Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
2.
Blood ; 117(24): 6509-19, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21502543

ABSTRACT

Exit from quiescence and reentry into cell cycle is essential for HSC self-renewal and regeneration. Skp2 is the F-box unit of the SCF E3-ligase that targets the CDK inhibitors (CKIs) p21(Cip1), p27(Kip1), p57(Kip2), and p130 for degradation. These CKIs inhibit the G(1) to S-phase transition of the cell cycle, and their deletion results in increased cell proliferation and decreased stem cell self-renewal. Skp2 deletion leads to CKIs stabilization inducing cell-cycle delay or arrest, and conversely, increased Skp2 expression is often found in cancers. Here, we show that SKP2 expression is increased in HSC and progenitors in response to hematopoietic stress from myelosuppression or after transplantation. At steady state, SKP2 deletion decreased the mitotic activity of HSC and progenitors resulting in enhanced HSC quiescence, increased HSC pool size, and maintenance. However, the inability to rapidly enter cell cycle greatly impaired the short-term repopulating potential of SKP2 null HSC and their ability to regenerate after myeloablative stress. Mechanistically, deletion of SKP2 in HSC and progenitors stabilized CKIs in vivo, particularly p27(Kip1), p57(Kip2), and p130. Our results demonstrate a previously unrecognized role for SKP2 in regulating HSC and progenitor expansion and hematopoietic regeneration after stress.


Subject(s)
Hematopoietic Stem Cells/physiology , Homeostasis/genetics , S-Phase Kinase-Associated Proteins/physiology , Stress, Physiological/physiology , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
3.
J Cell Physiol ; 202(2): 422-33, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15468068

ABSTRACT

PRIP-1 was isolated as a novel inositol 1,4,5-trisphosphate [Ins(1,4,5)P3] binding protein with a domain organization similar to phospholipase C-delta1 (PLC-delta1) but lacking the enzymatic activity. Further studies revealed that the pleckstrin homology (PH) domain of PRIP-1 is the region responsible for binding Ins(1,4,5)P3. In this study we aimed to clarify the role of PRIP-1 at the physiological concentration in Ins(1,4,5)P3-mediated Ca2+ signaling, as we had previously used COS-1 cells overexpressing PRIP-1 (Takeuchi et al., 2000, Biochem J 349:357-368). For this purpose we employed PRIP-1 knock out (PRIP-1-/-) mice generated previously (Kanematsu et al., 2002, EMBO J 21:1004-1011). The increase in free Ca2+ concentration in response to purinergic receptor stimulation was lower in primary cultured cortical neurons prepared from PRIP-1-/- mice than in those from wild type mice. The relative amounts of [3H]Ins(1,4,5)P3 measured in neurons labeled with [3H]inositol was also lower in cells from PRIP-1-/- mice. In contrast, PLC activities in brain cortex samples from PRIP-1-/- mice were not different from those in the wild type mice, indicating that the hydrolysis of Ins(1,4,5)P3 is enhanced in cells from PRIP-1-/- mice. In vitro analyses revealed that type1 inositol polyphosphate 5-phosphatase physically interacted with a PH domain of PRIP-1 (PRIP-1PH) and its enzyme activity was inhibited by PRIP-1PH. However, physical interaction with these two proteins did not appear to be the reason for the inhibition of enzyme activity, indicating that binding of Ins(1,4,5)P3 to the PH domain prevented its hydrolyzation. Together, these results indicate that PRIP-1 plays an important role in regulating the Ins(1,4,5)P3-mediated Ca2+ signaling by modulating type1 inositol polyphosphate 5-phosphatase activity through binding to Ins(1,4,5)P3.


Subject(s)
Calcium Signaling/physiology , Carrier Proteins/physiology , Inositol 1,4,5-Trisphosphate/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blood Proteins/genetics , Calcium/metabolism , Carrier Proteins/genetics , Cells, Cultured , Inositol Polyphosphate 5-Phosphatases , Mice , Neurons/metabolism , Phosphoproteins/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Structure, Tertiary/genetics , Sequence Homology
4.
Mol Cell Biol ; 24(24): 10868-81, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572689

