Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 8(17): 2794-801, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19652538

ABSTRACT

UV radiation causes a number of harmful events including growth delay, cell death and ultimately cancer. The reversal of such effects by concomitant exposure to visible light is a conserved mechanism which has been uncovered in many multi-cellular organisms. Here we show that light-dependent UV-tolerance is a cell autonomous phenomenon in zebrafish. In addition, we provide several lines of evidence indicating that light induction of 64PHR, a DNA repair enzyme, and the subsequent light-dependent DNA repair mediated by this enzyme are prerequisites for light-mediated UV tolerance. 64PHR is evolutionary related to and has a high degree of structural similarity to animal CRY, an essential circadian regulator. The zebrafish circadian clock is controlled by a cell-autonomous and light-dependent oscillator, where zCRY1a functions as an important mediator of light entrainment of the circadian clock. In this study, we show that light directly activates MAPK signaling cascades in zebrafish cells and we provide evidence that light-induced activation of these pathways controls the expression of two evolutionary-related genes, z64Phr and zCry1a, revealing that light-dependent DNA repair and the entrainment of circadian clock share common regulatory pathways.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , DNA Repair , Deoxyribodipyrimidine Photo-Lyase/metabolism , Light Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cells, Cultured , Cryptochromes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Eye Proteins/metabolism , MAP Kinase Signaling System , Period Circadian Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Time Factors , Ultraviolet Rays , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Biol Pharm Bull ; 32(7): 1183-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19571382

ABSTRACT

In the vertebrate circadian feedback loop, CLOCK:BMAL heterodimers induce the expression of Cry genes. The CRY proteins in turn inhibit CLOCK:BMAL-mediated transcription closing the negative feedback loop. Four CRYs, which all inhibit CLOCK:BMAL-mediated transcription, exist in zebrafish. Although these zebrafish Crys (zCry1a, 1b, 2a, and 2b) show a circadian pattern of expression, previous studies have indicated that the circadian oscillation of zCry1a could be CLOCK:BMAL-independent. Here we show that abrogation of CLOCK:BMAL-dependent transcription in zebrafish cells by the dominant negative zCLOCK3-DeltaC does not affect the circadian oscillation of zCry1a. Moreover, we provide several lines of evidence indicating that the extracellular signal-regulated kinase (ERK) signaling cascade modulates the circadian expression of zCry1a gene in constant darkness. Taken together, our data strongly support the notion that circadian oscillation of zCry1a is CLOCK:BMAL-independent and further indicate that mechanisms involving non-canonical clock genes could contribute to the circadian expression of zCry1a gene in a cell autonomous manner.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Circadian Rhythm/genetics , Trans-Activators/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , CLOCK Proteins , Cells, Cultured , Circadian Rhythm/physiology , Light , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Transcription, Genetic , Zebrafish/physiology
3.
J Biochem ; 137(2): 125-32, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15749826

ABSTRACT

Members of the IAP (inhibitor of apoptosis) family function as anti-apoptotic proteins by binding directly to caspase-3, -7, and -9 to inhibit their activities. During apoptosis, the activities of IAPs are relieved by a second mitochondria-derived caspase activator, named Smac/DIABLO. Some IAPs have a C-terminal RING finger domain that has been identified as the essential motif for the activity of ubiquitin ligase (E3). Here we show that X-linked IAP (XIAP) mediates the polyubiquitination of caspase-9 and Smac. The large subunit of mature caspase-9 was polyubiquitinated by XIAP in vitro, while procaspase-9 was not. Furthermore, the polyubiquitinated form of caspase-9 accumulated in an XIAP-dependent manner in intact cells. The ubiquitination of caspase-9 was significantly inhibited in the presence of mature Smac, whereas XIAP was also found to promote the polyubiquitination of cytosolic Smac both in vitro and in intact cells. These ubiquitination reactions require the RING finger domain of XIAP. These findings suggest that XIAP functions as ubiquitin ligase toward mature caspase-9 and Smac to inhibit apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , Mitochondrial Proteins/metabolism , Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Apoptosis Regulatory Proteins , Caspase 9 , Cells, Cultured , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein
4.
EMBO J ; 24(3): 452-63, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15660127

