Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Inflamm Res ; 56(10): 414-20, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18026698

ABSTRACT

OBJECTIVE: Inflammation is associated with the invasion of leukocytes into affected tissues and with the up-regulation of platelet activation and adhesion. Assuming that leukocyte accumulation is linked to platelet aggregation, the aim of our study was to examine the effects of selective platelet inhibition by the glycoprotein (GP) IIb/IIIa receptor antagonist Tirofiban on the leukocyte-endothelial cell interaction. MATERIAL AND METHODS: We used the model of antigen-induced arthritis (AiA) to induce inflammatory changes in the synovial microcirculation. Ex vivo labelled platelets and in vivo fluorescence-labelled leukocytes were visualized by intravital microscopy (IVM). C57/Bl6 mice were allocated to four groups; two control groups with saline or Tirofiban and two groups with AiA that also received either saline or Tirofiban (0.5 microg/g BW) intravenously. RESULTS: There was no significant change in platelet- or leukocyte- endothelial cell interaction in the endothelium in healthy control animals. In contrast, after selective inhibition of platelets, the platelet- and leukocyte-endothelial cell interaction was significantly reduced in arthritic mice and reached the level of the healthy control groups. CONCLUSION: Selective platelet inhibition by Tirofiban resulted in reduced leukocyte-endothelial cell interactions in AiA. Consequently, platelets contribute to leukocyte adhesion in AiA via GPIIb/IIIa and therefore platelet inhibition could become an additional therapy option in chronic arthritic disease.


Subject(s)
Arthritis, Experimental/drug therapy , Cell Communication/drug effects , Endothelial Cells/drug effects , Leukocytes/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Tyrosine/analogs & derivatives , Animals , Arthritis, Experimental/blood , Endothelial Cells/physiology , Female , Leukocytes/physiology , Mice , Mice, Inbred C57BL , Tirofiban , Tyrosine/pharmacology
2.
Gynecol Oncol ; 83(2): 292-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606087

ABSTRACT

OBJECTIVE: To determine if p27(Kip1) expression was altered in epithelial ovarian cancers as compared to normal ovarian surface epithelial (NOSE) cells and to determine if subcellular localization of p27(Kip1) was an important feature. METHODS: Thirteen tumor samples (1 Stage IC [early] and 12 Stage III/IV [advanced]) from patients with epithelial ovarian cancer and five NOSE samples were evaluated. Samples were surgically dissected to obtain an enriched population (90%) of cancer cells. The level of p27(Kip1) protein expression was determined by Western blot analysis. Actin was used as a loading control, and results were quantified by scanning densitometry using the ratio of the p27(Kip1) signal to the actin signal for comparison. To evaluate the subcellular localization of p27(Kip1), immunocytochemical staining was performed. Clinical pathological parameters were correlated to nuclear p27(Kip1) staining to establish if any association existed. RESULTS: When comparing the expression of p27(Kip1) between NOSE and ovarian cancer samples, only 2 of 13 ovarian cancer samples had altered p27(Kip1) expression. No correlation was found between the expression level of p27(Kip1) on Western blot and clinical pathological correlates. While no correlation between expression level of p27(Kip1) and subcellular localization was found, decreased nuclear staining (1+) was associated with shorter survivals using the log-rank test (P < 0.001). More importantly, in all tumor samples examined under the microscope, no nuclear p27(Kip1) staining was noted in cells that were undergoing mitosis. CONCLUSIONS: p27(Kip1) protein degradation may not be modified in ovarian cancer cells undergoing mitosis. Altered expression of p27(Kip1) is not an overwhelming feature in certain epithelial ovarian cancers. Decreased nuclear staining of p27(Kip1) is associated with poor survival in some epithelial ovarian cancers.


