Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1842(4): 665-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24463171

ABSTRACT

The lysosomal hydrolase galactocerebrosidase (GALC) catalyzes the removal of galactose from galactosylceramide and from other sphingolipids. GALC deficiency is responsible for globoid cell leukodystrophy (GLD), or Krabbe's disease, an early lethal inherited neurodegenerative disorder characterized by the accumulation of the neurotoxic metabolite psychosine in the central nervous system (CNS). The poor outcome of current clinical treatments calls for novel model systems to investigate the biological impact of GALC down-regulation and for the search of novel therapeutic strategies in GLD. Zebrafish (Danio rerio) represents an attractive vertebrate model for human diseases. Here, lysosomal GALC activity was demonstrated in the brain of zebrafish adults and embryos. Accordingly, we identified two GALC co-orthologs (named galca and galcb) dynamically co-expressed in CNS during zebrafish development. Both genes encode for lysosomal enzymes endowed with GALC activity. Single down-regulation of galca or galcb by specific antisense morpholino oligonucleotides results in a partial decrease of GALC activity in zebrafish embryos that was abrogated in double galca/galcb morphants. However, no psychosine accumulation was observed in galca/galcb double morphants. Nevertheless, double galca/galcb knockdown caused reduction and partial disorganization of the expression of the early neuronal marker neuroD and an increase of apoptotic events during CNS development. These observations provide new insights into the pathogenesis of GLD, indicating that GALC loss-of-function may have pathological consequences in developing CNS independent of psychosine accumulation. Also, they underscore the potentiality of the zebrafish system in studying the pathogenesis of lysosomal neurodegenerative diseases, including GLD.


Subject(s)
Galactosylceramidase/physiology , Leukodystrophy, Globoid Cell/etiology , Zebrafish/metabolism , Animals , Brain/embryology , Brain/enzymology , Cloning, Molecular , Disease Models, Animal , Galactosylceramidase/genetics , Humans , Leukodystrophy, Globoid Cell/enzymology , Zebrafish/embryology
2.
Dev Dyn ; 243(2): 299-314, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123392

ABSTRACT

BACKGROUND: The family of AP-1 complexes mediates protein sorting in the late secretory pathway and it is essential for the development of mammals. The ubiquitously expressed AP-1A complex consists of four adaptins γ1, ß1, µ1A, and σ1A. AP-1A mediates protein transport between the trans-Golgi network and early endosomes. The polarized epithelia AP-1B complex contains the µ1B-adaptin. AP-1B mediates specific transport of proteins from basolateral recycling endosomes to the basolateral plasma membrane of polarized epithelial cells. RESULTS: Analysis of the zebrafish genome revealed the existence of three µ1-adaptin genes, encoding µ1A, µ1B, and the novel isoform µ1C, which is not found in mammals. µ1C shows 80% sequence identity with µ1A and µ1B. The µ1C expression pattern largely overlaps with that of µ1A, while µ1B is expressed in epithelial cells. By knocking-down the synthesis of µ1A, µ1B and µ1C with antisense morpholino techniques we demonstrate that each of these µ1 adaptins is essential for zebrafish development, with µ1A and µ1C being involved in central nervous system development and µ1B in kidney, gut and liver formation. CONCLUSIONS: Zebrafish is unique in expressing three AP-1 complexes: AP-1A, AP-1B, and AP-1C. Our results demonstrate that they are not redundant and that each of them has specific functions, which cannot be fulfilled by one of the other isoforms. Each of the µ1 adaptins appears to mediate specific molecular mechanisms essential for early developmental processes, which depends on specific intracellular vesicular protein sorting pathways.


