Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Results Probl Cell Differ ; 67: 391-411, 2019.
Article in English | MEDLINE | ID: mdl-31435805

ABSTRACT

Centrosomes are tiny yet complex cytoplasmic structures that perform a variety of roles related to their ability to act as microtubule-organizing centers. Like the genome, centrosomes are single copy structures that undergo a precise semi-conservative replication once each cell cycle. Precise replication of the centrosome is essential for genome integrity, because the duplicated centrosomes will serve as the poles of a bipolar mitotic spindle, and any number of centrosomes other than two will lead to an aberrant spindle that mis-segregates chromosomes. Indeed, excess centrosomes are observed in a variety of human tumors where they generate abnormal spindles in situ that are thought to participate in tumorigenesis by driving genomic instability. At the heart of the centrosome is a pair of centrioles, and at the heart of centrosome duplication is the replication of this centriole pair. Centriole replication proceeds through a complex macromolecular assembly process. However, while centrosomes may contain as many as 500 proteins, only a handful of proteins have been shown to be essential for centriole replication. Our observations suggest that centriole replication is a modular, bottom-up process that we envision akin to building a house; the proper site of assembly is identified, a foundation is assembled at that site, and subsequent modules are added on top of the foundation. Here, we discuss the data underlying our view of modularity in the centriole assembly process, and suggest that non-essential centriole assembly factors take on greater importance in cancer cells due to their function in coordination between centriole modules, using the Monopolar spindles 1 protein kinase and its substrate Centrin 2 to illustrate our model.


Subject(s)
Centrioles/metabolism , Neoplasms/pathology , Cell Division , Humans , Spindle Apparatus
2.
Bioorg Med Chem ; 25(7): 2156-2166, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28259529

ABSTRACT

New targeted therapy approaches for certain subtypes of breast cancer, such as triple-negative breast cancers and other aggressive phenotypes, are desired. High levels of the mitotic checkpoint kinase Mps1/TTK have correlated with high histologic grade in breast cancer, suggesting a potential new therapeutic target for aggressive breast cancers (BC). Novel small molecules targeting Mps1 were designed by computer assisted docking analyses, and several candidate compounds were synthesized. These compounds were evaluated in anti-proliferative assays of a panel of 15 breast cancer cell lines and further examined for their ability to inhibit a variety of Mps1-dependent biological functions. The results indicate that the lead compounds have strong anti-proliferative potential through Mps1/TTK inhibition in both basal and luminal BC cell lines, exhibiting IC50 values ranging from 0.05 to 1.0µM. In addition, the lead compounds 1 and 13 inhibit Mps1 kinase enzymatic activity with IC50 values from 0.356µM to 0.809µM, and inhibited Mps1-associated cellular functions such as centrosome duplication and the spindle checkpoint in triple negative breast cancer cells. The most promising analog, compound 13, significantly decreased tumor growth in nude mice containing Cal-51 triple negative breast cancer cell xenografts. Using drug discovery technologies, computational modeling, medicinal chemistry, cell culture and in vivo assays, novel small molecule Mps1/TTK inhibitors have been identified as potential targeted therapies for breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Animals , Female , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
3.
Proc Natl Acad Sci U S A ; 113(28): 7828-33, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27339139