Subject(s)
Cell Cycle Proteins/metabolism , Ovarian Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Cycle Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/pathology , Ovary/cytology , Ovary/metabolism , Subcellular Fractions/metabolism , Survival Rate , Tumor Suppressor Proteins/biosynthesis
3.
J Biol Chem ; 276(11): 7859-66, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11096118

ABSTRACT

The IL-1 receptor-associated kinase (IRAK/mPLK) is linked to the regulation of nuclear factor-kappaB (NF-kappaB)-dependent gene expression. Here we describe a novel binding partner of IRAK/mPLK that we term SIMPL (signaling molecule that associates with the mouse pelle-like kinase). Overexpression of SIMPL leads to the activation of NF-kappaB-dependent promoters, and inactivation of SIMPL inhibits IRAK/mPLK as well as tumor necrosis factor receptor type I-induced NF-kappaB activity. Dominant inhibitory alleles of IkappaB kinase (IKKalpha or IKKbeta) block the activation of NF-kappaB by IRAK/mPLK and SIMPL. Furthermore, SIMPL binds IRAK/mPLK and the IKKs in vitro and in vivo. In the presence of antisense mRNA to SIMPL, the physical association between IRAK/mPLK and IKKbeta but not IRAK/mPLK and IKKalpha is greatly diminished. Moreover, dominant-negative SIMPL blocks IKKalpha- or IKKbeta-induced NF-kappaB activity. These results lead us to propose a model in which SIMPL functions to regulate NF-kappaB activity by linking IRAK/mPLK to IKKbeta/alpha-containing complexes.


Subject(s)
Antigens, CD/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/physiology , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/analysis , I-kappa B Kinase , Interleukin-1 Receptor-Associated Kinases , Molecular Sequence Data , Protein Kinases/analysis , Protein Kinases/physiology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/physiology , Receptors, Tumor Necrosis Factor, Type I , Transcriptional Activation , Two-Hybrid System Techniques
4.
Mol Cell Biol ; 20(21): 7845-52, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11027256

ABSTRACT

Ubiquitin-mediated degradation plays a crucial role in many fundamental biological pathways, including the mediation of cellular responses to changes in environmental conditions. A family of ubiquitin ligase complexes, called SCF complexes, found throughout eukaryotes, is involved in a variety of biological pathways. In Saccharomyces cerevisiae, an SCF complex contains a common set of components, namely, Cdc53p, Skp1p, and Hrt1p. Substrate specificity is defined by a variable component called an F-box protein. The F- box is a approximately 40-amino-acid motif that allows the F-box protein to bind Skp1p. Each SCF complex recognizes different substrates according to which F-box protein is associated with the complex. In yeasts, three SCF complexes have been demonstrated to associate with the ubiquitin-conjugating enzyme Cdc34p and have ubiquitin ligase activity. F-box proteins are not abundant and are unstable. As part of the SCF(Met30p) complex, the F-box protein Met30p represses methionine biosynthetic gene expression when availability of L-methionine is high. Here we demonstrate that in vivo SCF(Met30p) complex activity can be regulated by the abundance of Met30p. Furthermore, we provide evidence that Met30p abundance is regulated by the availability of L-methionine. We propose that the cellular responses mediated by an SCF complex are directly regulated by environmental conditions through the control of F-box protein stability.


Subject(s)
Cullin Proteins , Methionine/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Stem Cell Factor/metabolism , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Blotting, Western , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , F-Box Proteins , Flow Cytometry , Glutathione Transferase/metabolism , Ligases/chemistry , Ligases/metabolism , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptide Synthases/metabolism , Plasmids/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , S-Phase Kinase-Associated Proteins , SKP Cullin F-Box Protein Ligases , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Temperature , Time Factors , Ubiquitin-Protein Ligases , beta-Galactosidase/metabolism
5.
Mol Cell Biol ; 19(10): 6500-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490590