Subject(s)
Adaptor Protein Complex mu Subunits/metabolism , Embryonic Development/genetics , Zebrafish/embryology , trans-Golgi Network/metabolism , Acridine Orange , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Adaptor Protein Complex mu Subunits/genetics , Animals , Base Sequence , DNA Primers/genetics , Gene Knockdown Techniques , In Situ Hybridization , Molecular Sequence Data , Morpholinos/genetics , Phylogeny , Protein Subunits/genetics , Protein Transport/genetics , Protein Transport/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis , Zebrafish/metabolism
3.
BMC Biochem ; 12: 45, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21861893

ABSTRACT

BACKGROUND: Large surface loops contained within compact protein structures and not involved in catalytic process have been proposed as preferred regions for protein family evolution. These loops are subjected to lower sequence constraints and can evolve rapidly in novel structural variants. A good model to study this hypothesis is represented by sialidase enzymes. Indeed, the structure of sialidases is a ß-propeller composed by anti-parallel ß-sheets connected by loops that suit well with the rapid evolving loop hypothesis. These features prompted us to extend our studies on this protein family in birds, to get insights on the evolution of this class of glycohydrolases. RESULTS: Gallus gallus (Gg) genome contains one NEU3 gene encoding a protein with a unique 188 amino acid sequence mainly constituted by a peptide motif repeated six times in tandem with no homology with any other known protein sequence. The repeat region is located at the same position as the roughly 80 amino acid loop characteristic of mammalian NEU4. Based on molecular modeling, all these sequences represent a connecting loop between the first two highly conserved ß-strands of the fifth blade of the sialidase ß-propeller. Moreover this loop is highly variable in sequence and size in NEU3 sialidases from other vertebrates. Finally, we found that the general enzymatic properties and subcellular localization of Gg NEU3 are not influenced by the deletion of the repeat sequence. CONCLUSION: In this study we demonstrated that sialidase protein structure contains a surface loop, highly variable both in sequence and size, connecting two conserved ß-sheets and emerging on the opposite site of the catalytic crevice. These data confirm that sialidase family can serve as suitable model for the study of the evolutionary process based on rapid evolving loops, which may had occurred in sialidases. Giving the peculiar organization of the loop region identified in Gg NEU3, this protein can be considered of particular interest in such evolutionary studies and to get deeper insights in sialidase evolution.


Subject(s)
Chickens , Evolution, Molecular , Neuraminidase/chemistry , Neuraminidase/genetics , Amino Acid Sequence , Animals , COS Cells , Cattle , Chickens/genetics , Chlorocebus aethiops , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Neuraminidase/metabolism , Protein Structure, Secondary
4.
Dev Dyn ; 239(9): 2404-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20652956

ABSTRACT

Protein transport between the trans-Golgi network and endosomes is mediated by transport vesicles formed by the adaptor-protein complex AP-1, consisting of the adaptins γ1, ß1, µ1, σ1. Mammalia express µ1A ubiquitously and isoform µ1B in polarized epithelia. Mouse γ1 or µ1A 'knock out's revealed that AP-1 is indispensable for embryonic development. We isolated µ1A and µ1B from Danio rerio. Analysis of µ1A and µ1B expression revealed tissue-specific expression for either one during embryogenesis and in adult tissues in contrast to their expression in mammalia. µ1B transcript was detected in organs of endodermal derivation and "knock-down" experiments gave rise to embryos defective in formation of intestine, liver, and pronephric ducts. Development ceased at 7-8 dpf. µ1B is not expressed in murine liver, indicating loss of µ1B expression and establishment of alternative sorting mechanisms during mammalian development.


Subject(s)
Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Morphogenesis/physiology , Protein Isoforms/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/growth & development , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex mu Subunits/classification , Adaptor Protein Complex mu Subunits/genetics , Amino Acid Sequence , Animals , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genome , Humans , Mice , Molecular Sequence Data , Phenotype , Phylogeny , Protein Isoforms/classification , Protein Isoforms/genetics , Sequence Alignment , Tissue Distribution , Zebrafish/anatomy & histology , Zebrafish/genetics , Zebrafish Proteins/classification , Zebrafish Proteins/genetics
5.
Cell Biol Int ; 33(9): 1020-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524683

ABSTRACT

Sialidase Neu2 is a glycohydrolytic enzyme whose tissue distribution has been detected principally in differentiated skeletal muscle. In this study we show that Neu2 expression is absent in different embryonal and alveolar human tumor rhabdomyosarcoma (RMS) cells, which are genetically committed myoblasts characterized by delayed differentiation. Forced myogenic differentiation of an embryonal RMS cell line, as obtained via pharmacological and genetic p38 activation or via follistatin overexpression, was characterized by Neu2 loss of expression despite the significant rise of different muscle-specific markers, suggesting therefore that the defective myogenic program of RMS cells is accompanied by Neu2 suppression.