ABSTRACT

Faithful segregation of chromosomes to two daughter cells is regulated by the formation of a bipolar mitotic spindle and the spindle assembly checkpoint, ensuring proper spindle function. Here we show that the proper localization of the kinase Mps1 (monopolar spindle 1) is critical to both these processes. Separate elements in the Mps1 N-terminal extension (NTE) and tetratricopeptide repeat (TPR) domains govern localization to either the kinetochore or the centrosome. The third TPR (TPR3) and the TPR-capping helix (C-helix) are each sufficient to target Mps1 to the centrosome. TPR3 binds to voltage-dependent anion channel 3, but although this is sufficient for centrosome targeting of Mps1, it is not necessary because of the presence of the C-helix. A version of Mps1 lacking both elements cannot localize to or function at the centrosome, but maintains kinetochore localization and spindle assembly checkpoint function, indicating that TPR3 and the C-helix define a bipartite localization determinant that is both necessary and sufficient to target Mps1 to the centrosome but dispensable for kinetochore targeting. In contrast, elements required for kinetochore targeting (the NTE and first two TPRs) are dispensable for centrosomal localization and function. These data are consistent with a separation of Mps1 function based on localization determinants within the N terminus.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Kinetochores/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Tetratricopeptide Repeat , HeLa Cells , Humans , Mitosis
4.
J Cell Biol ; 212(6): 615-6, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26953355

ABSTRACT

The kinetochore-associated kinase Mps1 controls the spindle assembly checkpoint, but the regulation of its kinetochore recruitment and activity is unclear. In this issue, Isokane et al. (2016. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201408089) show that interaction with and phosphorylation of its substrate, ARHGEF17, regulates Mps1 kinetochore retention, suggesting an autoregulated, timer-like mechanism.


Subject(s)
Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Kinetochores/metabolism , Kinetochores/physiology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Spindle Apparatus/metabolism , Humans
5.
Mol Biol Cell ; 26(21): 3741-53, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26354417

ABSTRACT

Centrins are a family of small, calcium-binding proteins with diverse cellular functions that play an important role in centrosome biology. We previously identified centrin 2 and centrin 3 (Cetn2 and Cetn3) as substrates of the protein kinase Mps1. However, although Mps1 phosphorylation sites control the function of Cetn2 in centriole assembly and promote centriole overproduction, Cetn2 and Cetn3 are not functionally interchangeable, and we show here that Cetn3 is both a biochemical inhibitor of Mps1 catalytic activity and a biological inhibitor of centrosome duplication. In vitro, Cetn3 inhibits Mps1 autophosphorylation at Thr-676, a known site of T-loop autoactivation, and interferes with Mps1-dependent phosphorylation of Cetn2. The cellular overexpression of Cetn3 attenuates the incorporation of Cetn2 into centrioles and centrosome reduplication, whereas depletion of Cetn3 generates extra centrioles. Finally, overexpression of Cetn3 reduces Mps1 Thr-676 phosphorylation at centrosomes, and mimicking Mps1-dependent phosphorylation of Cetn2 bypasses the inhibitory effect of Cetn3, suggesting that the biological effects of Cetn3 are due to the inhibition of Mps1 function at centrosomes.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Amino Acid Sequence , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cells, Cultured , Centrioles/metabolism , Centrosome/metabolism , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism
6.
Cells ; 4(3): 331-53, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26264029

ABSTRACT

Centrosomes are major microtubule-organizing centers of animal cells that consist of two centrioles. In mitotic cells, centrosomes are duplicated to serve as the poles of the mitotic spindle, while in quiescent cells, centrosomes move to the apical membrane where the oldest centriole is transformed into a basal body to assemble a primary cilium. We recently showed that mitochondrial outer membrane porin VDAC3 localizes to centrosomes where it negatively regulates ciliogenesis. We show here that the other two family members, VDAC1 and VDAC2, best known for their function in mitochondrial bioenergetics, are also found at centrosomes. Like VDAC3, centrosomal VDAC1 is predominantly localized to the mother centriole, while VDAC2 localizes to centriolar satellites in a microtubule-dependent manner. Down-regulation of VDAC1 leads to inappropriate ciliogenesis, while its overexpression suppresses cilia formation, suggesting that VDAC1 and VDAC3 both negatively regulate ciliogenesis. However, this negative effect on ciliogenesis is not shared by VDAC2, which instead appears to promote maturation of primary cilia. Moreover, because overexpression of VDAC3 cannot compensate for depletion of VDAC1, our data suggest that while the entire VDAC family localizes to centrosomes, they have non-redundant functions in cilogenesis.

