilglucosaminilglicopéptido beta-1,4-Galactosiltransferasa/metabolismo
10.
Biochem J ; 400(1): 63-73, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16901266

RESUMEN

HS (heparan sulfate) is essential for normal embryonic development. This requirement is due to the obligatory role for HS in the signalling pathways of many growth factors and morphogens that bind to sulfated domains in the HS polymer chain. The sulfation patterning of HS is determined by a complex interplay of Golgi-located N- and O-sulfotransferases which sulfate the heparan precursor and cell surface endosulfatases that selectively remove 6-O-sulfates from mature HS chains. In the present study we generated single or double knock-out mice for the two murine endosulfatases mSulf1 and mSulf2. Detailed structural analysis of HS from mSulf1-/- fibroblasts showed a striking increase in 6-O-sulfation, which was not seen in mSulf2-/- HS. Intriguingly, the level of 6-O-sulfation in the double mSulf1-/-/2-/- HS was significantly higher than that observed in the mSulf1-/- counterpart. These data imply that mSulf1 and mSulf2 are functionally co-operative. Unlike their avian orthologues, mammalian Sulf activities are not restricted to the highly sulfated S-domains of HS. Mitogenesis assays with FGF2 (fibroblast growth factor 2) revealed that Sulf activity decreases the activating potential of newly-synthesized HS, suggesting an important role for these enzymes in cell growth regulation in embryonic and adult tissues.


Asunto(s)
Heparitina Sulfato/metabolismo , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Disacáridos/análisis , Epítopos/inmunología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Genotipo , Heparitina Sulfato/química , Heparitina Sulfato/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Estructura Molecular , Oligosacáridos/análisis , Sulfatasas/genética , Sulfatos/metabolismo , Sulfotransferasas/genética
11.
Mol Biol Cell ; 13(9): 3355-68, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12221139

RESUMEN

In LAMP-2-deficient mice autophagic vacuoles accumulate in many tissues, including liver, pancreas, muscle, and heart. Here we extend the phenotype analysis using cultured hepatocytes. In LAMP-2-deficient hepatocytes the half-life of both early and late autophagic vacuoles was prolonged as evaluated by quantitative electron microscopy. However, an endocytic tracer reached the autophagic vacuoles, indicating delivery of endo/lysosomal constituents to autophagic vacuoles. Enzyme activity measurements showed that the trafficking of some lysosomal enzymes to lysosomes was impaired. Immunoprecipitation of metabolically labeled cathepsin D indicated reduced intracellular retention and processing in the knockout cells. The steady-state level of 300-kDa mannose 6-phosphate receptor was slightly lower in LAMP-2-deficient hepatocytes, whereas that of 46-kDa mannose 6-phosphate receptor was decreased to 30% of controls due to a shorter half-life. Less receptor was found in the Golgi region and in vesicles and tubules surrounding multivesicular endosomes, suggesting impaired recycling from endosomes to the Golgi. More receptor was found in autophagic vacuoles, which may explain its shorter half-life. Our data indicate that in hepatocytes LAMP-2 deficiency either directly or indirectly leads to impaired recycling of 46-kDa mannose 6-phosphate receptors and partial mistargeting of a subset of lysosomal enzymes. Autophagic vacuoles may accumulate due to impaired capacity for lysosomal degradation.


Asunto(s)
Antígenos CD/fisiología , Hepatocitos/metabolismo , Lisosomas/metabolismo , Animales , Antígenos CD/metabolismo , Western Blotting , Catepsina D/metabolismo , Membrana Celular , Células Cultivadas , Endocitosis , Endosomas , Genotipo , Inmunohistoquímica , Metabolismo de los Lípidos , Proteínas de Membrana de los Lisosomas , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Fenotipo , Pruebas de Precipitina , Unión Proteica , Factores de Tiempo
12.
Mol Biol Cell ; 15(7): 3132-45, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15121881