Subject(s)
Muscle Development , Myoblasts, Skeletal/enzymology , Neuraminidase/metabolism , Rhabdomyosarcoma/enzymology , Caveolin 3/metabolism , Cell Differentiation , Cell Line, Tumor , Follistatin/metabolism , Humans , MAP Kinase Kinase 6/metabolism , Myoblasts, Skeletal/pathology , Myosin Heavy Chains/metabolism , Neuraminidase/genetics , Rhabdomyosarcoma/pathology , Tetradecanoylphorbol Acetate/pharmacology , Transfection
6.
Biochim Biophys Acta ; 1790(8): 817-28, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19371771

ABSTRACT

BACKGROUND: The sialidase Neu2 is a cytosolic enzyme which is fully expressed in mature muscle myofibers. METHODS: To investigate Neu2 expression during muscle atrophy, we employed an in vitro model consisting of terminally differentiated C2C12 myotubes exposed to different pro-atrophic stimuli that triggered catabolic pathways involved in proteasome activation or autophagy. RESULTS: Neu2 expression was unchanged in myotubes treated with TNF-alpha, a cytokine known to activate the proteasome. However, Neu2 transcript levels and enzymatic activity were downregulated in starved or dexamethasone-treated myotubes that showed proteosomal activation and several hallmarks of macroautophagy, such as formation of autophagosomes, the accumulation of LC3 dots and bulk degradation of long-lived proteins. Neu2 activity and protein levels were rescued upon cotreatment with the lysosomotropic agent NH4Cl, the autophagy inhibitor 3-methyladenine or cathepsin inhibitors, as well as by insulin administration, but were unaffected upon pharmacological inhibition of the proteasome. Moreover, HA- or GST-Neu2 recombinant fusion proteins were rapidly degraded in vitro by purified cathepsin L and B. Overall, we may conclude that Neu2 is degraded by lysosomal enzymes in myotubes undergoing autophagy-mediated atrophy. GENERAL SIGNIFICANCE: This study demonstrates that Neu2 enzyme degradation occurs in atrophic myotubes via macroautophagy and independently of proteasome activation.


Subject(s)
Autophagy , Cytosol/enzymology , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Neuraminidase/metabolism , Protein Processing, Post-Translational , Alkalies/metabolism , Animals , Atrophy , Autophagy/drug effects , Cathepsin B/metabolism , Cathepsin L , Cathepsins/metabolism , Cell Line , Cysteine Endopeptidases/metabolism , Cytosol/drug effects , Dexamethasone/pharmacology , Down-Regulation/drug effects , Insulin/pharmacology , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/ultrastructure , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure , Protein Processing, Post-Translational/drug effects , Rats , Tumor Necrosis Factor-alpha/pharmacology
7.
Biochem Biophys Res Commun ; 370(2): 376-81, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18384747

ABSTRACT

Sialidase Neu2 is an exoglycosidase that removes terminal sialic acids from glycolipids and glycoproteins. In this study, we investigated Neu2 expression during muscle hypertrophy and atrophy. Neu2 mRNA and enzymatic activity were significantly increased in hypertrophic myofibers. A rise in Neu2 activity was observed after constitutive activation of AKT or Igf-1 treatment as well as in myoblasts treated with vasopressin or trichostatin, an inhibitor of histone deacetylases. In contrast, myofiber atrophy obtained by dexamethasone treatment or starvation triggered a significant loss of Neu2 activity and was paralleled by downregulation of Neu2 transcript levels. Overall, we may conclude that Neu2 enzymatic activity is causally linked to proper muscle differentiation and growth.