ABSTRACT

The death domain-containing receptor superfamily and their respective downstream mediators control whether or not cells initiate apoptosis or activate NF-kappaB, events critical for proper immune system function. A screen for upstream activators of NF-kappaB identified a novel serine-threonine kinase capable of activating NF-kappaB and inducing apoptosis. Based upon domain organization and sequence similarity, this novel kinase, named mRIP3 (mouse receptor interacting protein 3), appears to be a new RIP family member. RIP, RIP2, and mRIP3 contain an N-terminal kinase domain that share 30 to 40% homology. In contrast to the C-terminal death domain found in RIP or the C-terminal caspase-recruiting domain found in RIP2, the C-terminal tail of mRIP3 contains neither motif and is unique. Despite this feature, overexpression of the mRIP3 C terminus is sufficient to induce apoptosis, suggesting that mRIP3 uses a novel mechanism to induce death. mRIP3 also induced NF-kappaB activity which was inhibited by overexpression of either dominant-negative NIK or dominant-negative TRAF2. In vitro kinase assays demonstrate that mRIP3 is catalytically active and has autophosphorylation site(s) in the C-terminal domain, but the mRIP3 catalytic activity is not required for mRIP3 induced apoptosis and NF-kappaB activation. Unlike RIP and RIP2, mRIP3 mRNA is expressed in a subset of adult tissues and is thus likely to be a tissue-specific regulator of apoptosis and NF-kappaB activity. While the lack of a dominant-negative mutant precludes linking mRIP3 to a known upstream regulator, characterizing the expression pattern and the in vitro functions of mRIP3 provides insight into the mechanism(s) by which cells modulate the balance between survival and death in a cell-type-specific manner.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , NF-kappa B/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Caspases/metabolism , DNA Damage , Fas-Associated Death Domain Protein , Gene Library , Genes, Reporter , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Precipitin Tests , Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases , Sequence Homology, Amino Acid , TNF Receptor-Associated Factor 2 , NF-kappaB-Inducing Kinase
6.
J Biol Chem ; 274(19): 13077-84, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224059

ABSTRACT

The innate immune response is an important defense against pathogenic agents. A component of this response is the NF-kappaB-dependent activation of genes encoding inflammatory cytokines such as interleukin-8 (IL-8) and cell adhesion molecules like E-selectin. Members of the serine/threonine innate immune kinase family of proteins have been proposed to mediate the innate immune response. One serine/threonine innate immune kinase family member, the mouse Pelle-like kinase/human interleukin-1 receptor-associated kinase (mPLK/IRAK), has been proposed to play an obligate role in promoting IL-1-mediated inflammation. However, it is currently unknown whether mPLK/IRAK catalytic activity is required for IL-1-dependent NF-kappaB activation. The present study demonstrates that mPLK/IRAK catalytic activity is not required for IL-1-mediated activation of an NF-kappaB-dependent signal. Intriguingly, catalytically inactive mPLK/IRAK inhibits type 1 tumor necrosis factor (TNF) receptor-dependent NF-kappaB activation. The pathway through which mPLK/IRAK mediates this TNF response is TRADD- and TRAF2-independent. Our data suggest that in addition to its role in IL-1 signaling, mPLK/IRAK is a component of a novel signal transduction pathway through which TNF R1 activates NF-kappaB-dependent gene expression.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Interleukin-1/metabolism , NF-kappa B/metabolism , Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigens, CD/metabolism , Cell Line , Humans , Interleukin-1 Receptor-Associated Kinases , Mice , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction
7.
Mol Cell Biol ; 19(3): 1759-67, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022863