ABSTRACT

The RAS-activated RAF-->MEK-->extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3'-kinase (PI3'-kinase)-->PDK1-->AKT signaling pathways are believed to cooperate to promote the proliferation of normal cells and the aberrant proliferation of cancer cells. To explore the mechanisms that underlie such cooperation, we have derived cells harboring conditionally active, steroid hormone-regulated forms of RAF and AKT. These cells permit the assessment of the biological and biochemical effects of activation of these protein kinases either alone or in combination with one another. Under conditions where activation of neither RAF nor AKT alone promoted S-phase progression, coactivation of both kinases elicited a robust proliferative response. Moreover, under conditions where high-level activation of RAF induced G(1) cell cycle arrest, activation of AKT bypassed the arrest and promoted S-phase progression. At the level of the cell cycle machinery, RAF and AKT cooperated to induce cyclin D1 and repress p27(Kip1) expression. Repression of p27(Kip1) was accompanied by a dramatic reduction in KIP1 mRNA and was observed in primary mouse embryo fibroblasts derived from mice either lacking SKP2 or expressing a T187A mutated form of p27(Kip1). Consistent with these observations, pharmacological inhibition of MEK or PI3'-kinase inhibited the effects of activated RAS on the expression of p27(Kip1) in NIH 3T3 fibroblasts and in a panel of bona fide human pancreatic cancer cell lines. Furthermore, we demonstrated that AKT activation led to sustained activation of cyclin/cdk2 complexes that occurred concomitantly with the removal of RAF-induced p21(Cip1) from cyclin E/cdk2 complexes. Cumulatively, these data strongly suggest that the RAF-->MEK-->ERK and PI3'K-->PDK-->AKT signaling pathways can cooperate to promote G(0)-->G(1)-->S-phase cell cycle progression in both normal and cancer cells.


Subject(s)
Cell Cycle , Cell Division , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , raf Kinases/metabolism , Animals , Benzamides/pharmacology , Blotting, Western , Butadienes/pharmacology , Cell Cycle Proteins/metabolism , Cell Extracts , Cell Line, Tumor , Cells, Cultured , Chromones/pharmacology , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Embryo, Mammalian/cytology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Microscopy, Fluorescence , Morpholines/pharmacology , NIH 3T3 Cells , Nitriles/pharmacology , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Retroviridae/genetics , Tumor Suppressor Proteins/metabolism , raf Kinases/genetics
5.
J Biol Chem ; 279(45): 46424-30, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15342634

ABSTRACT

The Skp2 oncoprotein belongs to the family of F-box proteins that function as substrate recognition factors for SCF (Skp1, cullin, F-box protein) E3 ubiquitin-ligase complexes. Binding of the substrate to the SCFSkp2 complex catalyzes the conjugation of ubiquitin molecules to the bound substrate, resulting in multi-ubiquitination and rapid degradation by the 26 S proteasome. Using Skp2 as bait in a yeast two-hybrid screen, we have identified UBP43 as a novel substrate for Skp2. UBP43 belongs to the family of ubiquitin isopeptidases and specifically cleaves ISG15, a ubiquitin-like molecule that is induced by cellular stresses, such as type 1 interferons (IFN), nephrotoxic damage, and bacterial infection. UBP43 was originally identified as an up-regulated gene in knock-in mice expressing an acute myelogenous leukemia fusion protein, AML1-ETO, as well as in melanoma cell lines treated with IFN-beta. The phenotype of UBP43 knockout mice includes shortened life span, hypersensitivity to IFN, and neuronal damage, suggesting that tight regulation of ISG15 conjugation is critical for normal cellular function. In this study, we demonstrate that UBP43 is ubiquitinated in vivo and accumulates in cells treated with proteasome inhibitors. We also show that Skp2 promotes UBP43 ubiquitination and degradation, resulting in higher levels of ISG15 conjugates. In Skp2-/- mouse cells, levels of UBP43 are consistently up-regulated, whereas levels of ISG15 conjugates are reduced. Our results demonstrate that the SCFSkp2 is involved in controlling UBP43 protein levels and may therefore play an important role in modulating type 1 IFN signaling.


