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










Publication year range
1.
PLoS One ; 9(10): e109800, 2014.
Article in English | MEDLINE | ID: mdl-25289695

ABSTRACT

Engagement of high-affinity immunoglobulin E receptors (FcεRI) activates two signaling pathways in mast cells. The Lyn pathway leads to recruitment of Syk and to calcium mobilization whereas the Fyn pathway leads to phosphatidylinositol 3-kinase recruitment. Mapping the connections between both pathways remains an important task to be completed. We previously reported that Phospholipid Scramblase 1 (PLSCR1) is phosphorylated on tyrosine after cross-linking FcεRI on RBL-2H3 rat mast cells, amplifies mast cell degranulation, and is associated with both Lyn and Syk tyrosine kinases. Here, analysis of the pathway leading to PLSCR1 tyrosine phosphorylation reveals that it depends on the FcRγ chain. FcεRI aggregation in Fyn-deficient mouse bone marrow-derived mast cells (BMMC) induced a more robust increase in FcεRI-dependent tyrosine phosphorylation of PLSCR1 compared to wild-type cells, whereas PLSCR1 phosphorylation was abolished in Lyn-deficient BMMC. Lyn association with PLSCR1 was not altered in Fyn-deficient BMMC. PLSCR1 phosphorylation was also dependent on the kinase Syk and significantly, but partially, dependent on detectable calcium mobilization. Thus, the Lyn/Syk/calcium axis promotes PLSCR1 phosphorylation in multiple ways. Conversely, the Fyn-dependent pathway negatively regulates it. This study reveals a complex regulation for PLSCR1 tyrosine phosphorylation in FcεRI-activated mast cells and that PLSCR1 sits at a crossroads between Lyn and Fyn pathways.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Phospholipid Transfer Proteins/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins c-fyn/immunology , Receptors, IgE/immunology , src-Family Kinases/immunology , Animals , Calcium/metabolism , Cell Degranulation/immunology , Cell Line , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Mast Cells/cytology , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phospholipid Transfer Proteins/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-fyn/genetics , Rats , Receptors, IgE/genetics , Signal Transduction , Syk Kinase , Tyrosine/metabolism , src-Family Kinases/genetics
2.
Blood ; 120(24): 4712-9, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23086752

ABSTRACT

Understanding the process of myeloid differentiation offers important insights into both normal and abnormal developmental processes but is limited by the dearth of experimental models. Here we show that myeloid progenitors can be derived from embryonic stem cells, immortalized, and applied to the study of the mechanisms underlying myeloid differentiation. The embryonic stem cell-derived myeloid progenitors, when immortalized with estrogen-regulated Hoxb8 protein, demonstrate normal karyotyping, are genetically tractable, and can be differentiated into functional neutrophils. Using this model, we identified mammalian target of rapamycin complex 1 as a critical regulator of myeloid differentiation. Together, our studies led to a convenient, karyotypically normal, and genetically manipulatable cellular system, which can be used to shed new light on the mechanisms for myeloid differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/cytology , Signal Transduction/physiology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Estradiol/pharmacology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Karyotype , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Phosphorylation/drug effects , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
J Leukoc Biol ; 90(2): 221-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21447647

ABSTRACT

PLSCR1-/- mice exhibit normal, steady-state hematologic parameters but impaired emergency granulopoiesis upon in vivo administration of G-CSF. The mechanism by which PLSCR1 contributes to G-CSF-induced neutrophil production is largely unknown. We now report that the expansion of bone marrow myelocytes upon in vivo G-CSF treatment is reduced in PLSCR1-/- mice relative to WT. Using SCF-ER-Hoxb8-immortalized myeloid progenitors to examine the progression of G-CSF-driven granulocytic differentiation in vitro, we found that PLSCR1 prolongs the period of mitotic expansion of proliferative granulocyte precursors, thereby giving rise to increased neutrophil production from their progenitors. This effect of PLSCR1 is blocked by a ΔNLS-PLSCR1, which prevents its nuclear import. By contrast, mutation that prevents the membrane association of PLSCR1 has minimal impact on the role of PLSCR1 in G-CSF-induced granulopoiesis. These data imply that the capacity of PLSCR1 to augment G-CSF-dependent production of mature neutrophils from myeloid progenitors is unrelated to its reported activities at the endofacial surface of the plasma membrane but does require entry of the protein into the nucleus, suggesting that this response is mediated through the observed effects of PLSCR1 on gene transcription.