ABSTRACT

Cyclin E, an activator of phospho-CDK2 (pCDK2), is important for cell cycle progression in metazoans and is frequently overexpressed in cancer cells. It is essential for entry to the cell cycle from G0 quiescent phase, for the assembly of prereplication complexes and for endoreduplication in megakaryotes and giant trophoblast cells. We report the crystal structure of pCDK2 in complex with a truncated cyclin E1 (residues 81-363) at 2.25 A resolution. The N-terminal cyclin box fold of cyclin E1 is similar to that of cyclin A and promotes identical changes in pCDK2 that lead to kinase activation. The C-terminal cyclin box fold shows significant differences from cyclin A. It makes additional interactions with pCDK2, especially in the region of the activation segment, and contributes to CDK2-independent binding sites of cyclin E. Kinetic analysis with model peptide substrates show a 1.6-fold increase in kcat for pCDK2/cyclin E1 (81-363) over kcat of pCDK2/cyclin E (full length) and pCDK2/cyclin A. The structural and kinetic results indicate no inherent substrate discrimination between pCDK2/cyclin E and pCDK2/cyclin A with model substrates.


Subject(s)
CDC2-CDC28 Kinases/chemistry , CDC2-CDC28 Kinases/metabolism , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , CDC2-CDC28 Kinases/genetics , Centrosome/metabolism , Crystallography, X-Ray , Cyclin E , Cyclin-Dependent Kinase 2 , Cyclins/chemistry , Cyclins/genetics , Cyclins/metabolism , DNA/genetics , Enzyme Activation , Humans , In Vitro Techniques , Kinetics , Models, Molecular , Molecular Sequence Data , Molecular Structure , Multiprotein Complexes , Oncogene Proteins/genetics , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
5.
Cell Motil Cytoskeleton ; 59(4): 249-63, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15499654

ABSTRACT

The function of Aurora-C kinase, a member of the Aurora kinase family identified in mammals, is currently unknown. We present evidence that Aurora-C, like Aurora-B kinase, is a chromosomal passenger protein localizing first to centromeres and then to the midzone of mitotic cells. Aurora-C transcript is expressed at a moderate level albeit about an order of magnitude lower than Aurora-B transcript in diploid human fibroblasts. The level of Aurora-C transcript is elevated in several human cancer cell types. Aurora-C and Aurora-B mRNA and protein expressions are maximally elevated during the G2/M phase but their expression profiles in synchronized cells reveal differential temporal regulation through the cell cycle with Aurora-C level peaking after that of Aurora-B during the later part of the M phase. Aurora-C, like Aurora-B, interacts with the inner centromere protein (INCENP) at the carboxyl terminal end spanning the conserved IN box domain. Competition binding assays and transfection experiments revealed that, compared with Aurora-C, Aurora-B has preferential binding affinity to INCENP and co-expression of the two in vivo interferes with INCENP binding, localization, and stability of these proteins. A kinase-dead mutant of Aurora-C had a dominant negative effect inducing multinucleation in a dose-dependent manner. siRNA mediated silencing of Aurora-C and Aurora-B also gave rise to multinucleated cells with the two kinases silenced at the same time displaying an additive effect. Finally, Aurora-C could rescue the Aurora-B silenced multinucleation phenotype, demonstrating that Aurora-C kinase function overlaps with and complements Aurora-B kinase function in mitosis.


Subject(s)
Cell Division/physiology , Chromosomes, Human , Fibroblasts/enzymology , Protein Serine-Threonine Kinases/metabolism , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Cell Division/genetics , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , G2 Phase/genetics , G2 Phase/physiology , HL-60 Cells , HeLa Cells , Humans , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Cells, Cultured
6.
J Cell Biol ; 166(2): 167-72, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15263015

ABSTRACT

Mitotic kinases of the Polo and Aurora families are key regulators of chromosome segregation and cytokinesis. Here, we have investigated the role of MKlp1 and MKlp2, two vertebrate mitotic kinesins essential for cytokinesis, in the spatial regulation of the Aurora B kinase. Previously, we have demonstrated that MKlp2 recruits Polo-like kinase 1 (Plk1) to the central spindle in anaphase. We now find that in MKlp2 but not MKlp1-depleted cells the Aurora B-INCENP complex remains at the centromeres and fails to relocate to the central spindle. MKlp2 exerts dual control over Aurora B localization, because it is a binding partner for Aurora B, and furthermore for the phosphatase Cdc14A. Cdc14A can dephosphorylate INCENP and may contribute to its relocation to the central spindle in anaphase. We propose that MKlp2 is involved in the localization of Plk1, Aurora B, and Cdc14A to the central spindle during anaphase, and that the integration of signaling by these proteins is necessary for proper cytokinesis.