7.
J Vis Exp ; (94)2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25548932

ABSTRACT

Centrosomes are small but important organelles that serve as the poles of mitotic spindle to maintain genomic integrity or assemble primary cilia to facilitate sensory functions in cells. The level of a protein may be regulated differently at centrosomes than at other .cellular locations, and the variation in the centrosomal level of several proteins at different points of the cell cycle appears to be crucial for the proper regulation of centriole assembly. We developed a quantitative fluorescence microscopy assay that measures relative changes in the level of a protein at centrosomes in fixed cells from different samples, such as at different phases of the cell cycle or after treatment with various reagents. The principle of this assay lies in measuring the background corrected fluorescent intensity corresponding to a protein at a small region, and normalize that measurement against the same for another protein that does not vary under the chosen experimental condition. Utilizing this assay in combination with BrdU pulse and chase strategy to study unperturbed cell cycles, we have quantitatively validated our recent observation that the centrosomal pool of VDAC3 is regulated at centrosomes during the cell cycle, likely by proteasome-mediated degradation specifically at centrosomes.


Subject(s)
Centrosome/chemistry , Fluorescent Antibody Technique , Microscopy, Fluorescence/methods , Proteins/analysis , Cell Cycle , Cilia/metabolism , Humans , Spindle Apparatus/metabolism
8.
Cell Cycle ; 12(5): 849-58, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23388454

ABSTRACT

Centrosomes serve to organize new centrioles in cycling cells, whereas in quiescent cells they assemble primary cilia. We have recently shown that the mitochondrial porin VDAC3 is also a centrosomal protein that is predominantly associated with the mother centriole and modulates centriole assembly by recruiting Mps1 to centrosomes. Here, we show that depletion of VDAC3 causes inappropriate ciliogenesis in cycling cells, while expression of GFP-VDAC3 suppresses ciliogenesis in quiescent cells. Mps1 also negatively regulates ciliogenesis, and the inappropriate ciliogenesis caused by VDAC3 depletion can be bypassed by targeting Mps1 to centrosomes independently of VDAC3. Thus, our data show that a VDAC3-Mps1 module at the centrosome promotes ciliary disassembly during cell cycle entry and suppresses cilia assembly in proliferating cells. Our data also suggests that VDAC3 might be a link between mitochondrial dysfunction and ciliopathies in mammalian cells.


Subject(s)
Cell Cycle Proteins/metabolism , Cilia/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Organogenesis , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Voltage-Dependent Anion Channels/metabolism , Cell Line , Centrosome/metabolism , Humans , Mitochondria/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Serum/metabolism
9.
Cancer Cell ; 22(5): 645-55, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23153537

ABSTRACT

How inflammation causes cancer is unclear. Interleukin-15 (IL-15) is a pro-inflammatory cytokine elevated in human large granular lymphocyte (LGL) leukemia. Mice overexpressing IL-15 develop LGL leukemia. Here, we show that prolonged in vitro exposure of wild-type (WT) LGL to IL-15 results in Myc-mediated upregulation of aurora kinases, centrosome aberrancies, and aneuploidy. Simultaneously, IL-15 represses miR-29b via induction of Myc/NF-κBp65/Hdac-1, resulting in Dnmt3b overexpression and DNA hypermethylation. All this is validated in human LGL leukemia. Adoptive transfer of WT LGL cultured with IL-15 led to malignant transformation in vivo. Drug targeting that reverses miR-29b repression cures otherwise fatal LGL leukemia. We show how excessive IL-15 initiates cancer and demonstrate effective drug targeting for potential therapy of human LGL leukemia.