Subject(s)
Endopeptidases/biosynthesis , Endopeptidases/genetics , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Gene Expression Regulation , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Interferon-beta/metabolism , Interferons/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Oncogene Proteins, Fusion/metabolism , Phenotype , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding , Protein Structure, Tertiary , RUNX1 Translocation Partner 1 Protein , Rats , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Time Factors , Transcription Factors/metabolism , Transfection , Transgenes , Two-Hybrid System Techniques , Ubiquitin/metabolism , Ubiquitin Thiolesterase , Up-Regulation
6.
Int Immunol ; 16(8): 1173-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15226272

ABSTRACT

Mice lacking Tyk2, Stat1 or Stat4, which are members of the Jak-Stat signaling cascade, were resistant to LPS-induced endotoxin shock. Interestingly, Tyk2-deficient mice had higher resistance to LPS challenge than mice lacking either Stat1 or Stat4. The activation of MAPK and NF-kappaB by LPS, and the production of TNF-alpha and IL-12 after LPS injection, were not abrogated by the absence of Tyk2, Stat1 or Stat4. In Stat1-deficient mice, the induction of IFN-beta by LPS in macrophages was severely reduced, although the serum level of IFN-gamma was elevated after LPS injection. In contrast, in Stat-4 deficient mice, the induction of IFN-beta by LPS was normal, but the serum level of IFN-gamma remained low after LPS injection. Interestingly, the induction of both IFN-beta and IFN-gamma by LPS was severely reduced in Tyk2-deficient mice. Therefore, Stat1 and Stat4 independently play substantial roles in the susceptibility to LPS. Tyk2 is essential for LPS-induced endotoxin shock, and this signaling pathway is transduced by the activation of Stat1 and Stat4.


Subject(s)
DNA-Binding Proteins/immunology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Protein-Tyrosine Kinases/immunology , Trans-Activators/immunology , Animals , Cytokines/blood , DNA-Binding Proteins/deficiency , Lipopolysaccharides/administration & dosage , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mice, Knockout , Protein-Tyrosine Kinases/deficiency , STAT1 Transcription Factor , STAT2 Transcription Factor , Shock, Septic/blood , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/immunology , Shock, Septic/pathology , TYK2 Kinase , Trans-Activators/deficiency
7.
J Neurochem ; 90(1): 231-44, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15198682

ABSTRACT

Over 100 mutants in superoxide dismutase 1 (SOD1) are reported in familial amyotrophic lateral sclerosis (ALS). However, the precise mechanism by which they are degraded through a ubiquitin-proteasomal pathway (UPP) remains unclear. Here, we report that heat-shock protein (Hsp) or heat-shock cognate (Hsc)70, and the carboxyl terminus of the Hsc70-interacting protein (CHIP), are involved in proteasomal degradation of mutant SOD1. Only mutant SOD1 interacted with Hsp/Hsc70 in vivo, and in vitro experiments revealed that Hsp/Hsc70 preferentially interacted with apo-SOD1 or dithiothreitol (DTT)-treated holo-SOD1, compared with metallated or oxidized forms. CHIP, a binding partner of Hsp/Hsc70, interacted only with mutant SOD1 and promoted its degradation. Both Hsp70 and CHIP promoted polyubiquitination of mutant SOD1-associated molecules, but not of mutant SOD1, indicating that mutant SOD1 is not a substrate of CHIP. Moreover, mutant SOD1-associated Hsp/Hsc70, a known substrate of CHIP, was polyubiquitinated in vivo, and polyubiquitinated Hsc70 by CHIP interacted with the S5a subunit of the 26S proteasome in vitro. Furthermore, CHIP was predominantly expressed in spinal neurons, and ubiquitinated inclusions in the spinal motor neurons of hSOD1(G93A) transgenic mice were CHIP-immunoreactive. Taken together, we propose a novel pathway in which ubiquitinated Hsp/Hsc70 might deliver mutant SOD1 to, and facilitate its degradation, at the proteasome.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , HSP70 Heat-Shock Proteins/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Superoxide Dismutase/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Cells, Cultured , Disease Models, Animal , HSC70 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/genetics , Humans , Inclusion Bodies/metabolism , Macromolecular Substances , Mice , Mice, Transgenic , Motor Neurons/metabolism , Mutation , Protein Subunits/metabolism , Superoxide Dismutase/genetics , Transfection , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Int Arch Allergy Immunol ; 134 Suppl 1: 25-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15166480

