Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 5: 17705, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26635098

ABSTRACT

To explore the general requirement of endothelial mTORC2 during embryonic and adolescent development, we knocked out the essential mTORC2 component Rictor in the mouse endothelium in the embryo, during adolescence and in endothelial cells in vitro. During embryonic development, Rictor knockout resulted in growth retardation and lethality around embryonic day 12. We detected reduced peripheral vascularization and delayed ossification of developing fingers, toes and vertebrae during this confined midgestational period. Rictor knockout did not affect viability, weight gain, and vascular development during further adolescence. However during this period, Rictor knockout prevented skin capillaries to gain larger and heterogeneously sized diameters and remodeling into tortuous vessels in response to FGF2. Rictor knockout strongly reduced extensive FGF2-induced neovascularization and prevented hemorrhage in FGF2-loaded matrigel plugs. Rictor knockout also disabled the formation of capillary-like networks by FGF2-stimulated mouse aortic endothelial cells in vitro. Low RICTOR expression was detected in quiescent, confluent mouse aortic endothelial cells, whereas high doses of FGF2 induced high RICTOR expression that was associated with strong mTORC2-specific protein kinase Cα and AKT phosphorylation. We demonstrate that the endothelial FGF-RICTOR axis is not required during endothelial quiescence, but crucial for midgestational development and sustained and extensive neovascularization in the adult.


Subject(s)
Carrier Proteins/biosynthesis , Embryonic Development/genetics , Fibroblast Growth Factor 2/genetics , Neovascularization, Physiologic/genetics , Animals , Carrier Proteins/genetics , Endothelium/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Developmental , Hemorrhage/genetics , Hemorrhage/pathology , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-akt/genetics , Rapamycin-Insensitive Companion of mTOR Protein , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
PLoS One ; 10(10): e0139853, 2015.
Article in English | MEDLINE | ID: mdl-26448642

ABSTRACT

INTRODUCTION: Sulf1 and Sulf2 are cell surface sulfatases, which remove specific 6-O-sulfate groups from heparan sulfate (HS) proteoglycans, resulting in modulation of various HS-dependent signaling pathways. Both Sulf1 and Sulf2 knockout mice show impairments in brain development and neurite outgrowth deficits in neurons. METHODOLOGY AND MAIN FINDINGS: To analyze the molecular mechanisms behind these impairments we focused on the postnatal cerebellum, whose development is mainly characterized by proliferation, migration, and neurite outgrowth processes of precursor neurons. Primary cerebellar granule cells isolated from Sulf1 or Sulf2 deficient newborns are characterized by a reduction in neurite length and cell survival. Furthermore, Sulf1 deficiency leads to a reduced migration capacity. The observed impairments in cell survival and neurite outgrowth could be correlated to Sulf-specific interference with signaling pathways, as shown for FGF2, GDNF and NGF. In contrast, signaling of Shh, which determines the laminar organization of the cerebellar cortex, was not influenced in either Sulf1 or Sulf2 knockouts. Biochemical analysis of cerebellar HS demonstrated, for the first time in vivo, Sulf-specific changes of 6-O-, 2-O- and N-sulfation in the knockouts. Changes of a particular HS epitope were found on the surface of Sulf2-deficient cerebellar neurons. This epitope showed a restricted localization to the inner half of the external granular layer of the postnatal cerebellum, where precursor cells undergo final maturation to form synaptic contacts. CONCLUSION: Sulfs introduce dynamic changes in HS proteoglycan sulfation patterns of the postnatal cerebellum, thereby orchestrating fundamental mechanisms underlying brain development.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Neurites/physiology , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Female , Fibroblast Growth Factor 2/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Hedgehog Proteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurons/metabolism , Signal Transduction , Sulfatases/deficiency , Sulfatases/genetics , Sulfotransferases/deficiency , Sulfotransferases/genetics
3.
PLoS One ; 8(8): e69642, 2013.
Article in English | MEDLINE | ID: mdl-23950901