ABSTRACT

Posttranslational modification of a protein by ubiquitin usually results in rapid degradation of the ubiquitinated protein by the proteasome. The transfer of ubiquitin to substrate is a multistep process. Cdc4p is a component of a ubiquitin ligase that tethers the ubiquitin-conjugating enzyme Cdc34p to its substrates. Among the domains of Cdc4p that are crucial for function are the F-box, which links Cdc4p to Cdc53p through Skp1p, and the WD-40 repeats, which are required for binding the substrate for Cdc34p. In addition to Cdc4p, other F-box proteins, including Grr1p and Met30p, may similarly act together with Cdc53p and Skp1p to function as ubiquitin ligase complexes. Because the relative abundance of these complexes, known collectively as SCFs, is important for cell viability, we have sought evidence of mechanisms that modulate F-box protein regulation. Here we demonstrate that the abundance of Cdc4p is subject to control by a peptide segment that we term the R-motif (for "reduced abundance"). Furthermore, we show that binding of Skp1p to the F-box of Cdc4p inhibits R-motif-dependent degradation of Cdc4p. These results suggest a general model for control of SCF activities.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins , Fungal Proteins/metabolism , Ubiquitin-Protein Ligases , Binding Sites , Cell Cycle , Cell Cycle Proteins/genetics , Fungal Proteins/genetics , Glutathione Transferase/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S-Phase Kinase-Associated Proteins , Signal Transduction , Structure-Activity Relationship , Transfection
8.
Mol Cell Biol ; 18(9): 4994-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9710583

ABSTRACT

MyoD is a basic helix-loop-helix transcription factor involved in the activation of genes encoding skeletal muscle-specific proteins. Independent of its ability to transactivate muscle-specific genes, MyoD can also act as a cell cycle inhibitor. MyoD activity is regulated by transcriptional and posttranscriptional mechanisms. While MyoD can be found phosphorylated, the functional significance of this posttranslation modification has not been established. MyoD contains several consensus cyclin-dependent kinase (CDK) phosphorylation sites. In these studies, we examined whether a link could be established between MyoD activity and phosphorylation at putative CDK sites. Site-directed mutagenesis of potential CDK phosphorylation sites in MyoD revealed that S200 is required for MyoD hyperphosphorylation as well as the normally short half-life of the MyoD protein. Additionally, we determined that turnover of the MyoD protein requires the proteasome and Cdc34 ubiquitin-conjugating enzyme activity. Results of these studies demonstrate that hyperphosphorylated MyoD is targeted for rapid degradation by the ubiquitin pathway. The targeted degradation of MyoD following CDK phosphorylation identifies a mechanism through which MyoD activity can be regulated coordinately with the cell cycle machinery (CDK2 and CDK4) and/or coordinately with the cellular transcriptional machinery (CDK7, CDK8, and CDK9).


Subject(s)
Cell Nucleus/metabolism , Cyclin-Dependent Kinases/metabolism , MyoD Protein/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Cycle , Cell Line , Kinetics , Mice , Mutagenesis, Site-Directed , MyoD Protein/chemistry , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine , Transcriptional Activation , Transfection
9.
Genes Dev ; 12(7): 914-26, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9531531

ABSTRACT

The RUB1/NEDD-8 family of ubiquitin-related genes is widely represented among eukaryotes. Here we report that Cdc53p in Saccharomyces cerevisiae, a member of the Cullin family of proteins, is stably modified by the covalent attachment of a single Rub1p molecule. Two genes have been identified that are required for Rub1p conjugation to Cdc53p. The first gene, designated ENR2, encodes a protein with sequence similarity to the amino-terminal half of the ubiquitin-activating enzyme. By analogy with Aos1p, we infer that Enr2p functions in a bipartite Rub1p-activating enzyme. The second gene is SKP1, shown previously to be required for some ubiquitin-conjugation events. A deletion allele of ENR2 is lethal with temperature-sensitive alleles of cdc34 and enhances the phenotypes of cdc4, cdc53, and skp1, strongly implying that Rub1p conjugation to Cdc53p is required for optimal assembly or function of the E3 complex SCFCdc4. Consistent with this model, both enr2delta and an allele of Cdc53p that is not Rub1p modified, render cells sensitive to alterations in the levels of Cdc4p, Cdc34p, and Cdc53p.