Subject(s)
Chromosomal Instability , DNA Methylation , Gene Expression Regulation, Neoplastic , Interleukin-15/genetics , Leukemia, Large Granular Lymphocytic/genetics , Aneuploidy , Animals , Cell Transformation, Neoplastic/genetics , Centrosome/physiology , Chromosome Segregation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Humans , Interleukin-15/metabolism , Leukemia, Large Granular Lymphocytic/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , DNA Methyltransferase 3B
10.
Cell Cycle ; 11(19): 3666-78, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22935710

ABSTRACT

Centrioles are duplicated during S-phase to generate the two centrosomes that serve as mitotic spindle poles during mitosis. The centrosomal pool of the Mps1 kinase is important for centriole assembly, but how Mps1 is delivered to centrosomes is unknown. Here we have identified a centrosome localization domain within Mps1 and identified the mitochondrial porin VDAC3 as a protein that binds to this region of Mps1. Moreover, we show that VDAC3 is present at the mother centriole and modulates centriole assembly by recruiting Mps1 to centrosomes.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Voltage-Dependent Anion Channels/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Transport , Protein-Tyrosine Kinases/chemistry , RNA, Small Interfering/metabolism
11.
J Biol Chem ; 286(51): 43660-43667, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22045814

ABSTRACT

Ufd2 is a U-box-containing ubiquitylation enzyme that promotes ubiquitin chain assembly on substrates. The physiological function of Ufd2 remains poorly understood. Here, we show that ubiquitylation and degradation of the cell cycle kinase Mps1, a known target of the anaphase-promoting complex E3, require Ufd2 enzyme. Yeast cells lacking UFD2 exhibit altered chromosome stability and several spindle-related phenotypes, expanding the biological function of Ufd2. We demonstrate that Ufd2-mediated Mps1 degradation is conserved in humans. Our results underscore the significance of Ufd2 in proteolysis and further suggest that Ufd2-like enzymes regulate far more substrates than previously envisioned.


Subject(s)
Candida albicans/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Fungal , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Bone Marrow Cells/metabolism , Candida albicans/metabolism , Cell Line, Tumor , Humans , Lectins/chemistry , Male , Mice , Mitosis , Proteolysis , Ubiquitin/chemistry , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligases/chemistry
12.
Cell Div ; 6: 9, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21492451

ABSTRACT

The Mps1 protein kinase is an intriguing and controversial player in centriole assembly. Originally shown to control duplication of the budding yeast spindle pole body, Mps1 is present in eukaryotes from yeast to humans, the nematode C. elegans being a notable exception, and has also been shown to regulate the spindle checkpoint and an increasing number of cellular functions relating to genomic stability. While its function in the spindle checkpoint appears to be both universally conserved and essential in most organisms, conservation of its originally described function in spindle pole duplication has proven controversial, and it is less clear whether Mps1 is essential for centrosome duplication outside of budding yeast. Recent studies of Mps1 have identified at least two distinct functions for Mps1 in centriole assembly, while simultaneously supporting the notion that Mps1 is dispensable for the process. However, the fact that at least one centrosomal substrate of Mps1 is conserved from yeast to humans down to the phosphorylation site, combined with evidence demonstrating the exquisite control exerted over centrosomal Mps1 levels suggest that the notion of being essential may not be the most important of distinctions.

14.
Cell Cycle ; 10(7): 1031-1030, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-28927327

ABSTRACT

Comment on: Mattison CP, et al. Cell Cycle 2011; 10:783-93.