Subject(s)
Muscle Development , Myoblasts/enzymology , Myoblasts/pathology , Neuraminidase/metabolism , Animals , Atrophy/chemically induced , Cell Differentiation/genetics , Dexamethasone/pharmacology , Hypertrophy , Muscle Development/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Myoblasts/drug effects , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Rats
8.
FEBS Lett ; 581(26): 5099-104, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17935719

ABSTRACT

Caveolin-3 (Cav-3) is the main scaffolding protein present in myofiber caveolae. We transfected C2C12 myoblasts with dominant negative forms of Cav-3, P104L or DeltaTFT, respectively, which cause the limb-girdle muscular dystrophy 1-C. Both these forms triggered Cav-3 loss during C2C12 cell differentiation. The P104L mutation reduced myofiber formation by impaired AKT signalling, accompanied by dramatic expression of the E3 ubiquitin ligase Atrogin. On the other hand, the DeltaTFT mutation triggered hypertrophic myotubes sustained by prolonged AKT activation, but independent of increased levels of follistatin and interleukin 4 expression. These data suggest that separated mutations within the same dystrophy-related gene may cause muscle degeneration through different mechanisms.


Subject(s)
Caveolin 3/metabolism , Cell Differentiation , Myoblasts/cytology , Animals , Caveolin 3/genetics , Humans , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Myoblasts/metabolism , Phenotype , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Biochem J ; 408(2): 211-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17708748

ABSTRACT

Sialidase NEU3 is also known as the plasma-membrane-associated form of mammalian sialidases, exhibiting a high substrate specificity towards gangliosides. In this respect, sialidase NEU3 modulates cell-surface biological events and plays a pivotal role in different cellular processes, including cell adhesion, recognition and differentiation. At the moment, no detailed studies concerning the subcellular localization of NEU3 are available, and the mechanism of its association with cellular membranes is still unknown. In the present study, we have demonstrated that sialidase NEU3, besides its localization at the plasma membrane, is present in intracellular structures at least partially represented by a subset of the endosomal compartment. Moreover, we have shown that NEU3 present at the plasma membrane is internalized and locates then to the recycling endosomal compartment. The enzyme is associated with the outer leaflet of the plasma membrane, as shown by selective cell-surface protein biotinylation. This evidence is in agreement with the ability of NEU3 to degrade gangliosides inserted into the plasma membrane of adjacent cells. Moreover, the mechanism of the protein association with the lipid bilayer was elucidated by carbonate extraction. Under these experimental conditions, we have succeeded in solubilizing NEU3, thus demonstrating that the enzyme is a peripheral membrane protein. In addition, Triton X-114 phase separation demonstrates further the hydrophilic nature of the protein. Overall, these results provide important information about the biology of NEU3, the most studied member of the mammalian sialidase family.


Subject(s)
Cell Membrane/enzymology , Endosomes/enzymology , Membrane Proteins/metabolism , Neuraminidase/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endosomes/metabolism , HeLa Cells , Humans , Membrane Proteins/chemistry , Neuraminidase/chemistry
10.
Biochem J ; 408(3): 395-406, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17708749

ABSTRACT

Sialidases remove sialic acid residues from various sialo-derivatives. To gain further insights into the biological roles of sialidases in vertebrates, we exploited zebrafish (Danio rerio) as an animal model. A zebrafish transcriptome- and genome-wide search using the sequences of the human NEU polypeptides as templates revealed the presence of seven different genes related to human sialidases. neu1 and neu4 are the putative orthologues of the mammalian sialidases NEU1 and NEU4 respectively. Interestingly, the remaining genes are organized in clusters located on chromosome 21 and are all more closely related to mammalian sialidase NEU3. They were thus named neu3.1, neu3.2, neu3.3, neu3.4 and neu3.5. Using RT-PCR (reverse transcription-PCR) we detected transcripts for all genes, apart from neu3.4, and whole-mount in situ hybridization experiments show a localized expression pattern in gut and lens for neu3.1 and neu4 respectively. Transfection experiments in COS7 (monkey kidney) cells demonstrate that Neu3.1, Neu3.2, Neu3.3 and Neu4 zebrafish proteins are sialidase enzymes. Neu3.1, Neu3.3 and Neu4 are membrane-associated and show a very acidic pH optimum below 3.0, whereas Neu3.2 is a soluble sialidase with a pH optimum of 5.6. These results were further confirmed by subcellular localization studies carried out using immunofluorescence. Moreover, expression in COS7 cells of these novel zebrafish sialidases (with the exception of Neu3.2) induces a significant modification of the ganglioside pattern, consistent with the results obtained with membrane-associated mammalian sialidases. Overall, the redundancy of sialidases together with their expression profile and their activity exerted on gangliosides of living cells indicate the biological relevance of this class of enzymes in zebrafish.