Subject(s)
Cell Cycle Proteins/metabolism , Cullin Proteins , Fungal Proteins/genetics , Genes, cdc/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/chemistry , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Cell Cycle Proteins/genetics , Fungal Proteins/metabolism , Ligases/genetics , Ligases/metabolism , Molecular Sequence Data , Protein Processing, Post-Translational , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
10.
J Biol Chem ; 273(7): 4040-5, 1998 Feb 13.
Article in English | MEDLINE | ID: mdl-9461595

ABSTRACT

The CDC34 gene of the yeast Saccharomyces cerevisiae encodes a ubiquitin-conjugating protein that transfers ubiquitin onto substrates to signal rapid degradation via the proteasome. Cdc34p has been implicated in signaling the destruction of a variety of substrates including the cyclin-dependent kinase inhibitor, Sic1p, which must be degraded for cells to enter S-phase. Mutants lacking CDC34 activity fail to degrade Sic1p and fail to enter S-phase, a phenotype that is also shared with cells lacking CDC4 and CDC53 activity. Here we demonstrate that Cdc4p, Cdc34p, and Cdc53p interact in vivo. We have mapped a Cdc4p/Cdc53p-binding region on Cdc34p; this region is essential for S-phase entry and thus the association of these three proteins is required for Sic1p degradation. All three proteins migrate in gel filtration to sizes that greatly exceed their actual size suggesting that they form stable associations with other proteins and we observe Cdc4p, Cdc34p, and Cdc53p fractionating into overlapping families of high molecular weight complexes. Finally, we demonstrate that Cdc4p, Cdc34p, and Cdc53p are stable throughout the cell cycle and that Cdc34p permanently resides as part of a complex throughout the cell cycle. This suggests that all Cdc34p substrates are ubiquitinated by a similar high molecular weight complex.


Subject(s)
Cell Cycle Proteins/metabolism , Cullin Proteins , F-Box Proteins , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases , Anaphase-Promoting Complex-Cyclosome , Binding Sites , Cell Cycle/genetics , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor Proteins , Cysteine Endopeptidases/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/genetics , Ligases/chemistry , Macromolecular Substances , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , Protein Binding , Saccharomyces cerevisiae/genetics , Signal Transduction/physiology , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism
11.
Mol Gen Genet ; 256(4): 365-75, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9393434

ABSTRACT

In Saccharomyces cerevisiae, entry into S phase requires the activation of the protein kinase Cdc28p through binding with cyclin Clb5p or Clb6p, as well as the destruction of the cyclin-dependent kinase inhibitor Sic1p. Mutants that are defective in this activation event arrest after START, with unreplicated DNA and multiple, elongated buds. These mutants include cells defective in CDC4, CDC34 or CDC53, as well as cells that have lost all CLB function. Here we describe mutations in another gene, CAK1, that lead to a similar arrest. Cells that are defective in CAK1 are inviable and arrest with a single nucleus and multiple, elongated buds. CAK1 encodes a protein kinase most closely related to the Cdc2p family of protein kinases. Mutations that lead to the production of an inactive kinase that can neither autophosphorylate, nor phosphorylate Cdc28p in vitro are also incapable of rescuing a cell with a deletion of CAK1. These results underscore the importance of the Cak1p protein kinase activity in cell cycle progression.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Animals , CDC28 Protein Kinase, S cerevisiae/metabolism , DNA Mutational Analysis , Molecular Sequence Data , Phenotype , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/chemistry , Rabbits , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Cyclin-Dependent Kinase-Activating Kinase
12.
Yeast ; 13(3): 233-40, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9090052

ABSTRACT

Although the entire DNA sequence of the yeast genome has been determined, the functions of nearly a third of the identified genes are unknown. Recently, we described a collection of mutants, each with a transposon-tagged disruption in an essential gene in Saccharomyces cerevisiae. Identification of these essential genes and characterization of their mutant phenotypes should help assign functions to these thousands of novel genes, and since each mutation in our collection is physically marked by the uniform, unique DNA sequence of the transposable element, it should be possible to use the polymerase chain reaction (PCR) to amplify the DNA adjacent to the transposon. However, existing PCR methods include steps that make their use on a large scale cumbersome. In this report, we describe a semi-random, two-step PCR protocol, ST-PCR. This method is simpler and more specific than current methods, requiring only genomic DNA and two pairs of PCR primers, and involving two successive PCR reactions. Using this method, we have rapidly and easily identified the essential genes identified by several of our mutants.


Subject(s)
DNA Transposable Elements/genetics , DNA, Fungal/genetics , Genes, Fungal/physiology , Polymerase Chain Reaction/methods , Saccharomyces cerevisiae/genetics , Cloning, Molecular , Genome, Fungal , Sequence Analysis, DNA
13.
Mol Cell Biol ; 16(12): 6634-43, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8943317

ABSTRACT

Regulation of cell cycle progression occurs in part through the targeted degradation of both activating and inhibitory subunits of the cyclin-dependent kinases. During G1, CDC4, encoding a WD-40 repeat protein, and CDC34, encoding a ubiquitin-conjugating enzyme, are involved in the destruction of these regulators. Here we describe evidence indicating that CDC53 also is involved in this process. Mutations in CDC53 cause a phenotype indistinguishable from those of cdc4 and cdc34 mutations, numerous genetic interactions are seen between these genes, and the encoded proteins are found physically associated in vivo. Cdc53p defines a large family of proteins found in yeasts, nematodes, and humans whose molecular functions are uncharacterized. These results suggest a role for this family of proteins in regulating cell cycle proliferation through protein degradation.


Subject(s)
CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Cell Cycle , F-Box Proteins , G1 Phase/genetics , S Phase/genetics , Saccharomyces cerevisiae/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases , Amino Acid Sequence , Base Sequence , Gene Expression Regulation , Molecular Sequence Data , Mutation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins , Sequence Alignment
14.
J Biol Chem ; 271(30): 17609-12, 1996 Jul 26.
Article in English | MEDLINE | ID: mdl-8663605

ABSTRACT

The NF-kappaB/c-Rel proteins are a family of evolutionarily conserved transcription factors activated during development that in the adult, mediate many processes including the immune response. A high degree of sequence similarity is shared between the NF-kappaB/c-Rel family of transcription factors and the Drosophila Dorsal protein as well as between its cytoplasmic inhibitor, IkappaBalpha, and the Drosophila Cactus protein. Genetic analyses of Dorsal have defined components of a signaling pathway for Dorsal activation, including a serine/threonine kinase, Pelle, placed upstream of Dorsal and Cactus. We demonstrate that this pathway is likely to be conserved in mammals by the isolation of a cDNA that encodes a novel mouse protein highly related to Pelle, mPLK (mouse Pelle-like protein kinase). Expression of mPLK mRNA is developmentally regulated in the mouse and in adult tissue mPLK expression is greatest in the liver, a tissue that expresses a high level of NF-kappaB. Recombinant mPLK produced in bacteria is a protein kinase capable of autophosphorylating and phosphorylating IkappaBalpha.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila Proteins , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cloning, Molecular , Interleukin-1 Receptor-Associated Kinases , Mice , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Sequence Homology, Amino Acid , Signal Transduction , Tissue Distribution
15.
Cell ; 86(2): 263-74, 1996 Jul 26.
Article in English | MEDLINE | ID: mdl-8706131

ABSTRACT

We have identified the yeast and human homologs of the SKP1 gene as a suppressor of cdc4 mutants and as a cyclin F-binding protein. Skp1p indirectly binds cyclin A/Cdk2 through Skp2p, and directly binds Skp2p, cyclin F, and Cdc4p through a novel structural motif called the F-box. SKP1 is required for ubiquitin-mediated proteolysis of Cin2p, Clb5p, and the Cdk inhibitor Sic1p, and provides a link between these molecules and the proteolysis machinery. A large number of proteins contain the F-box motif and are thereby implicated in the ubiquitin pathway. Different skp1 mutants arrest cells in either G1 or G2, suggesting a connection between regulation of proteolysis in different stages of the cycle.