ABSTRACT

Mast cells are recognized not only as the major effector cells of type I hypersensitivity reactions but also as an important player of innate immune response against bacterial infection. Type I IFNs are also involved in the response against bacterial infection. However, the role of type I IFNs and their associated Janus kinase Tyk2 in mast cell functions remains to be determined. In this study, we addressed this issue using Tyk2-deficient (Tyk2(-/-)) bone marrow-derived mast cells (BMMCs). When BMMCs from wild-type (WT) mice were stimulated with IFN-alpha, they expressed mRNA for IFN-gamma-inducible protein 10 (IP-10) and monocyte chemoattractant protein-5 (MCP-5). Interestingly, IFN-alpha-induced expression of IP-10 and MCP-5 was severely decreased in Tyk2(-/-) BMMCs. In addition, IFN-alpha-induced Stat1 phosphorylation was decreased in Tyk2(-/-) BMMCs. On the other hand, IFN-alpha-induced Stat1 phosphorylation and IP-10 and MCP-5 expression were normal in Tyk2(-/-) fibroblasts. These results indicate that IFN-alpha induces the expression of TNF-alpha and the chemokines IP-10 and MCP-5 in mast cells and thatTyk2 plays a nonredundant role in IFN-alpha signaling in mast cells.


Subject(s)
Gene Expression Regulation/immunology , Interferon-alpha/pharmacology , Mast Cells/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/immunology , Animals , Cells, Cultured , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Flow Cytometry , Interferon-alpha/immunology , Mast Cells/immunology , Mice , Monocyte Chemoattractant Proteins/biosynthesis , Phosphorylation , Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor , TYK2 Kinase , Trans-Activators/drug effects , Trans-Activators/metabolism
9.
Exp Hematol ; 31(12): 1317-22, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14662340

ABSTRACT

OBJECTIVE: Limitin, an interferon-like cytokine, suppresses B lymphopoiesis through ligation of the interferon-alpha/beta (IFN-alpha/beta) receptor. The aim of this study was to examine the intracellular signal transduction pathways activated by limitin. MATERIALS AND METHODS: The effects of limitin on cell growth, the activation of Jak kinase and Stat proteins, and the induction of interferon regulatory factor-1 (IRF-1) and Daxx were examined using the mouse pre-B-cell line 18.81, wild-type, and Tyk2-deficient mouse bone marrow cells. In addition, the change of localization of the Daxx protein after limitin treatment in wild-type and Tyk2-deficient mice was examined. RESULTS: Limitin phosphorylates Tyk2, Jak1, Stat1, and Stat2 and rapidly induces IRF-1 mRNA production. Phosphorylation of Stat1 by limitin is partially dependent on Tyk2. Suppression of B-cell growth by limitin, however, is severely impaired in the absence of Tyk2, whereas it is unaffected by the absence of Stat1. Limitin also induces the expression and nuclear translocation of Daxx, which is essential for IFN-alpha-induced inhibition of B-lymphocyte development. The absence of Tyk2 abrogates this induction of Daxx expression and nuclear translocation. CONCLUSIONS: Limitin suppresses B-cell growth through activation of Tyk2, resulting in the up-regulation and nuclear translocation of Daxx. This limitin-mediated signaling pathway does not require Stat1.


Subject(s)
B-Lymphocytes/cytology , Carrier Proteins/metabolism , Immunoglobulins/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins/pharmacology , Nuclear Proteins/metabolism , Proteins/metabolism , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , B-Lymphocytes/drug effects , Bone Marrow Cells , Carrier Proteins/biosynthesis , Cell Division/drug effects , Cells, Cultured , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Janus Kinase 1 , Mice , Mice, Knockout , Molecular Chaperones , Nuclear Proteins/biosynthesis , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , STAT1 Transcription Factor , TYK2 Kinase , Trans-Activators/metabolism , Up-Regulation/drug effects
10.
Br J Haematol ; 123(3): 528-35, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14617019