Subject(s)
Isoenzymes/metabolism , Neuraminidase/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA Primers , DNA, Complementary , In Situ Hybridization , Isoenzymes/genetics , Molecular Sequence Data , Neuraminidase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/enzymology , Zebrafish
11.
Biochem Biophys Res Commun ; 357(1): 314-8, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17418092

ABSTRACT

Caveolin-3 (Cav-3) is a muscle-specific membrane protein crucial for myoblast differentiation, as loss of the protein due to mutations within the gene causes an autosomal dominant form of limb girdle muscular dystrophy 1-c. Here we show that along with p38 activity the PI3-kinase/AKT/mTOR pathway is required for proper Cav-3 up-regulation during muscle differentiation and hypertrophy, as confirmed by the marked increase of Cav-3 expression in hypertrophied C2C12 cells transfected with an activated form of AKT. Accordingly, Cav-3 expression was further increased during hypertrophy of L6C5 myoblasts treated with Arg(8)-vasopressin and in hypertrophic muscles of MLC/mIGF-1 transgenic mice. In contrast, Cav-3 expression was down-regulated in C2C12 myotubes exposed to atrophic stimuli such as starvation or treatment with dexamethasone. This study clearly suggests that Cav-3 expression is causally linked to the maturation of muscle phenotype and it is tightly regulated by hypertrophic and atrophic stimuli.


Subject(s)
Caveolin 3/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myoblasts/metabolism , Myoblasts/pathology , Animals , Cell Line , Hypertrophy/metabolism , Hypertrophy/pathology , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Statistics as Topic
12.
FEBS J ; 273(16): 3709-21, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16869890

ABSTRACT

Cytosolic sialidase (neuraminidase 2; Neu2) is an enzyme whose expression increases during myoblast differentiation. Here we show that insulin-like growth factor 1 (IGF1)-induced hypertrophy of myoblasts notably increases Neu2 synthesis by activation of the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (P13K/AKT/mTOR) pathway, whereas the proliferative effect mediated by activation of the extracellular regulated kinase 1/2 (ERK1/2) pathway negatively contributed to Neu2 activity. Accordingly, the differentiation L6MLC/IGF-1 cell line, in which the forced postmitotic expression of insulin-like growth factor 1 stimulates a dramatic hypertrophy, was accompanied by a stronger Neu2 increase. Indeed, the hypertrophy induced by transfection of a constitutively activated form of AKT was able to induce high Neu2 activity in C2C12 cells, whereas the transfection of a kinase-inactive form of AKT prevented myotube formation, triggering Neu2 downregulation. Neu2 expression was strictly correlated with IGF-1 signaling also in C2 myoblasts overexpressing the insulin-like growth factor 1 binding protein 5 and therefore not responding to endogenously produced insulin-like growth factor 1. Although Neu2-transfected myoblasts exhibited stronger differentiation, we demonstrated that Neu2 overexpression does not override the block of differentiation mediated by PI3 kinase and mTOR inhibitors. Finally, Neu2 overexpression did not modify the ganglioside pattern of C2C12 cells, suggesting that glycoproteins might be the target of Neu2 activity. Taken together, our data demonstrate that IGF-1-induced differentiation and hypertrophy are driven, at least in part, by Neu2 upregulation and further support the significant role of cytosolic sialidase in myoblasts.