Subject(s)
Cell Proliferation , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Leukopoiesis/drug effects , Phospholipid Transfer Proteins/physiology , Active Transport, Cell Nucleus , Animals , Mice , Mice, Knockout , Mitosis , Myeloid Progenitor Cells/cytology , Neutrophils/cytology , Phospholipid Transfer Proteins/deficiency , Transcription, Genetic
4.
Bioinformatics ; 25(2): 159-62, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19010806

ABSTRACT

MOTIVATION: Phospholipid scramblases (PLSCRs) constitute a family of cytoplasmic membrane-associated proteins that were identified based upon their capacity to mediate a Ca(2+)-dependent bidirectional movement of phospholipids across membrane bilayers, thereby collapsing the normally asymmetric distribution of such lipids in cell membranes. The exact function and mechanism(s) of these proteins nevertheless remains obscure: data from several laboratories now suggest that in addition to their putative role in mediating transbilayer flip/flop of membrane lipids, the PLSCRs may also function to regulate diverse processes including signaling, apoptosis, cell proliferation and transcription. A major impediment to deducing the molecular details underlying the seemingly disparate biology of these proteins is the current absence of any representative molecular structures to provide guidance to the experimental investigation of their function. RESULTS: Here, we show that the enigmatic PLSCR family of proteins is directly related to another family of cellular proteins with a known structure. The Arabidopsis protein At5g01750 from the DUF567 family was solved by X-ray crystallography and provides the first structural model for this family. This model identifies that the presumed C-terminal transmembrane helix is buried within the core of the PLSCR structure, suggesting that palmitoylation may represent the principal membrane anchorage for these proteins. The fold of the PLSCR family is also shared by Tubby-like proteins. A search of the PDB with the HHpred server suggests a common evolutionary ancestry. Common functional features also suggest that tubby and PLSCR share a functional origin as membrane tethered transcription factors with capacity to modulate phosphoinositide-based signaling.


Subject(s)
Cell Membrane/enzymology , Membrane Proteins/chemistry , Phospholipid Transfer Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Arabidopsis Proteins/chemistry , Crystallography, X-Ray , Databases, Protein , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
5.
Biochim Biophys Acta ; 1771(9): 1177-85, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17590392

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is a member of PLSCR gene family that has been implicated in multiple cellular processes including movement of phospholipids, gene regulation, immuno-activation, and cell proliferation/apoptosis. In the present study, we identified PLSCR1 as a positive intracellular acute phase protein that is upregulated by LPS in liver, heart, and adipose tissue, but not skeletal muscle. LPS administration resulted in a marked increase in PLSCR1 mRNA and protein levels in the liver. This stimulation occurred rapidly (within 2 h), and was very sensitive to LPS (half-maximal response at 0.1 microg/mouse). Moreover, two other APR-inducers, zymosan and turpentine, also produced significant increases in PLSCR1 mRNA and protein levels, indicating that PLSCR1 was stimulated in a number of models of the APR. To determine signaling pathways by which LPS stimulated PLSCR1, we examined the effect of proinflammatory cytokines in vitro and in vivo. TNFalpha, IL-1beta, and IL-6 all stimulated PLSCR1 in cultured Hep B3 hepatocytes, whereas only TNFalpha stimulated PLSCR1 in cultured 3T3-L1 adipocytes, suggesting cell type-specific effects of cytokines. Furthermore, the LPS-stimulated increase in liver PLSCR1 mRNA was greatly attenuated by 80% in TNFalpha and IL-1beta receptor null mice as compared to wild-type controls. In contrast, PLSCR1 levels in adipose tissue were induced to a similar extent in TNFalpha and IL-1beta receptor null mice and controls. These results indicate that maximal stimulation of PLSCR1 by LPS in liver required TNFalpha and/or IL-1beta, whereas the stimulation of PLSCR1 in adipose tissue is not dependent on TNFalpha and/or IL-1beta. These data provide evidence that PLSCR1 is a positive intracellular acute phase protein with a tissue-specific mechanism for up-regulation.