ABSTRACT

Interferon (IFN)-alpha and IFN-gamma suppress the growth of haematopoietic progenitor cells. IFN-alpha activates Janus kinase-1 (Jak1) and Tyrosine kinase-2 (Tyk2), followed by the phosphorylation of the signal transducers and activators of transcription, Stat1 and Stat2. IFN-gamma activates Jak1 and Jak2, followed by the activation of Stat1. Activated Stats bind the promoter regions of IFN-inducible genes. We evaluated the role of Tyk2 and Stat1 in the IFN-mediated inhibition of haematopoietic progenitor cell growth. While IFN-alpha (1000 U/ml) suppressed the number of granulocyte-macrophage colony-forming units (CFU-GM) or erythroid burst-forming units (BFU-E) from wild-type mouse bone marrow cells, this suppression was partially inhibited by a deficiency in Tyk2 and completely inhibited by a deficiency in Stat1. High levels of IFN-alpha (10,000 U/ml) suppressed the CFU-GM or BFU-E obtained from Stat1-deficient mice, but did not suppress this growth in cells from Tyk2-deficient mice. Stat1 was phosphorylated by IFN-alpha in Tyk2-deficient cells, although the level of phosphorylation was weaker than that observed in wild type mice. Thus, the inhibitory signal on haematopoietic progenitor cells mediated by IFN-alpha may be transduced by two signalling pathways, one regulated by Tyk2 and the other dependent on Stat1. IFN-gamma also suppressed the number of CFU-GM or BFU-E, and this pathway was mediated by IFN-gamma in a Stat1-dependent manner, independently of Tyk2.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Interferons/pharmacology , Intracellular Fluid/immunology , Protein-Tyrosine Kinases , Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Cell Division/physiology , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , Depression, Chemical , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Knockout , Phosphorylation , Proteins/genetics , STAT1 Transcription Factor , TYK2 Kinase , Trans-Activators/genetics
11.
Cancer Res ; 63(7): 1623-30, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12670914

ABSTRACT

Accumulated evidence suggests that connexin43 (Cx43) serves as a tumor-suppressing gene. We have previously shownA. B. that Cx43 suppressed the G(1)-S phase cell cycle transition via increasing the level of p27 (Zhang, Y. W., et al., Oncogene, 20: 4138-4149, 2001). Here we report that Cx43 inhibited expression of Skp2, the human F-box protein that regulates p27 ubiquitination. This reduction was attributed to an increased degradation of Skp2. The Cx43 antisense oligonucleotide blocked this inhibitory effect of Cx43 on Skp2 expression and led to p27 down-regulation. In contrast, the antisense oligonucleotide of Skp2 induced a further increase in the level of p27. However, ectopic expression of Skp2 reversed the Cx43-induced Skp2 reduction, p27 accumulation, and cell proliferation inhibition. Cx43 increased p27 expression only in the SKP2 +/+ mouse embryo fibroblasts (MEFs), but not in the SKP2 -/- MEFs, indicating that Skp2 plays a critical role in the Cx43-induced p27 up-regulation. We also show that both Skp2 and p27 are required for Cx43 to inhibit cell proliferation, in that Cx43 hardly inhibited cell proliferation of the SKP2 -/- and p27 -/- MEFs, whereas it clearly did both in the SKP2 +/- and in the p27 +/- MEFs. Our findings suggest a new route for Cx43 to inhibit tumor growth by linking it with the key cell cycle regulators.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Connexin 43/physiology , Animals , COS Cells , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Division/physiology , Chlorocebus aethiops , Connexin 43/genetics , Cyclin-Dependent Kinase Inhibitor p27 , HeLa Cells , Humans , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Rats , S-Phase Kinase-Associated Proteins , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
12.
Biochem Biophys Res Commun ; 302(4): 635-45, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12646216

ABSTRACT

Ubiquitin-protein ligases (E3s) determine the substrate specificity of ubiquitylation and, until recently, had been classified into two families, the HECT and RING-finger families. The U-box is a domain of approximately 70 amino acids that is present in proteins from yeast to humans. The prototype U-box protein, yeast Ufd2, was identified as a ubiquitin chain assembly factor (E4) that cooperates with a ubiquitin-activating enzyme (E1), a ubiquitin-conjugating enzyme (E2), and an E3 to catalyze the formation of a ubiquitin chain on artificial substrates. We recently showed that mammalian U-box proteins, in conjunction with an E1 and an E2, mediate polyubiquitylation in the absence of a HECT type or RING-finger type E3. U-box proteins have thus been defined as a third family of E3s. We here review recent progress in the characterization of U-box proteins and of their role in the quality control system that underlies the cellular stress response to the intracellular accumulation of abnormal proteins.