RESUMEN

Mice double deficient in LAMP-1 and -2 were generated. The embryos died between embryonic days 14.5 and 16.5. An accumulation of autophagic vacuoles was detected in many tissues including endothelial cells and Schwann cells. Fibroblast cell lines derived from the double-deficient embryos accumulated autophagic vacuoles and the autophagy protein LC3II after amino acid starvation. Lysosomal vesicles were larger and more peripherally distributed and showed a lower specific density in Percoll gradients in double deficient when compared with control cells. Lysosomal enzyme activities, cathepsin D processing and mannose-6-phosphate receptor expression levels were not affected by the deficiency of both LAMPs. Surprisingly, LAMP-1 and -2 deficiencies did not affect long-lived protein degradation rates, including proteolysis due to chaperone-mediated autophagy. The LAMP-1/2 double-deficient cells and, to a lesser extent, LAMP-2 single-deficient cells showed an accumulation of unesterified cholesterol in endo/lysosomal, rab7, and NPC1 positive compartments as well as reduced amounts of lipid droplets. The cholesterol accumulation in LAMP-1/2 double-deficient cells could be rescued by overexpression of murine LAMP-2a, but not by LAMP-1, highlighting the more prominent role of LAMP-2. Taken together these findings indicate partially overlapping functions for LAMP-1 and -2 in lysosome biogenesis, autophagy, and cholesterol homeostasis.


Asunto(s)
Antígenos CD/fisiología , Colesterol/metabolismo , Vesículas Citoplasmáticas/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Células Cultivadas , Colesterol/análisis , Vesículas Citoplasmáticas/inmunología , Vesículas Citoplasmáticas/ultraestructura , Embrión de Mamíferos/metabolismo , Fibroblastos/química , Fibroblastos/inmunología , Fibroblastos/metabolismo , Filipina/análisis , Filipina/química , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Membrana de los Lisosomas , Lisosomas/enzimología , Lisosomas/ultraestructura , Ratones , Ratones Noqueados , Proteína Niemann-Pick C1 , Proteínas/análisis , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
13.
J Neurosci ; 25(28): 6539-49, 2005 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16014715

RESUMEN

Mice with alpha-mannosidase gene inactivation provide an experimental model for alpha-mannosidosis, a lysosomal storage disease with severe neuropsychological and psychopathological complications. Neurohistological alterations in these mice were similar to those in patients and included vacuolations and axonal spheroids in the CNS and peripheral nervous system. Vacuolation was most prominent and evenly distributed in neuronal perikarya of the hippocampal CA2 and CA3 regions, whereas CA1 and dentate gyrus were weakly or not affected. Field potential recordings from CA1 region in hippocampal slices showed enhanced theta burst-induced long-term potentiation (LTP) in alpha-mannosidase-deficient mice. Longitudinal assessment in age-matched alpha-mannosidase-deficient and wild-type littermates, using an extended test battery, demonstrated a neurocognitive and psychotiform profile that may relate to the psychopathological alterations in clinical alpha-mannosidosis. Brainstem auditory-evoked potentials and basic neuromotor abilities were not impaired and did not deteriorate with age. Exploratory and conflict tests revealed consistent decreases in exploratory activity and emotional blunting in the knock-out group. alpha-Mannosidosis mice were also impaired in aversively motivated learning and acquisition of signal-shock associations. Acquisition and reversal learning in the water maze task, passive avoidance learning in the step-through procedure, as well as emotional response conditioning in an operant procedure were all impaired. Acquisition or shaping of an appetitive instrumental conditioning task was unchanged. Appetitive odor discrimination learning was only marginally impaired during shaping, whereas both the discrimination and reversal subtasks were normal. We propose that prominent storage and enhanced LTP in hippocampus have contributed to these specific behavioral alterations in alpha-mannosidase-deficient mice.


Asunto(s)
Conducta Animal , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Potenciación a Largo Plazo/fisiología , alfa-Manosidosis/psicología , Animales , Conducta Apetitiva , Reacción de Prevención , Condicionamiento Operante , Discriminación en Psicología , Emociones , Potenciales Evocados Auditivos del Tronco Encefálico , Potenciales Postsinápticos Excitadores , Conducta Exploratoria , Femenino , Fuerza de la Mano , Hipocampo/patología , Humanos , Discapacidades para el Aprendizaje/genética , Lisosomas/enzimología , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Sistema Nervioso/patología , Neuronas/ultraestructura , Desempeño Psicomotor , Olfato , Vacuolas/ultraestructura , alfa-Manosidasa/genética , alfa-Manosidosis/genética , alfa-Manosidosis/fisiopatología
14.
FEBS Lett ; 580(24): 5747-52, 2006 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17007843