Subject(s)
Insulin-Like Growth Factor I/physiology , Myoblasts/cytology , Myoblasts/metabolism , Neuraminidase/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Cytosol/enzymology , Enzyme Inhibitors/pharmacology , Hypertrophy , Insulin-Like Growth Factor I/pharmacology , Mice , Myoblasts/drug effects , Neuraminidase/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Up-Regulation
13.
FASEB J ; 20(8): 1227-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16645048

ABSTRACT

Ceramide is a key lipid molecule necessary to regulate some cellular processes, including apoptosis and cell differentiation. In this context, its production has been shown to occur via sphingomyelin hydrolysis or sphingosine acylation. Here, we show that in human fibroblasts, plasma membrane ceramide is also produced from ganglioside GM3 by detachment of sugar units. Membrane-bound glycosylhydrolases have a role in this process. In fact, the production of ceramide from GM3 has been observed even under experimental conditions able to block endocytosis or lysosomal activity, and the overexpression of the plasma membrane ganglioside sialidase Neu3 corresponded to a higher production of ceramide in the plasma membrane. The increased activity of Neu3 was paralleled by an increase of GM3 synthase mRNA and GM3 synthase activity. Neu3-overexpressing fibroblasts were characterized by a reduced proliferation rate and higher basal number of apoptotic cells in comparison with wild-type cells. A similar behavior was observed when normal fibroblasts were treated with exogenous C2-ceramide.


Subject(s)
Cell Membrane/enzymology , Ceramides/metabolism , Fibroblasts/enzymology , G(M3) Ganglioside/metabolism , Neuraminidase/metabolism , Animals , Apoptosis , Cells, Cultured , Fibroblasts/cytology , Humans , Mice , Neuraminidase/analysis , Neuraminidase/genetics , RNA, Messenger/metabolism , Sphingolipids/chemistry
14.
J Biol Chem ; 279(17): 16989-95, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14970224

ABSTRACT

We describe herein the enzyme behavior of MmNEU3, the plasma membrane-associated sialidase from mouse (Mus musculus). MmNEU3 is localized at the plasma membrane as demonstrated directly by confocal microscopy analysis. In addition, administration of the radiolabeled ganglioside GD1a to MmNEU3-transfected cells, under conditions that prevent lysosomal activity, led to its hydrolysis into ganglioside GM1, further indicating the plasma membrane topology of MmNEU3. Metabolic labeling with [1-(3)H]sphingosine allowed the characterization of the ganglioside patterns of COS-7 cells. MmNEU3 expression in COS-7 cells led to an extensive modification of the cell ganglioside pattern, i.e. GM3 and GD1a content was decreased to about one-third compared with mock-transfected cells. At the same time, a 35% increase in ganglioside GM1 content was observed. Mixed culture of MmNEU3-transfected cells with [1-(3)H]sphingosine-labeled cells demonstrates that the enzyme present at the cell surface is able to recognize gangliosides exposed on the membrane of nearby cells. Under these experimental conditions, the extent of ganglioside pattern changes was a function of MmNEU3 transient expression. Overall, the variations in GM3, GD1a, and GM1 content were very similar to those observed in the case of [1-(3)H]sphingosine-labeled MmNEU3-transfected cells, indicating that the enzyme mainly exerted its activity toward ganglioside substrates present at the surface of neighboring cells. These results indicate that the plasma membrane-associated sialidase MmNEU3 is able to hydrolyze ganglioside substrates in intact living cells at a neutral pH, mainly through cell-to-cell interactions.


Subject(s)
Gangliosides/metabolism , Neuraminidase/physiology , Animals , COS Cells , Cattle , Cell Communication , Cell Membrane/metabolism , Cells, Cultured , Coculture Techniques , G(M1) Ganglioside/chemistry , Gangliosides/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Lipid Metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Neuraminidase/metabolism , Sphingosine/chemistry , Transfection
15.
J Biol Chem ; 279(5): 3169-79, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14613940

ABSTRACT

Recombinant human cytosolic sialidase (HsNEU2), expressed in Escherichia coli, was purified to homogeneity, and its substrate specificity was studied. HsNEU2 hydrolyzed 4-methylumbelliferyl alpha-NeuAc, alpha 2-->3 sialyllactose, glycoproteins (fetuin, alpha-acid glycoprotein, transferrin, and bovine submaxillary gland mucin), micellar gangliosides GD1a, GD1b, GT1b, and alpha 2-->3 paragloboside, and vesicular GM3. alpha 2-->6 sialyllactose, colominic acid, GM1 oligosaccharide, whereas micellar GM2 and GM1 were resistant. The optimal pH was 5.6, kinetics Michaelis-Menten type, V(max) varying from 250 IU/mg protein (GD1a) to 0.7 IU/mg protein (alpha(1)-acid glycoprotein), and K(m) in the millimolar range. HsNEU2 was activated by detergents (Triton X-100) only with gangliosidic substrates; the change of GM3 from vesicular to mixed micellar aggregation led to a 8.5-fold V(max) increase. HsNEU2 acted on gangliosides (GD1a, GM1, and GM2) at nanomolar concentrations. With these dispersions (studied in detailed on GM1), where monomers are bound to the tube wall or dilutedly associated (1:2000, mol/mol) to Triton X-100 micelles, the V(max) values were 25 and 72 microIU/mg protein, and K(m) was 10 and 15 x 10(-9) m, respectively. Remarkably, GM1 and GM2 were recognized only as monomers. HsNEU2 worked at pH 7.0 with an efficiency (compared with that at pH 5.6) ranging from 4% (on GD1a) to 64% (on alpha(1)-acid glycoprotein), from 7% (on GD1a) to 45% (on GM3) in the presence of Triton X-100, and from 30 to 40% on GM1 monomeric dispersion. These results show that HsNEU2 differentially recognizes the type of sialosyl linkage, the aglycone part of the substrate, and the supramolecular organization (monomer/micelle/vesicle) of gangliosides. The last ability might be relevant in sialidase interactions with gangliosides under physiological conditions.


Subject(s)
G(M1) Ganglioside/chemistry , Neuraminidase/chemistry , Recombinant Proteins/chemistry , Animals , Cattle , Detergents/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , G(M1) Ganglioside/metabolism , Gangliosides/chemistry , Glycolipids/chemistry , Glycoproteins/chemistry , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Micelles , Molecular Sequence Data , Neuraminidase/metabolism , Octoxynol/pharmacology , Temperature , Time Factors
16.
Neurochem Res ; 27(7-8): 649-63, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12374200

ABSTRACT

This review summarizes the recent research development on mammalian sialidase molecular cloning. Sialic acid-containing compounds are involved in several physiological processes, and sialidases, as glycohydrolytic enzymes that remove sialic acid residues, play a pivotal role as well. Sialidases hydrolyze the nonreducing, terminal sialic acid linkage in various natural substrates, such as glycoproteins, glycolipids, gangliosides, and polysaccharides. Mammalian sialidases are present in several tissues/organs and cells with a typical subcellular distribution: they are the lysosomal, the cytosolic, and the plasma membrane-associated sialidases. Starting in 1993, 12 different mammalian sialidases have been cloned and sequenced. A comparison of their amino acid sequences revealed the presence of highly conserved regions. These conserved regions are shared with viral and microbial sialidases that have been characterized at three-dimensional structural level, allowing us to perform the molecular modeling of the mammalian proteins and suggesting a monophyletic origin of the sialidase enzymes. Overall, the availability of the cDNA species encoding mammalian sialidases is an important step leading toward a comprehensive picture of the relationships between the structure and biological function of these enzymes.


Subject(s)
Neuraminidase/genetics , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cell Membrane/enzymology , Cloning, Molecular , Humans , Mammals , Molecular Sequence Data , Mucolipidoses/enzymology , Mutagenesis, Site-Directed , Neoplasms/enzymology , Neuraminidase/chemistry , Neuraminidase/physiology , Sequence Homology, Amino Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...