Subject(s)
Ligases/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Humans , Ligases/chemistry , Ligases/classification , Ligases/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Multigene Family , Phylogeny , Protein Folding , Protein Structure, Tertiary , Saccharomyces cerevisiae/physiology , Sequence Alignment , Ubiquitin-Protein Ligases
13.
Genes Cells ; 8(4): 311-24, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12653960

ABSTRACT

BACKGROUND: Cell lines that stably over-express protein kinase C (PKC) delta frequently show a decrease in growth rate and saturation density, leading to the hypothesis that PKC delta has a negative effect on cell proliferation. However, the mode of PKC delta activation, the cell cycle stage requiring PKC delta activity, and the exact role of PKC delta at that stage remains unknown. RESULTS: Here we show that the treatment of quiescent fibroblasts with serum activates PKC delta at two distinct time points, within 10 min after serum treatment, and for a longer duration between 6 and 10 h. This biphasic activation correlates with the phosphorylation of Thr-505 at the activation loop of PKC delta. Importantly, an inhibitor of PKC delta, rottlerin, suppresses the biphasic activation of PKC delta, and suppression of the second phase of PKC delta activation is sufficient for the suppression of DNA synthesis. Consistent with this, the transient over-expression of PKC delta mutant molecules lacking kinase activity suppresses serum-induced DNA synthesis. These results imply that PKC delta plays a positive role in cell cycle progression. While the over-expression of PKC delta enhances serum-induced DNA synthesis, this was not observed for PKC epsilon. Similar experiments using a series of PKCdelta/ epsilon chimeras showed that the carboxyl-terminal 51 amino acids of PKC delta are responsible for the stimulatory effect. On the other hand, the over-expression of PKC delta suppresses cell entry into M-phase, being consistent with the previous studies based on stable over-expressors. CONCLUSIONS: We conclude that PKC delta plays a role in the late-G1 phase through the positive regulation of cell-cycle progression, in addition to negative regulation of the entry into M-phase.


Subject(s)
DNA/biosynthesis , G1 Phase/physiology , Protein Kinase C/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Cells, Cultured , Chimera , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Fluorescent Antibody Technique , In Vitro Techniques , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta , Protein Kinase C-epsilon , Rats , Threonine/metabolism , Up-Regulation
14.
Rinsho Shinkeigaku ; 43(11): 906-8, 2003 Nov.
Article in Japanese | MEDLINE | ID: mdl-15152500

ABSTRACT

Machado-Joseph disease (MJD)/Spinocerebellar ataxia type 3 (SCA3) is neurodegenerative disease which is caused by polyglutamine expansion in a responsible gene product, MJD1/Ataxin3. MJD1 has now been shown to undergo ubiquitylation and degradation by proteasome-dependent pathway. MJD1 with expanded polyglutamine tract was more resistant to degradation than normal MJD1. We established an in vitro system of ubiquitylation of MJD1, thereby biochemically purified activity to mediate polyubiquitylation of MJD1 from rabbit reticulocyte lysate. An AAA-family ATPase VCP was isolated from the active fraction, and found to binds to MJD1. Furthermore, UFD2a, a mammalian ubiquitin-chain assembly factor (E4), associated with VCP and induced polyubiquitylation of MJD1. UFD2a markedly promoted ubiquitylation and degradation of MJD1 with expanded polyglutamine tract, resulting in the clearance of MJD1 protein. In contrast, dominant-negative mutant UFD2a reduced the degradation rate of MJD1, leading to the formation of intracellular aggregation. In Drosophila model, overexpression of UFD2a significantly suppressed the neurodegeneration induced by expression of MJD1 with expanded polyglutamine tract. These findings suggest that E4 is a rate-limiting factor of degradation of pathologic polyglutamine-containing proteins, and may give a potential tool for gene therapy to control the clinical conditions of MJD.


Subject(s)
Machado-Joseph Disease/genetics , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Ubiquitin-Protein Ligases/physiology , Adenosine Triphosphatases/metabolism , Animals , Ataxin-3 , Cysteine Endopeptidases/metabolism , Genetic Therapy , Humans , Machado-Joseph Disease/therapy , Multienzyme Complexes/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins , Peptides/genetics , Proteasome Endopeptidase Complex , Repressor Proteins , Transcription Factors , Trinucleotide Repeat Expansion , Ubiquitin-Protein Ligases/genetics , Ubiquitins/metabolism
15.
Pharmacogenetics ; 12(8): 621-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12439222