RESUMEN

Recently, we and others identified the 66.3-kDa protein as one of several putative novel lysosomal matrix proteins by analyzing mannose 6-phosphate receptors binding proteins [Kollmann K., Mutenda K.E., Balleininger M., Eckermann E., von Figura K., Schmidt B., Lübke T. (2005) Identification of novel lysosomal matrix proteins by proteome analysis. Proteomics 5(15), 3966-3678, Sleat D.E., Lackland H., Wang Y., Sohar I., Xiao G., Li H., Lobel P. (2005) The human brain mannose 6-phosphate glycoproteome: a complex mixture composed of multiple isoforms of many soluble lysosomal proteins. Proteomics. 5(6), 1520-1532]. Here, we describe the expression of the mouse 66.3-kDa protein in HT1080 cells in which it is synthesized as a precursor of about 75kDa and subsequently processed by limited proteolysis to mature polypeptides accumulating in the lysosomal compartment. The lysosomal localisation of the endogenous 66.3-kDa protein was verified by indirect immunofluorescence in mouse embryonic fibroblasts and by subcellular fractionation of tyloxapol-filled mouse liver lysosomes. Northern blot analysis reveals high transcriptional levels in testis, liver and kidney, whereas Western blot analysis shows high protein levels in brain, heart, lung and spleen. Interestingly, in mouse the endogenous 66.3-kDa protein is processed in a highly tissue-dependent manner to mature forms.


Asunto(s)
Glicoproteínas/metabolismo , Lisosomas/metabolismo , Animales , Línea Celular Tumoral , Glicoproteínas/genética , Glicoproteínas/aislamiento & purificación , Glicosilación , Humanos , Ratones , Peso Molecular , Especificidad de Órganos , Procesamiento Proteico-Postraduccional , Especificidad por Sustrato
15.
J Neuropathol Exp Neurol ; 64(6): 513-22, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15977643

RESUMEN

Among the autophagic vacuolar myopathies (AVMs), a subgroup is characterized pathologically by unusual autophagic vacuoles with sarcolemmal features (AVSF) and includes Danon disease and X-linked myopathy with excessive autophagy. The diagnostic importance and detailed morphologic features of AVSF in different AVMs have not been well established, and the mechanism of AVSF formation is not known. To address these issues, we have performed detailed histologic studies of myopathies with AVSF and other AVMs. In Danon disease and related AVMs, at the light microscopic level, autophagic vacuoles appeared to be accumulations of lysosomes, which, by electron microscopy consisted of clusters of autophagic vacuoles, indicative of autolysosomes. Some autolysosomes were surrounded by membranes with sarcolemmal proteins, acetylcholinesterase activity, and basal lamina. In Danon disease, the number of fibers with AVSF increased linearly with age while the number with autolysosomal accumulations decreased slightly, suggesting that AVSF are produced secondarily in response to autolysosomes. Most of the AVSF form enclosed spaces, indicating that the vacuolar membranes may be formed in situ rather than through sarcolemmal indentation. This unique intracytoplasmic membrane structure was not found in other AVMs. In conclusion, AVSF with acetylcholinesterase activity are autolysosomes surrounded by secondarily generated intracytoplasmic sarcolemma-like structure and delineates a subgroup of AVMs.


Asunto(s)
Autofagia/fisiología , Músculo Esquelético , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Sarcolema/patología , Vacuolas , Acetilcolinesterasa/metabolismo , Adolescente , Adulto , Animales , Antígenos CD/genética , Bungarotoxinas/metabolismo , Niño , Preescolar , Glicoproteínas/metabolismo , Humanos , Inmunohistoquímica/métodos , Lactante , Proteínas de Membrana de los Lisosomas , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Inmunoelectrónica/métodos , Modelos Biológicos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Enfermedades Musculares/clasificación , Enfermedades Musculares/metabolismo , Sarcolema/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura
16.
FEBS J ; 282(17): 3262-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26077311