Subject(s)
Anaphase , Centromere/metabolism , Kinesins/physiology , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Zebrafish Proteins , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Humans , Macromolecular Substances , Metaphase , Microtubule-Associated Proteins/physiology , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Transport , Protein Tyrosine Phosphatases , Proto-Oncogene Proteins , Polo-Like Kinase 1
7.
J Cell Biol ; 166(2): 179-91, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15249581

ABSTRACT

The chromosomal passenger complex of Aurora B kinase, INCENP, and Survivin has essential regulatory roles at centromeres and the central spindle in mitosis. Here, we describe Borealin, a novel member of the complex. Approximately half of Aurora B in mitotic cells is complexed with INCENP, Borealin, and Survivin; and Borealin binds Survivin and INCENP in vitro. A second complex contains Aurora B and INCENP, but no Borealin or Survivin. Depletion of Borealin by RNA interference delays mitotic progression and results in kinetochore-spindle misattachments and an increase in bipolar spindles associated with ectopic asters. The extra poles, which apparently form after chromosomes achieve a bipolar orientation, severely disrupt the partitioning of chromosomes in anaphase. Borealin depletion has little effect on histone H3 serine10 phosphorylation. These results implicate the chromosomal passenger holocomplex in the maintenance of spindle integrity and suggest that histone H3 serine10 phosphorylation is performed by an Aurora B-INCENP subcomplex.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Spindle Apparatus , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Division , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes , Humans , Inhibitor of Apoptosis Proteins , Macromolecular Substances , Microtubule-Associated Proteins/metabolism , Mutation , Neoplasm Proteins , Recombinant Fusion Proteins/genetics , Sequence Alignment , Survivin
8.
Curr Opin Genet Dev ; 14(1): 29-36, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15108802

ABSTRACT

Aurora kinases play critical roles in chromosome segregation and cell division. They are implicated in the centrosome cycle, spindle assembly, chromosome condensation, microtubule-kinetochore attachment, the spindle checkpoint and cytokinesis. Aurora kinases are regulated through phosphorylation, the binding of specific partners and ubiquitin-dependent proteolysis. Several Aurora substrates have been identified and their roles are being elucidated. The deregulation of Aurora kinases impairs spindle assembly, checkpoint function and cell division, causing missegregation of individual chromosomes or polyploidization accompanied by centrosome amplification. Aurora kinases are frequently overexpressed in cancers and the identification of Aurora A as a cancer-susceptibility gene provides a strong link between mitotic errors and carcinogenesis.


Subject(s)
Chromosome Segregation/genetics , Gene Expression Regulation, Neoplastic , Mitosis/genetics , Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Aurora Kinases , Cell Division , Genetic Predisposition to Disease , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/genetics
9.
Mol Biol Cell ; 14(8): 3325-41, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12925766

ABSTRACT

The function of the Aurora B kinase at centromeres and the central spindle is crucial for chromosome segregation and cytokinesis, respectively. Herein, we have investigated the regulation of human Aurora B by its complex partners inner centromere protein (INCENP) and survivin. We found that overexpression of a catalytically inactive, dominant-negative mutant of Aurora B impaired the localization of the entire Aurora B/INCENP/survivin complex to centromeres and the central spindle and severely disturbed mitotic progression. Similar results were also observed after depletion, by RNA interference, of either Aurora B, INCENP, or survivin. These data suggest that Aurora B kinase activity and the formation of the Aurora B/INCENP/survivin complex both contribute to its proper localization. Using recombinant proteins, we found that Aurora B kinase activity was stimulated by INCENP and that the C-terminal region of INCENP was sufficient for activation. Under identical assay conditions, survivin did not detectably influence kinase activity. Human INCENP was a substrate of Aurora B and mass spectrometry identified three consecutive residues (threonine 893, serine 894, and serine 895) containing at least two phosphorylation sites. A nonphosphorylatable mutant (TSS893-895AAA) was a poor activator of Aurora B, demonstrating that INCENP phosphorylation is important for kinase activation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinases , Cell Division , Cells, Cultured , Centromere/metabolism , Cloning, Molecular , Enzyme Activation , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Mutation , Neoplasm Proteins , Phosphorylation , RNA Interference , Survivin
10.
Biochem Biophys Res Commun ; 302(4): 869-72, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12646252