ABSTRACT

Corneal epithelial wound repair involves the migration of epithelial cells to cover the defect followed by the proliferation of the cells to restore thickness. Heparan sulfate proteoglycans (HSPGs) are ubiquitous extracellular molecules that bind to a plethora of growth factors, cytokines, and morphogens and thereby regulate their signaling functions. Ligand binding by HS chains depends on the pattern of four sulfation modifications, one of which is 6-O-sulfation of glucosamine (6OS). SULF1 and SULF2 are highly homologous, extracellular endosulfatases, which post-synthetically edit the sulfation status of HS by removing 6OS from intact chains. The SULFs thereby modulate multiple signaling pathways including the augmentation of Wnt/ß-catenin signaling. We found that wounding of mouse corneal epithelium stimulated SULF1 expression in superficial epithelial cells proximal to the wound edge. Sulf1⁻/⁻, but not Sulf2⁻/⁻, mice, exhibited a marked delay in healing. Furthermore, corneal epithelial cells derived from Sulf1⁻/⁻ mice exhibited a reduced rate of migration in repair of a scratched monolayer compared to wild-type cells. In contrast, human primary corneal epithelial cells expressed SULF2, as did a human corneal epithelial cell line (THCE). Knockdown of SULF2 in THCE cells also slowed migration, which was restored by overexpression of either mouse SULF2 or human SULF1. The interchangeability of the two SULFs establishes their capacity for functional redundancy. Knockdown of SULF2 decreased Wnt/ß-catenin signaling in THCE cells. Extracellular antagonists of Wnt signaling reduced migration of THCE cells. However in SULF2- knockdown cells, these antagonists exerted no further effects on migration, consistent with the SULF functioning as an upstream regulator of Wnt signaling. Further understanding of the mechanistic action of the SULFs in promoting corneal repair may lead to new therapeutic approaches for the treatment of corneal injuries.


Subject(s)
Cornea/cytology , Heparitin Sulfate/metabolism , Sulfatases/metabolism , Sulfotransferases/metabolism , Wound Healing , Animals , Cell Movement , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Signal Transduction , Sulfatases/genetics , Sulfotransferases/genetics , Up-Regulation , Wnt Proteins/metabolism , beta Catenin/metabolism
4.
J Biol Chem ; 288(42): 30019-30028, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-23986440

ABSTRACT

The human sulfatase family has 17 members, 13 of which have been characterized biochemically. These enzymes specifically hydrolyze sulfate esters in glycosaminoglycans, sulfolipids, or steroid sulfates, thereby playing key roles in cellular degradation, cell signaling, and hormone regulation. The loss of sulfatase activity has been linked to severe pathophysiological conditions such as lysosomal storage disorders, developmental abnormalities, or cancer. A novel member of this family, arylsulfatase K (ARSK), was identified bioinformatically through its conserved sulfatase signature sequence directing posttranslational generation of the catalytic formylglycine residue in sulfatases. However, overall sequence identity of ARSK with other human sulfatases is low (18-22%). Here we demonstrate that ARSK indeed shows desulfation activity toward arylsulfate pseudosubstrates. When expressed in human cells, ARSK was detected as a 68-kDa glycoprotein carrying at least four N-glycans of both the complex and high-mannose type. Purified ARSK turned over p-nitrocatechol and p-nitrophenyl sulfate. This activity was dependent on cysteine 80, which was verified to undergo conversion to formylglycine. Kinetic parameters were similar to those of several lysosomal sulfatases involved in degradation of sulfated glycosaminoglycans. An acidic pH optimum (~4.6) and colocalization with LAMP1 verified lysosomal functioning of ARSK. Further, it carries mannose 6-phosphate, indicating lysosomal sorting via mannose 6-phosphate receptors. ARSK mRNA expression was found in all tissues tested, suggesting a ubiquitous physiological substrate and a so far non-classified lysosomal storage disorder in the case of ARSK deficiency, as shown before for all other lysosomal sulfatases.


Subject(s)
Arylsulfatases , Lysosomes/enzymology , Arylsulfatases/biosynthesis , Arylsulfatases/chemistry , Arylsulfatases/genetics , Arylsulfatases/isolation & purification , Gene Expression , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/isolation & purification , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Substrate Specificity/genetics
5.
Cell Physiol Biochem ; 30(4): 1083-96, 2012.
Article in English | MEDLINE | ID: mdl-23202547