RESUMEN

C α-formylglycine (FGly) is the catalytic residue of sulfatases in eukaryotes. It is generated by a unique post-translational modification catalysed by the FGly-generating enzyme (FGE) in the endoplasmic reticulum. FGE oxidizes a cysteine residue within the conserved CxPxR sequence motif of nascent sulfatase polypeptides to FGly. Here we show that this oxidation is strictly dependent on molecular oxygen (O2) and consumes 1 mol O2 per mol FGly formed. For maximal activity FGE requires an O2 concentration of 9% (105 µM). Sustained FGE activity further requires the presence of a thiol-based reductant such as DTT. FGly is also formed in the absence of DTT, but its formation ceases rapidly. Thus inactivated FGE accumulates in which the cysteine pair Cys336/Cys341 in the catalytic site is oxidized to form disulfide bridges between either Cys336 and Cys341 or Cys341 and the CxPxR cysteine of the sulfatase. These results strongly suggest that the Cys336/Cys341 pair is directly involved in the O2 -dependent conversion of the CxPxR cysteine to FGly. The available data characterize eukaryotic FGE as a monooxygenase, in which Cys336/Cys341 disulfide bridge formation donates the electrons required to reduce one oxygen atom of O2 to water while the other oxygen atom oxidizes the CxPxR cysteine to FGly. Regeneration of a reduced Cys336/Cys341 pair is accomplished in vivo by a yet unknown reductant of the endoplasmic reticulum or in vitro by DTT. Remarkably, this monooxygenase reaction utilizes O2 without involvement of any activating cofactor.


Asunto(s)
Alanina/análogos & derivados , Glicina/análogos & derivados , Oxigenasas de Función Mixta/metabolismo , Oxígeno/metabolismo , Sulfatasas/metabolismo , Alanina/química , Alanina/metabolismo , Animales , Baculoviridae/genética , Biocatálisis , Dominio Catalítico , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Ditiotreitol/química , Pruebas de Enzimas , Expresión Génica , Glicina/química , Glicina/metabolismo , Humanos , Cinética , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Oxígeno/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera , Sulfatasas/química , Sulfatasas/genética
17.
Gene ; 316: 47-56, 2003 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-14563551

RESUMEN

Recently, the human C(alpha)-formylglycine (FGly)-generating enzyme (FGE), whose deficiency causes the autosomal-recessively transmitted lysosomal storage disease multiple sulfatase deficiency (MSD), has been identified. In sulfatases, FGE posttranslationally converts a cysteine residue to FGly, which is part of the catalytic site and is essential for sulfatase activity. FGE is encoded by the sulfatase modifying factor 1 (SUMF1) gene, which defines a new gene family comprising orthologs from prokaryotes to higher eukaryotes. The genomes of E. coli, S. cerevisiae and C. elegans lack SUMF1, indicating a phylogenetic gap and the existence of an alternative FGly-generating system. The genomes of vertebrates including mouse, man and pufferfish contain a sulfatase modifying factor 2 (SUMF2) gene encoding an FGE paralog of unknown function. SUMF2 evolved from a single exon SUMF1 gene as found in diptera prior to divergent intron acquisition. In several prokaryotic genomes, the SUMF1 gene is cotranscribed with genes encoding sulfatases which require FGly modification. The FGE protein contains a single domain that is made up of three highly conserved subdomains spaced by nonconserved sequences of variable lengths. The similarity among the eukaryotic FGE orthologs varies between 72% and 100% for the three subdomains and is highest for the C-terminal subdomain, which is a hotspot for mutations in MSD patients.


Asunto(s)
Alanina/análogos & derivados , Células Eucariotas/enzimología , Glicina/análogos & derivados , Filogenia , Células Procariotas/enzimología , Procesamiento Proteico-Postraduccional , Sulfatasas/genética , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Dominio Catalítico/genética , Secuencia Conservada/genética , Células Eucariotas/metabolismo , Glicina/genética , Glicina/metabolismo , Humanos , Datos de Secuencia Molecular , Familia de Multigenes/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Células Procariotas/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Sulfatasas/metabolismo
18.
Transplantation ; 78(5): 758-61, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371683

RESUMEN

Cytotoxic T lymphocytes (CTL) play an important role in the rejection of allogeneic cells and organs. CTL secrete granzymes and perforin as cytotoxic effector molecules. The mannose 6-phosphate receptor (Mpr)300 has been reported to function as receptor for granzyme B on target cells and to be essential for the rejection of allogeneic cells in vivo. Using mouse embryonal fibroblasts from Mpr300 and Mpr46 knockout mice, we show that both Mpr 300 and Mpr46 are dispensable on target cells for lysis and apoptosis mediated by alloreactive CTL in vitro and for allorejection in vivo. In agreement with a postulated function of Mpr300 as a tumor suppressor gene, deficiency of Mpr300 appears to promote cellular proliferation and tumorigenicity but not resistance to allorejection.