15.
Mol Biol Cell ; 21(24): 4361-72, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980622

ABSTRACT

The nondegradable Mps1(Δ12/13) protein drives centriole overproduction, suggesting that Mps1 phosphorylates a subset of centrosomal proteins to drive the assembly of new centrioles. Here we identify three Mps1 phosphorylation sites within the centriolar protein Centrin 2 (Cetn2). Although centrioles can be assembled in the absence of Cetn2, centriole assembly is attenuated in the absence of Cetn2. While wild-type Cetn2 can compensate for this attenuation, a nonphosphorylatable version cannot. In addition, overexpressing Cetn2 causes Mps1-dependent centriole overproduction that requires each of the three Mps1 phosphorylation sites within Cetn2 and is greatly exacerbated by mimicking phosphorylation at any of these sites. Wild-type Cetn2 generates excess foci that are competent as mitotic spindle poles in HsSas-6-depleted cells, suggesting that Cetn2 can organize a subset of centriolar proteins independently of cartwheels. However, centriole overproduction caused by a phosphomimetic Cetn2 mutant requires HsSas-6, suggesting that Cetn2 phosphorylation stimulates the canonical centriole assembly pathway. Moreover, in the absence of Cetn2, Mps1(Δ12/13) cannot drive the production of mature centrioles capable of recruiting γ-Tubulin, and a nonphosphorylatable Cetn2 mutant cannot compensate for this defect and exacerbates Cetn2 depletion. Together, our data suggest that Mps1-dependent phosphorylation of Cetn2 stimulates the canonical centriole assembly pathway.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Line , Centrioles/physiology , Centrioles/ultrastructure , Centrosome/metabolism , Centrosome/physiology , Humans , Molecular Sequence Data , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases , RNA, Small Interfering , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Tubulin/metabolism
16.
Mol Biol Cell ; 21(22): 3878-89, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20861309

ABSTRACT

Extra centrosomes are found in many tumors, and their appearance is an early event that can generate aberrant mitotic spindles and aneuploidy. Because the failure to appropriately degrade the Mps1 protein kinase correlates with centrosome overproduction in tumor-derived cells, defects in the factors that promote Mps1 degradation may contribute to extra centrosomes in tumors. However, while we have recently characterized an Mps1 degradation signal, the factors that regulate Mps1 centrosomal Mps1 are unknown. Antizyme (OAZ), a mediator of ubiquitin-independent degradation and a suspected tumor suppressor, was recently shown to localize to centrosomes and modulate centrosome overproduction, but the known OAZ substrates were not responsible for its effect on centrosomes. We have found that OAZ exerts its effect on centrosomes via Mps1. OAZ promotes the removal of Mps1 from centrosomes, and centrosome overproduction caused by reducing OAZ activity requires Mps1. OAZ binds to Mps1 via the Mps1 degradation signal and modulates the function of Mps1 in centrosome overproduction. Moreover, OAZ regulates the canonical centrosome duplication cycle, and reveals a function for Mps1 in procentriole assembly. Together, our data suggest that OAZ restrains the assembly of centrioles by controlling the levels of centrosomal Mps1 through the Cdk2-regulated Mps1 degradation signal.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases , Proteins/genetics , RNA Interference
17.
Environ Mol Mutagen ; 50(8): 654-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19274768

ABSTRACT

Centrosomes are microtubule-organizing centers that must be precisely duplicated before mitosis. Centrosomes regulate mitotic spindle assembly, and the presence of excess centrosomes leads to the production of aberrant mitotic spindles which generate chromosome segregation errors. Many human tumors possess excess centrosomes that lead to the production of abnormal spindles in situ. In some tumors, these extra centrosomes appear before aneuploidy, suggesting that defects in centrosome duplication might promote genomic instability and tumorigenesis. The Mps1 protein kinase is required for centrosome duplication, and preventing the proteasome-dependent degradation of Mps1 at centrosomes increases its local concentration and causes the production of excess centrosomes during a prolonged S-phase. Here, we show that Mps1 degradation is misregulated in two tumor-derived cell lines, and that the failure to appropriately degrade Mps1 correlates with the ability of these cells to produce extra centrosomes during a prolonged S-phase. In the 21NT breast-tumor derived cell line, a mutant Mps1 protein that is normally constitutively degraded can accumulate at centrosomes and perturb centrosome duplication, suggesting that these cells have a defect in the mechanisms that target Mps1 to the proteasome. In contrast, the U2OS osteosarcoma cell line expresses a nondegradable form of Mps1, which we show causes the dose-dependent over duplication of centrioles even at very low levels of expression. Our data demonstrate that defects in Mps1 degradation can occur through multiple mechanisms, and suggest that Mps1 may provide a link between the control of centrosome duplication and genomic instability.