ABSTRACT

Drosophila Numb protein functions as an antagonist against Notch signal. The expression of this protein is asymmetrical in divided cells and thought to be involved in the neural cell differentiation and/or cell fate. Human homologue of Numb (hNumb) was cloned as Mdm2-binding protein by yeast two-hybrid screening. Since Mdm2 is an oncoprotein and has ubiquitin ligase activity toward tumor suppressor p53, we assessed to find out whether Mdm2 ubiquitinylates the hNumb protein. The recombinant hNumb expressed in Sf-9 cells using baculovirus protein expression system bound to Mdm2 in vitro. When hNumb was subjected to in vitro ubiquitinylation assay system, which contains E1, E2, or UbcH5c, and Mdm2, hNumb was ubiquitinylated as efficiently as the p53 protein. However, when the Ring-finger domain mutant of Mdm2 was used in place of wild-type Mdm2, hNumb was not ubiquitinylated. Furthermore, when U2OS cells were co-transfected with hNumb and Mdm2, the hNumb protein was ubiquitinylated and degraded. These data strongly suggest that Mdm2 functions as the ubiquitin ligase toward hNumb and that it induces its degradation in intact cells.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Animals , Genes, Reporter , Humans , Protein Binding , Proto-Oncogene Proteins c-mdm2 , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Two-Hybrid System Techniques , Ubiquitin/metabolism
11.
J Biol Chem ; 277(51): 50131-6, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12393906

ABSTRACT

Mdm2, a ubiquitin ligase that acts on p53, is regulated by sumoylation. In the current study, we identify the enzymes responsible for the sumoylation of Mdm2. When mammalian cells are co-transfected with cDNAs encoding Mdm2 and PIAS1 or PIASxbeta (protein inhibitor of activated STAT) as sumoylation enzymes, Mdm2 is highly sumoylated. Mdm2 is also sumoylated in an in vitro system containing PIASxbeta, PIAS1, and RanBP2. When several lysine residues of Mdm2 were sequentially mutated to arginine, the K182R mutant was not sumoylated in intact cells; however, in the in vitro system this mutant was sumoylated by PIAS1, PIASxbeta, and RanBP2 as efficiently as the wild-type Mdm2 protein. Lysine residues 182 and 185 map within the nuclear localization signal of Mdm2. A K185R mutant of Mdm2 is sumoylated in intact cells, whereas a K182R protein is not. Only a Mdm2 protein bearing the K182R mutation is localized exclusively in the cytoplasm. Because RanBP2 is a nuclear pore protein and PIAS proteins are localized within the nucleus, our data suggest that Mdm2 is sumoylated during nuclear translocation by RanBP2 and then further sumoylated once in the nucleus by PIASxbeta and PIAS1.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA, Complementary/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Microscopy, Fluorescence , Molecular Chaperones , Mutation , Plasmids/metabolism , Protein Binding , Protein Inhibitors of Activated STAT , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
12.
EMBO J ; 21(4): 483-92, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11847097

ABSTRACT

Aberrations in centrosome numbers have long been implicated in aneuploidy and tumorigenesis, but their origins are unknown. Here we have examined how overexpression of Aurora-A kinase causes centrosome amplification in cultured cells. We show that excess Aurora-A does not deregulate centrosome duplication but gives rise to extra centrosomes through defects in cell division and consequent tetraploidization. Over expression of other mitotic kinases (Polo-like kinase 1 and Aurora-B) also causes multinucleation and concomitant increases in centrosome numbers. Absence of a p53 checkpoint exacerbates this phenotype, providing a plausible explanation for the centrosome amplification typical of p53-/- cells. We propose that errors during cell division, combined with the inability to detect the resulting hyperploidy, constitute a major cause for numerical centrosome aberrations in tumors.


Subject(s)
Centrosome , Gene Expression Regulation, Enzymologic , Polyploidy , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/physiology , Animals , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Cell Line , Cricetinae , Humans , Mice , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...