ABSTRACT

BACKGROUND: PIM1 is a constitutively active serine-threonine kinase regulating cell survival and proliferation. Increased PIM1 expression has been correlated with cancer metastasis by facilitating migration and anti-adhesion. Endothelial cells play a pivotal role in these processes by contributing a barrier to the blood stream. Here, we investigated whether PIM1 regulates mouse aortic endothelial cell (MAEC) monolayer integrity. METHODS: Pim1-/-MAEC were isolated from Pim1 knockout mice and used in trypsinization-, wound closure assays, electrical cell-substrate sensing, immunostaining, cDNA transfection and as RNA source for microarray analysis. RESULTS: Pim1-/-MAEC displayed decreased migration, slowed cell detachment and increased electrical resistance across the endothelial monolayer. Reintroduction of Pim1- cDNA into Pim1-/-MAEC significantly restored wildtype adhesive characteristics. Pim1-/--MAEC displayed enhanced focal adhesion and adherens junction structures containing vinculin and ß-catenin, respectively. Junctional molecules such as Cadherin 13 and matrix components such as Collagen 6a3 were highly upregulated in Pim1-/- cells. Intriguingly, extracellular matrix deposited by Pim1-/- cells alone was sufficient to induce the hyperadhesive phenotype in wildtype endothelial cells. CONCLUSION: Loss of Pim1 induces a strong adhesive phenotype by enhancing endothelial cell-cell and cell-matrix adhesion by the deposition of a specific extracellular matrix. Targeting PIM1 function therefore might be important to promote endothelial barrier integrity.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Adherens Junctions/metabolism , Animals , Aorta/cytology , Cell Adhesion , Cell Movement , Cells, Cultured , Focal Adhesions/metabolism , Gene Deletion , Humans , Mice , Mice, Knockout , Proto-Oncogene Proteins c-pim-1/metabolism , Transfection , Wound Healing
6.
Biochem Biophys Res Commun ; 429(1-2): 24-30, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23131564

ABSTRACT

The PIM serine/threonine kinases and the mTOR/AKT pathway integrate growth factor signaling and promote cell proliferation and survival. They both share phosphorylation targets and have overlapping functions, which can partially substitute for each other. In cancer cells PIM kinases have been reported to produce resistance to mTOR inhibition by rapamycin. Tumor growth depends highly on blood vessel infiltration into the malignant tissue and therefore on endothelial cell proliferation. We therefore investigated how the PIM1 kinase modulates growth inhibitory effects of rapamycin in mouse aortic endothelial cells (MAEC). We found that proliferation of MAEC lacking Pim1 was significantly more sensitive to rapamycin inhibition, compared to wildtype cells. Inhibition of mTOR and AKT in normal MAEC resulted in significantly elevated PIM1 protein levels in the cytosol and in the nucleus. We observed that truncation of the C-terminal part of Pim1 beyond Ser 276 resulted in almost exclusive nuclear localization of the protein. Re-expression of this Pim1 deletion mutant significantly increased the proliferation of Pim1(-/-) cells when compared to expression of the wildtype Pim1 cDNA. Finally, overexpression of the nuclear localization mutant and the wildtype Pim1 resulted in complete resistance to growth inhibition by rapamycin. Thus, mTOR inhibition-induced nuclear accumulation of PIM1 or expression of a nuclear C-terminal PIM1 truncation mutant is sufficient to increase endothelial cell proliferation, suggesting that nuclear localization of PIM1 is important for resistance of MAEC to rapamycin-mediated inhibition of proliferation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/enzymology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-pim-1/metabolism , Sirolimus/pharmacology , Animals , Cells, Cultured , Endothelial Cells/drug effects , Mice , Proto-Oncogene Proteins c-pim-1/genetics , Sequence Deletion
7.
Proc Natl Acad Sci U S A ; 109(26): 10310-5, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22689975

ABSTRACT

Deficiency of glycosaminoglycan (GAG) degradation causes a subclass of lysosomal storage disorders called mucopolysaccharidoses (MPSs), many of which present with severe neuropathology. Critical steps in the degradation of the GAG heparan sulfate remain enigmatic. Here we show that the lysosomal arylsulfatase G (ARSG) is the long-sought glucosamine-3-O-sulfatase required to complete the degradation of heparan sulfate. Arsg-deficient mice accumulate heparan sulfate in visceral organs and the central nervous system and develop neuronal cell death and behavioral deficits. This accumulated heparan sulfate exhibits unique nonreducing end structures with terminal N-sulfoglucosamine-3-O-sulfate residues, allowing diagnosis of the disorder. Recombinant human ARSG is able to cleave 3-O-sulfate groups from these residues as well as from an authentic 3-O-sulfated N-sulfoglucosamine standard. Our results demonstrate the key role of ARSG in heparan sulfate degradation and strongly suggest that ARSG deficiency represents a unique, as yet unknown form of MPS, which we term MPS IIIE.


Subject(s)
Arylsulfatases/antagonists & inhibitors , Mucopolysaccharidoses/etiology , Sulfatases/metabolism , Animals , Behavior, Animal , Mice , Mucopolysaccharidoses/enzymology
8.
J Biol Chem ; 287(15): 12217-29, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22351753

ABSTRACT

Cell surface heparan sulfate (HS) is an essential regulator of cell signaling and development. HS traps signaling molecules, like Wnt in the glycosaminoglycan side chains of HS proteoglycans (HSPGs), and regulates their functions. Endosulfatases Sulf1 and Sulf2 are secreted at the cell surface to selectively remove 6-O-sulfate groups from HSPGs, thereby modifying the affinity of cell surface HSPGs for its ligands. This study provides molecular evidence for the functional roles of HSPG sulfation and desulfation in dentinogenesis. We show that odontogenic cells are highly sulfated on the cell surface and become desulfated during their differentiation to odontoblasts, which produce tooth dentin. Sulf1/Sulf2 double null mutant mice exhibit a thin dentin matrix and short roots combined with reduced expression of dentin sialophosphoprotein (Dspp) mRNA, encoding a dentin-specific extracellular matrix precursor protein, whereas single Sulf mutants do not show such defective phenotypes. In odontoblast cell lines, Dspp mRNA expression is potentiated by the activation of the Wnt canonical signaling pathway. In addition, pharmacological interference with HS sulfation promotes Dspp mRNA expression through activation of Wnt signaling. On the contrary, the silencing of Sulf suppresses the Wnt signaling pathway and subsequently Dspp mRNA expression. We also show that Wnt10a protein binds to cell surface HSPGs in odontoblasts, and interference with HS sulfation decreases the binding affinity of Wnt10a for HSPGs, which facilitates the binding of Wnt10a to its receptor and potentiates the Wnt signaling pathway, thereby up-regulating Dspp mRNA expression. These results demonstrate that Sulf-mediated desulfation of cellular HSPGs is an important modification that is critical for the activation of the Wnt signaling in odontoblasts and for production of the dentin matrix.


Subject(s)
Dentinogenesis , Heparitin Sulfate/physiology , Animals , Cells, Cultured , Dentin/growth & development , Dentin/metabolism , Dentin/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Mice , Mice, Knockout , Molar/growth & development , Molar/metabolism , Molar/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Odontoblasts/metabolism , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Sulfatases/genetics , Sulfatases/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Tooth Abnormalities/enzymology , Tooth Abnormalities/genetics , Transcription, Genetic , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Proteins/physiology , Wnt Signaling Pathway
9.
J Neurosci ; 30(32): 10784-98, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20702708

ABSTRACT

The neural cell adhesion molecule NCAM plays important functional roles not only during nervous system development, but also in the adult after injury and in synaptic plasticity. Homophilic binding of NCAM triggers intracellular signaling events resulting in cellular responses such as neurite outgrowth that require NCAM palmitoylation-dependent raft localization and activation of the nonreceptor tyrosine kinases fyn and fak. In this study, we show that stimulation of NCAM by a function-triggering NCAM antibody results in proteolytic processing of NCAM and fak. The C-terminal fragment of NCAM, consisting of the intracellular domain, the transmembrane domain, and a stub of the extracellular domain, and the N-terminal fragment of fak are imported into the nucleus. NCAM-stimulated fak activation, generation, and nuclear import of NCAM and fak fragments as well as neurite outgrowth are abolished by mutation of the calmodulin binding motif in the intracellular domain of NCAM that is responsible for the calcium-dependent binding of calmodulin to NCAM. This mutation interferes neither with NCAM cell surface expression, palmitoylation, and raft localization nor with fyn activation. The way by which the transmembrane NCAM fragment reaches the nucleus in a calmodulin- and calcium-dependent manner is by endocytotic transport via the endoplasmic reticulum and the cytoplasm. The generation and nuclear import of NCAM and phosphorylated fak fragments resulting from NCAM stimulation may represent a signal pathway activating cellular responses in parallel or in association with classical kinase- and phosphorylation-dependent signaling cascades.


Subject(s)
Calmodulin/metabolism , Cell Nucleolus/metabolism , Focal Adhesion Kinase 1/metabolism , Neural Cell Adhesion Molecules/physiology , Neurites/physiology , Neurons/cytology , Protein Interaction Domains and Motifs/physiology , Analysis of Variance , Animals , Anthraquinones/metabolism , Antibodies/pharmacology , Benzimidazoles/pharmacology , Biotinylation/methods , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Cell Nucleolus/drug effects , Cell Nucleolus/genetics , Cells, Cultured , Cerebellum/cytology , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Focal Adhesion Kinase 1/chemistry , Gene Expression Regulation/genetics , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Mutagenesis, Site-Directed/methods , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/immunology , Protein Binding/genetics , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/genetics , Surface Plasmon Resonance/methods , Transfection/methods
10.
J Biol Chem ; 284(38): 25714-22, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19596853

ABSTRACT

Studies on glycosaminoglycans and proteoglycans (PGs) have been hampered by difficulties in isolation and analysis by traditional methods that are laborious and lack sensitivity and throughput. Here we demonstrate a simple method for rapid isolation of proteoglycans (RIP) employing phenol/guanidine/chloroform reagent to purify heparan sulfate (HS) PGs quantitatively from various tissues and cells. We further show that this generic purification methodology, when applied in concert with a BODIPY fluorescent label, permits structural analyses on RIP-purified HS at approximately 1,000-fold higher sensitivity than standard UV detection methods and approximately 10-100-fold higher sensitivity than previous fluorescence detection methods. The utility of RIP-BODIPY methodology was demonstrated by rapid profiling of HS structural composition from small tissue samples, multiple mouse organs, and as little as a few thousand cultured cells. It was also used to generate novel insights into in vivo structural changes in HS from Sulf1 knock-out mice for the first time that differed significantly from previous observations limited to tissue culture experiments. RIP was also applied to purify HS for bioassay testing, exemplified by cell assays of fibroblast growth factor signaling activation; this generated data from 2-O-sulfotransferase knock-out mice and revealed an unexpected deficiency in fibroblast growth factor activation by HS from heterozygous mice. These data demonstrate that RIP will underpin emerging efforts to develop glycomics profiling strategies for HS and other glycosaminoglycans to explore their structure-function relationships in complex biological systems.


Subject(s)
Heparitin Sulfate/analysis , Heparitin Sulfate/isolation & purification , 3T3 Cells , Animals , Boron Compounds/chemistry , Female , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Heparitin Sulfate/metabolism , Mice , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfotransferases/genetics , Sulfotransferases/metabolism
11.
J Cell Mol Med ; 13(11-12): 4505-21, 2009.
Article in English | MEDLINE | ID: mdl-20394677

ABSTRACT

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.


Subject(s)
Behavior, Animal , Brain/embryology , Brain/enzymology , Neuronal Plasticity , Neurons/enzymology , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Animals, Newborn , Embryo Loss/enzymology , Embryo Loss/pathology , Embryo Loss/physiopathology , Extracellular Space/enzymology , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/ultrastructure , Hydrocephalus/complications , Hydrocephalus/enzymology , Hydrocephalus/pathology , Hydrocephalus/physiopathology , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Nervous System Malformations/complications , Nervous System Malformations/enzymology , Nervous System Malformations/physiopathology , Neurites/enzymology , Neurites/pathology , Neurons/pathology , Phenotype , Sulfatases/deficiency , Sulfotransferases/deficiency , Synaptic Transmission/physiology
12.
Dev Dyn ; 237(2): 339-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18213582

ABSTRACT

Modification of the sulfation pattern of heparan sulfate (HS) during organ development is thought to regulate binding and signal transduction of several growth factors. The secreted sulfatases, Sulf1 and Sulf2, desulfate HS on 6-O-positions extracellularly. We show that both sulfatases are expressed in overlapping patterns during embryonic skeletal development. Analysis of compound mutants of Sulf1 and Sulf2 derived from gene trap insertions and targeted null alleles revealed subtle but distinct skeletal malformations including reduced bone length, premature vertebrae ossification and fusions of sternebrae and tail vertebrae. Molecular analysis of endochondral ossification points to a function of Sulf1 and Sulf2 in delaying the differentiation of endochondral bones. Penetrance and severity of the phenotype increased with reduced numbers of functional alleles indicating redundant functions of both sulfatases. The mild skeletal phenotype of double mutants suggests a role for extracellular modification of 6-O-sulfation in fine-tuning rather than regulating the development of skeletal structures.


Subject(s)
Bone and Bones/embryology , Heparitin Sulfate/metabolism , Osteogenesis/physiology , Signal Transduction/physiology , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , DNA Primers/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mutation/genetics , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sulfatases/genetics , Sulfotransferases/genetics
13.
J Biotechnol ; 129(2): 290-307, 2007 Apr 30.
Article in English | MEDLINE | ID: mdl-17337080

ABSTRACT

Heparan sulfate (HS) is a cell surface carbohydrate polymer modified with sulfate moieties whose highly ordered composition is central to directing specific cell signaling events. The ability of the cell to generate these information rich glycans with such specificity has opened up a new field of "heparanomics" which seeks to understand the systems involved in generating these cell type and developmental stage specific HS sulfation patterns. Unlike other instances where biological information is encrypted as linear sequences in molecules such as DNA, HS sulfation patterns are generated through a non-template driven process. Thus, deciphering the sulfation code and the dynamic nature of its generation has posed a new challenge to system biologists. The recent discovery of two sulfatases, Sulf1 and Sulf2, with the unique ability to edit sulfation patterns at the cell surface, has opened up a new dimension as to how we understand the regulation of HS sulfation patterning and pattern-dependent cell signaling events. This review will focus on the functional relationship between HS sulfation patterning and biological processes. Special attention will be given to Sulf1 and Sulf2 and how these key editing enzymes might act in concert with the HS biosynthetic enzymes to generate and regulate specific HS sulfation patterns in vivo. We will further explore the use of knock out mice as biological models for understanding the dynamic systems involved in generating HS sulfation patterns and their biological relevance. A brief overview of new technologies and innovations summarizes advances in the systems biology field for understanding non-template molecular networks and their influence on the "heparanome".


Subject(s)
Cell Communication/physiology , Heparan Sulfate Proteoglycans/metabolism , Animals , Heparan Sulfate Proteoglycans/genetics , Mice , Mice, Knockout , Sulfatases/genetics , Sulfatases/metabolism , Systems Biology
14.
Biochem J ; 400(1): 63-73, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16901266

ABSTRACT

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.


Subject(s)
Heparitin Sulfate/metabolism , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Disaccharides/analysis , Epitopes/immunology , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genotype , Heparitin Sulfate/chemistry , Heparitin Sulfate/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Structure , Oligosaccharides/analysis , Sulfatases/genetics , Sulfates/metabolism , Sulfotransferases/genetics
15.
J Neurochem ; 98(1): 78-88, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805798

ABSTRACT

The transmembrane and multidomain neural cell adhesion molecule (NCAM) plays important functional roles in the developing and adult nervous system. NCAM is proteolytically processed and appears in soluble forms in the cerebrospinal fluid and in serum under normal and pathological conditions. In this report, we present evidence that the metalloprotease a disintegrin and a metalloprotease (ADAM)17/tumour necrosis factor alpha converting enzyme (TACE) cleaves the polysialylated as well as the non-polysialylated transmembrane isoforms of NCAM, whereas the glycophosphatidylinositol-linked isoform of NCAM is not proteolytically cleaved. A truncated, enzymatically inactive mutant of TACE did not result in release of the NCAM110 cleavage product. Proteolytic cleavage was enhanced by a calmodulin-specific inhibitor and the actin-destabilizing agents cytochalasin D and latrunculin B. In contrast, the microtubule-stabilizing agent colchicine or microtubule-destabilizing agent paclitaxel did not affect the release of the 110-kDa fragment of NCAM. Neurite outgrowth from cerebellar microexplants was inhibited in the presence of the metalloprotease inhibitor GM 6001 on substrate-coated NCAM, but not on poly-l-lysine. Upon transfection of hippocampal neurones with an enzymatically inactive mutant of TACE, NCAM-stimulated neurite outgrowth was inhibited without affecting neurite outgrowth on poly-l-lysine, showing that proteolytic processing of NCAM by the metalloprotease TACE is involved in NCAM-mediated neurite outgrowth.


Subject(s)
ADAM Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/physiology , Neurons/cytology , Neurons/physiology , ADAM Proteins/deficiency , ADAM17 Protein , Actins/metabolism , Animals , Blotting, Western/methods , Brain/cytology , Calmodulin/metabolism , Cells, Cultured , Cerebellum/cytology , Cricetinae , Cricetulus , Metalloproteases/pharmacology , Mice , Mice, Inbred C57BL , Molecular Biology/methods , Neurites/drug effects , Neuroblastoma , Neurons/drug effects , Transfection/methods
16.
J Biol Chem ; 278(12): 10381-8, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12529374

ABSTRACT

The transmembrane and multidomain neural adhesion molecule L1 plays important functional roles in the developing and adult nervous system. L1 is proteolytically processed at two distinct sites within the extracellular domain, leading to the generation of different fragments. In this report, we present evidence that the proprotein convertase PC5A is the protease that cleaves L1 in the third fibronectin type III domain, whereas the proprotein convertases furin, PC1, PC2, PACE4, and PC7 are not effective in cleaving L1. Analysis of mutations revealed Arg(845) to be the site of cleavage generating the N-terminal 140-kDa fragment. This fragment was present in the hippocampus, which expresses PC5A, but was not detectable in the cerebellum, which does not express PC5A. The 140-kDa L1 fragment was found to be tightly associated with the full-length 200-kDa L1 molecule. The complex dissociated from the membrane upon cleavage by a protease acting at a more membrane-proximal site of full-length L1. This proteolytic cleavage was inhibited by the metalloprotease inhibitor GM 6001 and enhanced by a calmodulin inhibitor. L1-dependent neurite outgrowth of cerebellar neurons was inhibited by GM 6001, suggesting that proteolytic processing of L1 by a metalloprotease is involved in neurite outgrowth.


Subject(s)
Metalloendopeptidases/physiology , Neural Cell Adhesion Molecule L1/metabolism , Serine Endopeptidases/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Calmodulin/antagonists & inhibitors , Cerebellum/metabolism , Dimerization , Fibronectins/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neural Cell Adhesion Molecule L1/chemistry , Neurites/physiology , Proprotein Convertase 5 , Tumor Cells, Cultured
17.
Biochem J ; 362(Pt 2): 433-42, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11853552

ABSTRACT

Syncollin, a novel pancreatic zymogen granule protein, is present on the luminal side of the granule membrane. To address the function of syncollin, we searched for putative binding partners. Cross-linking experiments with purified syncollin, and granule content and membrane proteins revealed a direct interaction between syncollin and GP-2, a major glycosylphosphatidylinositol (GPI)-anchored membrane glycoprotein. An interaction was also observed when cross-linking was performed with recombinant GP-2. In addition, syncollin could be cross-linked to itself, supporting the suggestion that it exists as a homo-oligomer. Cleavage of the GPI anchor of GP-2 by treatment of granule membranes with phosphatidylinositol-specific phospholipase C had no effect on the membrane attachment of syncollin, indicating that it is not mediated exclusively via an interaction with GP-2. Syncollin was found to be associated with detergent-insoluble cholesterol/glycolipid-enriched complexes. These complexes floated to the lighter fractions of sucrose-density gradients and also contained GP-2, the lectin ZG16p, sulphated matrix proteoglycans and the soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptors (SNAREs) syntaxin 3 and synaptobrevin 2. Our results indicate that membrane-associated syncollin is a component of lipid rafts, where it interacts both with GP-2 and membrane lipids. We suggest that the syncollin-GP-2 complex might play a role in signal transduction across the granule membrane.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Pancreas/metabolism , Secretory Vesicles/metabolism , Animals , Binding Sites , Carrier Proteins/isolation & purification , Carrier Proteins/ultrastructure , Cross-Linking Reagents , Glycosylphosphatidylinositols/metabolism , Membrane Glycoproteins/isolation & purification , Membrane Proteins/isolation & purification , Membrane Proteins/ultrastructure , Mice , Microscopy, Electron , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Secretory Vesicles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...