Subject(s)
Acute-Phase Reaction , Enzyme Induction , Isoenzymes , Phospholipid Transfer Proteins , 3T3-L1 Cells , Adipose Tissue/enzymology , Animals , Female , Genes, Immediate-Early , Humans , Interleukin-1beta/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lipopolysaccharides/immunology , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family , Muscle, Skeletal/enzymology , Myocardium/enzymology , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Solvents , Tumor Necrosis Factor-alpha/metabolism , Turpentine/metabolism , Zymosan/immunology
6.
Genome Biol ; 8(3): R38, 2007.
Article in English | MEDLINE | ID: mdl-17355638

ABSTRACT

BACKGROUND: The obesity epidemic has prompted the search for candidate genes capable of influencing adipose function. One such candidate, that encoding phospholipid scramblase 3 (PLSCR3), was recently identified, as genetic deletion of it led to lipid accumulation in abdominal fat pads and changes characteristic of metabolic syndrome. Because adipose tissue is increasingly recognized as an endocrine organ, capable of releasing small molecules that modulate disparate physiological processes, we examined the plasma from wild-type, Plscr1-/-, Plscr3-/- and Plscr1&3-/- mice. Using an untargeted comprehensive metabolite profiling approach coupled with targeted gene expression analyses, the perturbed biochemistry and functional redundancy of PLSCR proteins was assessed. RESULTS: Nineteen metabolites were differentially and similarly regulated in both Plscr3-/- and Plscr1&3-/- animals, of which five were characterized from accurate mass, tandem mass spectrometry data and their correlation to the Metlin database as lysophosphatidylcholine (LPC) species enriched with C16:1, C18:1, C20:3, C20:5 and C22:5 fatty acids. No significant changes in the plasma metabolome were detected upon elimination of PLSCR1, indicating that increases in pro-inflammatory lipids are specifically associated with the obese state of Plscr3-deficient animals. Correspondingly, increases in white adipose lipogenic gene expression confirm a role for PLSCR3 in adipose lipid metabolism. CONCLUSION: The untargeted profiling of circulating metabolites suggests no detectable functional redundancies between PLSCR proteins; however, this approach simultaneously identified previously unrecognized lipid metabolites that suggest a novel molecular link between obesity, inflammation and the downstream consequences associated with PLSCR3-deficiency.


Subject(s)
Inflammation/etiology , Lipid Metabolism , Obesity/etiology , Phospholipid Transfer Proteins/deficiency , Adipose Tissue, White/metabolism , Animals , Fatty Acids/analysis , Gene Expression Profiling , Lysophosphatidylcholines/chemistry , Mice , Mice, Knockout , Phospholipid Transfer Proteins/physiology , Tandem Mass Spectrometry
7.
J Biol Chem ; 280(41): 35062-8, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16091359

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is a multiply palmitoylated, endofacial membrane protein originally identified based on its capacity to promote accelerated transbilayer phospholipid movement in response to Ca(2+). Recent evidence suggests that this protein also participates in cell response to various growth factors and cytokines, influencing myeloid differentiation, tumor growth, and the antiviral activity of interferon. Whereas plasma membrane PLSCR1 was shown to be required for normal recruitment and activation of Src kinase by stimulated cell surface growth factor receptors, PLSCR1 was also found to traffic into the nucleus and to tightly bind to genomic DNA, suggesting a possible additional nuclear function. We now report evidence that PLSCR1 directly binds to the 5'-promoter region of the inositol 1,4,5-triphosphate receptor type 1 gene (IP3R1) to enhance expression of the receptor. Probing a CpG island genomic library with PLSCR1 as bait identified four clones with avidity for PLSCR1, including a 191-bp fragment of the IP3R1 promoter. Using electrophoretic mobility shift and transcription reporter assays, the PLSCR1-binding site in IP3R1 was mapped to residues (-101)GTAACCATGTGGA(-89), and the segment spanning Met(86)-Glu(118) in PLSCR1 was identified to mediate its transcriptional activity. The significance of this interaction between PLSCR1 and IP3R1 in situ was confirmed by comparing levels of IP3R1 mRNA and protein in matched cells that either expressed or were deficient in PLSCR1. These data suggest that in addition to its role at the plasma membrane, effects of PLSCR1 on cell proliferative and maturational responses may also relate to alterations in expression of cellular IP3 receptors.


Subject(s)
Calcium Channels/genetics , Gene Expression Regulation, Enzymologic , Membrane Glycoproteins/genetics , Phospholipid Transfer Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Antiviral Agents/pharmacology , Base Sequence , Binding Sites , Blotting, Northern , Blotting, Western , Calcium/metabolism , Calcium Channels/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Cloning, Molecular , CpG Islands , DNA, Complementary/metabolism , Fibroblasts/metabolism , Gene Deletion , Glutathione Transferase/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors , Mice , Molecular Sequence Data , Phospholipids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , src-Family Kinases/metabolism
8.
Blood ; 104(12): 3731-8, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15308560

ABSTRACT

Although phospholipid scramblase 1 (PLSCR1) was originally identified based on its capacity to promote transbilayer movement of membrane phospholipids, subsequent studies also provided evidence for its role in cell proliferation, maturation, and apoptosis. In this report, we investigate the potential role of PLSCR1 in leukemic cell differentiation. We show that all-trans retinoic acid (ATRA), an effective differentiation-inducing agent of acute promyelocytic leukemic (APL) cells, can elevate PLSCR1 expression in ATRA-sensitive APL cells NB4 and HL60, but not in maturation-resistant NB4-LR1 cells. ATRA- and phorbol 12-myristate 13-acetate (PMA)-induced monocytic differentiation is accompanied by increased PLSCR1 expression, whereas only a slight or no elevation of PLSCR1 expression is observed in U937 cells differentiated with dimethyl sulfoxide (DMSO), sodium butyrate, or vitamin D3. Cell differentiation with ATRA and PMA, but not with vitamin D3 or DMSO, results in phosphorylation of protein kinase Cdelta (PKCdelta), and the PKCdelta-specific inhibitor rottlerin nearly eliminates the ATRA- and PMA-induced expression of PLSCR1, while ectopic expression of a constitutively active form of PKCdelta directly increases PLSCR1 expression. Finally, decreasing PLSCR1 expression with small interfering RNA inhibits ATRA/PMA-induced differentiation. Taken together, these results suggest that as a protein induced upon PKCdelta activation, PLSCR1 is required for ATRA- and PMA-triggered leukemic cell differentiation.


Subject(s)
Leukemia/pathology , Membrane Proteins/genetics , Phospholipid Transfer Proteins/genetics , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Humans , Membrane Proteins/metabolism , Phospholipid Transfer Proteins/metabolism , Phosphorylation , Protein Kinase C-delta , RNA, Small Interfering/pharmacology , Up-Regulation
9.
J Virol ; 78(17): 8983-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15308695

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is an interferon (IFN)- and growth factor-inducible, calcium-binding protein that either inserts into the plasma membrane or binds DNA in the nucleus depending on its state of palmyitoylation. In certain hematopoietic cells, PLSCR1 is required for normal maturation and terminal differentiation from progenitor cells as regulated by select growth factors, where it promotes recruitment and activation of Src kinases. PLSCR1 is a substrate of Src (and Abl) kinases, and transcription of the PLSCR1 gene is regulated by the same growth factor receptor pathways in which PLSCR1 potentiates afferent signaling. The marked transcriptional upregulation of PLSCR1 by IFNs led us to explore whether PLSCR1 plays an analogous role in cellular responses to IFN, with specific focus on antiviral activities. Accordingly, human cells in which PLSCR1 expression was decreased with short interfering RNA were rendered relatively insensitive to the antiviral activity of IFNs, resulting in higher titers of vesicular stomatitis virus (VSV) and encephalomyocarditis virus. Similarly, VSV replicated to higher titers in mouse PLSCR1(-/-) embryonic fibroblasts than in identical cells transduced to express PLSCR1. PLSCR1 inhibited accumulation of primary VSV transcripts, similar to the effects of IFN against VSV. The antiviral effect of PLSCR1 correlated with increased expression of a subset of IFN-stimulated genes (ISGs), including ISG15, ISG54, p56, and guanylate binding proteins. Our results suggest that PLSCR1, which is itself an ISG-encoded protein, provides a mechanism for amplifying and enhancing the IFN response through increased expression of a select subset of potent antiviral genes.


Subject(s)
Antiviral Agents/pharmacology , Carrier Proteins/metabolism , Gene Expression Regulation/drug effects , Interferons/pharmacology , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Proteins/metabolism , Vesicular stomatitis Indiana virus/physiology , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Encephalomyocarditis virus/physiology , Fibroblasts/virology , Gene Deletion , Gene Expression Profiling , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , STAT1 Transcription Factor , Trans-Activators/metabolism , Vesicular stomatitis Indiana virus/genetics , Virus Replication/physiology , eIF-2 Kinase/metabolism
10.
Proc Natl Acad Sci U S A ; 101(36): 13296-301, 2004 Sep 07.
Article in English | MEDLINE | ID: mdl-15328404

ABSTRACT

The phospholipid scramblases (PLSCR1 to PLSCR4) are a structurally and functionally unique class of proteins, which are products of a tetrad of genes conserved from Caenorhabditis elegans to humans. The best characterized member of this family, PLSCR1, is implicated in the remodeling of the transbilayer distribution of plasma membrane phospholipids but is also required for normal signaling through select growth factor receptors. Mice with targeted deletion of PLSCR1 display perinatal granulocytopenia due to defective response of hematopoietic precursors to granulocyte colony-stimulating factor and stem cell factor. To gain insight into the biologic function of another member of the PLSCR family, we investigated mice with targeted deletion of PLSCR3, a protein that like PLSCR1 is expressed in many blood cells but which, by contrast to PLSCR1, is also highly expressed in fat and muscle. PLSCR3(-/-) mice at 2 months of age displayed aberrant accumulation of abdominal fat when maintained on standard rodent chow, which was accompanied by insulin resistance, glucose intolerance, and dyslipidemia. Primary adipocytes and cultured bone-marrow-derived macrophages from PLSCR3(-/-) mice were engorged with neutral lipid, and adipocytes displayed defective responses to exogenous insulin. Plasma of PLSCR3(-/-) mice was elevated in non-high-density lipoproteins, cholesterol, triglycerides, nonesterified fatty acids, and leptin, whereas adiponectin was low. These data suggest that the expression of PLSCR3 may be required for normal adipocyte and/or macrophage maturation or function and raise the possibility that deletions or mutations affecting the PLSCR3(-/-) gene locus may contribute to the risk for lipid-related disorders in humans.


Subject(s)
Adipose Tissue/pathology , Carrier Proteins/physiology , Hyperlipidemias/etiology , Insulin Resistance , Intercellular Signaling Peptides and Proteins , Membrane Proteins/physiology , Phospholipid Transfer Proteins , Phospholipids/metabolism , Adiponectin , Animals , Cells, Cultured , Glucose/metabolism , Hematopoietic Stem Cells/physiology , Insulin/pharmacology , Leptin/blood , Mice , Proteins/analysis , Proteins/physiology
11.
Biochemistry ; 43(12): 3518-26, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15035622

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is a multiply palmitoylated, Ca(2+)-binding, endofacial plasma membrane protein originally identified by its capacity to accelerate transbilayer movement of membrane phospholipids. We recently reported that when palmitoylation of PLSCR1 does not occur, it is localized to the nucleus rather than the plasma membrane. Nuclear localization of PLSCR1 was also observed upon induction of its de novo synthesis by cytokines such as interferon alpha that activate the PLSCR1 gene. Despite its capacity to enter the nucleus, its sequence does not predict a nuclear localization signal. To gain insight into the mechanism and potential significance of nuclear PLSCR1, we investigated the conditions required for its import and retention in the nucleus. We show that nuclear localization of PLSCR1 is dependent on cytosolic factors and energy. Furthermore, we show that PLSCR1 is specifically transported into the nucleus by the importin alpha/beta import pathway, and binds directly and with high affinity to importin alpha. Analysis of deletion mutants suggested that the NLS of PLSCR1 is between residues 242 and 290 and, furthermore, that a peptide within this region encompassing residues (257)GKISKHWTGI(266) is sufficient for nuclear import when conjugated to BSA. In addition, in intact cells, mutation of positively charged amino acids within this putative NLS in the full-length protein completely blocked its entry into the nucleus, consistent with its role in targeting PLSCR1 to the nucleus. Release of PLSCR1 from the nucleus was only observed after treatment of cells with both detergent and an elevated NaCl concentration, or following DNase treatment of the nucleus, suggesting ionic interactions of PLSCR1 with a nuclear component bound to genomic DNA or directly with genomic DNA. Purified PLSCR1 was also found to bind directly to a genomic DNA-cellulose conjugate, and its elution from DNA also required an elevated NaCl concentration. These data support a mechanism of receptor-mediated nuclear import of PLSCR1 and suggest a potential nuclear function for this plasma membrane protein.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , DNA/metabolism , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus/genetics , Amino Acid Sequence , Animals , Buffers , Carrier Proteins/genetics , Carrier Proteins/physiology , Cattle , Cell Line , Cell Nucleus/genetics , DNA/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Molecular Sequence Data , Nuclear Envelope/metabolism , Octoxynol , Peptide Fragments/metabolism , Peptide Fragments/physiology , Rats , Receptors, Cell Surface/genetics , Signal Transduction/genetics , alpha Karyopherins/metabolism , alpha Karyopherins/physiology , beta Karyopherins/physiology , ran GTP-Binding Protein/physiology
12.
J Biol Chem ; 278(39): 37413-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-12871937

ABSTRACT

Phospholipid scramblase (PLSCR1) is a multiply palmitoylated, calcium-binding endofacial membrane protein proposed to mediate transbilayer movement of plasma membrane phospholipids. PLSCR1 is a component of membrane lipid rafts and has been shown to both physically and functionally interact with activated epidermal growth factor (EGF) receptors and other raft-associated cell surface receptors. Cell stimulation by EGF results in Tyr phosphorylation of PLSCR1, its association with both Shc and EGF receptors, and rapid cycling of PLSCR1 between plasma membrane and endosomal compartments. We now report evidence that upon EGF stimulation, PLSCR1 is phosphorylated by c-Src, within the tandem repeat sequence 68VYNQPVYNQP77. The in vivo interaction between PLSCR1 and Shc requires the Src-mediated phosphorylation on tyrosines 69 and 74. In in vitro pull down studies, phosphorylated PLSCR1 was found to bind directly to Shc through the phosphotyrosine binding domain. Consistent with the potential role of PLSCR1 in growth factor signaling pathways, granulocyte precursors derived from mice deficient in PLSCR1 show impaired proliferation and maturation under cytokine stimulation. Using PLSCR1-/- embryonic fibroblasts and kidney epithelial cells, we now demonstrate that deletion of PLSCR1 from the plasma membrane reduces the activation of c-Src by EGF, implying that PLSCR1 normally facilitates receptor-dependent activation of this kinase. We propose that PLSCR1, through its interaction with Shc, promotes Src kinase activation through the EGF receptor.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Membrane Proteins/physiology , Phospholipid Transfer Proteins , Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , CSK Tyrosine-Protein Kinase , Carrier Proteins/chemistry , Cell Line , Enzyme Activation , Humans , Membrane Proteins/chemistry , Phosphorylation , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/metabolism , src-Family Kinases
13.
Biochemistry ; 42(5): 1227-33, 2003 Feb 11.
Article in English | MEDLINE | ID: mdl-12564925