Subject(s)
Cell Cycle Proteins/physiology , Centrosome , Genomic Instability , Protein Serine-Threonine Kinases/physiology , Alleles , Blotting, Western , Cell Line, Tumor , Humans , Protein-Tyrosine Kinases , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Biol Cell ; 18(11): 4457-69, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17804818

ABSTRACT

Supernumerary centrosomes promote the assembly of abnormal mitotic spindles in many human tumors. In human cells, overexpression of the cyclin-dependent kinase (Cdk)2 partner cyclin A during a prolonged S phase produces extra centrosomes, called centrosome reduplication. Cdk2 activity protects the Mps1 protein kinase from proteasome-mediated degradation, and we demonstrate here that Mps1 mediates cyclin A-dependent centrosome reduplication. Overexpression of cyclin A or a brief proteasome inhibition increases the centrosomal levels of Mps1, whereas depletion of Cdk2 leads to the proteasome-dependent loss of Mps1 from centrosomes only. When a Cdk2 phosphorylation site within Mps1 (T468) is mutated to alanine, Mps1 cannot accumulate at centrosomes or participate in centrosome duplication. In contrast, phosphomimetic mutations at T468 or deletion of the region surrounding T468 prevent the proteasome-dependent removal of Mps1 from centrosomes in the absence of Cdk2 activity. Moreover, cyclin A-dependent centrosome reduplication requires Mps1, and these stabilizing Mps1 mutations cause centrosome reduplication, bypassing cyclin A. Together, our data demonstrate that the region surrounding T468 contains a motif that regulates the accumulation of Mps1 at centrosomes. We suggest that phosphorylation of T468 attenuates the degradation of Mps1 at centrosomes and that preventing this degradation is necessary and sufficient to cause centrosome reduplication in human cells.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Centrosome/drug effects , Conserved Sequence , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein-Tyrosine Kinases , Sequence Alignment , Signal Transduction
19.
Curr Biol ; 14(24): R1058-60, 2004 Dec 29.
Article in English | MEDLINE | ID: mdl-15620641

ABSTRACT

Analysis of a mutation in the Drosophila Mps1 ortholog further demonstrates the universality of Mps1 function in the spindle checkpoint, but suggests Mps1 function in centrosome duplication might not be so conserved. The work also contributes new additions to a list of Mps1 functions that continues to grow with each new study.


Subject(s)
Cell Cycle Proteins/physiology , Chromosome Segregation/physiology , Genes, cdc/physiology , Protein Kinases/physiology , Spindle Apparatus/physiology , Animals , Cell Cycle Proteins/genetics , Cell Division/genetics , Centrosome/physiology , Drosophila , Mutation/genetics , Protein Kinases/genetics
20.
Cell Cycle ; 3(4): 439-42, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14963409

ABSTRACT

Cell cycle events must be faithfully executed and properly integrated to ensure genetic stability. The Mps1 family of protein kinases has recently emerged as a critical regulator of genetic stability, because they regulate several processes central to mitotic fidelity. The spindle checkpoint monitors alignment of mitotic chromosomes, and centrosomes control cell cycle entry, mitotic spindle assembly, and cytokinesis. Several studies have shown that vertebrate orthologues of budding yeast Mps1p regulate the spindle checkpoint. More recently it has been demonstrated that human Mps1 is also required for centrosome duplication, normal mitotic progression, and cytokinesis.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Kinases/metabolism , Animals , Cell Cycle , Cell Division , Centrosome/metabolism , Chromosomes/ultrastructure , Gene Expression Regulation , Humans , Kinetochores/ultrastructure , Mitosis , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , RNA, Small Interfering/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomycetales , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...