Subject(s)
Cell Cycle Proteins/chemistry , F-Box Proteins , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases , Ubiquitins/physiology , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Base Sequence , Binding Sites/physiology , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cyclins/chemistry , Cyclins/genetics , Cyclins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Fungal Proteins/physiology , G1 Phase/physiology , G2 Phase/physiology , Gene Dosage , Gene Expression/physiology , Humans , Ligases/genetics , Mitosis/physiology , Molecular Sequence Data , Molecular Structure , Mutation/physiology , Protein Structure, Tertiary , Proteins/metabolism , S Phase/physiology , S-Phase Kinase-Associated Proteins , Saccharomyces cerevisiae Proteins , Yeasts/genetics
16.
Genetics ; 143(1): 119-27, 1996 May.
Article in English | MEDLINE | ID: mdl-8722767

ABSTRACT

Mutations in GLC7, the gene encoding the type 1 protein phosphatase catalytic subunit, cause a variety of abberrant phenotypes in yeast, such as impaired glycogen synthesis and relief of glucose repression of the expression of some genes. Loss of function of the REG1/HEX2 gene, necessary for glucose repression of several genes, was found to suppress the glycogen-deficient phenotype of the glc7-1 allele. Deletion of REG1 in a wild-type background led to overaccumulation of glycogen as well as slow growth and an enlarged cell size. However, loss of REG1 did not suppress other phenotypes associated with GLC7 mutations, such as inability to sporulate or, in cells bearing the glc7Y-170 allele, lack of growth at 14 degrees. The effect of REG1 deletion on glycogen accumulation is not simply due to derepression of glucose-repressed genes, although it does require the presence of SNF1, which encodes a protein kinase essential for expression of glucose-repressed genes and for glycogen accumulation. We propose that REG1 has a role in controlling glycogen accumulation.


Subject(s)
Fungal Proteins/genetics , Genes, Fungal , Phosphoprotein Phosphatases/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Base Sequence , DNA Primers , Fungal Proteins/biosynthesis , Gene Deletion , Genetic Complementation Test , Genotype , Glycogen/metabolism , Molecular Sequence Data , Mutagenesis , Mutagenesis, Insertional , Phosphoprotein Phosphatases/biosynthesis , Polymerase Chain Reaction , Protein Phosphatase 1 , Restriction Mapping
17.
Mol Cell Biol ; 16(2): 677-84, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8552096

ABSTRACT

The Cdc34 (Ubc3) ubiquitin-conjugating enzyme from Saccharomyces cerevisiae plays an essential role in the progression of cells from the G1 to S phase of the cell division cycle. Using a high-copy suppression strategy, we have identified a yeast gene (UBS1) whose elevated expression suppresses the conditional cell cycle defects associated with cdc34 mutations. The UBS1 gene encodes a 32.2-kDa protein of previously unknown function and is identical in sequence to a genomic open reading frame on chromosome II (GenBank accession number Z36034). Several lines of evidence described here indicate that Ubs1 functions as a general positive regulator of Cdc34 activity. First, overexpression of UBS1 suppresses not only the cell proliferation and morphological defects associated with cdc34 mutants but also the inability of cdc34 mutant cells to degrade the general amino acid biosynthesis transcriptional regulator, Gcn4. Second, deletion of the UBS1 gene profoundly accentuates the cell cycle defect when placed in combination with a cdc34 temperature-sensitive allele. Finally, a comparison of the Ubs1 and Cdc34 polypeptide sequences reveals two noncontiguous regions of similarity, which, when projected onto the three-dimensional structure of a ubiquitin-conjugating enzyme, define a single region situated on its surface. While cdc34 mutations corresponding to substitutions outside this region are suppressed by UBS1 overexpression, Ubs1 fails to suppress amino acid substitutions made within this region. Taken together with other findings, the allele specificity exhibited by UBS1 expression suggests that Ubs1 regulates Cdc34 by interaction or modification.


Subject(s)
Cell Cycle/genetics , DNA-Binding Proteins , Fungal Proteins/genetics , Genes, Fungal , Genes, Regulator , Ligases/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Base Sequence , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/growth & development , Sequence Homology, Amino Acid , Suppression, Genetic , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
18.
Genetics ; 142(1): 39-50, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8770583

ABSTRACT

The yeast Saccharomyces cerevisiae reproduces by budding, and many genes are required for proper bud development. Mutations in some of these genes cause cells to die with an unusual terminal morphology-elongated or otherwise aberrantly shaped buds. To gain insight into bud development, we set out to identify novel genes that encode proteins required for proper bud morphogenesis. Previous studies screened collections of conditional mutations to identify genes required for essential functions, including bud formation. However, genes that are not susceptible to the generation of mutations that cause a conditional phenotype will not be identified in such screens. To identify a more comprehensive collection of mutants, we used transposon mutagenesis to generate a large collection of lethal disruption mutations. This collection was used to identify 209 mutants with disruptions that cause an aberrant terminal bud morphology. The disruption mutations in 33 of these mutants identify three previously uncharacterized genes as essential, and the mutant phenotypes suggest roles for their products in bud morphogenesis.


Subject(s)
DNA Transposable Elements , Genes, Fungal , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , DNA Primers/genetics , DNA, Fungal/genetics , Fungal Proteins/genetics , Genes, Lethal , Genomic Library , Haploidy , Molecular Sequence Data , Mutagenesis, Insertional , Open Reading Frames , Phenotype , Saccharomyces cerevisiae/growth & development , Sequence Tagged Sites , Spores, Fungal/genetics
19.
Prog Cell Cycle Res ; 2: 115-27, 1996.
Article in English | MEDLINE | ID: mdl-9552389

ABSTRACT

Genetic and biochemical data indicate that ubiquitin-mediated proteolysis is involved in the regulated turnover of proteins required for controlling cell cycle progression. In general, mutations in some genes that encode proteins involved in the ubiquitin pathway cause cell cycle defects and affect the turnover of cell cycle regulatory proteins. Furthermore, some cell cycle regulatory proteins are short-lived, ubiquitinated, and degraded by the ubiquitin pathway. This review will examine how the ubiquitin pathway plays a role in regulating progression from the G1 to the S phase of the cell cycle, as well as the G2 to M phase transition.


Subject(s)
Cell Cycle/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligase Complexes , Ubiquitins/metabolism , Anaphase-Promoting Complex-Cyclosome , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor Proteins , Cyclins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Fungal Proteins/metabolism , Genes, Fungal , Ligases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Saccharomyces cerevisiae/genetics , Ubiquitin-Protein Ligases
20.
Mol Cell Biol ; 15(12): 6632-40, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8524228

ABSTRACT

Glycogen, a branched polymer of glucose, is a storage molecule whose accumulation is under rigorous nutritional control in many cells. We report the identification of two Saccharomyces cerevisiae genes, GLG1 and GLG2, whose products are implicated in the biogenesis of glycogen. These genes encode self-glucosylating proteins that in vitro can act as primers for the elongation reaction catalyzed by glycogen synthase. Over a region of 258 residues, the Glg proteins have 55% sequence identify to each other and approximately 33% identity to glycogenin, a mammalian protein postulated to have a role in the initiation of glycogen biosynthesis. Yeast cells defective in either GLG1 or GLG2 are similar to the wild type in their ability to accumulate glycogen. Disruption of both genes results in the inability of the cells to synthesize glycogen despite normal levels of glycogen synthase. These results suggest that a self-glucosylating protein is required for glycogen biosynthesis in a eukaryotic cell. The activation state of glycogen synthase in glg1 glg2 cells is suppressed, suggesting that the Glg proteins may additionally influence the phosphorylation state of glycogen synthase.


Subject(s)
Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycogen/biosynthesis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , 1,4-alpha-Glucan Branching Enzyme/genetics , 1,4-alpha-Glucan Branching Enzyme/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Primers , Genes, Fungal , Genotype , Glycogen Synthase/metabolism , Kinetics , Molecular Sequence Data , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Sequence Tagged Sites
SELECTION OF CITATIONS
SEARCH DETAIL
...