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is a Ca(2+)-binding, endofacial plasma membrane protein thought to contribute to the transbilayer movement of phosphatidylserine and other membrane phospholipids that is observed upon influx of calcium into the cytosol. Expression of PLSCR1 is markedly induced by interferon and other cytokines, and PLSCR1-/- bone marrow cells exhibit defective myeloid proliferation and differentiation in response to stimulation by select growth factors, implying that PLSCR1 also functions in cytokine signaling or response pathways. PLSCR1 is multiply palmitoylated and partitions into membrane lipid raft domains. We have now identified the Cys-rich sequence (184)CCCPCC(189) in PLSCR1 as required for palmitoylation of the polypeptide. Mutation of these five cysteines abrogates PLSCR1 trafficking to the plasma membrane and results in virtually all of the expressed protein localizing to the nucleus. Consistent with this observation, cell treatment with the palmitoylation inhibitor, 2-bromo-palmitate, results in a marked redistribution of endogenous PLSCR1 from plasma membrane to nucleus. In a small percentage of untreated cells, predominantly nuclear localization of PLSCR1 is also observed. Furthermore, PLSCR1 is also found in the nucleus following its cytokine-induced expression. These data suggest that under the circumstance of rapid biosynthesis in response to gene induction by cytokines, PLSCR1 traffics into the nucleus, implying a potential nuclear function for this protein.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Nucleus/enzymology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Palmitic Acid/chemistry , Palmitic Acid/metabolism , Phospholipid Transfer Proteins , Phospholipids/chemistry , Phospholipids/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Animals , Antigens/biosynthesis , Antigens/chemistry , Antigens/genetics , Antigens/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cysteine/chemistry , Cysteine/genetics , Cytokines/physiology , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed , Palmitates/pharmacology , Palmitic Acid/antagonists & inhibitors , Transfection , Tumor Cells, Cultured
14.
Blood ; 99(11): 4030-8, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12010804

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is an endofacial plasma membrane protein proposed to participate in transbilayer movement of phosphatidylserine and other phospholipids. In addition to its putative role in the reorganization of plasma membrane phospholipids, PLSCR1 is a substrate of intracellular kinases that imply its possible participation in diverse signaling pathways underlying proliferation, differentiation, or apoptosis. Because PLSCR1 is prominently expressed in a variety of blood cells, we evaluated PLSCR activity in platelets and erythrocytes, and cytokine-dependent growth of hematopoietic precursor cells, of PLSCR1 knock-out mice. Adult PLSCR1(-/-) mice showed no obvious hematologic or hemostatic abnormality, and blood cells from these animals normally mobilized phosphatidylserine to the cell surface upon stimulation. Whereas blood cell counts in adult PLSCR1(-/-) mice were normal, in both fetus and newborn animals neutrophil counts were significantly depressed relative to age-matched wild type (WT). Furthermore, when compared with WT, hematopoietic precursor cells from PLSCR1(-/-) mice showed defective colony formation and impaired differentiation to mature granulocytes as stimulated by stem cell factor and granulocyte colony-stimulating factor (G-CSF). By contrast, PLSCR1(-/-) cells showed normal colony formation stimulated by interleukin-3 or granulocyte-macrophage CSF, and expansion of megakaryocytic and erythroid progenitors by thrombopoietin or erythropoietin was unaffected. Stem cell factor and G-CSF were also found to induce marked increases in PLSCR1 levels in WT cells. Consistent with in vitro assays, PLSCR1(-/-) mice treated with G-CSF showed less than 50% of the granulocytosis observed in identically treated WT mice. These data provide direct evidence that PLSCR1 functionally contributes to cytokine-regulated cell proliferation and differentiation and suggest it is required for normal myelopoiesis.


Subject(s)
Blood Platelets/physiology , Carrier Proteins/metabolism , Growth Substances/pharmacology , Hematopoiesis/physiology , Hemostasis/physiology , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Animals , Animals, Newborn , Apoptosis/drug effects , Bleeding Time , Bone Marrow Cells/cytology , Ca(2+) Mg(2+)-ATPase/deficiency , Ca(2+) Mg(2+)-ATPase/genetics , Ca(2+) Mg(2+)-ATPase/metabolism , Carrier Proteins/genetics , Cells, Cultured , Colony-Forming Units Assay , Fetus , Gene Expression Regulation, Enzymologic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocytes/physiology , Hematopoiesis/drug effects , Liver/cytology , Liver/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Platelet Activation , Reference Values , Stem Cell Factor/pharmacology
15.
Biochemistry ; 41(20): 6338-45, 2002 May 21.
Article in English | MEDLINE | ID: mdl-12009895

ABSTRACT

We have identified physical and functional interactions between the epidermal growth factor (EGF) receptor and phospholipid scramblase 1 (PLSCR1), an endofacial plasma membrane protein proposed to affect phospholipid organization. PLSCR1, a palmitoylated protein, was found to partition with the EGF receptor in membrane lipid rafts. Cell stimulation with EGF transiently elevated Tyr-phosphorylation of PLSCR1, peaking at 5 min. Although PLSCR1 is a known substrate of c-Abl [Sun, J., et al. (2001) J.Biol. Chem. 276, 28984-28990], the Abl inhibitor STI571 did not substantially affect its EGF-dependent phosphorylation, suggesting PLSCR1 is a substrate of the EGF receptor kinase, or another EGF-activated kinase. Coinciding with phosphorylation, there was a transient increase in physical association of PLSCR1 with both the EGF receptor and the adapter protein Shc, as determined by immunoprecipitation and Western blotting. Confocal immunofluorescence analysis revealed that EGF initiates rapid internalization of both the EGF receptor and PLSCR1, with trafficking into both distinct and common endosomal pools. These data also suggested that whereas the EGF receptor is ultimately degraded, much of the endocytosed PLSCR1 is recycled to the cell surface within 3 h after EGF treatment. Consistent with this interpretation, Western blotting revealed neither ubiquitination nor proteolysis of PLSCR1 under these conditions, whereas the ubiquitination and degradation of the EGF receptor were readily confirmed. Finally, stimulation with EGF was also found to markedly increase the total cellular expression of PLSCR1, suggesting that in addition to its initial interactions with activated EGF receptor, PLSCR1 may also contribute to posttranscriptional effector pathway(s) mediating the cellular response to EGF.


Subject(s)
Carrier Proteins/metabolism , ErbB Receptors/metabolism , Membrane Microdomains/enzymology , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Phospholipids/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Endocytosis/physiology , Epidermal Growth Factor/pharmacology , Humans , KB Cells , Membrane Microdomains/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Phosphorylation , Phosphotyrosine/metabolism , Proto-Oncogene Proteins c-abl/physiology
16.
Cancer Res ; 62(2): 397-402, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11809687

ABSTRACT

Phospholipid scramblase 1 (PLSCR1) is an IFN-inducible, endofacial plasma membrane protein that has been proposed to mediate accelerated transbilayer movement of plasma membrane phospholipids in cells exposed to elevated cytoplasmic [Ca (2+)]. The marked transcriptional up-regulation of this gene by IFN in a wide variety of cell types suggested that PLSCR1 might also contribute to biological effects associated with IFN. To study the potential contribution of cellular PLSCR1 to the antiproliferative and tumor-suppressive activities of IFN, PLSCR1 cDNA was stably expressed in the human ovarian cancer cell line HEY1B, and the growth of these cells was compared with matched vector transfected controls both in vitro and in vivo. Whereas we detected no difference in either growth rate or morphology between PLSCR1-transfected cells and vector controls during in vitro culture in serum, when these cells were implanted s.c. into athymic nude mice, we observed a marked suppression of tumor development from cells transfected to express elevated levels of PLSCR1. Tumors from the PLSCR1-transfected cells were greatly reduced in size, showed increased infiltration of leukocytes and macrophages, and appeared to undergo differentiation to a more uniform and spindle-shaped morphology that markedly contrasted the highly undifferentiated and pleiomorphic cell shape normally observed for HEY1B cells in vitro or for vector-transfected control HEY1B cells both in vitro and in vivo. These data suggest that the up-regulation of PLSCR1 expression in tumor cells exposed to IFN may contribute to the observed tumor-suppressive action of this cytokine.


Subject(s)
Carrier Proteins/physiology , Membrane Proteins/physiology , Ovarian Neoplasms/pathology , Phospholipid Transfer Proteins , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Division/physiology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Female , Gene Expression , Humans , Macrophages/pathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neutrophils/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...