ABSTRACT

Aldehyde dehydrogenase (ALDH) 2 plays a major role in the detoxification of aldehyde and is known to be responsible for alcohol preference. A diminished enzyme activity due to mutation of the Aldh2 gene is associated with high alcohol sensitivity and a low alcohol tolerance in humans. The genomic background distinguishing an alcohol preference and avoidance in various inbred mouse strains is not clear. We created Aldh2-negative mice by transgenic knockout of the Aldh2 gene into the high alcohol preference C57BL/6 background. The Aldh2 gene targeting (Aldh-/-) mice exhibited an alcohol avoidance characteristic. After free-choice ethanol and water drinking, brain and liver acetaldehyde concentrations of Aldh2-/- mice were almost equal to those of wild-type (Aldh2+/+) mice although the Aldh2-/- mice drank less ethanol than the Aldh2+/+ mice. This result indicates that a direct effect of the Aldh2 genotype plays an important role on alcohol preference and acetaldehyde concentration in the brain is correlated with alcohol avoidance. This highlights the potential benefits of alcoholism and alcohol-related disease research in the animal model of ALDH2 alleles.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Ethanol/administration & dosage , Acetaldehyde/blood , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Base Sequence , DNA Primers , Ethanol/blood , Genotype , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
J Immunol ; 169(9): 4707-11, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391177

ABSTRACT

IFN-alpha inhibits B lymphocyte development, and the nuclear protein Daxx has been reported to be essential for this biological activity. We show in this study that IFN-alpha inhibits the clonal proliferation of B lymphocyte progenitors in response to IL-7 in wild-type, but not in tyk2-deficient, mice. In addition, the IFN-alpha-induced up-regulation and nuclear translocation of Daxx are completely abrogated in the absence of tyk2. Therefore, tyk2 is directly involved in IFN-alpha signaling for the induction and translocation of Daxx, which may result in B lymphocyte growth arrest and/or apoptosis.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Carrier Proteins/metabolism , Growth Inhibitors/physiology , Immunosuppressive Agents/pharmacology , Interferon-alpha/physiology , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/physiology , Proteins/physiology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Co-Repressor Proteins , Coculture Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Interleukin-7/antagonists & inhibitors , Interleukin-7/physiology , Mice , Mice, Knockout , Molecular Chaperones , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Proteins/genetics , TYK2 Kinase
17.
Mol Cell ; 10(1): 55-67, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12150907

ABSTRACT

Unfolded Pael receptor (Pael-R) is a substrate of the E3 ubiquitin ligase Parkin. Accumulation of Pael-R in the endoplasmic reticulum (ER) of dopaminergic neurons induces ER stress leading to neurodegeneration. Here, we show that CHIP, Hsp70, Parkin, and Pael-R formed a complex in vitro and in vivo. The amount of CHIP in the complex was increased during ER stress. CHIP promoted the dissociation of Hsp70 from Parkin and Pael-R, thus facilitating Parkin-mediated Pael-R ubiquitination. Moreover, CHIP enhanced Parkin-mediated in vitro ubiquitination of Pael-R in the absence of Hsp70. Furthermore, CHIP enhanced the ability of Parkin to inhibit cell death induced by Pael-R. Taken together, these results indicate that CHIP is a mammalian E4-like molecule that positively regulates Parkin E3 activity.


Subject(s)
Ligases/metabolism , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/genetics , Animals , Blotting, Western , Cell Death , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , Humans , Ligases/genetics , Macromolecular Substances , Male , Mice , Models, Biological , Parkinsonian Disorders/pathology , Protein Binding , Protein Folding , Protein Transport , Rats , Rats, Wistar , Substantia Nigra/ultrastructure , Transfection , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
18.
Cancer Res ; 62(13): 3819-25, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12097295

ABSTRACT

Reduced expression level of p27, a cyclin-dependent kinase inhibitor, is associated with high aggressiveness and poor prognosis of various malignant tumors, including gastric carcinoma. S-phase kinase-associated protein 2 (Skp2), a member of the F-box family of substrate-recognition subunits of Skp1-Cullin-F-box ubiquitin-protein ligase complexes, is necessary for p27 ubiquitination and degradation. In the present study, we examined the clinical and biological significance of Skp2 expression in human gastric carcinoma and the relationship between the expression of Skp2 and p27. Northern blot analysis showed that Skp2 mRNA was overexpressed in carcinoma tissues (P < 0.05), and the high Skp2 expression group showed significantly poorer prognosis in 98 patients with gastric carcinoma (P < 0.05). Immunohistochemical analysis showed that Skp2 protein was expressed predominantly in carcinoma cells. We also found an inverse correlation between the expression of Skp2 mRNA and p27 protein in vivo (P < 0.01). To analyze the biological behavior of Skp2, we established stably Skp2-transfected gastric carcinoma cell lines. Western blot analysis showed that Skp2-transfected cells expressed lower levels of p27 protein than the control cells. Skp2-transfected cells showed significantly higher levels of growth rate (P < 0.05), percentage of bromodeoxyuridine-positive cells after serum starvation (P < 0.01), resistance to apoptosis induction by actinomycin D treatment (P < 0.05), and invasion potential (P < 0.01) than the control cells. These findings indicate that Skp2 expression can modulate the malignant phenotype of gastric carcinoma, possibly via p27 proteolysis. Skp2 can play an important role in gastric carcinoma progression and would be a novel target for the treatment of gastric carcinoma as well as a strong prognostic marker.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Division/genetics , Cell Movement/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Dactinomycin/pharmacology , Female , Gastric Mucosa/metabolism , Gene Expression , Humans , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , S-Phase Kinase-Associated Proteins , Stomach Neoplasms/genetics , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
19.
Blood ; 99(6): 2094-9, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11877284