Asunto(s)
Trasplante de Células , Rechazo de Injerto/patología , Receptor IGF Tipo 2/deficiencia , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis , Fibroblastos/patología , Fibroblastos/fisiología , Rechazo de Injerto/inmunología , Ratones , Ratones Noqueados , Linfocitos T Citotóxicos/patología , Trasplante Homólogo/patología
19.
J Mass Spectrom ; 38(1): 80-6, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12526009

RESUMEN

C(alpha)-Formylglycine, the catalytic amino acid residue in the active site of sulfatases, is generated by post-translational modification of a cysteine or serine residue. We describe a highly sensitive procedure for the detection of C(alpha)-formylglycine-containing peptides in tryptic digests of sulfatase proteins. The protocol is based on the formation of hydrazone derivatives of C(alpha)-formylglycine-containing peptides when using dinitrophenylhydrazine as a matrix for matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS). The hydrazone derivatives desorb and ionize with high efficiency and can be detected in the sub-femtomole range. The presence of C(alpha)-formylglycine is indicated by a mass increment of 180.13 u, corresponding to the hydrazone moiety, and also by a unique C-terminal fragment ion, characteristic of sulfatases, that becomes prominent in MALDI post-source decay mass spectra of the hydrazone derivatives.


Asunto(s)
Alanina/análogos & derivados , Glicina/análogos & derivados , Glicina/análisis , Sulfatasas/química , Alanina/análisis , Secuencia de Aminoácidos , Glicina/química , Klebsiella/enzimología , Datos de Secuencia Molecular , Pseudomonas/enzimología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Sulfatasas/metabolismo , Tripsina/metabolismo
20.
Biosci Rep ; 22(5-6): 513-21, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12635848

RESUMEN

Mannose 6-phosphate receptor (MPR 300) protein was earlier affinity purified on phosphomannan gel from the membrane extracts of whole animal acetone powder of a mollusc, unio, in the presence of EDTA (Udaya Lakshmi, Y., Radha, Y., Hille-Rehfeld, A., von Figura, K., and Siva Kumar, N. (1999) Biosci. Rep. 19:403-409). In the present study we demonstrate that the unio also contains the putative mannose 6-phosphate receptor (MPR 46) that can be purified on the same gel in presence of divalent metal ions (10 mM each of calcium, manganese, and magnesium), and in the absence of sodium chloride and at pH 6.5. Chicken and Fish cell MPR 46 proteins were purified under these conditions (Siva Kumar, N., Udaya Lakshmi, Y., Hille-Rehfeld, A., and von Figura, K. (1999) Comp. Biochem. & PhysioL 123B:261-265). The authenticity of the receptor is further confirmed by its ability to react with the MSC1 antibody that is specific for MPR 46 protein. Additional evidence for the presence of MPR 46 in molluscs could be obtained by metabolic labeling of mollusc cells Biomphalaria glabrata (Bg cells) with [35S] methionine and cysteine, and passing the labeled membrane extract on phosphomannan gel (at pH 6.5 and 7.0). On elution with mannose 6-phosphate, followed by immunoprecipitation of the column fractions, we identified the putative MPR 46 protein in the Bg cells. When Bg cell MPR 46 was deglycosylated along with chicken MPR 46 (control) both species yielded a single polypeptide corresponding to molecular mass of 26 kDa, suggesting that both contain the same receptor protein.


Asunto(s)
Biomphalaria/metabolismo , Manosafosfatos/metabolismo , Receptor IGF Tipo 2/aislamiento & purificación , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos/fisiología , Biomphalaria/citología , Membrana Celular/química , Electroforesis en Gel de Poliacrilamida , Evolución Molecular , Peso Molecular , Filogenia , Receptor IGF Tipo 2/genética , Fracciones Subcelulares/química , Vertebrados/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA