Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 34(28): 3711-27, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25241900

ABSTRACT

Glioblastoma is the most common and lethal primary malignant brain tumor in adults. The tumor suppressor gene PTEN is deleted, mutated or hypermethylated in more than 60% of glioblastoma cases resulting in hyperactivation of the phosphoinositide 3-kinase pathway, which leads to sustained PI(3,4,5)P3 signaling, and thereby hyperactivation of Akt and other effectors. PI(3,4,5)P3 is also hydrolyzed to PI(3,4)P2 by inositol polyphosphate 5-phosphatases such as SKIP, but the role this pathway has in glioblastoma is unknown. Microarray expression profiling of SKIP in human glioblastoma has revealed both increased and decreased SKIP gene expression. Here we have screened PTEN-deficient glioblastoma for SKIP protein expression by immunohistochemistry and report that SKIP expression is increased in some cases or decreased relative to normal brain. Using the U-87MG PTEN-deficient cell line we show that SKIP knockdown did not further enhance cell proliferation or survival. However, SKIP overexpression in U-87MG cells suppressed anchorage-independent cell growth and growth factor-induced PI(3,4,5)P3/Akt signaling. Although, SKIP knockdown did not affect cell proliferation or survival, cell migration was significantly retarded, associated with significantly increased PI(4,5)P2 signals, and decreased phosphorylation of the actin-regulatory protein cofilin, a PI(4,5)P2-binding protein. Notably, overexpression of SKIP also inhibited migration of U-87MG cells to a similar degree as observed with PTEN reconstitution, however, via distinct mechanisms. PTEN reconstitution promoted sustained lamellipodia generation and focal adhesion formation. In contrast, SKIP overexpression reduced sustained lamellipodia formation, talin incorporation into focal adhesions and recruitment of PI(4,5)P2-binding proteins to the plasma membrane. Notably, analysis of two independent ONCOMINE microarray data sets revealed a significant correlation between increased SKIP mRNA expression in glioblastoma and improved long-term survival. Therefore, SKIP expression in glioblastoma may affect the local invasion of PTEN-deficient tumors.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , PTEN Phosphohydrolase/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/enzymology , Glioblastoma/genetics , Humans , MAP Kinase Signaling System , Oligonucleotide Array Sequence Analysis , Survival Analysis
2.
J Cell Biol ; 155(6): 1065-79, 2001 Dec 10.
Article in English | MEDLINE | ID: mdl-11739414

ABSTRACT

SHIP-2 is a phosphoinositidylinositol 3,4,5 trisphosphate (PtdIns[3,4,5]P3) 5-phosphatase that contains an NH2-terminal SH2 domain, a central 5-phosphatase domain, and a COOH-terminal proline-rich domain. SHIP-2 negatively regulates insulin signaling. In unstimulated cells, SHIP-2 localized in a perinuclear cytosolic distribution and at the leading edge of the cell. Endogenous and recombinant SHIP-2 localized to membrane ruffles, which were mediated by the COOH-terminal proline-rich domain. To identify proteins that bind to the SHIP-2 proline-rich domain, yeast two-hybrid screening was performed, which isolated actin-binding protein filamin C. In addition, both filamin A and B specifically interacted with SHIP-2 in this assay. SHIP-2 coimmunoprecipitated with filamin from COS-7 cells, and association between these species did not change after epidermal growth factor stimulation. SHIP-2 colocalized with filamin at Z-lines and the sarcolemma in striated muscle sections and at membrane ruffles in COS-7 cells, although the membrane ruffling response was reduced in cells overexpressing SHIP-2. SHIP-2 membrane ruffle localization was dependent on filamin binding, as SHIP-2 was expressed exclusively in the cytosol of filamin-deficient cells. Recombinant SHIP-2 regulated PtdIns(3,4,5)P3 levels and submembraneous actin at membrane ruffles after growth factor stimulation, dependent on SHIP-2 catalytic activity. Collectively these studies demonstrate that filamin-dependent SHIP-2 localization critically regulates phosphatidylinositol 3 kinase signaling to the actin cytoskeleton.


Subject(s)
Actins/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Animals , COS Cells/drug effects , COS Cells/enzymology , Cell Membrane/enzymology , Contractile Proteins/genetics , Epidermal Growth Factor/pharmacology , Filamins , Humans , Inositol Polyphosphate 5-Phosphatases , Melanoma , Mice , Microfilament Proteins/genetics , Molecular Sequence Data , Muscle Fibers, Skeletal/chemistry , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Myocardium/chemistry , Myocardium/cytology , Oligonucleotide Probes , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/analysis , Phosphoric Monoester Hydrolases/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques , Yeasts
3.
J Biol Chem ; 276(10): 7643-53, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11116155

ABSTRACT

The budding yeast Saccharomyces cerevisiae has four inositol polyphosphate 5-phosphatase (5-phosphatase) genes, INP51, INP52, INP53, and INP54, all of which hydrolyze phosphatidylinositol (4,5)-bisphosphate. INP54 encodes a protein of 44 kDa which consists of a 5-phosphatase domain and a C-terminal leucine-rich tail, but lacks the N-terminal SacI domain and proline-rich region found in the other three yeast 5-phosphatases. We report that Inp54p belongs to the family of tail-anchored proteins and is localized to the endoplasmic reticulum via a C-terminal hydrophobic tail. The hydrophobic tail comprises the last 13 amino acids of the protein and is sufficient to target green fluorescent protein to the endoplasmic reticulum. Protease protection assays demonstrated that the N terminus of Inp54p is oriented toward the cytoplasm of the cell, with the C terminus of the protein also exposed to the cytosol. Null mutation of INP54 resulted in a 2-fold increase in secretion of a reporter protein, compared with wild-type yeast or cells deleted for any of the SacI domain-containing 5-phosphatases. We propose that Inp54p plays a role in regulating secretion, possibly by modulating the levels of phosphatidylinositol (4,5)-bisphosphate on the cytoplasmic surface of the endoplasmic reticulum membrane.


Subject(s)
Endoplasmic Reticulum/metabolism , Phosphoric Monoester Hydrolases/biosynthesis , Amino Acid Sequence , Cell Membrane/metabolism , Cloning, Molecular , Cytoplasm/metabolism , Green Fluorescent Proteins , Inositol Polyphosphate 5-Phosphatases , Luminescent Proteins/metabolism , Microscopy, Confocal , Models, Biological , Molecular Sequence Data , Mutation , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Time Factors , Water/metabolism
4.
Mol Cell Biol ; 20(24): 9376-90, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11094088

ABSTRACT

The Saccharomyces cerevisiae inositol polyphosphate 5-phosphatases (Inp51p, Inp52p, and Inp53p) each contain an N-terminal Sac1 domain, followed by a 5-phosphatase domain and a C-terminal proline-rich domain. Disruption of any two of these 5-phosphatases results in abnormal vacuolar and plasma membrane morphology. We have cloned and characterized the Sac1-containing 5-phosphatases Inp52p and Inp53p. Purified recombinant Inp52p lacking the Sac1 domain hydrolyzed phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] and PtdIns(3, 5)P(2). Inp52p and Inp53p were expressed in yeast as N-terminal fusion proteins with green fluorescent protein (GFP). In resting cells recombinant GFP-tagged 5-phosphatases were expressed diffusely throughout the cell but were excluded from the nucleus. Following hyperosmotic stress the GFP-tagged 5-phosphatases rapidly and transiently associated with actin patches, independent of actin, in both the mother and daughter cells of budding yeast as demonstrated by colocalization with rhodamine phalloidin. Both the Sac1 domain and proline-rich domains were able to independently mediate translocation of Inp52p to actin patches, following hyperosmotic stress, while the Inp53p proline-rich domain alone was sufficient for stress-mediated localization. Overexpression of Inp52p or Inp53p, but not catalytically inactive Inp52p, which lacked PtdIns(4,5)P(2) 5-phosphatase activity, resulted in a dramatic reduction in the repolarization time of actin patches following hyperosmotic stress. We propose that the osmotic-stress-induced translocation of Inp52p and Inp53p results in the localized regulation of PtdIns(3,5)P(2) and PtdIns(4,5)P(2) at actin patches and associated plasma membrane invaginations. This may provide a mechanism for regulating actin polymerization and cell growth as an acute adaptive response to hyperosmotic stress.


Subject(s)
Actins/metabolism , Cell Membrane Structures/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoric Monoester Hydrolases/metabolism , Saccharomyces cerevisiae/enzymology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cloning, Molecular , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Gene Deletion , Genes, Reporter , Inositol Polyphosphate 5-Phosphatases , Marine Toxins/pharmacology , Microscopy, Confocal , Microscopy, Fluorescence , Osmotic Pressure , Phosphoric Monoester Hydrolases/genetics , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Thiazoles/pharmacology , Thiazolidines , Time Factors
5.
J Biol Chem ; 275(47): 37055-61, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10962003

ABSTRACT

Inositol polyphosphate 5-phosphatases (5-phosphatase) hydrolyze the 5-position phosphate from the inositol ring of phosphatidylinositol-derived signaling molecules; however, the mechanism of catalysis is only partially characterized. These enzymes play critical roles in regulating cell growth, apoptosis, intracellular calcium oscillations, and post-synaptic vesicular trafficking. The UCLA fold recognition server (threader) predicted that the conserved 300-amino acid catalytic domain, common to all 5-phosphatases, adopts the fold of the apurinic/apyrimidinic (AP) base excision repair endonucleases. PSI-BLAST searches of GENPEPT, using the amino acid sequence of AP endonuclease exonuclease III, identified all members of the 5-phosphatase family with highly significant scores. A sequence alignment between exonuclease III and all known 5-phosphatases revealed six highly conserved motifs containing residues that corresponded to the catalytic residues in the AP endonucleases. Mutation of each of these residues to alanine in the mammalian 43-kDa, or yeast Inp52p 5-phosphatase, resulted in complete loss of enzyme activity. We predict the 5-phosphatase enzymes share a similar mechanism of catalysis to the AP endonucleases, consistent with other common functional similarities such as an absolute requirement for magnesium for activity. Based on this analysis, functional roles have been assigned to conserved residues in all 5-phosphatase enzymes.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA Repair , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Animals , Catalysis , Crystallography, X-Ray , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Gene Library , Humans , Inositol Polyphosphate 5-Phosphatases , Models, Molecular , Molecular Sequence Data , Protein Conformation , Rats , Sequence Alignment
6.
J Biol Chem ; 275(31): 24052-64, 2000 Aug 04.
Article in English | MEDLINE | ID: mdl-10806194

ABSTRACT

The inositol-polyphosphate 5-phosphatase enzyme family removes the 5-position phosphate from both inositol phosphate and phosphoinositide signaling molecules. We have cloned and characterized a novel 5-phosphatase, which demonstrates a restricted substrate specificity and tissue expression. The 3.9-kb cDNA predicts for a 72-kDa protein with an N-terminal proline rich domain, a central 5-phosphatase domain, and a C-terminal CAAX motif. The 3. 9-kilobase mRNA showed a restricted expression but was abundant in testis and brain. Antibodies against the sequence detected a 72-kDa protein in the testis in the detergent-insoluble fraction. Indirect immunofluorescence of the Tera-1 cell line using anti-peptide antibodies to the 72-kDa 5-phosphatase demonstrated that the enzyme is predominantly located to the Golgi. Expression of green fluorescent protein-tagged 72-kDa 5-phosphatase in COS-7 cells revealed that the enzyme localized predominantly to the Golgi, mediated by the N-terminal proline-rich domain, but not the C-terminal CAAX motif. In vitro, the protein inserted into microsomal membranes on the cytoplasmic face of the membrane. Immunoprecipitated recombinant 72-kDa 5-phosphatase hydrolyzed phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3, 5-bisphosphate, forming phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol 3-phosphate, respectively. We propose that the novel 5-phosphatase hydrolyzes phosphatidylinositol 3,4, 5-trisphosphate and phosphatidylinositol 3,5-bisphosphate on the cytoplasmic Golgi membrane and thereby may regulate Golgi-vesicular trafficking.


Subject(s)
Golgi Apparatus/enzymology , Phosphoric Monoester Hydrolases/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Brain/enzymology , Cell Compartmentation , Cell Polarity , Cloning, Molecular , Female , In Situ Hybridization , Inositol Polyphosphate 5-Phosphatases , Male , Mice , Molecular Sequence Data , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Sorting Signals , RNA, Messenger/isolation & purification , Substrate Specificity , Testis/enzymology , Tissue Distribution
7.
J Biol Chem ; 274(38): 27083-91, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480922

ABSTRACT

We have cloned and characterized a novel isoform of the skeletal muscle LIM protein 1 (SLIM1), designated SLIMMER. SLIM1 contains an N-terminal single zinc finger followed by four LIM domains. SLIMMER is identical to SLIM1 over the first three LIM domains but contains a novel C-terminal 96 amino acids with three potential bipartite nuclear localization signals, a putative nuclear export sequence, and 27 amino acids identical to the RBP-J binding region of KyoT2, a murine isoform of SLIM1. SLIM1 localized to the cytosol of Sol8 myoblasts and myotubes. SLIMMER was detected in the nucleus of myoblasts and, following differentiation into myotubes, was exclusively cytosolic. Recombinant green fluorescent protein-SLIM1 localized to the cytoplasm and associated with focal adhesions and actin filaments in COS-7 cells, while green fluorescent protein-SLIMMER was predominantly nuclear. SLIMMER truncation mutants revealed that the first nuclear localization signal mediates nuclear localization. The addition of the proposed nuclear export sequence decreased the level of exclusively nuclear expression and increased cytosolic SLIMMER expression in COS-7 cells. The leucine-rich nuclear export signal was required for the export of SLIMMER from the nucleus of myoblasts to the cytoplasm of myotubes. Collectively, these results suggest distinct roles for SLIM1 and SLIMMER in focal adhesions and nuclear-cytoplasmic communication.


Subject(s)
Muscle Proteins/metabolism , Muscle, Skeletal/chemistry , Zinc Fingers , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Bone Marrow Cells/chemistry , COS Cells , Cell Adhesion , Cell Nucleus/metabolism , Cloning, Molecular , Cytoplasm/metabolism , Cytoskeleton/metabolism , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Molecular Sequence Data , Muscle Proteins/genetics , Transfection
8.
J Mol Cell Cardiol ; 31(4): 837-43, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10329211

ABSTRACT

LIM proteins perform critical roles in development and tissue differentiation. The skeletal muscle LIM protein 1 (SLIM1) comprises four and a half LIM domains. Northern blot analysis demonstrated high level expression of SLIM1 mRNA in adult human skeletal muscle with intermediate expression in adult heart and lower expression in other tissues. Western blot analysis using specific affinity-purified anti-SLIM1 antipeptide antibodies demonstrated a 32 kDa polypeptide in the aorta and atria of rabbit heart, but not in vena cava, interventricular septum or ventricular muscle. SLIM1 was also demonstrated in rabbit skeletal muscle. In situ hybridization of whole mouse embryos confirmed the cardiac expression of SLIM1 was restricted to the cardiac outflow tract from embryonic day 8.5-11. No expression was seen in atrial or ventricular muscle. SLIM1 mRNA was also demonstrated in the hindbrain, neural tube and somites. The localized expression of SLIM1 to the outflow tract of the embryonic heart implies an important role for the protein in the development of this region and possibly in congenital heart anomalies involving the separation and formation of the aortic and pulmonary trunks.


Subject(s)
Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Adult , Animals , Base Sequence , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Tissue Distribution
9.
Am J Hematol ; 49(1): 15-20, 1995 May.
Article in English | MEDLINE | ID: mdl-7741133

ABSTRACT

Chromosomal in situ hybridization (ISH) has extended the scope of cytogenetic analysis to nondividing cells by the use of chromosome-specific probes detected by nonisotopic techniques. This provides a rapid and sensitive method for identifying chromosomes in interphase cells, and is useful in gauging engraftment following bone marrow transplantation, particularly when the number of cells obtained is minimal. We have performed ISH using a Y-heterochromatin-specific probe to monitor patients with malignant hematological disease who have received a sex-mismatched transplant. The results have been compared with those obtained from concurrently performed standard cytogenetic analysis. Host cells were detected by interphase cytogenetics in all patients posttransplant, at times varying from 28-1,825 days, whereas routine analysis detected host cells in only 4 patients, 3 of whom were found to be in relapse. The significance of the persistence of host cells is unknown, but it does not appear to indicate impending relapse.


Subject(s)
Bone Marrow Transplantation/pathology , Chromosomes, Human , Adolescent , Adult , Aged , Anemia/therapy , Cells, Cultured , Cytogenetics/methods , Female , Hodgkin Disease/therapy , Humans , In Situ Hybridization , Interphase , Leukemia/therapy , Male , Middle Aged , Monitoring, Physiologic , Reference Values , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...