ABSTRACT

Tyk2 is activated in response to interleukin-12 (IL-12) and is essential for IL-12-induced T-cell function, including interferon-gamma (IFN-gamma) production and Th1 cell differentiation. Because IL-12 is a stimulatory factor for natural killer (NK) cell-mediated cytotoxicity, we examined whether tyk2 is required for IL-12-induced NK cell activity. IL-12-induced NK cell activity in cells from tyk2-deficient mice was drastically reduced compared to that in cells from wild-type mice. IL-18 shares its biologic functions with IL-12. However, the molecular mechanism of IL-18 signaling, which activates an IL-1 receptor-associated kinase and nuclear translocation of nuclear factor-kappaB, is different from that of IL-12. We next examined whether biologic functions induced by IL-18 are affected by the absence of tyk2. NK cell activity and IFN-gamma production induced by IL-18 were reduced by the absence of tyk2. Moreover, the synergistic effect of IL-12 and IL-18 for the production of IFN-gamma was also abrogated by the absence of tyk2. This was partially due to the absence of any up-regulation of the IL-18 receptor treated with IL-12, and it might suggest the presence of the cross-talk between Jak-Stat and mitogen-activated protein kinase pathways in cytokine signaling.


Subject(s)
Interleukin-12/physiology , Interleukin-18/physiology , Protein-Tyrosine Kinases , Proteins/physiology , Signal Transduction/drug effects , Animals , Gene Expression Regulation/drug effects , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Interleukin-18 Receptor alpha Subunit , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Count , Mice , Mice, Knockout , Proteins/genetics , Proteins/pharmacology , Receptors, Interleukin/metabolism , Receptors, Interleukin-18 , TYK2 Kinase
20.
Biochem Biophys Res Commun ; 290(5): 1399-407, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11820777

ABSTRACT

The SCF complex is a type of ubiquitin-protein ligase (E3) that consists of invariable components, including Skp1, Cdc53/Cul1, and Rbx1, as well as variable components known as F-box proteins. Using a yeast two-hybrid system, we isolated six proteins that interact with Schizosaccharomyces pombe Skp1. Among them, Pof10 is a novel F-box protein consisting of 662 amino acids, harboring the F-box domain required for the binding to Skp1 and followed by four WD40 repeats. Overexpression of Pof10 in fission yeast resulted in loss of viability with marked morphological changes that are similar to those in pop1 mutant yeast. Coexpression of Skp1 with Pof10 prevented the lethality, suggesting that the lethality from Pof10 overexpression results from the sequestration of Skp1 from other F-box proteins including Pop1. Whereas most F-box proteins show rapid turnover, Pof10 has a remarkably long half-life in vivo and has been shown to be localized predominantly in cytoplasm. These results suggest that the stable F-box protein Pof10 might target abundant cytoplasmic proteins for degradation in fission yeast.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , F-Box Proteins , Fungal Proteins/chemistry , Fungal Proteins/isolation & purification , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/isolation & purification , Schizosaccharomyces/chemistry , Amino Acid Sequence , Base Sequence , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , Fungal Proteins/biosynthesis , Fungal Proteins/genetics , Gene Deletion , Half-Life , Molecular Sequence Data , Peptide Synthases/metabolism , S-Phase Kinase-Associated Proteins , SKP Cullin F-Box Protein Ligases , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/biosynthesis , Schizosaccharomyces pombe Proteins/genetics , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL